Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Gen Hosp Psychiatry ; 86: 103-107, 2024.
Article in English | MEDLINE | ID: mdl-38181710

ABSTRACT

OBJECTIVE: Posttraumatic stress symptoms (PSS) due to acute cardiac events are common and may lead patients to avoid secondary prevention behaviors. However, patients' daily experience of cardiac event-induced PSS has not been studied after a potentially traumatic cardiac hospitalization. METHOD: In an observational cohort study, 108 mostly male patients with coronary heart disease were recruited after evaluation for suspected acute coronary syndrome (ACS). One month later, PSS were assessed via telephone-administered PTSD Checklist for DSM-5 (PCL-5). The exposure of interest was elevated (PCL-5 ≥ 20) vs. non-elevated PSS (PCL-5 ≤ 5). The occurrence and severity of cardiac-related intrusive thoughts were assessed 5 times daily for 2 weeks via electronic surveys on a wrist-worn device. RESULTS: Moderate-to-severe intrusive thoughts were experienced by 48.1% of patients but more commonly by elevated-PSS (n = 36; 66.7%) than non-elevated-PSS (n = 72; 38.9%) patients. After adjustment for demographic and clinical characteristics, elevated- vs. non-elevated-PSS patients had a 9-fold higher odds of experiencing a moderate-to-severe intrusive thought during each 2-h assessment interval (adjusted OR = 9.14, 95% CI [2.99, 27.92], p < .01). After adjustment, intrusive thoughts on a 0-to-6 point scale were over two times as intense for elevated-PSS vs. non-elevated-PSS patients. CONCLUSIONS: Intrusive thoughts about cardiac risk were common in patients recently evaluated for ACS, but much more prevalent and intense in those with elevated vs non-elevated PSS.


Subject(s)
Acute Coronary Syndrome , Stress Disorders, Post-Traumatic , Humans , Male , Female , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/diagnosis , Ecological Momentary Assessment , Cognition , Acute Coronary Syndrome/epidemiology , Acute Coronary Syndrome/complications , Cohort Studies
2.
Health Psychol ; 43(1): 34-40, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37917470

ABSTRACT

OBJECTIVE: Threat perceptions during evaluation for acute coronary syndrome (ACS) in the emergency department (ED) predict posttraumatic stress symptoms (PSS). It is unknown how health insurance status affects threat perceptions. We tested whether lacking health insurance is associated with higher threat perceptions and PSS in patients with suspected ACS in the ED and whether threat perceptions mediate associations between lack of health insurance and subsequent PSS. METHOD: Patients in the Columbia University Irving Medical Center ED with suspected ACS enrolled in an observational cohort study of psychological and cardiovascular outcomes. A multivariable linear regression model tested health insurance status as the predictor of ED threat perceptions and PSS 1-month posthospitalization, adjusting for age, gender, education, Charlson Comorbidity Index, and Global Registry of Acute Coronary Events risk score. A bootstrapped mediation model tested health insurance status as the predictor, PSS 1-month posthospitalization as the outcome, and ED threat perceptions as the mediator, with the same covariates. RESULTS: Of 1,741 patients with suspected ACS in the ED (Mage = 61.01 years, SD = 13.27; 47.1% women), a plurality identified as "Other" race (36.1%), Black (23.9%), and White (22.4%), and 10.3% of patients were uninsured. Lack of health insurance was associated with greater threat perceptions, b = -0.16, 95% CI [-0.26, -0.06], p = .002. Threat perceptions mediated the association between lack of health insurance and higher 1-month PSS, indirect effect = -1.04, 95% CI [-1.98, -0.17]. CONCLUSIONS: Lacking health insurance may heighten threat perceptions during ACS evaluation, which may put patients at risk of developing PSS. (PsycInfo Database Record (c) 2023 APA, all rights reserved).


