Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Publication year range
1.
Ned Tijdschr Geneeskd ; 1672023 03 08.
Article in Dutch | MEDLINE | ID: mdl-36920315

ABSTRACT

BACKGROUND: Cat scratch disease (CSD) is caused by Bartonella henselae infection. CSD is usually characterized by self-limiting regional lymphadenopathy. However, sometimes CSD presents as a disseminated disease with multiple organ involvement. CASE DESCRIPTIONS: We describe two patients with CSD. Patient A, an 18-year old woman, was referred because of fatigue, a subfebrile temperature and axillary lymphadenopathy. Patient B, a 50-year old man, visited the emergency ward with fever, back pain and painful inguinal lymphadenopathy. MRI showed osteitis of vertebrae and hepatic abcesses. In both patients symptoms started after being scratched by a cat and both were tested positive for infection with Bartonella henselae. Patient B was treated with antibiotics. Both patients made a full recovery. CONCLUSION: Recent contact with a cat in a patient with unexplained fever and lymphadenopathy raises the possibility of CSD. Diagnosis can be confirmed by serologic testing, histopathology or PCR. Antimicrobial treatment must be considered in all cases.


Subject(s)
Bartonella henselae , Cat-Scratch Disease , Lymphadenopathy , Humans , Cat-Scratch Disease/complications , Cat-Scratch Disease/diagnosis , Cat-Scratch Disease/drug therapy , Anti-Bacterial Agents/therapeutic use , Lymphadenopathy/diagnosis , Lymphadenopathy/etiology , Liver
2.
J Nucl Med ; 61(12): 1839-1844, 2020 12.
Article in English | MEDLINE | ID: mdl-32358092

ABSTRACT

18F-BMS-986192, an adnectin-based human programmed cell death ligand 1 (PD-L1) tracer, was developed to noninvasively determine whole-body PD-L1 expression by PET. We evaluated the usability of 18F-BMS-986192 PET to detect different PD-L1 expression levels and therapy-induced changes in PD-L1 expression in tumors. Methods: In vitro binding assays with 18F-BMS-986192 were performed on human tumor cell lines with different total cellular and membrane PD-L1 protein expression levels. Subsequently, PET imaging was performed on immunodeficient mice xenografted with these cell lines. The mice were treated with interferon γ (IFNγ) intraperitoneally for 3 d or with the mitogen-activated protein kinase kinase inhibitor selumetinib by oral gavage for 24 h. Afterward, 18F-BMS-986192 was administered intravenously, followed by a 60-min dynamic PET scan. Tracer uptake was expressed as percentage injected dose per gram of tissue. Tissues were collected to evaluate ex vivo tracer biodistribution and to perform flow cytometric, Western blot, and immunohistochemical tumor analyses. Results:18F-BMS-986192 uptake reflected PD-L1 membrane levels in tumor cell lines, and tumor tracer uptake in mice was associated with PD-L1 expression measured immunohistochemically. In vitro IFNγ treatment increased PD-L1 expression in the tumor cell lines and caused up to a 12-fold increase in tracer binding. In vivo, IFNγ affected neither PD-L1 tumor expression measured immunohistochemically nor 18F-BMS-986192 tumor uptake. In vitro, selumetinib downregulated cellular and membrane levels of PD-L1 in tumor cells by 50% as measured by Western blotting and flow cytometry. In mice, selumetinib lowered cellular, but not membrane, PD-L1 levels of tumors, and consequently, no treatment-induced change in 18F-BMS-986192 tumor uptake was observed. Conclusion:18F-BMS-986192 PET imaging allows detection of membrane-expressed PD-L1 as soon as 60 min after tracer injection. The tracer can discriminate a range of tumor cell PD-L1 membrane expression levels.


Subject(s)
B7-H1 Antigen/metabolism , Gene Expression Regulation , Molecular Imaging/methods , Peptide Fragments , Positron-Emission Tomography/methods , Animals , Cell Line, Tumor , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/metabolism , Humans , Mice , Radioactive Tracers , Tissue Distribution
3.
J Pathol ; 249(1): 52-64, 2019 09.
Article in English | MEDLINE | ID: mdl-30972766

ABSTRACT

Immune checkpoint inhibitors targeting programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) have improved the survival of patients with non-small cell lung cancer (NSCLC). Still, many patients do not respond to these inhibitors. PD-L1 (CD274) expression, one of the factors that influences the efficacy of immune checkpoint inhibitors, is dynamic. Here, we studied the regulation of PD-L1 expression in NSCLC without targetable genetic alterations in EGFR, ALK, BRAF, ROS1, MET, ERBB2 and RET. Analysis of RNA sequencing data from these NSCLCs revealed that inferred IFNγ, EGFR and MAPK signaling correlated with CD274 gene expression in lung adenocarcinoma. In a representative lung adenocarcinoma cell line panel, stimulation with EGF or IFNγ increased CD274 mRNA and PD-L1 protein and membrane levels, which were further enhanced by combining EGF and IFNγ. Similarly, tumor cell PD-L1 membrane levels increased after coculture with activated peripheral blood mononuclear cells. Inhibition of the MAPK pathway, using EGFR inhibitors cetuximab and erlotinib or the MEK 1 and 2 inhibitor selumetinib, prevented EGF- and IFNγ-induced CD274 mRNA and PD-L1 protein and membrane upregulation, but had no effect on IFNγ-induced MHC-I upregulation. Interestingly, although IFNγ increases transcriptional activity of CD274, MAPK signaling also increased stabilization of CD274 mRNA. In conclusion, MAPK pathway activity plays a key role in EGF- and IFNγ-induced PD-L1 expression in lung adenocarcinoma without targetable genetic alterations and may present a target to improve the efficacy of immunotherapy. © 2019 The Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma of Lung/enzymology , B7-H1 Antigen/metabolism , Lung Neoplasms/enzymology , Mitogen-Activated Protein Kinases/metabolism , A549 Cells , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Antineoplastic Agents/pharmacology , B7-H1 Antigen/genetics , Coculture Techniques , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/pharmacology , Humans , Interferon-gamma/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Protein Kinase Inhibitors/pharmacology , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
4.
Pharmacol Ther ; 156: 90-101, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26388292

ABSTRACT

It is increasingly acknowledged that breast cancer can be an immunogenic disease. Immunogenicity appears to differ between subtypes. For instance, in triple negative breast cancer (TNBC) and HER2-positive breast cancer tumor infiltrating lymphocytes (TILs) are prognostic and predictive for response to chemotherapy containing anthracyclines, but in other subtypes they are not. Preclinical evidence suggests important immune based mechanisms of conventional chemotherapeutics, in particular anthracyclines. Early clinical studies with monoclonal antibodies targeting programmed death protein 1, programmed death-ligand 1 and cytotoxic T-lymphocyte-associated antigen 4 have shown anti-tumor efficacy. Tumor vaccines designed to increase the body's own anti-tumor immunity have shown an increased anti-tumor immunity, however clinical efficacy has not yet been demonstrated. Novel strategies will likely follow. In light of the increased interest in immune modulation, this review focuses on predictive immune-based biomarkers, immune-mediated effects from conventional therapies, as well as recent results and ongoing studies concerning immunotherapies in breast cancer.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/therapy , Anthracyclines/therapeutic use , Antibodies, Monoclonal/immunology , Biomarkers , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CTLA-4 Antigen/immunology , Cancer Vaccines/administration & dosage , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Programmed Cell Death 1 Receptor/immunology , Triple Negative Breast Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...