Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Blood ; 137(13): 1777-1791, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33075814

ABSTRACT

Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia characterized by granulomatous lesions containing pathological CD207+ dendritic cells (DCs) with persistent MAPK pathway activation. Standard-of-care chemotherapies are inadequate for most patients with multisystem disease, and optimal strategies for relapsed and refractory disease are not defined. The mechanisms underlying development of inflammation in LCH lesions, the role of inflammation in pathogenesis, and the potential for immunotherapy are unknown. Analysis of the immune infiltrate in LCH lesions identified the most prominent immune cells as T lymphocytes. Both CD8+ and CD4+ T cells exhibited "exhausted" phenotypes with high expression of the immune checkpoint receptors. LCH DCs showed robust expression of ligands to checkpoint receptors. Intralesional CD8+ T cells showed blunted expression of Tc1/Tc2 cytokines and impaired effector function. In contrast, intralesional regulatory T cells demonstrated intact suppressive activity. Treatment of BRAFV600ECD11c LCH mice with anti-PD-1 or MAPK inhibitor reduced lesion size, but with distinct responses. Whereas MAPK inhibitor treatment resulted in reduction of the myeloid compartment, anti-PD-1 treatment was associated with reduction in the lymphoid compartment. Notably, combined treatment with MAPK inhibitor and anti-PD-1 significantly decreased both CD8+ T cells and myeloid LCH cells in a synergistic fashion. These results are consistent with a model that MAPK hyperactivation in myeloid LCH cells drives recruitment of functionally exhausted T cells within the LCH microenvironment, and they highlight combined MAPK and checkpoint inhibition as a potential therapeutic strategy.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Histiocytosis, Langerhans-Cell/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Drug Synergism , Histiocytosis, Langerhans-Cell/pathology , Humans , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/antagonists & inhibitors
2.
Blood Adv ; 4(1): 87-99, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31899802

ABSTRACT

Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder that is characterized by the inflammatory lesions with pathogenic CD1a+CD207+ dendritic cells (DCs). BRAFV600E and other somatic activating MAPK gene mutations have been identified in differentiating bone marrow and blood myeloid cells, but the origin of the LCH lesion CD1a+CD207+ DCs and mechanisms of lesion formation remain incompletely defined. To identify candidate LCH CD1a+CD207+ DC precursor populations, gene-expression profiles of LCH lesion CD1a+CD207+ DCs were first compared with established gene signatures from human myeloid cell subpopulations. Interestingly, the CD1c+ myeloid DC (mDC) gene signature was most enriched in the LCH CD1a+CD207+ DC transcriptome. Additionally, the BRAFV600E allele was not only localized to CD1a+CD207- DCs and CD1a+CD207+ DCs, but it was also identified in CD1c+ mDCs in LCH lesions. Transcriptomes of CD1a+CD207- DCs were nearly indistinguishable from CD1a+CD207+ DCs (both CD1a+CD207low and CD1a+CD207high subpopulations). Transcription profiles of LCH lesion CD1a+CD207+ DCs and peripheral blood CD1c+ mDCs from healthy donors were compared to identify potential LCH DC-specific biomarkers: HLA-DQB2 expression was significantly increased in LCH lesion CD1a+CD207+ DCs compared with circulating CD1c+ mDCs from healthy donors. HLA-DQB2 antigen was identified on LCH lesion CD1a+CD207- DCs and CD1a+CD207+ DCs as well as on CD1c+(CD1a+CD207-) mDCs, but it was not identified in any other lesion myeloid subpopulations. HLA-DQB2 expression was specific to peripheral blood of patients with BRAFV600E+ peripheral blood mononuclear cells, and HLA-DQB2+CD1c+ blood cells were highly enriched for the BRAFV600E in these patients. These data support a model in which blood CD1c+HLA-DQB2+ mDCs with activated ERK migrate to lesion sites where they differentiate into pathogenic CD1a+CD207+ DCs.