Subject(s)
Acute Coronary Syndrome , Stress Disorders, Post-Traumatic , Humans , Female , Middle Aged , Male , Stress Disorders, Post-Traumatic/diagnosis , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/psychology , Acute Coronary Syndrome/psychology , Cohort Studies , Risk Factors , Emergency Service, Hospital , Insurance, Health
5.
Neurooncol Adv ; 4(1): vdac130, 2022.
Article in English | MEDLINE | ID: mdl-36071925

ABSTRACT

Background: EGFR targeting antibody-drug conjugates (ADCs) are highly effective against EGFR-amplified tumors, but poor distribution across the blood-brain barrier (BBB) limits their efficacy in glioblastoma (GBM) when administered systemically. We studied whether convection-enhanced delivery (CED) can be used to safely infuse ADCs into orthotopic patient-derived xenograft (PDX) models of EGFRvIII mutant GBM. Methods: The efficacy of the EGFR-targeted ADCs depatuxizumab mafodotin (Depatux-M) and Serclutamab talirine (Ser-T) was evaluated in vitro and in vivo. CED was performed in nontumor and tumor-bearing mice. Immunostaining was used to evaluate ADC distribution, pharmacodynamic effects, and normal cell toxicity. Results: Dose-finding studies in orthotopic GBM6 identified single infusion of 2 µg Ser-T and 60 µg Depatux-M as safe and effective associated with extended survival prolongation (>300 days and 95 days, respectively). However, with serial infusions every 21 days, four Ser-T doses controlled tumor growth but was associated with lethal toxicity approximately 7 days after the final infusion. Limiting dosing to two infusions in GBM108 provided profound median survival extension of over 200 days. In contrast, four Depatux-M CED doses were well tolerated and significantly extended survival in both GBM6 (158 days) and GBM108 (310 days). In a toxicity analysis, Ser-T resulted in a profound loss in NeuN+ cells and markedly elevated GFAP staining, while Depatux-M was associated only with modest elevation in GFAP staining. Conclusion: CED of Depatux-M is well tolerated and results in extended survival in orthotopic GBM PDXs. In contrast, CED of Ser-T was associated with a much narrower therapeutic window.

6.
Neuro Oncol ; 24(8): 1261-1272, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35231103

ABSTRACT

BACKGROUND: RBBP4 activates transcription by histone acetylation, but the partner histone acetyltransferases are unknown. Thus, we investigated the hypothesis that RBBP4 interacts with p300 in a complex in glioblastoma (GBM). METHODS: shRNA silencing of RBBP4 or p300 and RNAseq was used to identify genes co-regulated by RBBP4 and p300 in GBM43 patient-derived xenograft (PDX). RBBP4/p300 complex was demonstrated using proximity ligation assay (PLA) and ChIPseq delineated histone H3 acetylation and RBBP4/p300 complex binding in promoters/enhancers. Temozolomide (TMZ)-induced DNA double strand breaks (DSBs) were evaluated by γ-H2AX and proliferation by CyQuant and live cell monitoring assays. In vivo efficacy was based on survival of mice with orthotopic tumors. RESULTS: shRBBP4 and shp300 downregulated 4768 genes among which 1485 (31%) were commonly downregulated by both shRNAs, while upregulated genes were 2484, including 863 (35%) common genes. The pro-survival genes were the top-ranked among the downregulated genes, including C-MYC. RBBP4/p300 complex was demonstrated in the nucleus, and shRBBP4 or shp300 significantly sensitized GBM cells to TMZ compared to the control shNT in vitro (P < .05). Moreover, TMZ significantly prolonged the survival of mice bearing GBM22-shRBBP4 orthotopic tumors compared with control shNT tumors (median shNT survival 52 days vs. median shRBBP4 319 days; P = .001). CREB-binding protein (CBP)/p300 inhibitor CPI-1612 suppressed H3K27Ac and RBBP4/p300 complex target proteins, including C-MYC, and synergistically sensitized TMZ in vitro. Pharmacodynamic evaluation confirmed brain penetration by CPI-1612 supporting further investigation to evaluate efficacy to sensitize TMZ. CONCLUSIONS: RBBP4/p300 complex is present in GBM cells and is a potential therapeutic target.


