Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Publication year range
1.
Cell ; 171(5): 1042-1056.e10, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29056344

ABSTRACT

We present an extensive assessment of mutation burden through sequencing analysis of >81,000 tumors from pediatric and adult patients, including tumors with hypermutation caused by chemotherapy, carcinogens, or germline alterations. Hypermutation was detected in tumor types not previously associated with high mutation burden. Replication repair deficiency was a major contributing factor. We uncovered new driver mutations in the replication-repair-associated DNA polymerases and a distinct impact of microsatellite instability and replication repair deficiency on the scale of mutation load. Unbiased clustering, based on mutational context, revealed clinically relevant subgroups regardless of the tumors' tissue of origin, highlighting similarities in evolutionary dynamics leading to hypermutation. Mutagens, such as UV light, were implicated in unexpected cancers, including sarcomas and lung tumors. The order of mutational signatures identified previous treatment and germline replication repair deficiency, which improved management of patients and families. These data will inform tumor classification, genetic testing, and clinical trial design.


Subject(s)
Neoplasms/genetics , Adult , Child , Cluster Analysis , DNA Polymerase II/genetics , DNA Polymerase III/genetics , DNA Replication , Humans , Mutation , Neoplasms/classification , Neoplasms/pathology , Neoplasms/therapy , Poly-ADP-Ribose Binding Proteins/genetics
2.
Acta Neuropathol ; 140(5): 765-776, 2020 11.
Article in English | MEDLINE | ID: mdl-32895736

ABSTRACT

Replication repair deficiency (RRD) leading to hypermutation is an important driving mechanism of high-grade glioma (HGG) occurring predominantly in the context of germline mutations in RRD-associated genes. Although HGG presents specific patterns of DNA methylation corresponding to oncogenic mutations, this has not been well studied in replication repair-deficient tumors. We analyzed 51 HGG arising in the background of gene mutations in RRD utilizing either 450 k or 850 k methylation arrays. These were compared with HGG not known to be from patients with RRD. RRD HGG harboring secondary mutations in glioma genes such as IDH1 and H3F3A displayed a methylation pattern corresponding to these methylation subgroups. Strikingly, RRD HGG lacking these known secondary mutations clustered together with an incompletely described group of HGG previously labeled "Wild type-C" or "Paediatric RTK 1". Independent analysis of two comparator HGG cohorts showed that other RRD/hypermutant tumors clustered within these subgroups, suggesting that undiagnosed RRD may be driving some HGG clustering in this location. RRD HGG displayed a unique CpG Island Demethylator Phenotype in contrast to the CpG Island Methylator Phenotype described in other cancers. Hypomethylation was enriched at gene promoters with prominent demethylation in genes and pathways critical to cellular survival including cell cycle, gene expression, cellular metabolism, and organization. These data suggest that methylation arrays may provide diagnostic information for the detection of RRD HGG. Furthermore, our findings highlight the unique natural selection pressures in these highly dysregulated, hypermutant cancers and provide the novel impact of hypermutation and RRD on the cancer epigenome.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation/genetics , DNA Repair-Deficiency Disorders/genetics , DNA Repair/genetics , Glioma/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Repair-Deficiency Disorders/complications , Female , Germ-Line Mutation , Humans , Male , Young Adult
3.
Cancer Invest ; 38(6): 349-355, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32441531

ABSTRACT

Background: Meningiomas represent ∼30% of primary central nervous system (CNS) tumors. Although advances in surgery and radiotherapy have significantly improved survival, there remains an important subset of patients whose tumors have more aggressive behavior and are refractory to conventional therapy. Recent advances in molecular genetics and epigenetics suggest that this aggressive behavior may be due to the deletion of the DNA repair and tumor suppressor gene, CHEK2, neurofibromatosis Type 2 (NF2) mutation on chromosome 22q12, and genetic abnormalities in multiple RTKs including FGFRs. Management of higher-grade meningiomas, such as anaplastic meningiomas (AM: WHO grade III), is truly challenging and there isn't an established chemotherapy option. We investigate the effect of active multi tyrosine receptor kinase inhibitor Dovitinib at stopping AM cell growth in in vitro with either frequent codeletion or mutated CHEK2 and NF2 gene.Methods: Treatment effects were assessed using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, western blot analysis, caspases assay, and DNA fragmentation assay.Results: Treatment of CH157MN and IOMM-Lee cells with Dovitinib suppressed multiple angiokinases-mainly FGFRs, leading to suppression of downstream signaling by RAS-RAF-MAPK molecules and PI3K-AKT molecules which are involved in cell proliferation, cell survival, and tumor invasion. Furthermore, Dovitinib induced apoptosis via downregulation of survival proteins (Bcl-XL), and over-expression of apoptotic factors (Bax and caspase-3) regardless of CHEK2 and NF2 mutation status.Conclusions: This study establishes the groundwork for the development of Dovitinib as a therapeutic agent for high-grade AM with either frequent codeletion or mutated CHEK2 and NF2, an avenue with high translational potential.