Subject(s)
Histiocytosis, Langerhans-Cell , Leukocytes, Mononuclear , Antigens, CD/genetics , Antigens, CD1/genetics , Biomarkers , Dendritic Cells , Glycoproteins , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/genetics , Humans , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Myeloid Cells
3.
Pediatr Hematol Oncol ; 35(5-6): 362-368, 2018.
Article in English | MEDLINE | ID: mdl-30468406

ABSTRACT

Tumor necrosis factor alpha (TNF-α) is produced in Langerhans cell histiocytosis (LCH) lesions and is elevated in plasma of patients with active LCH. It has been postulated that TNF-α may play a role in the pathophysiology of LCH. Etanercept, an anti-TNF-α antibody, has been used in TNF-modulated diseases such as rheumatoid arthritis (RA). We conducted a phase II study to determine the efficacy of etanercept for patients with refractory or relapsed LCH. Five LCH patients who had failed at least 2 prior treatments (range 2-9) received etanercept at a dose of 0.4 mg/kg twice weekly for up to a total of 24 doses. Disease response was assessed at 4 and 8 weeks. None of the five patients had improvement in their disease with etanercept treatment. Three progressed at week 4 and 1 progressed at week 8. One subject died after 3 weeks of treatment from disease progression. During the study, only one drug-related toxicity was noted which spontaneously resolved. The study was concluded early due to lack of response to etanercept and insufficient accrual rate. This data suggests that etanercept as given in this study may not be effective for relapsed or refractory LCH. However, the number of patients treated was not adequate enough to power this study and it is possible that a different dose and regimen of etanercept may be required to successfully treat this disease.


Subject(s)
Etanercept/administration & dosage , Histiocytosis, Langerhans-Cell/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adolescent , Adult , Child, Preschool , Etanercept/adverse effects , Female , Humans , Infant , Male
4.
J Thorac Cardiovasc Surg ; 156(3): 1139-1148.e3, 2018 09.
Article in English | MEDLINE | ID: mdl-30029780

ABSTRACT

OBJECTIVE: We sought to develop a 3-D printing-based simulator for teaching extended septal myectomy to trainees in cardiothoracic surgery (clinical postgraduate year 4-7). This procedure is difficult to teach because of generally unfamiliar and highly variable anatomy, limited visibility for the assistant, and significant potential complications. METHODS: A curriculum using multimedia didactics and 3-D print-based patient-specific surgical simulation was implemented. Six identical 3-D prints were constructed for each of 5 consecutive patients. Preoperative septal myectomy was performed on each printed heart by an attending surgeon and 5 residents. Model myectomy specimen volumes were measured according to liquid displacement. All print resections were videotaped and blindly evaluated by 3 attending surgeons. Pre- and post-test evaluations, and a survey tool were also used to evaluate the curriculum. RESULTS: Baseline myectomy resection volumes differed significantly (attending 15 cm3 vs resident 3.1 cm3; P < .05). Residents resected increasingly larger volumes of tissue over the course of the study. Initial resection volume (compared with faculty) increased by 0.82 cm3 per resection (95% confidence interval, 0.37-1.3 cm3; P < .0001). Total resection volume (compared with faculty) increased by 3.6 cm3 per resection (95% confidence interval, 2.4-4.9 cm3; P < .0001). The residents' survey assessment of the simulator was favorable. CONCLUSIONS: A patient-specific 3-D printing-based simulation module shows promise as a tool to augment and improve cardiothoracic resident training in septal myectomy. The residents were quickly able to perform resections on par with the attending. Residents rated the simulator favorably. Each resident benefited by experiencing the variable anatomy of 5 separate patient-specific models.