Subject(s)
Brain Neoplasms , E1A-Associated p300 Protein , Glioblastoma , Retinoblastoma-Binding Protein 4 , Acetylation , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 4/genetics , Retinoblastoma-Binding Protein 4/metabolism , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
7.
Neuro Oncol ; 24(3): 384-395, 2022 03 12.
Article in English | MEDLINE | ID: mdl-34232318

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is an incurable disease with few approved therapeutic interventions. Radiation therapy (RT) and temozolomide (TMZ) remain the standards of care. The efficacy and optimal deployment schedule of the orally bioavailable small-molecule tumor checkpoint controller lisavanbulin alone, and in combination with, standards of care were assessed using a panel of IDH-wildtype GBM patient-derived xenografts. METHODS: Mice bearing intracranial tumors received lisavanbulin +/-RT +/-TMZ and followed for survival. Lisavanbulin concentrations in plasma and brain were determined by liquid chromatography with tandem mass spectrometry, while flow cytometry was used for cell cycle analysis. RESULTS: Lisavanbulin monotherapy showed significant benefit (P < .01) in 9 of 14 PDXs tested (median survival extension 9%-84%) and brain-to-plasma ratios of 1.3 and 1.6 at 2- and 6-hours postdose, respectively, validating previous data suggesting significant exposure in the brain. Prolonged lisavanbulin dosing from RT start until moribund was required for maximal benefit (GBM6: median survival lisavanbulin/RT 90 vs. RT alone 69 days, P = .0001; GBM150: lisavanbulin/RT 143 days vs. RT alone 73 days, P = .06). Similar observations were seen with RT/TMZ combinations (GBM39: RT/TMZ/lisavanbulin 502 days vs. RT/TMZ 249 days, P = .0001; GBM26: RT/TMZ/lisavanbulin 172 days vs. RT/TMZ 121 days, P = .04). Immunohistochemical analyses showed a significant increase in phospho-histone H3 with lisavanbulin treatment (P = .01). CONCLUSIONS: Lisavanbulin demonstrated excellent brain penetration, significant extension of survival alone or in RT or RT/TMZ combinations, and was associated with mitotic arrest. These data provide a strong clinical rationale for testing lisavanbulin in combination with RT or RT/TMZ in GBM patients.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/pathology , Glioblastoma/pathology , Heterografts , Humans , Mice , Microtubules/metabolism , Microtubules/pathology , Temozolomide/therapeutic use
8.
Neuro Oncol ; 23(12): 2066-2075, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34107029

ABSTRACT

BACKGROUND: Appropriately designed preclinical patient-derived xenograft (PDX) experiments are important to accurately inform human clinical trials. There is little experimental design guidance regarding choosing the number of PDX lines to study, and the number of mice within each PDX line. METHODS: Retrospective data from IDH-wildtype glioblastoma preclinical experiments evaluating a uniform regimen of fractionated radiation (RT), temozolomide (TMZ) chemotherapy, and concurrent RT/TMZ across 27 PDX lines were used to evaluate experimental designs and empirically estimate statistical power for ANOVA and Cox regression. RESULTS: Increasing the number of PDX lines resulted in more precise and reproducible estimates of effect size. To achieve 80% statistical power using ANOVA, experiments using a single PDX line required subsampling of 6 mice per PDX for each treatment group to detect a difference in survival of 135 days, and 9 mice per PDX to detect a difference of 100 days. Alternatively, a design that used 10 PDX lines had greater than 80% power to detect a difference of 135 days with a single mouse per PDX per treatment group, a difference of 100 days with 2 mice per PDX per treatment, and 35 days with more than 10 mice per PDX per treatment. Power for Cox regression was slightly smaller than ANOVA for very small experiments regardless of effect size and slightly higher than ANOVA for detecting a smaller effect size of 35 days difference in survival for moderate-to-large experiments. CONCLUSIONS: Experimental designs using few mice across many PDX lines can provide robust results and account for inter-tumor variability.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Cell Line, Tumor , Mice , Research Design , Retrospective Studies , Temozolomide , Xenograft Model Antitumor Assays
9.
Neuro Oncol ; 23(12): 2042-2053, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34050676