Subject(s)
Benzimidazoles/pharmacology , Checkpoint Kinase 2/genetics , Meningioma/drug therapy , Neurofibromin 2/genetics , Quinolones/pharmacology , Apoptosis/drug effects , Caspase 3/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Meningioma/genetics , Meningioma/pathology , Mutation/genetics , Neoplasm Staging , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction/drug effects , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
4.
J Transl Med ; 16(1): 142, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29843811

ABSTRACT

BACKGROUND: Standard therapy for glioblastoma includes surgery, radiotherapy, and temozolomide. This Phase 3 trial evaluates the addition of an autologous tumor lysate-pulsed dendritic cell vaccine (DCVax®-L) to standard therapy for newly diagnosed glioblastoma. METHODS: After surgery and chemoradiotherapy, patients were randomized (2:1) to receive temozolomide plus DCVax-L (n = 232) or temozolomide and placebo (n = 99). Following recurrence, all patients were allowed to receive DCVax-L, without unblinding. The primary endpoint was progression free survival (PFS); the secondary endpoint was overall survival (OS). RESULTS: For the intent-to-treat (ITT) population (n = 331), median OS (mOS) was 23.1 months from surgery. Because of the cross-over trial design, nearly 90% of the ITT population received DCVax-L. For patients with methylated MGMT (n = 131), mOS was 34.7 months from surgery, with a 3-year survival of 46.4%. As of this analysis, 223 patients are ≥ 30 months past their surgery date; 67 of these (30.0%) have lived ≥ 30 months and have a Kaplan-Meier (KM)-derived mOS of 46.5 months. 182 patients are ≥ 36 months past surgery; 44 of these (24.2%) have lived ≥ 36 months and have a KM-derived mOS of 88.2 months. A population of extended survivors (n = 100) with mOS of 40.5 months, not explained by known prognostic factors, will be analyzed further. Only 2.1% of ITT patients (n = 7) had a grade 3 or 4 adverse event that was deemed at least possibly related to the vaccine. Overall adverse events with DCVax were comparable to standard therapy alone. CONCLUSIONS: Addition of DCVax-L to standard therapy is feasible and safe in glioblastoma patients, and may extend survival. Trial registration Funded by Northwest Biotherapeutics; Clinicaltrials.gov number: NCT00045968; https://clinicaltrials.gov/ct2/show/NCT00045968?term=NCT00045968&rank=1 ; initially registered 19 September 2002.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , Cancer Vaccines/immunology , Dendritic Cells/immunology , Glioblastoma/immunology , Glioblastoma/therapy , Adult , Aged , Brain Neoplasms/diagnosis , Cancer Vaccines/adverse effects , Endpoint Determination , Female , Glioblastoma/diagnosis , Humans , Male , Middle Aged , Prognosis , Survival Analysis , Treatment Outcome , Young Adult
5.
J Transl Med ; 16(1): 179, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29958537

ABSTRACT

Following publication of the original article [1], the authors reported an error in the spelling of one of the author names. In this Correction the incorrect and correct author names are indicated and the author name has been updated in the original publication. The authors also reported an error in the Methods section of the original article. In this Correction the incorrect and correct versions of the affected sentence are indicated. The original article has not been updated with regards to the error in the Methods section.

