Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
medRxiv ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38947015

ABSTRACT

INTRODUCTION: Normal pressure hydrocephalus (NPH) patients undergoing cortical shunting frequently show early AD pathology on cortical biopsy, which is predictive of progression to clinical AD. The objective of this study was to use samples from this cohort to identify CSF biomarkers for AD-related CNS pathophysiologic changes using tissue and fluids with early pathology, free of post-mortem artifact. METHODS: We analyzed Simoa, proteomic, and metabolomic CSF data from 81 patients with previously documented pathologic and transcriptomic changes. RESULTS: AD pathology on biopsy correlates with CSF ß-amyloid-40/42, neurofilament light chain (NfL), and phospho-tau-181(p-tau181)/ß-amyloid-42, while several gene expression modules correlate with NfL. Proteomic analysis highlights 7 core proteins that correlate with pathology and gene expression changes on biopsy, and metabolomic analysis of CSF identifies disease-relevant groups that correlate with biopsy data.. DISCUSSION: As additional biomarkers are added to AD diagnostic panels, our work provides insight into the CNS pathophysiology these markers are tracking.

2.
Res Sq ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38585856

ABSTRACT

Intratumoral heterogeneity poses a significant challenge to the diagnosis and treatment of glioblastoma (GBM). This heterogeneity is further exacerbated during GBM recurrence, as treatment-induced reactive changes produce additional intratumoral heterogeneity that is ambiguous to differentiate on clinical imaging. There is an urgent need to develop non-invasive approaches to map the heterogeneous landscape of histopathological alterations throughout the entire lesion for each patient. We propose to predictively fuse Magnetic Resonance Imaging (MRI) with the underlying intratumoral heterogeneity in recurrent GBM using machine learning (ML) by leveraging image-localized biopsies with their associated locoregional MRI features. To this end, we develop BioNet, a biologically-informed neural network model, to predict regional distributions of three tissue-specific gene modules: proliferating tumor, reactive/inflammatory cells, and infiltrated brain tissue. BioNet offers valuable insights into the integration of multiple implicit and qualitative biological domain knowledge, which are challenging to describe in mathematical formulations. BioNet performs significantly better than a range of existing methods on cross-validation and blind test datasets. Voxel-level prediction maps of the gene modules by BioNet help reveal intratumoral heterogeneity, which can improve surgical targeting of confirmatory biopsies and evaluation of neuro-oncological treatment effectiveness. The non-invasive nature of the approach can potentially facilitate regular monitoring of the gene modules over time, and making timely therapeutic adjustment. These results also highlight the emerging role of ML in precision medicine.

3.
Nat Commun ; 14(1): 2586, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142563

ABSTRACT

Glioblastoma (GBM) diffusely infiltrates the brain and intermingles with non-neoplastic brain cells, including astrocytes, neurons and microglia/myeloid cells. This complex mixture of cell types forms the biological context for therapeutic response and tumor recurrence. We used single-nucleus RNA sequencing and spatial transcriptomics to determine the cellular composition and transcriptional states in primary and recurrent glioma and identified three compositional 'tissue-states' defined by cohabitation patterns between specific subpopulations of neoplastic and non-neoplastic brain cells. These tissue-states correlated with radiographic, histopathologic, and prognostic features and were enriched in distinct metabolic pathways. Fatty acid biosynthesis was enriched in the tissue-state defined by the cohabitation of astrocyte-like/mesenchymal glioma cells, reactive astrocytes, and macrophages, and was associated with recurrent GBM and shorter survival. Treating acute slices of GBM with a fatty acid synthesis inhibitor depleted the transcriptional signature of this pernicious tissue-state. These findings point to therapies that target interdependencies in the GBM microenvironment.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/pathology , Prognosis , Brain Neoplasms/pathology , Glioma/genetics , Astrocytes/metabolism , Tumor Microenvironment/genetics
4.
Lancet Oncol ; 23(11): 1409-1418, 2022 11.
Article in English | MEDLINE | ID: mdl-36243020