ABSTRACT

BACKGROUND: Antibody drug conjugates (ADCs) targeting the epidermal growth factor receptor (EGFR), such as depatuxizumab mafodotin (Depatux-M), is a promising therapeutic strategy for glioblastoma (GBM) but recent clinical trials did not demonstrate a survival benefit. Understanding the mechanisms of failure for this promising strategy is critically important. METHODS: PDX models were employed to study efficacy of systemic vs intracranial delivery of Depatux-M. Immunofluorescence and MALDI-MSI were performed to detect drug levels in the brain. EGFR levels and compensatory pathways were studied using quantitative flow cytometry, Western blots, RNAseq, FISH, and phosphoproteomics. RESULTS: Systemic delivery of Depatux-M was highly effective in nine of 10 EGFR-amplified heterotopic PDXs with survival extending beyond one year in eight PDXs. Acquired resistance in two PDXs (GBM12 and GBM46) was driven by suppression of EGFR expression or emergence of a novel short-variant of EGFR lacking the epitope for the Depatux-M antibody. In contrast to the profound benefit observed in heterotopic tumors, only two of seven intrinsically sensitive PDXs were responsive to Depatux-M as intracranial tumors. Poor efficacy in orthotopic PDXs was associated with limited and heterogeneous distribution of Depatux-M into tumor tissues, and artificial disruption of the BBB or bypass of the BBB by direct intracranial injection of Depatux-M into orthotopic tumors markedly enhanced the efficacy of drug treatment. CONCLUSIONS: Despite profound intrinsic sensitivity to Depatux-M, limited drug delivery into brain tumor may have been a key contributor to lack of efficacy in recently failed clinical trials.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunoconjugates , Pharmaceutical Preparations , Antibodies, Monoclonal, Humanized , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Humans
10.
Gen Hosp Psychiatry ; 70: 98-102, 2021.
Article in English | MEDLINE | ID: mdl-33774490

ABSTRACT

OBJECTIVE: Post-traumatic stress disorder (PTSD) can develop after a life-threatening medical event. According to the enduring somatic treat (EST) model, internal somatic cues (e.g., rapid heart rate) may contribute to symptoms of stroke/TIA-induced PTSD. To address this possibility, the present study tested the association of stroke-induced disability with PTSD symptoms in patients treated for stroke or transient ischemic attack (TIA). METHOD: Participants (n = 300) were drawn from an observational cohort study examining PTSD symptoms in patients admitted to the NewYork Presbyterian Hospital between 2015 and 2017 for a stroke/TIA. Patients self-reported acute stress symptoms in-person approximately 3 days post-stroke/TIA and PTSD symptoms via telephone one month later. Severity of stroke symptoms (i.e., stroke disability) was evaluated using the NIH Stroke Scale prior to hospital discharge. RESULTS: Stroke disability had a significant, positive association with acute stress symptoms early post-stroke/TIA, B = 0.46, se = 0.15, p = .002, and with PTSD symptoms one month later, B = 0.56, se = 0.19, p = .003. CONCLUSIONS: Stroke disability is positively associated with both acute distress and PTSD symptoms one month later following a stroke/TIA, supporting the hypothesis that internal somatic symptoms contribute to the development stroke/TIA-induced PTSD symptoms.