7.
Tumour Biol ; 37(6): 7525-34, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26684801

ABSTRACT

Radiation-induced necrosis (RN) is a relatively common side effect of radiation therapy for glioblastoma. However, the molecular mechanisms involved and the ways RN mechanisms differ from regulated cell death (apoptosis) are not well understood. Here, we compare the molecular mechanism of cell death (apoptosis or necrosis) of C6 glioma cells in both in vitro and in vivo (C6 othotopically allograft) models in response to low and high doses of X-ray radiation. Lower radiation doses were used to induce apoptosis, while high-dose levels were chosen to induce radiation necrosis. Our results demonstrate that active caspase-8 in this complex I induces apoptosis in response to low-dose radiation and inhibits necrosis by cleaving RIP1 and RI. When activation of caspase-8 was reduced at high doses of X-ray radiation, the RIP1/RIP3 necrosome complex II is formed. These complexes induce necrosis through the caspase-3-independent pathway mediated by calpain, cathepsin B/D, and apoptosis-inducing factor (AIF). AIF has a dual role in apoptosis and necrosis. At high doses, AIF promotes chromatinolysis and necrosis by interacting with histone H2AX. In addition, NF-κB, STAT-3, and HIF-1 play a crucial role in radiation-induced inflammatory responses embedded in a complex inflammatory network. Analysis of inflammatory markers in matched plasma and cerebrospinal fluid (CSF) isolated from in vivo specimens demonstrated the upregulation of chemokines and cytokines during the necrosis phase. Using RIP1/RIP3 kinase specific inhibitors (Nec-1, GSK'872), we also establish that the RIP1-RIP3 complex regulates programmed necrosis after either high-dose radiation or TNF-α-induced necrosis requires RIP1 and RIP3 kinases. Overall, our data shed new light on the relationship between RIP1/RIP3-mediated programmed necrosis and AIF-mediated caspase-independent programmed necrosis in glioblastoma.


Subject(s)
Gamma Rays/adverse effects , Glioblastoma/radiotherapy , Necrosis/metabolism , Necrosis/pathology , Protein Serine-Threonine Kinases/metabolism , Radiation Injuries/metabolism , Radiation Injuries/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Blotting, Western , Caspases , Cell Proliferation , Glioblastoma/metabolism , Glioblastoma/pathology , Immunoenzyme Techniques , Male , Necrosis/etiology , Radiation Injuries/etiology , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Tumour Biol ; 36(9): 7027-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25864108

ABSTRACT

Recurrent meningiomas constitute an uncommon but significant problem after standard (surgery and radiation) therapy failure. Current chemotherapies (hydroxyurea, RU-486, and interferon-α) are only of marginal benefit. There is an urgent need for more effective treatments for meningioma patients who have failed surgery and radiation therapy. Limonin, Tangeritin, Zerumbone, 6-Gingerol, Ganoderic Acid A, and Ganoderic Acid DM are some of the plant derivatives that have anti-tumorgenic properties and cause cell death in meningioma cells in vitro. Due to its ease of administration, long-term tolerability, and low incidence of long-term side effects, we explored its potential as a therapeutic agent against meningiomas by examining their efficacy in vitro against meningioma cells. Treatment effects were assessed using MTT assay, Western blot analysis, caspases assay, and DNA fragmentation assay. Results indicated that treatments of IOMM-Lee and CH157MN meningioma cells with Limonin, Tangeritin, Zerumbone, 6-Gingerol, Ganoderic Acid A, and Ganoderic Acid DM induced apoptosis with enhanced phosphorylation of glycogen synthase kinase 3 ß (GSK3ß) via inhibition of the Wnt5/ß-catenin pathway. These drugs did not induce apoptosis in normal human neurons. Other events in apoptosis included downregulation of tetraspanin protein (TSPAN12), survival proteins (Bcl-XL and Mcl-1), and overexpression apoptotic factors (Bax and caspase-3). These results provide preliminary strong evidence that medicinal plants containing Limonin, Tangeritin, 6-Gingerol, Zerumbone, Ganoderic Acid A, and Ganoderic Acid DM can be applied to high-grade meningiomas as a therapeutic agent, and suggests that further in vivo studies are necessary to explore its potential as a therapeutic agent against malignant meningiomas.


Subject(s)
Catechols/administration & dosage , Fatty Alcohols/administration & dosage , Flavones/administration & dosage , Heptanoic Acids/administration & dosage , Lanosterol/analogs & derivatives , Limonins/administration & dosage , Meningioma/drug therapy , Sesquiterpenes/administration & dosage , Triterpenes/administration & dosage , Apoptosis/drug effects , Catechols/chemistry , Cell Line, Tumor , DNA Fragmentation/drug effects , Fatty Alcohols/chemistry , Flavones/chemistry , Glycogen Synthase Kinase 3/biosynthesis , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Heptanoic Acids/chemistry , Humans , Lanosterol/administration & dosage , Lanosterol/chemistry , Limonins/chemistry , Meningioma/genetics , Meningioma/pathology , Sesquiterpenes/chemistry , Triterpenes/chemistry , Wnt Signaling Pathway/drug effects
9.
J Natl Compr Canc Netw ; 11(7): 745-9; quiz 750, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23847213