ABSTRACT

BACKGROUND: Topotecan is cytotoxic to glioma cells but is clinically ineffective because of drug delivery limitations. Systemic delivery is limited by toxicity and insufficient brain penetrance, and, to date, convection-enhanced delivery (CED) has been restricted to a single treatment of restricted duration. To address this problem, we engineered a subcutaneously implanted catheter-pump system capable of repeated, chronic (prolonged, pulsatile) CED of topotecan into the brain and tested its safety and biological effects in patients with recurrent glioblastoma. METHODS: We did a single-centre, open-label, single-arm, phase 1b clinical trial at Columbia University Irving Medical Center (New York, NY, USA). Eligible patients were at least 18 years of age with solitary, histologically confirmed recurrent glioblastoma showing radiographic progression after surgery, radiotherapy, and chemotherapy, and a Karnofsky Performance Status of at least 70. Five patients had catheters stereotactically implanted into the glioma-infiltrated peritumoural brain and connected to subcutaneously implanted pumps that infused 146 µM topotecan 200 µL/h for 48 h, followed by a 5-7-day washout period before the next infusion, with four total infusions. After the fourth infusion, the pump was removed and the tumour was resected. The primary endpoint of the study was safety of the treatment regimen as defined by presence of serious adverse events. Analyses were done in all treated patients. The trial is closed, and is registered with ClinicalTrials.gov, NCT03154996. FINDINGS: Between Jan 22, 2018, and July 8, 2019, chronic CED of topotecan was successfully completed safely in all five patients, and was well tolerated without substantial complications. The only grade 3 adverse event related to treatment was intraoperative supplemental motor area syndrome (one [20%] of five patients in the treatment group), and there were no grade 4 adverse events. Other serious adverse events were related to surgical resection and not the study treatment. Median follow-up was 12 months (IQR 10-17) from pump explant. Post-treatment tissue analysis showed that topotecan significantly reduced proliferating tumour cells in all five patients. INTERPRETATION: In this small patient cohort, we showed that chronic CED of topotecan is a potentially safe and active therapy for recurrent glioblastoma. Our analysis provided a unique tissue-based assessment of treatment response without the need for large patient numbers. This novel delivery of topotecan overcomes limitations in delivery and treatment response assessment for patients with glioblastoma and could be applicable for other anti-glioma drugs or other CNS diseases. Further studies are warranted to determine the effect of this drug delivery approach on clinical outcomes. FUNDING: US National Institutes of Health, The William Rhodes and Louise Tilzer Rhodes Center for Glioblastoma, the Michael Weiner Glioblastoma Research Into Treatment Fund, the Gary and Yael Fegel Foundation, and The Khatib Foundation.


Subject(s)
Glioblastoma , Glioma , Humans , Topotecan/adverse effects , Glioblastoma/drug therapy , Convection , Neoplasm Recurrence, Local/drug therapy , Glioma/pathology
5.
Neuro Oncol ; 24(1): 78-87, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34214170

ABSTRACT

BACKGROUND: Gliomas comprise the most common type of primary brain tumor, are highly invasive, and often fatal. IDH-mutated gliomas are particularly challenging to image and there is currently no clinically accepted method for identifying the extent of tumor burden in these neoplasms. This uncertainty poses a challenge to clinicians who must balance the need to treat the tumor while sparing healthy brain from iatrogenic damage. The purpose of this study was to investigate the feasibility of using resting-state blood oxygen level-dependent (BOLD) functional magnetic resonance imaging (fMRI) to detect glioma-related asynchrony in vascular dynamics for distinguishing tumor from healthy brain. METHODS: Twenty-four stereotactically localized biopsies were obtained during open surgical resection from ten treatment-naïve patients with IDH-mutated gliomas who received standard-of-care preoperative imaging as well as echo-planar resting-state BOLD fMRI. Signal intensity for BOLD asynchrony and standard-of-care imaging was compared to cell counts of total cellularity (H&E), tumor density (IDH1 & Sox2), cellular proliferation (Ki67), and neuronal density (NeuN), for each corresponding sample. RESULTS: BOLD asynchrony was directly related to total cellularity (H&E, P = 4 × 10-5), tumor density (IDH1, P = 4 × 10-5; Sox2, P = 3 × 10-5), cellular proliferation (Ki67, P = .002), and inversely related to neuronal density (NeuN, P = 1 × 10-4). CONCLUSIONS: Asynchrony in vascular dynamics, as measured by resting-state BOLD fMRI, correlates with tumor burden and provides a radiographic delineation of tumor boundaries in IDH-mutated gliomas.