Subject(s)
Ischemic Attack, Transient , Stress Disorders, Post-Traumatic , Stroke , Cohort Studies , Humans , Ischemic Attack, Transient/epidemiology , Stress Disorders, Post-Traumatic/epidemiology , Stroke/epidemiology
12.
J Surg Oncol ; 121(5): 863-872, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31902142

ABSTRACT

Despite the fact laparoscopic liver resections (LLR) for cholangiocarcinoma is still limited, this systematic review addressed surgical and oncological outcomes of LLR to treat both perihilar cholangiocarcinoma (pCCA) and intrahepatic cholangiocarcinoma (iCCA). Five comparative and 20 noncomparative studies were found. Regarding iCCA, LLR had lower blood loss and less need for Pringle maneuver. However, open liver resections (OLR) were performed more for major hepatectomies, with better lymphadenectomy rates and higher number of harvested lymph nodes. High heterogeneity and selection bias were suggested for iCCA studies.


Subject(s)
Bile Duct Neoplasms/surgery , Cholangiocarcinoma/surgery , Hepatectomy/methods , Laparoscopy , Blood Loss, Surgical , Humans , Lymph Node Excision
13.
Clin Cancer Res ; 26(5): 1094-1104, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31852831

ABSTRACT

PURPOSE: Glioblastoma is the most frequent and lethal primary brain tumor. Development of novel therapies relies on the availability of relevant preclinical models. We have established a panel of 96 glioblastoma patient-derived xenografts (PDX) and undertaken its genomic and phenotypic characterization. EXPERIMENTAL DESIGN: PDXs were established from glioblastoma, IDH-wildtype (n = 93), glioblastoma, IDH-mutant (n = 2), diffuse midline glioma, H3 K27M-mutant (n = 1), and both primary (n = 60) and recurrent (n = 34) tumors. Tumor growth rates, histopathology, and treatment response were characterized. Integrated molecular profiling was performed by whole-exome sequencing (WES, n = 83), RNA-sequencing (n = 68), and genome-wide methylation profiling (n = 76). WES data from 24 patient tumors was compared with derivative models. RESULTS: PDXs recapitulate many key phenotypic and molecular features of patient tumors. Orthotopic PDXs show characteristic tumor morphology and invasion patterns, but largely lack microvascular proliferation and necrosis. PDXs capture common and rare molecular drivers, including alterations of TERT, EGFR, PTEN, TP53, BRAF, and IDH1, most at frequencies comparable with human glioblastoma. However, PDGFRA amplification was absent. RNA-sequencing and genome-wide methylation profiling demonstrated broad representation of glioblastoma molecular subtypes. MGMT promoter methylation correlated with increased survival in response to temozolomide. WES of 24 matched patient tumors showed preservation of most genetic driver alterations, including EGFR amplification. However, in four patient-PDX pairs, driver alterations were gained or lost on engraftment, consistent with clonal selection. CONCLUSIONS: Our PDX panel captures the molecular heterogeneity of glioblastoma and recapitulates many salient genetic and phenotypic features. All models and genomic data are openly available to investigators.


Subject(s)
Biomarkers, Tumor/genetics , Exome Sequencing/methods , Genotype , Glioblastoma/classification , Glioblastoma/genetics , Mutation , Phenotype , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/classification , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , ErbB Receptors/genetics , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Male , Mice , Middle Aged , Neoplasm Staging , Promoter Regions, Genetic , Survival Rate , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , Young Adult
14.
Mol Med ; 25(1): 49, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31726966