ABSTRACT

Many effective therapeutic options are available for patients with chronic myelogenous leukemia (CML). Imatinib, a first-generation tyrosine kinase inhibitor (TKI), is one of several options for patients who present with CML, whether in chronic phase, accelerated phase, or blast crisis. Although CML is very responsive to the selective TKIs, with response rates in chronic phase of greater than 90%, unusual presentations have been documented. Response rates for patients with CML in accelerated phase and blast crisis are notably lower to both first- and second-generation TKIs. This report presents a recent case of a young woman with newly diagnosed CML who presented with an accelerated phase isolated central nervous system (CNS) relapse after standard imatinib therapy, who initially experienced a complete hematologic response. Further treatment options, and monitoring of disease response, are discussed. Aggressive strategies, such as intrathecal chemotherapy, change in tyrosine kinase inhibitor to one with increased CNS penetration, and consideration of allogenic stem cell transplantation, are potential therapeutic modalities. Prophylaxis of the CNS in patients deemed at risk is an area requiring further study.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/secondary , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Central Nervous System Neoplasms/drug therapy , Cerebrospinal Fluid/cytology , Female , Humans , Injections, Spinal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Recurrence , Treatment Outcome , Young Adult
10.
J Clin Oncol ; 41(6): 1285-1295, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36260832

ABSTRACT

PURPOSE: To assess whether reirradiation (re-RT) and concurrent bevacizumab (BEV) improve overall survival (OS) and/or progression-free survival (PFS), compared with BEV alone in recurrent glioblastoma (GBM). The primary objective was OS, and secondary objectives included PFS, response rate, and treatment adverse events (AEs) including delayed CNS toxicities. METHODS: NRG Oncology/RTOG1205 is a prospective, phase II, randomized trial of re-RT and BEV versus BEV alone. Stratification factors included age, resection, and Karnofsky performance status (KPS). Patients with recurrent GBM with imaging evidence of tumor progression ≥ 6 months from completion of prior chemo-RT were eligible. Patients were randomly assigned 1:1 to re-RT, 35 Gy in 10 fractions, with concurrent BEV IV 10 mg/kg once in every 2 weeks or BEV alone until progression. RESULTS: From December 2012 to April 2016, 182 patients were randomly assigned, of whom 170 were eligible. Patient characteristics were well balanced between arms. The median follow-up for censored patients was 12.8 months. There was no improvement in OS for BEV + RT, hazard ratio, 0.98; 80% CI, 0.79 to 1.23; P = .46; the median survival time was 10.1 versus 9.7 months for BEV + RT versus BEV alone. The median PFS for BEV + RT was 7.1 versus 3.8 months for BEV, hazard ratio, 0.73; 95% CI, 0.53 to 1.0; P = .05. The 6-month PFS rate improved from 29.1% (95% CI, 19.1 to 39.1) for BEV to 54.3% (95% CI, 43.5 to 65.1) for BEV + RT, P = .001. Treatment was well tolerated. There were a 5% rate of acute grade 3+ treatment-related AEs and no delayed high-grade AEs. Most patients died of recurrent GBM. CONCLUSION: To our knowledge, NRG Oncology/RTOG1205 is the first prospective, randomized multi-institutional study to evaluate the safety and efficacy of re-RT in recurrent GBM using modern RT techniques. Overall, re-RT was shown to be safe and well tolerated. BEV + RT demonstrated a clinically meaningful improvement in PFS, specifically the 6-month PFS rate but no difference in OS.


Subject(s)
Brain Neoplasms , Glioblastoma , Re-Irradiation , Humans , Bevacizumab , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Re-Irradiation/adverse effects , Prospective Studies , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects
11.
Neuro Oncol ; 25(1): 137-145, 2023 01 05.
Article in English | MEDLINE | ID: mdl-35657335

