Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Neuroradiol J ; : 19714009241242642, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38565221

ABSTRACT

BACKGROUND AND PURPOSE: Perivascular spaces (PVS) are interstitial fluid-filled spaces surrounding blood vessels traversing the deep gray nuclei and white matter of the brain. These are commonly encountered on CT and MR imaging and are generally asymptomatic and of no clinical significance. However, occasional changes in the size of focal PVS, for example, when enlarging, may mimic pathologies including neoplasms and infections, hence potentially confounding radiological interpretation. Given these potential diagnostic issues, we sought to better characterize common clinical and imaging features of focal PVS demonstrating size fluctuations. MATERIALS AND METHODS: Upon institutional approval, we retrospectively identified 4 cases demonstrating PVS with size changes at our institution. To supplement our cases, we also performed a literature review, which identified an additional 14 cases. Their clinical and imaging data were analyzed to identify characteristic features. RESULTS: Of the 18 total cases (including the 4 institutional cases), 10 cases increased and 8 decreased in size. These focal PVS ranged from 0.4-4.5 cm in size. Whereas a decrease in size did not represent a diagnostic issue, focal increase in size of PVS led to concerning differential diagnoses in at least 30% of the radiology reports. These enlarging PVS were most found in the basal ganglia and temporal lobe, and in patients with previous brain radiation treatment. CONCLUSION: Focal size change of PVS can occur, especially years after brain radiation treatment. Being cognizant of this benign finding is important to consider in the differential diagnosis to avoid undue patient anxiety or unnecessary medical intervention.

2.
BJU Int ; 133(6): 656-664, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38506328

ABSTRACT

OBJECTIVE: To determine the prevalence of 'spin' (i.e., reporting practices that distort the interpretation of results by positively reflecting negative findings or downplaying potential harms) strategies and level of spin in urological observational studies and whether the use of spin has changed over time. MATERIALS AND METHODS: MEDLINE and Embase were searched to identify observational studies comparing therapeutic interventions in the top five urology journals and major urological subspecialty journals, published between 2000 and 2001, 2010 and 2011, and 2020 and 2021. RESULTS: A total of 235 studies were included. Spin was identified in 81% of studies, with a median of two strategies per study. The most commonly used strategies were inadequate implication for clinical practice (30%), causal language or causal claim (29%), and use of linguistic spin (29%). Moderate to high levels of spin were found in 55% of conclusions. From 2000 to 2020, the average number of strategies used has significantly decreased each decade (H = 27.459, P < 0.001), and the median level of spin in conclusions was significantly lower in studies published in the 2020s and 2010s than in the 2000s (H = 11.649, P = 0.003). CONCLUSIONS: Our results suggest that 81% of urological observational studies comparing therapeutic interventions contained spin. Over the past two decades, the use of spin has significantly declined, but this remains an area for improvement, with 70% of included studies published in the 2020s employing spin. Medical writing should scrupulously avoid words or phrases that are not supported by data in the manuscript.


Subject(s)
Urology , Humans , Observational Studies as Topic
3.
Article in English | MEDLINE | ID: mdl-38521092

ABSTRACT

BACKGROUND AND PURPOSE: Interest in artificial intelligence (AI) and machine learning (ML) has been growing in neuroradiology, but there is limited knowledge on how this interest has manifested into research and specifically, its qualities and characteristics. This study aims to characterize the emergence and evolution of AI/ML articles within neuroradiology and provide a comprehensive overview of the trends, challenges, and future directions of the field. MATERIALS AND METHODS: We performed a bibliometric analysis of the American Journal of Neuroradiology (AJNR): the journal was queried for original research articles published since inception (Jan. 1, 1980) to Dec. 3, 2022 that contained any of the following key terms: "machine learning", "artificial intelligence", "radiomics", "deep learning", "neural network", "generative adversarial network", "object detection", or "natural language processing". Articles were screened by two independent reviewers, and categorized into Statistical Modelling (Type 1), AI/ML Development (Type 2), both representing developmental research work but without a direct clinical integration, or End-user Application (Type 3) which is the closest surrogate of potential AI/ML integration into day-to-day practice. To better understand the limiting factors to Type 3 articles being published, we analyzed Type 2 articles as they should represent the precursor work leading to Type 3. RESULTS: A total of 182 articles were identified with 79% being non-integration focused (Type 1 n = 53, Type 2 n = 90) and 21% (n = 39) being Type 3. The total number of articles published grew roughly five-fold in the last five years, with the non-integration focused articles mainly driving this growth. Additionally, a minority of Type 2 articles addressed bias (22%) and explainability (16%). These articles were primarily led by radiologists (63%), with most of them (60%) having additional postgraduate degrees. CONCLUSIONS: AI/ML publications have been rapidly increasing in neuroradiology with only a minority of this growth being attributable to end-user application. Areas identified for improvement include enhancing the quality of Type 2 articles, namely external validation, and addressing both bias and explainability. These results ultimately provide authors, editors, clinicians, and policymakers important insights to promote a shift towards integrating practical AI/ML solutions in neuroradiology. ABBREVIATIONS: AI = artificial intelligence; ML = machine learning.