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is the most commonly used chemotherapeutic agent used to treat glioblastoma (GBM), which causes significant DNA damage to highly proliferative cells. Our observations have added to accumulating evidence that TMZ induces stress-responsive cellular programs known to promote cell survival, including autophagy. As such, targeting these survival pathways may represent new vulnerabilities of GBM after treatment with TMZ. METHODS: Using the T98G human glioma cell line, we assessed the molecular signaling associated with TMZ treatment, the cellular consequences of using the pan-PI3K inhibitor PX-866, and performed clonogenic assays to determine the effect sequential treatment of TMZ and PX-866 had on colony formation. Additionally, we also use subcutaneous GBM patient derived xenograft (PDX) tumors to show relative LC3 protein expression and correlations between survival pathways and molecular markers which dictate clinical responsiveness to TMZ. RESULTS: Here, we report that TMZ can induce autophagic flux in T98G glioma cells. GBM patient-derived xenograft (PDX) tumors treated with TMZ also display an increase in the autophagosome marker LC3 II. Additionally, O6-methylguanine-DNA-methyltransferase (MGMT) expression correlates with PI3K/AKT activity, suggesting that patients with inherent resistance to TMZ (MGMT-high) would benefit from PI3K/AKT inhibitors in addition to TMZ. Accordingly, we have identified that the blood-brain barrier (BBB) penetrant pan-PI3K inhibitor, PX-866, is an early-stage inhibitor of autophagic flux, while maintaining its ability to inhibit PI3K/AKT signaling in glioma cells. Lastly, due to the induction of autophagic flux by TMZ, we provide evidence for sequential treatment of TMZ followed by PX-866, rather than combined co-treatment, as a means to shut down autophagy-induced survival in GBM cells and to enhance apoptosis. CONCLUSIONS: The understanding of how TMZ induces survival pathways, such as autophagy, may offer new therapeutic vulnerabilities and opportunities to use sequential inhibition of alternate pro-survival pathways that regulate autophagy. As such, identification of additional ways to inhibit TMZ-induced autophagy could enhance the efficacy of TMZ.


Subject(s)
Autophagy/drug effects , Glioblastoma/metabolism , Gonanes/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Temozolomide/pharmacology , Apoptosis/drug effects , Brain Neoplasms/metabolism , Cell Line, Tumor , Humans , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects
15.
Neuro Oncol ; 21(11): 1458-1469, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31346613

ABSTRACT

BACKGROUND: Glioblastoma (GBM) represents an aggressive cancer type with a median survival of only 14 months. With fewer than 5% of patients surviving 5 years, comprehensive profiling of these rare patients could elucidate prognostic biomarkers that may confer better patient outcomes. We utilized multiple molecular approaches to characterize the largest patient cohort of isocitrate dehydrogenase (IDH)-wildtype GBM long-term survivors (LTS) to date. METHODS: Retrospective analysis was performed on 49 archived formalin-fixed paraffin embedded tumor specimens from patients diagnosed with GBM at the Mayo Clinic between December 1995 and September 2013. These patient samples were subdivided into 2 groups based on survival (12 LTS, 37 short-term survivors [STS]) and subsequently examined by mutation sequencing, copy number analysis, methylation profiling, and gene expression. RESULTS: Of the 49 patients analyzed in this study, LTS were younger at diagnosis (P = 0.016), more likely to be female (P = 0.048), and MGMT promoter methylated (UniD, P = 0.01). IDH-wildtype STS and LTS demonstrated classic GBM mutations and copy number changes. Pathway analysis of differentially expressed genes showed LTS enrichment for sphingomyelin metabolism, which has been linked to decreased GBM growth, invasion, and angiogenesis. STS were enriched for DNA repair and cell cycle control networks. CONCLUSIONS: While our findings largely report remarkable similarity between these LTS and more typical STS, unique attributes were observed in regard to altered gene expression and pathway enrichment. These attributes may be valuable prognostic markers and are worth further examination. Importantly, this study also underscores the limitations of existing biomarkers and classification methods in predicting patient prognosis.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Survivors/statistics & numerical data , Adult , Aged , Aged, 80 and over , DNA Methylation , Epigenesis, Genetic , Female , Follow-Up Studies , Gene Expression Profiling , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Transcriptome , Young Adult
16.
PLoS One ; 14(2): e0208341, 2019.
Article in English | MEDLINE | ID: mdl-30811507