ABSTRACT

BACKGROUND: Three- and five-year progression-free survival (PFS) for low-risk meningioma managed with surgery and observation reportedly exceeds 90%. Herewith we summarize outcomes for low-risk meningioma patients enrolled on NRG/RTOG 0539. METHODS: This phase II trial allocated patients to one of three groups per World Health Organization grade, recurrence status, and resection extent. Low-risk patients had either gross total (GTR) or subtotal resection (STR) for a newly diagnosed grade 1 meningioma and were observed after surgery. The primary endpoint was 3-year PFS. Adverse events (AEs) were scored using Common Terminology Criteria for Adverse Events (CTCAE) version 3. RESULTS: Among 60 evaluable patients, the median follow-up was 9.1 years. The 3-, 5-, and 10-year rates were 91.4% (95% CI, 84.2 to 98.6), 89.4% (95% CI, 81.3 to 97.5), 85.0% (95% CI, 75.3 to 94.7) for PFS and 98.3% (95% CI, 94.9 to 100), 98.3%, (95% CI, 94.9 to 100), 93.8% (95% CI, 87.0 to 100) for overall survival (OS), respectively. With centrally confirmed GTR, 3/5/10y PFS and OS rates were 94.3/94.3/87.6% and 97.1/97.1/90.4%. With STR, 3/5/10y PFS rates were 83.1/72.7/72.7% and 10y OS 100%. Five patients reported one grade 3, four grade 2, and five grade 1 AEs. There were no grade 4 or 5 AEs. CONCLUSIONS: These results prospectively validate high PFS and OS for low-risk meningioma managed surgically but raise questions regarding optimal management following STR, a subcohort that could potentially benefit from adjuvant therapy.


Subject(s)
Meningeal Neoplasms , Meningioma , Humans , Meningioma/surgery , Radiotherapy, Adjuvant/methods , Progression-Free Survival , Risk , Meningeal Neoplasms/surgery , Retrospective Studies
12.
JAMA Oncol ; 9(1): 112-121, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36394838

ABSTRACT

Importance: Glioblastoma is the most lethal primary brain cancer. Clinical outcomes for glioblastoma remain poor, and new treatments are needed. Objective: To investigate whether adding autologous tumor lysate-loaded dendritic cell vaccine (DCVax-L) to standard of care (SOC) extends survival among patients with glioblastoma. Design, Setting, and Participants: This phase 3, prospective, externally controlled nonrandomized trial compared overall survival (OS) in patients with newly diagnosed glioblastoma (nGBM) and recurrent glioblastoma (rGBM) treated with DCVax-L plus SOC vs contemporaneous matched external control patients treated with SOC. This international, multicenter trial was conducted at 94 sites in 4 countries from August 2007 to November 2015. Data analysis was conducted from October 2020 to September 2021. Interventions: The active treatment was DCVax-L plus SOC temozolomide. The nGBM external control patients received SOC temozolomide and placebo; the rGBM external controls received approved rGBM therapies. Main Outcomes and Measures: The primary and secondary end points compared overall survival (OS) in nGBM and rGBM, respectively, with contemporaneous matched external control populations from the control groups of other formal randomized clinical trials. Results: A total of 331 patients were enrolled in the trial, with 232 randomized to the DCVax-L group and 99 to the placebo group. Median OS (mOS) for the 232 patients with nGBM receiving DCVax-L was 19.3 (95% CI, 17.5-21.3) months from randomization (22.4 months from surgery) vs 16.5 (95% CI, 16.0-17.5) months from randomization in control patients (HR = 0.80; 98% CI, 0.00-0.94; P = .002). Survival at 48 months from randomization was 15.7% vs 9.9%, and at 60 months, it was 13.0% vs 5.7%. For 64 patients with rGBM receiving DCVax-L, mOS was 13.2 (95% CI, 9.7-16.8) months from relapse vs 7.8 (95% CI, 7.2-8.2) months among control patients (HR, 0.58; 98% CI, 0.00-0.76; P < .001). Survival at 24 and 30 months after recurrence was 20.7% vs 9.6% and 11.1% vs 5.1%, respectively. Survival was improved in patients with nGBM with methylated MGMT receiving DCVax-L compared with external control patients (HR, 0.74; 98% CI, 0.55-1.00; P = .03). Conclusions and Relevance: In this study, adding DCVax-L to SOC resulted in clinically meaningful and statistically significant extension of survival for patients with both nGBM and rGBM compared with contemporaneous, matched external controls who received SOC alone. Trial Registration: ClinicalTrials.gov Identifier: NCT00045968.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Temozolomide/therapeutic use , Prospective Studies , Brain Neoplasms/pathology , Recurrence , Dendritic Cells/pathology , Vaccination
13.
World Neurosurg ; 161: e61-e74, 2022 05.
Article in English | MEDLINE | ID: mdl-35032716