Subject(s)
Brain Neoplasms , Glioma , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Glioma/diagnostic imaging , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Magnetic Resonance Imaging , Mutation , Oxygen Saturation , Tumor Burden
6.
Nat Commun ; 12(1): 5659, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580300

ABSTRACT

Early Alzheimer's disease (AD) pathology can be found in cortical biopsies taken during shunt placement for Normal Pressure Hydrocephalus. This represents an opportunity to study early AD pathology in living patients. Here we report RNA-seq data on 106 cortical biopsies from this patient population. A restricted set of genes correlate with AD pathology in these biopsies, and co-expression network analysis demonstrates an evolution from microglial homeostasis to a disease-associated microglial phenotype in conjunction with increasing AD pathologic burden, along with a subset of additional astrocytic and neuronal genes that accompany these changes. Further analysis demonstrates that these correlations are driven by patients that report mild cognitive symptoms, despite similar levels of biopsy ß-amyloid and tau pathology in comparison to patients who report no cognitive symptoms. Taken together, these findings highlight a restricted set of microglial and non-microglial genes that correlate with early AD pathology in the setting of subjective cognitive decline.


Subject(s)
Alzheimer Disease/complications , Cerebral Cortex/pathology , Cognitive Dysfunction/immunology , Gene Regulatory Networks/immunology , Hydrocephalus, Normal Pressure/immunology , Age of Onset , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Astrocytes/immunology , Astrocytes/pathology , Biopsy , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/genetics , Cognitive Dysfunction/pathology , Female , Humans , Hydrocephalus, Normal Pressure/genetics , Hydrocephalus, Normal Pressure/pathology , Hydrocephalus, Normal Pressure/surgery , Male , Microglia/immunology , Microglia/pathology , Neuropsychological Tests , RNA-Seq , Retrospective Studies
7.
Adv Parasitol ; 112: 51-76, 2021.
Article in English | MEDLINE | ID: mdl-34024359

ABSTRACT

Urogenital schistosomiasis remains a major global challenge. Optimal management of this infection depends upon imaging-based assessment of sequelae. Although established imaging modalities such as ultrasonography, plain radiography, magnetic resonance imaging (MRI), narrow band imaging, and computerized tomography (CT) have been used to determine tissue involvement by urogenital schistosomiasis, newer refinements in associated technologies may lead to improvements in patient care. Moreover, application of investigational imaging methods such as confocal laser endomicroscopy and two-photon microscopy in animal models of urogenital schistosomiasis are likely to contribute to our understanding of this infection's pathogenesis. This review discusses prior use of imaging in patients with urogenital schistosomiasis and experimentally infected animals, the advantages and limitations of these modalities, the latest radiologic developments relevant to this infection, and a proposed future diagnostic standard of care for management of afflicted patients.


Subject(s)
Schistosomiasis haematobia/diagnostic imaging , Animals , Humans , Magnetic Resonance Imaging , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Narrow Band Imaging , Tomography, X-Ray Computed , Ultrasonography , Urinary Bladder/diagnostic imaging , Urinary Bladder/parasitology , Urogenital System/parasitology
8.
Langenbecks Arch Surg ; 406(2): 401-404, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33547483

ABSTRACT

PURPOSE: Acquiring SARS-CoV-2 infection for uninfected patients undergoing surgical procedures following a COVID positive (COVID+) patient is of significant concern, both for patients seeking medical care in hospital settings and for management of surgical services during pandemic times. METHODS: Using data identifying all COVID+ surgical patients during the initial pandemic peak in New York City (March 15 to May 15, 2020), we analyzed the rate of postoperative symptomatic SARS-CoV-2 infection in COVID negative (COVID-) patients undergoing surgery in the same operating room within 48 h, thus determining nosocomial symptomatic infection rate attributable to COVID operating room exposure. RESULTS: Five COVID- patients directly followed a COVID+ patient, while 19 patients were exposed to COVID+ operating rooms within 24 h. By 48 h, 21 additional patients were exposed. No exposed patients acquired symptomatic SARS-CoV-2 infection postoperatively. CONCLUSION: With implementation of infection prevention and control procedures in the operating room under local pandemic conditions, our findings suggest that the risk of acquiring SARS-CoV-2 infection, when following a COVID+ patient in the same operating room, is very low.