4.
Clin Cancer Res ; 27(21): 6054-6064, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34376537

ABSTRACT

PURPOSE: The development of allogeneic chimeric antigen receptor (CAR) T-cell therapies for off-the-shelf use is a major goal that faces two main immunologic challenges, namely the risk of graft-versus-host disease (GvHD) induction by the transferred cells and the rejection by the host immune system limiting their persistence. In this work we assessed the direct and indirect antitumor effect of allogeneic CAR-engineered invariant natural killer T (iNKT) cells, a cell population without GvHD-induction potential that displays immunomodulatory properties. EXPERIMENTAL DESIGN: After assessing murine CAR iNKT cells direct antitumor effects in vitro and in vivo, we employed an immunocompetent mouse model of B-cell lymphoma to assess the interaction between allogeneic CAR iNKT cells and endogenous immune cells. RESULTS: We demonstrate that allogeneic CAR iNKT cells exerted potent direct and indirect antitumor activity when administered across major MHC barriers by inducing tumor-specific antitumor immunity through host CD8 T-cell cross-priming. CONCLUSIONS: In addition to their known direct cytotoxic effect, allogeneic CAR iNKT cells induce host CD8 T-cell antitumor responses, resulting in a potent antitumor effect lasting longer than the physical persistence of the allogeneic cells. The utilization of off-the-shelf allogeneic CAR iNKT cells could meet significant unmet needs in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Immunotherapy, Adoptive/methods , Natural Killer T-Cells , Neoplasms/genetics , Neoplasms/therapy , Allogeneic Cells , Animals , Mice
5.
Oncoimmunology ; 9(1): 1757360, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32923113

ABSTRACT

Recent advances in novel immune strategies, particularly chimeric antigen receptor (CAR)-bearing T-cells, have shown limited efficacy against glioblastoma (GBM) in clinical trials. We currently have an incomplete understanding of how these emerging therapies integrate with the current standard of care, specifically radiation therapy (RT). Additionally, there is an insufficient number of preclinical studies monitoring these therapies with high spatiotemporal resolution. To address these limitations, we report the first longitudinal fluorescence-based intravital microscopy imaging of CAR T-cells within an orthotopic GBM preclinical model to illustrate the necessity of RT for complete therapeutic response. Additionally, we detail the first usage of murine-derived CAR T-cells targeting the disialoganglioside GD2 in an immunocompetent tumor model. Cell culture assays demonstrated substantial GD2 CAR T-cell-mediated killing of murine GBM cell lines SB28 and GL26 induced to overexpress GD2. Complete antitumor response in advanced syngeneic orthotopic models of GBM was achieved only when a single intravenous dose of GD2 CAR T-cells was following either sub-lethal whole-body irradiation or focal RT. Intravital microscopy imaging successfully visualized CAR T-cell homing and T-cell mediated apoptosis of tumor cells in real-time within the tumor stroma. Findings indicate that RT allows for rapid CAR T-cell extravasation from the vasculature and expansion within the tumor microenvironment, leading to a more robust and lasting immunologic response. These exciting results highlight potential opportunities to improve intravenous adoptive T-cell administration in the treatment of GBM through concurrent RT. Additionally, they emphasize the need for advancements in immunotherapeutic homing to and extravasation through the tumor microenvironment.


Subject(s)
Glioblastoma , Animals , Cell Line, Tumor , Glioblastoma/radiotherapy , Immunotherapy, Adoptive , Intravital Microscopy , Mice , Receptors, Antigen, T-Cell , T-Lymphocytes , Tumor Microenvironment , Xenograft Model Antitumor Assays
6.
SLAS Discov ; 23(7): 603-612, 2018 08.
Article in English | MEDLINE | ID: mdl-29634393

ABSTRACT

High-throughput flow cytometry is an attractive platform for the analysis of adoptive cellular therapies such as chimeric antigen receptor T cell therapy (CAR-T) because it allows for the concurrent measurement of T cell-dependent cellular cytotoxicity (TDCC) and the functional characterization of engineered T cells with respect to percentage of CAR transduction, T cell phenotype, and measurement of T cell function such as activation in a single assay. The use of adherent tumor cell lines can be challenging in these flow-based assays. Here, we present the development of a high-throughput flow-based assay to measure TDCC for a CAR-T construct co-cultured with multiple adherent tumor cell lines. We describe optimal assay conditions (such as adherent cell dissociation techniques to minimize impact on cell viability) that result in robust cytotoxicity assays. In addition, we report on the concurrent use of T cell transduction and activation antibody panels (CD25) that provide further dissection of engineered T cell function. In conclusion, we present the development of a high-throughput flow cytometry method allowing for in vitro interrogation of solid tumor, targeting CAR-T cell-mediated cytotoxicity, CAR transduction, and engineered T cell characterization in a single assay.