ABSTRACT

BACKGROUND: Glioblastoma (GB) is an aggressive tumor showing extensive intertumoral and intratumoral heterogeneity. Several possible reasons contribute to the historical inability to develop effective therapeutic strategies for treatment of GB. One such challenge is the inability to consistently procure high-quality biologically preserved specimens for use in molecular research and patient-derived xenograft model development. No scientifically derived standardized method exists for intraoperative tissue collection specifically designed with the fragility of RNA in mind. METHODS: In this investigation, we set out to characterize matched specimens from 6 GB patients comparing the traditional handling and collection processes of intraoperative tissue used in most neurosurgical operating rooms versus an automated resection, collection, and biological preservation system (APS) which captures, preserves, and biologically maintains tissue in a prescribed and controlled microenvironment. Matched specimens were processed in parallel at various time points and temperatures, evaluating viability, RNA and protein concentrations, and isolation of GB cell lines. RESULTS: We found that APS-derived GB slices stored in an APS modified medium remained viable and maintained high-quality RNA and protein concentration for up to 24 hours. CONCLUSIONS: Our results showed that primary GB cell cultures derived in this manner had improved growth over widely used collection and preservation methods. By implementing an automated intraoperative system, we also eliminated inconsistencies in methodology of tissue collection, handling and biological preservation, establishing a repeatable and standardized practice that does not require additional staff or a laboratory technician to manage it.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Pilot Projects , Preservation, Biological , RNA , Tissue Preservation/methods , Tumor Microenvironment
14.
Clin Neurol Neurosurg ; 216: 107218, 2022 05.
Article in English | MEDLINE | ID: mdl-35367747

ABSTRACT

Few cases have been reported of the diagnosis and treatment of glioblastoma (GB) during pregnancy. Subsequently, surgical, medical, and obstetrical management of complicated primary central nervous system malignancy in antepartum and postpartum patients remains under-investigated. The authors report the case of a 24-year-old female patient who developed generalized tonic-clonic seizures and focal neurologic deficits. MRI imaging (3T Skyra, Siemens, Erlangen, Germany) revealed an intracranial mass suspicious for malignant tumor and surgical resection under awake sedation was scheduled. The patient was incidentally found to be in her first trimester of pregnancy. Using neuronavigation, neurophysiologic monitoring, and conscious sedation the tumor was debulked successfully and histopathologic analysis confirmed giant cell glioblastoma, WHO Grade IV, 1p/19q intact, IDH wild-type, with NF1 p.Y2285fs and RB1 p.S318fs somatic mutations. Post-surgical oncologic management continued with fractioned radiotherapy and use of the Optune® device. The patient underwent uncomplicated cesarean section at 34-weeks gestation, the child remains healthy and the patient remains disease-disease free at 1-year. Thus, this case presents an approach to management of complicated GBM during first trimester pregnancy.


Subject(s)
Brain Neoplasms , Glioblastoma , Child , Humans , Pregnancy , Female , Young Adult , Adult , Glioblastoma/diagnostic imaging , Glioblastoma/surgery , Glioblastoma/genetics , Brain Neoplasms/pathology , Wakefulness , Cesarean Section , Craniotomy/methods
15.
Nat Med ; 28(1): 125-135, 2022 01.
Article in English | MEDLINE | ID: mdl-34992263

ABSTRACT

Cancers arising from germline DNA mismatch repair deficiency or polymerase proofreading deficiency (MMRD and PPD) in children harbour the highest mutational and microsatellite insertion-deletion (MS-indel) burden in humans. MMRD and PPD cancers are commonly lethal due to the inherent resistance to chemo-irradiation. Although immune checkpoint inhibitors (ICIs) have failed to benefit children in previous studies, we hypothesized that hypermutation caused by MMRD and PPD will improve outcomes following ICI treatment in these patients. Using an international consortium registry study, we report on the ICI treatment of 45 progressive or recurrent tumors from 38 patients. Durable objective responses were observed in most patients, culminating in a 3 year survival of 41.4%. High mutation burden predicted response for ultra-hypermutant cancers (>100 mutations per Mb) enriched for combined MMRD + PPD, while MS-indels predicted response in MMRD tumors with lower mutation burden (10-100 mutations per Mb). Furthermore, both mechanisms were associated with increased immune infiltration even in 'immunologically cold' tumors such as gliomas, contributing to the favorable response. Pseudo-progression (flare) was common and was associated with immune activation in the tumor microenvironment and systemically. Furthermore, patients with flare who continued ICI treatment achieved durable responses. This study demonstrates improved survival for patients with tumors not previously known to respond to ICI treatment, including central nervous system and synchronous cancers, and identifies the dual roles of mutation burden and MS-indels in predicting sustained response to immunotherapy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , DNA Repair/genetics , DNA Replication/genetics , Germ-Line Mutation , Adolescent , Adult , Biomarkers, Tumor , Child , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Male , Neoplasms/drug therapy , Prospective Studies , Retrospective Studies , Survival Analysis , Tumor Microenvironment , Young Adult
16.
J Clin Neurophysiol ; 38(2): 143-148, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-31856045