ABSTRACT

The DNA repair protein O6-methylguanine DNA methyltransferase (MGMT) strongly influences the effectiveness of cancer treatment with chemotherapeutic alkylating agents, and MGMT status in cancer cells could potentially contribute to tailored therapies for individual patients. However, the promoter methylation and immunohistochemical assays presently used for measuring MGMT in clinical samples are indirect, cumbersome and sometimes do not accurately report MGMT activity. Here we directly compare the accuracy of 6 analytical methods, including two fluorescent reporter assays, against the in vitro MGMT activity assay that is considered the gold standard for measuring MGMT DNA repair capacity. We discuss the relative advantages of each method. Our data indicate that two recently developed fluorescence-based assays measure MGMT activity accurately and efficiently, and could provide a functional dimension to clinical efforts to identify patients who are likely to benefit from alkylating chemotherapy.


Subject(s)
Biological Assay/methods , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Fluorescent Dyes/administration & dosage , Tumor Suppressor Proteins/genetics , Antineoplastic Agents, Alkylating/pharmacology , Cell Line , DNA Methylation/drug effects , DNA Repair/genetics , Humans , Promoter Regions, Genetic/genetics
17.
Rev Sci Instrum ; 89(6): 063508, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29960581

ABSTRACT

Plasma Facing Components (PFC) temperature measurement is mandatory to ensure safe high power and long pulse tokamak operation. IR thermography systems which are widely used in magnetic fusions devices become challenged with the choice of tungsten as a PFC material in the ITER tokamak, mainly due to emissivity uncertainties and reflection issues in a hot environment. Embedded temperature measurements are foreseen to cross-check the IR thermography measurements. Fiber Bragg grating sensors are potentially of great interest for this application because they are immune to electromagnetic interference and allow the measurement of a large number of temperature spots on a single fiber. Four optical fiber temperature sensing probes, each of them including 11 regenerated fiber Bragg gratings equally spaced by 12.5 mm (equivalent to one ITER-like tungsten monoblock), have been specifically designed and manufactured for the WEST project (W-tungsten Environment and Steady State Tokamak). The four probes are embedded in W-coated graphite components at two different distances from the surface, 3.5 mm and 7 mm, to cover a wide range of temperatures up to 900 °C. This paper addresses the design and integration issues and the qualification and performance assessment performed in the laboratory. It also shows the first measurements of this new diagnostic achieved in a tokamak environment during baking of the machine and during early diverted plasma exposure.

18.
Mol Cancer Ther ; 17(9): 1893-1901, 2018 09.
Article in English | MEDLINE | ID: mdl-29970480

ABSTRACT

Controversy exists surrounding whether heterogeneous disruption of the blood-brain barrier (BBB), as seen in glioblastoma (GBM), leads to adequate drug delivery sufficient for efficacy in GBM. This question is especially important when using potent, targeted agents that have a poor penetration across an intact BBB. Efficacy of the murine double minute-2 (MDM2) inhibitor SAR405838 was tested in patient-derived xenograft (PDX) models of GBM. In vitro efficacy of SAR405838 was evaluated in PDX models with varying MDM2 expression and those with high (GBM108) and low (GBM102) expression were evaluated for flank and orthotopic efficacy. BBB permeability, evaluated using TexasRed-3 kDa dextran, was significantly increased in GBM108 through VEGFA overexpression. Drug delivery, MRI, and orthotopic survival were compared between BBB-intact (GBM108-vector) and BBB-disrupted (GBM108-VEGFA) models. MDM2-amplified PDX lines with high MDM2 expression were sensitive to SAR405838 in comparison with MDM2 control lines in both in vitro and heterotopic models. In contrast with profound efficacy observed in flank xenografts, SAR405838 was ineffective in orthotopic tumors. Although both GBM108-vector and GBM108-VEGFA readily imaged on MRI following gadolinium contrast administration, GBM108-VEGFA tumors had a significantly enhanced drug and gadolinium accumulation, as determined by MALDI-MSI. Enhanced drug delivery in GBM108-VEGFA translated into a marked improvement in orthotopic efficacy. This study clearly shows that limited drug distribution across a partially intact BBB may limit the efficacy of targeted agents in GBM. Brain penetration of targeted agents is a critical consideration in any precision medicine strategy for GBM. Mol Cancer Ther; 17(9); 1893-901. ©2018 AACR.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Indoles/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Spiro Compounds/pharmacology , Xenograft Model Antitumor Assays , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Indoles/pharmacokinetics , Male , Mice , Proto-Oncogene Proteins c-mdm2/metabolism , Spiro Compounds/pharmacokinetics , Survival Analysis , Treatment Outcome , Tumor Suppressor Protein p53/metabolism
19.
Nat Commun ; 9(1): 2949, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054476