Subject(s)
Cytotoxicity, Immunologic , Flow Cytometry , High-Throughput Screening Assays , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , Cell Line, Tumor , Flow Cytometry/methods , Humans , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Transduction, Genetic
7.
Nat Med ; 24(5): 572-579, 2018 05.
Article in English | MEDLINE | ID: mdl-29662203

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) and other diffuse midline gliomas (DMGs) with mutated histone H3 K27M (H3-K27M)1-5 are aggressive and universally fatal pediatric brain cancers 6 . Chimeric antigen receptor (CAR)-expressing T cells have mediated impressive clinical activity in B cell malignancies7-10, and recent results suggest benefit in central nervous system malignancies11-13. Here, we report that patient-derived H3-K27M-mutant glioma cell cultures exhibit uniform, high expression of the disialoganglioside GD2. Anti-GD2 CAR T cells incorporating a 4-1BBz costimulatory domain 14 demonstrated robust antigen-dependent cytokine generation and killing of DMG cells in vitro. In five independent patient-derived H3-K27M+ DMG orthotopic xenograft models, systemic administration of GD2-targeted CAR T cells cleared engrafted tumors except for a small number of residual GD2lo glioma cells. To date, GD2-targeted CAR T cells have been well tolerated in clinical trials15-17. Although GD2-targeted CAR T cell administration was tolerated in the majority of mice bearing orthotopic xenografts, peritumoral neuroinflammation during the acute phase of antitumor activity resulted in hydrocephalus that was lethal in a fraction of animals. Given the precarious neuroanatomical location of midline gliomas, careful monitoring and aggressive neurointensive care management will be required for human translation. With a cautious multidisciplinary clinical approach, GD2-targeted CAR T cell therapy for H3-K27M+ diffuse gliomas of pons, thalamus and spinal cord could prove transformative for these lethal childhood cancers.


Subject(s)
Brain Neoplasms/immunology , Gangliosides/metabolism , Glioma/immunology , Histones/metabolism , Immunotherapy, Adoptive , Lysine/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Humans , Methylation , Mice , Xenograft Model Antitumor Assays
8.
J Immunol ; 193(8): 3835-41, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25281753

ABSTRACT

Programmed death ligand 1 (PD-L1, also known as B7 homolog 1 or CD274) is a major obstacle to antitumor immunity because it tolerizes/anergizes tumor-reactive T cells by binding to its receptor programmed death-1 (CD279), renders tumor cells resistant to CD8(+) T cell- and FasL-mediated lysis, and tolerizes T cells by reverse signaling through T cell-expressed CD80. PD-L1 is abundant in the tumor microenvironment, where it is expressed by many malignant cells, as well as by immune cells and vascular endothelial cells. The critical role of PD-L1 in obstructing antitumor immunity has been demonstrated in multiple animal models and in recent clinical trials. This article reviews the mechanisms by which PD-L1 impairs antitumor immunity and discusses established and experimental strategies for maintaining T cell activation in the presence of PD-L1-expressing cells in the tumor microenvironment.


Subject(s)
B7-1 Antigen/immunology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Apoptosis/immunology , Fas Ligand Protein/immunology , Humans , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Signal Transduction/immunology , Tumor Microenvironment/immunology
9.
Cancer Immunol Res ; 2(7): 610-5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24819296

ABSTRACT

Tumor cells use various methods of immunosuppression to overcome antitumor immunity. One such method is that of programmed death ligand-1 (PD-L1 or B7-H1), which upon binding its receptor PD-1 on T cells triggers apoptotic death of the activated T cells. Overexpression of the costimulatory molecule CD80 on PD-L1(+) tumor cells, or inclusion of a soluble form of CD80 (CD80-Fc), maintains the activation of PD-1(+)-activated T cells. Using T cells from CD28-deficient mice and antibodies to block CD28 on human T cells, we now report that a soluble form of CD80 mediates this effect by simultaneously neutralizing PD-1-PD-L1-mediated immunosuppression and by providing CD80-CD28 costimulation, and is more effective than antibodies to PD-L1 or PD-1 in maintaining IFNγ production by PD-1(+) activated T cells. Therefore, soluble CD80 may be a more effective therapeutic than these checkpoint antibodies for facilitating the development and maintenance of antitumor immunity because it has the dual functions of preventing PD-L1-mediated immunosuppression and simultaneously delivering the second signal for T-cell activation.