ABSTRACT

INTRODUCTION: Metastases to the brain (MB) occur in up to 30% of adults with cancer; of these, 15% to 35% may have seizures. We investigated clinical and pathologic associations with seizure and EEG findings in patients with MB, given the sparse literature in this area. METHODS: We performed a retrospective chart review of adults with pathologically confirmed MB treated at a large tertiary care center between April 8, 2006, and December 14, 2018. Primary outcomes were odds of "chart-documented seizure" (CDS) in the full sample and EEG-captured seizure or any epileptiform discharges among those monitored on EEG. RESULTS: We studied 187 patients with MB, of whom 55 (28.3%) were monitored on EEG. We found an overall CDS prevalence of 29.4% and an EEG-captured seizure of 18.9% among patients monitored on EEG. Of those monitored on EEG, 47.2% had epileptiform discharges. Adenocarcinoma pathology was associated with lower odds of CDS (odds ratio [OR] 0.50, 95% CI 0.26-0.96) and EEG-captured seizure (OR 0.09, 95% CI 0.01-0.87) versus other pathologies. When modeled separately, melanoma pathology was associated with CDS (OR 4.45, 95% CI 1.58-12.57) versus other pathologies. Hemorrhagic MB were associated with any epileptiform discharges (OR 5.50, 95% CI 1.65-18.37), regardless of pathology modeled. Increasing size of the largest dimension of the largest MB was associated with lower odds of CDS (OR 0.68, 95% CI 0.52-0.89 when adenocarcinoma modeled, OR 0.69, 95% CI 0.53-0.91 when melanoma modeled). CONCLUSIONS: Seizures and epileptiform discharges are common in patients with MB. Tumor size and pathology were significantly associated with CDS. Larger studies are needed for further analysis.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/epidemiology , Electroencephalography/methods , Seizures/diagnosis , Seizures/epidemiology , Adult , Aged , Brain Neoplasms/physiopathology , Brain Neoplasms/secondary , Female , Humans , Male , Middle Aged , Prevalence , Retrospective Studies , Seizures/physiopathology
17.
Cancer Discov ; 11(5): 1176-1191, 2021 05.
Article in English | MEDLINE | ID: mdl-33355208

ABSTRACT

Although replication repair deficiency, either by mismatch repair deficiency (MMRD) and/or loss of DNA polymerase proofreading, can cause hypermutation in cancer, microsatellite instability (MSI) is considered a hallmark of MMRD alone. By genome-wide analysis of tumors with germline and somatic deficiencies in replication repair, we reveal a novel association between loss of polymerase proofreading and MSI, especially when both components are lost. Analysis of indels in microsatellites (MS-indels) identified five distinct signatures (MS-sigs). MMRD MS-sigs are dominated by multibase losses, whereas mutant-polymerase MS-sigs contain primarily single-base gains. MS deletions in MMRD tumors depend on the original size of the MS and converge to a preferred length, providing mechanistic insight. Finally, we demonstrate that MS-sigs can be a powerful clinical tool for managing individuals with germline MMRD and replication repair-deficient cancers, as they can detect the replication repair deficiency in normal cells and predict their response to immunotherapy. SIGNIFICANCE: Exome- and genome-wide MSI analysis reveals novel signatures that are uniquely attributed to mismatch repair and DNA polymerase. This provides new mechanistic insight into MS maintenance and can be applied clinically for diagnosis of replication repair deficiency and immunotherapy response prediction.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Cell Transformation, Neoplastic , DNA Mismatch Repair , DNA-Directed DNA Polymerase , Gene Expression Regulation, Neoplastic , Microsatellite Instability , Neoplasms/genetics , Humans , Exome Sequencing
18.
J Neurol Sci ; 418: 117102, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32866816