Subject(s)
COVID-19/prevention & control , COVID-19/transmission , Cross Infection/prevention & control , Infection Control/organization & administration , Operating Rooms , Postoperative Complications/virology , COVID-19/diagnosis , Cross Infection/diagnosis , Cross Infection/virology , Humans , Postoperative Complications/diagnosis , Postoperative Complications/prevention & control , Retrospective Studies , Risk Assessment
9.
Ann Surg ; 273(1): 41-48, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33156061

ABSTRACT

OBJECTIVE: To determine the risk of acquiring perioperative COVID-19 infection in previously COVID-19 negative patients. SUMMARY OF BACKGROUND DATA: During the initial peak of the COVID-19 pandemic, there was significant concern of hospital acquired COVID-19 infections. Medical centers rapidly implemented systems to minimize perioperative transmission, including routine preoperative testing, patient isolation, and enhanced cleaning. METHODS: In this retrospective cohort study, medical records of all adult patients who underwent surgery at our quaternary, acute care hospital between March 15 and May 15, 2020 were reviewed. The risk of preoperatively negative patients developing symptomatic COVID-19 within 2-14 days postoperatively was determined. Surgical characteristics, outcomes, and complications were compared between those with and without acquired perioperative COVID-19 infection. RESULTS: Among 501 negative patients undergoing index surgeries, 9 (1.8%) developed symptomatic COVID-19 in the postoperative period; all occurred before implementation of routine preoperative testing [9/243, 3.7% vs 0/258, 0%, odds ratio (OR): 0.048, P = 0.036]. No patient who was polymerase-chain-reaction negative on the day of surgery (n = 170) developed postoperative infection. Perioperative infection was associated with preoperative diabetes (OR: 3.70, P = 0.042), cardiovascular disease (OR: 3.69, P = 0.043), angiotensin receptor blocker use (OR: 6.58, P = 0.004), and transplant surgery (OR: 11.00, P = 0.002), and multiple complications, readmission (OR: 5.50, P = 0.029) and death (OR: 12.81, P = 0.001). CONCLUSIONS: During the initial peak of the COVID-19 pandemic, there was minimal risk of acquiring symptomatic perioperative COVID-19 infection, especially after the implementation of routine preoperative testing. However, perioperative COVID-19 infection was associated with poor postoperative outcome.


Subject(s)
COVID-19/epidemiology , Elective Surgical Procedures , Pandemics , Perioperative Care/methods , Postoperative Complications/epidemiology , Risk Assessment/methods , SARS-CoV-2 , Adolescent , Adult , Aged , Comorbidity , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Retrospective Studies , United States/epidemiology , Young Adult
10.
J Neurooncol ; 143(3): 457-464, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31054100

ABSTRACT

BACKGROUND: Prior studies have highlighted infratentorial tumor location as a prognostic factor for solitary fibrous tumor (SFT) and hemangiopericytoma (HPC) of the central nervous system (CNS), and spinal location is considered a positive prognostic factor for other tumors of the CNS. While SFT/HPC of the CNS is known to frequently arise from the spinal meninges, there are no case series that report outcomes for spinally located CNS tumors, and their prognosis in relation to intracranial and other CNS-located tumors is unknown. OBJECTIVE: To investigate outcomes for patients with SFT/HPC of the spinal meninges. METHODS: The Surveillance, Epidemiology, and End-Results Program was used to identify patients with SFT/HPC within the CNS from 1993-2015. We retrospectively analyzed the relationship between tumor location (spinal vs. Brain and other CNS) and survival. RESULTS: We identified 551 cases of CNS SFT/HPC, 64 (11.6%) of which were primary tumors of the spinal meninges. Spinal tumors were more likely than brain and other CNS tumors to be SFT vs. HPC (37.5 vs. 12%, p < 0.001), benign (42.2 vs. 20.3%, p < 0.001), and less than 5 cm (53.1 vs. 35.7%, p < 0.001). The 10-year survival rates for spinal and brain/other CNS tumors were 85 and 58%, respectively. Median survival time was significantly longer for spinal tumors (median survival not reached vs. 138 months, p = 0.03, HR = 0.41 [95% CI 0.18-0.94]). On multivariable analysis, spinal tumor location was associated with improved survival over tumors located in the brain and other CNS (HR = 0.36 [95% CI 0.15-0.89], p = 0.03). CONCLUSION: Spinal tumor location is associated with improved survival in patients with SFT/HPC of the CNS. Larger institutional studies are necessary to characterize the relationship between tumor location and other relevant factors such as presentation and amenability to gross-total resection and adjuvant radiotherapy. Future studies exploring optimal management of spinally located tumors are also needed.