Subject(s)
B7-1 Antigen/immunology , B7-H1 Antigen/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Animals , CD28 Antigens/deficiency , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Tumor Cells, Cultured , Tumor Escape/immunology
10.
J Immunol ; 191(5): 2829-36, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918985

ABSTRACT

Many tumor cells escape anti-tumor immunity through their expression of programmed death ligand-1 (PDL1 or B7-H1), which interacts with T cell-expressed PD1 and results in T cell apoptosis. We previously reported that transfection of human tumor cells with a membrane-bound form of the human costimulatory molecule CD80 prevented PD1 binding and restored T cell activation. We now report that a membrane-bound form of murine CD80 similarly reduces PDL1-PD1-mediated suppression by mouse tumor cells and that a soluble protein consisting of the extracellular domains of human or mouse CD80 fused to the Fc domain of IgG1 (CD80-Fc) overcomes PDL1-mediated suppression by human and mouse tumor cells, respectively. T cell activation experiments with human and mouse tumor cells indicate that CD80-Fc facilitates T cell activation by binding to PDL1 to inhibit PDL1-PD1 interactions and by costimulating through CD28. CD80-Fc is more effective in preventing PD1-PDL1-mediated suppression and restoring T cell activation compared with treatment with mAb to either PD1 or PDL1. These studies identify CD80-Fc as an alternative and potentially more efficacious therapeutic agent for overcoming PDL1-induced immune suppression and facilitating tumor-specific immunity.


Subject(s)
B7-1 Antigen/immunology , B7-H1 Antigen/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Animals , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/metabolism , Transfection
11.
J Immunol ; 186(12): 6822-9, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21555531

ABSTRACT

Programmed death ligand 1 (PDL1, or B7-H1) is expressed constitutively or is induced by IFN-γ on the cell surface of most human cancer cells and acts as a "molecular shield" by protecting tumor cells from T cell-mediated destruction. Using seven cell lines representing four histologically distinct solid tumors (lung adenocarcinoma, mammary carcinoma, cutaneous melanoma, and uveal melanoma), we demonstrate that transfection of human tumor cells with the gene encoding the costimulatory molecule CD80 prevents PDL1-mediated immune suppression by tumor cells and restores T cell activation. Mechanistically, CD80 mediates its effects through its extracellular domain, which blocks the cell surface expression of PDL1 but does not prevent intracellular expression of PDL1 protein. These studies demonstrate a new role for CD80 in facilitating antitumor immunity and suggest new therapeutic avenues for preventing tumor cell PDL1-induced immune suppression.


Subject(s)
Antigens, CD/immunology , B7-1 Antigen/immunology , T-Lymphocytes/immunology , B7-1 Antigen/administration & dosage , B7-1 Antigen/genetics , B7-H1 Antigen , Cell Line, Tumor , Gene Expression/drug effects , Humans , Immunotherapy/methods , Lymphocyte Activation/immunology , Transfection
12.
Ethn Dis ; 20(1 Suppl 1): S1-101-3, 2010.
Article in English | MEDLINE | ID: mdl-20521395

ABSTRACT

Lead toxicity has been associated with its ability to interact and damage DNA. However, its molecular mechanisms of action are not fully understood. In vitro studies in our laboratory indicated that lead nitrate (PbNO3) induces cytotoxicity and oxidative stress to human liver carcinoma (HepG2) cells in a dose-dependent manner. In this research, we hypothesized that n-acetyl-cysteine (NAC), a known antioxidant compound, affords protection against lead-induced cell death associated with genotoxic damage. To test this hypothesis, HepG2 cells were treated either with a physiologic dose of NAC, NAC plus PbNO3, or PbNO3 alone, followed by incubation in humidified 5% CO2 incubator at 37 degrees C for 48 hr. The cell viability was determined by trypan blue exclusion test. The degree of DNA damage was detected by micro gel electrophoresis (comet) assay. Our results showed that lead exposure induces a substantial cytotoxicity as well as a significant genotoxicity to HepG2 cells. However, co-treatment with a physiologic dose (500 microM) of NAC slightly increases cell viability, and significantly reduced (P < .05) the degree of DNA damage. Hence, NAC treatment may be a promising therapeutic candidate for chemoprevention against lead toxicity, based on its ability to scavenge free radicals.


Subject(s)
Acetylcysteine/pharmacology , DNA Damage/drug effects , Free Radical Scavengers/pharmacology , Lead/toxicity , Cell Survival/drug effects , Comet Assay , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Oxidative Stress/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...