ABSTRACT

Temozolomide (TMZ) therapy is the standard of care for patients with glioblastoma (GBM). Clinical studies have shown that elevated levels of DNA repair protein O (6)-methylguanine-DNA methyltransferase (MGMT) or deficiency/defect of DNA mismatch repair (MMR) genes is associated with TMZ resistance in some, but not all, GBM tumors. Another reason for GBM treatment failure is signal redundancy due to coactivation of several functionally linked receptor tyrosine kinases (RTKs), including anaplastic lymphoma kinase (ALK) and c-Met (hepatocyte growth factor receptor). As such, these tyrosine kinases serve as potential targets for GBM therapy. Thus, we tested two novel drugs: INC280 (Capmatinib: a highly selective c-Met receptor tyrosine kinase-RTK inhibitor) and LDK378 (Ceritinib: a highly selective anaplastic lymphoma kinase-ALK inhibitor), aiming to overcome TMZ resistance in MGMT-unmethylated GBM cells in in vitro cell culture models. Treatments were examined using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, caspase-3 assay and western blot analysis. Results obtained from our experiments demonstrated that preconditioning with INC280 and LDK378 drugs exhibit increased MMR protein expression, specifically MMR protein MLH1 (MutL Homolog 1) and MSH6 (MutS Homolog 6) and sensitized TMZ in MGMT-unmethylated GBM cells via suppression of ALK and c-Met expression. INC280 and LDK378 plus TMZ also induced apoptosis by modulating downstream signaling of PI3K/AKT/STAT3. Taken together, this data indicates that co-inhibition of ALK and c-MET can enhance growth inhibitory effects in MGMT-unmethylated cells and enhance TMZ sensitivity in-vitro, suggesting c-Met inhibitors combined with ALK-targeting provide a therapeutic benefit in MGMT-unmethylated GBM patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Pharmaceutical Preparations , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Benzamides , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Humans , Imidazoles , Phosphatidylinositol 3-Kinases , Pyrimidines , Sulfones , Temozolomide/pharmacology , Temozolomide/therapeutic use , Triazines , Tumor Suppressor Proteins/genetics
19.
Neuro Oncol ; 22(9): 1249-1261, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32391559

ABSTRACT

In oncology, "immunotherapy" is a broad term encompassing multiple means of utilizing the patient's immune system to combat malignancy. Prominent among these are immune checkpoint inhibitors, cellular therapies including chimeric antigen receptor T-cell therapy, vaccines, and oncolytic viruses. Immunotherapy for glioblastoma (GBM) has had mixed results in early trials. In this context, the past, present, and future of immune oncology for the treatment of GBM was discussed by clinical, research, and thought leaders as well as patient advocates at the first annual Remission Summit in 2019. The goal was to use current knowledge (published and unpublished) to identify possible causes of treatment failures and the best strategies to advance immunotherapy as a treatment modality for patients with GBM. The discussion focuses on past failures, current limitations, failure analyses, and proposed best practices moving forward.


Subject(s)
Brain Neoplasms , Glioblastoma , Oncolytic Viruses , Adult , Brain Neoplasms/therapy , Glioblastoma/therapy , Humans , Immunotherapy
20.
Neurooncol Adv ; 2(1): vdaa155, 2020.
Article in English | MEDLINE | ID: mdl-33392505

ABSTRACT

BACKGROUND: Giant cell glioblastoma (gcGBM) is a rare histologic subtype of glioblastoma characterized by numerous bizarre multinucleate giant cells and increased reticulin deposition. Compared with conventional isocitrate dehydrogenase (IDH)-wildtype glioblastomas, gcGBMs typically occur in younger patients and are generally associated with an improved prognosis. Although prior studies of gcGBMs have shown enrichment of genetic events, such as TP53 alterations, no defining aberrations have been identified. The aim of this study was to evaluate the genomic profile of gcGBMs to facilitate more accurate diagnosis and prognostication for this entity. METHODS: Through a multi-institutional collaborative effort, we characterized 10 gcGBMs by chromosome studies, single nucleotide polymorphism microarray analysis, and targeted next-generation sequencing. These tumors were subsequently compared to the genomic and epigenomic profile of glioblastomas described in The Cancer Genome Atlas (TCGA) dataset. RESULTS: Our analysis identified a specific pattern of genome-wide massive loss of heterozygosity (LOH) driven by near haploidization in a subset of glioblastomas with giant cell histology. We compared the genomic signature of these tumors against that of all glioblastomas in the TCGA dataset (n = 367) and confirmed that our cohort of gcGBMs demonstrated a significantly different genomic profile. Integrated genomic and histologic review of the TCGA cohort identified 3 additional gcGBMs with a near haploid genomic profile. CONCLUSIONS: Massive LOH driven by haploidization represents a defining molecular hallmark of a subtype of gcGBM. This unusual mechanism of tumorigenesis provides a diagnostic genomic hallmark to evaluate in future cases, may explain reported differences in survival, and suggests new therapeutic vulnerabilities.

SELECTION OF CITATIONS
SEARCH DETAIL