Subject(s)
Hemangiopericytoma/mortality , Solitary Fibrous Tumors/mortality , Spinal Neoplasms/mortality , Spinal Neoplasms/pathology , Female , Follow-Up Studies , Hemangiopericytoma/pathology , Hemangiopericytoma/surgery , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , SEER Program , Solitary Fibrous Tumors/pathology , Solitary Fibrous Tumors/surgery , Spinal Neoplasms/surgery , Survival Rate
12.
Genome Med ; 10(1): 57, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30041684

ABSTRACT

BACKGROUND: Despite extensive molecular characterization, we lack a comprehensive understanding of lineage identity, differentiation, and proliferation in high-grade gliomas (HGGs). METHODS: We sampled the cellular milieu of HGGs by profiling dissociated human surgical specimens with a high-density microwell system for massively parallel single-cell RNA-Seq. We analyzed the resulting profiles to identify subpopulations of both HGG and microenvironmental cells and applied graph-based methods to infer structural features of the malignantly transformed populations. RESULTS: While HGG cells can resemble glia or even immature neurons and form branched lineage structures, mesenchymal transformation results in unstructured populations. Glioma cells in a subset of mesenchymal tumors lose their neural lineage identity, express inflammatory genes, and co-exist with marked myeloid infiltration, reminiscent of molecular interactions between glioma and immune cells established in animal models. Additionally, we discovered a tight coupling between lineage resemblance and proliferation among malignantly transformed cells. Glioma cells that resemble oligodendrocyte progenitors, which proliferate in the brain, are often found in the cell cycle. Conversely, glioma cells that resemble astrocytes, neuroblasts, and oligodendrocytes, which are non-proliferative in the brain, are generally non-cycling in tumors. CONCLUSIONS: These studies reveal a relationship between cellular identity and proliferation in HGG and distinct population structures that reflects the extent of neural and non-neural lineage resemblance among malignantly transformed cells.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Single-Cell Analysis , Transcriptome , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Cell Transformation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Neuroglia/pathology , Neurons/pathology
13.
J Parasitol ; 101(5): 542-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26186584

ABSTRACT

Although Syrian golden hamsters are widely used as hosts for experimental infection by Schistosoma haematobium , surprisingly little is known about the course of infection and associated intensity (as defined by measures of parasite burden). As such, we sought to define inexpensive, simple, noninvasive, and accurate methods for assessing and predicting the severity of disease in S. haematobium -infected hamsters in order to prevent premature hamster sacrifice and unexpected morbidity and mortality. Through monitoring the weight and behavior of infected hamsters, we determined that the weight-loss patterns of infected hamsters are highly correlated with commonly used measures of the severity of infection (i.e., numbers of eggs passed in the stool, worm burdens, and total egg yields). In contrast, we found no significant correlation between hamster weight-loss patterns and egg yields from liver and intestinal tissues. Our findings suggest that a more complex relationship exists among worm burden, fecundity, and egg passage in the feces than previously appreciated. Regardless, our data may be useful for workers seeking to optimize harvests of S. haematobium eggs and worms from infected hamsters for downstream applications.


Subject(s)
Disease Models, Animal , Mesocricetus/parasitology , Rodent Diseases/parasitology , Schistosomiasis haematobia/veterinary , Animals , Body Weight , Bulinus , Cricetinae , Feces/parasitology , Male , Mesocricetus/anatomy & histology , Parasite Egg Count , Schistosoma haematobium/physiology , Schistosomiasis haematobia/parasitology , Urogenital System/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...