ABSTRACT

Temozolomide (TMZ) was used for the treatment of glioblastoma (GBM) for over a decade, but its treatment benefits are limited by acquired resistance, a process that remains incompletely understood. Here we report that an enhancer, located between the promoters of marker of proliferation Ki67 (MKI67) and O6-methylguanine-DNA-methyltransferase (MGMT) genes, is activated in TMZ-resistant patient-derived xenograft (PDX) lines and recurrent tumor samples. Activation of the enhancer correlates with increased MGMT expression, a major known mechanism for TMZ resistance. We show that forced activation of the enhancer in cell lines with low MGMT expression results in elevated MGMT expression. Deletion of this enhancer in cell lines with high MGMT expression leads to a dramatic reduction of MGMT and a lesser extent of Ki67 expression, increased TMZ sensitivity, and impaired proliferation. Together, these studies uncover a mechanism that regulates MGMT expression, confers TMZ resistance, and potentially regulates tumor proliferation.


Subject(s)
DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/genetics , Glioblastoma/metabolism , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA Methylation , Drug Resistance, Neoplasm/drug effects , E1A-Associated p300 Protein/genetics , Gene Deletion , HEK293 Cells , Heterografts , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Promoter Regions, Genetic/genetics
20.
Front Oncol ; 8: 670, 2018.
Article in English | MEDLINE | ID: mdl-30723695

ABSTRACT

Prognosis of patients with glioblastoma (GBM) remains dismal despite maximal surgical resection followed by aggressive chemo-radiation therapy. Almost every GBM, regardless of genotype, relapses as aggressive recurrent disease. Sensitization of GBM cells to chemo-radiation is expected to extend survival of patients with GBM by enhancing treatment efficacy. The PARP family of enzymes has a pleiotropic role in DNA repair and metabolism and has emerged as an attractive target for sensitization of cancer cells to genotoxic therapies. However, despite promising results from a number of preclinical studies, progress of clinical trials involving PARP inhibitors (PARPI) has been slower in GBM as compared to other malignancies. Preclinical in vivo studies have uncovered limitations of PARPI-mediated targeting of base excision repair, considered to be the likely mechanism of sensitization for temozolomide (TMZ)-resistant GBM. Nevertheless, PARPI remain a promising sensitizing approach for at least a subset of GBM tumors that are inherently sensitive to TMZ. Our PDX preclinical trial has helped delineate MGMT promoter hyper-methylation as a biomarker of the PARPI veliparib-mediated sensitization. In clinical trials, MGMT promoter hyper-methylation now is being studied as a potential predictive biomarker not only for response to TMZ therapy alone, but also PARPI-mediated sensitization of TMZ therapy. Besides the combination approach being investigated, IDH1/2 mutant gliomas associated with 2-hydroxygluterate (2HG)-mediated homologous recombination (HR) defect may potentially benefit from PARPI monotherapy. In this article, we discuss existing results and provide additional data in support of potential alternative mechanisms of sensitization that would help identify potential biomarkers for PARPI-based therapeutic approaches to GBM.

SELECTION OF CITATIONS
SEARCH DETAIL
...