Subject(s)
Cardiac Surgical Procedures/education , Curriculum , Heart Septum/surgery , Printing, Three-Dimensional , Simulation Training/methods , Teaching Materials , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/surgery , Heart Septum/anatomy & histology , Humans
5.
Mol Ther ; 26(5): 1354-1365, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29606504

ABSTRACT

Glioblastoma (GBM) is the least treatable type of brain tumor, afflicting over 15,000 people per year in the United States. Patients have a median survival of 16 months, and over 95% die within 5 years. The chemokine receptor ACKR3 is selectively expressed on both GBM cells and tumor-associated blood vessels. High tumor expression of ACKR3 correlates with poor prognosis and potential treatment resistance, making it an attractive therapeutic target. We engineered a single chain FV-human FC-immunoglobulin G1 (IgG1) antibody, X7Ab, to target ACKR3 in human and mouse GBM cells. We used hydrodynamic gene transfer to overexpress the antibody, with efficacy in vivo. X7Ab kills GBM tumor cells and ACKR3-expressing vascular endothelial cells by engaging the cytotoxic activity of natural killer (NK) cells and complement and the phagocytic activity of macrophages. Combining X7Ab with TMZ allows the TMZ dosage to be lowered, without compromising therapeutic efficacy. Mice treated with X7Ab and in combination with TMZ showed significant tumor reduction by MRI and longer survival overall. Brain-tumor-infiltrating leukocyte analysis revealed that X7Ab enhances the activation of M1 macrophages to support anti-tumor immune response in vivo. Targeting ACKR3 with immunotherapeutic monoclonal antibodies (mAbs) in combination with standard of care therapies may prove effective in treating GBM.


Subject(s)
Antibodies, Monoclonal/pharmacology , Glioblastoma/immunology , Glioblastoma/metabolism , Receptors, CXCR/antagonists & inhibitors , Temozolomide/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antibody Affinity/immunology , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Drug Synergism , Glioblastoma/diagnosis , Glioblastoma/mortality , Humans , Magnetic Resonance Imaging , Mice , Mortality , Protein Binding/immunology , Receptors, CXCR/metabolism , Xenograft Model Antitumor Assays
6.
J Thorac Cardiovasc Surg ; 153(1): 132-140, 2017 01.
Article in English | MEDLINE | ID: mdl-27650000

ABSTRACT

OBJECTIVE: Static 3-dimensional printing is used for operative planning in cases that involve difficult anatomy. An interactive 3D print allowing deliberate surgical practice would represent an advance. METHODS: Two patients with hypertrophic cardiomyopathy had 3-dimensional prints constructed preoperatively. Stereolithography files were generated by segmentation of chest computed tomographic scans. Prints were made with hydrogel material, yielding tissue-like models that can be surgically manipulated. Septal myectomy of the print was performed preoperatively in the simulation laboratory. Volumetric measures of print and patient resected specimens were compared. An assessment tool was developed and used to rate the utility of this process. Clinical and echocardiographic data were reviewed. RESULTS: There was congruence between volumes of print and patient resection specimens (patient 1, 3.5 cm3 and 3.0 cm3, respectively; patient 2, 4.0 cm3 and 4.0 cm3, respectively). The prints were rated useful (3.5 and 3.6 on a 5-point Likert scale) for preoperative visualization, planning, and practice. Intraoperative echocardiographic assessment showed adequate relief of left ventricular outflow tract obstruction (patient 1, 80 mm Hg to 18 mm Hg; patient 2, 96 mm Hg to 9 mm Hg). Both patients reported symptomatic improvement (New York Heart Association functional class III to class I). CONCLUSIONS: Three-dimensional printing of interactive hypertrophic cardiomyopathy heart models allows for patient-specific preoperative simulation. Resection volume relationships were congruous on both specimens and suggest evidence of construct validity. This model also holds educational promise for simulation of a low-volume, high-risk operation that is traditionally difficult to teach.


Subject(s)
Cardiomyopathy, Hypertrophic/surgery , Heart Failure/etiology , Models, Anatomic , Models, Cardiovascular , Patient-Specific Modeling , Printing, Three-Dimensional , Uterine Myomectomy/methods , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/diagnostic imaging , Cardiomyopathy, Hypertrophic/physiopathology , Echocardiography, Transesophageal , Female , Heart Failure/diagnosis , Heart Failure/physiopathology , Humans , Middle Aged , Recovery of Function , Tomography, X-Ray Computed , Treatment Outcome
7.
Blood ; 128(21): 2533-2537, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27729324

ABSTRACT

Langerhans cell histiocytosis (LCH) is characterized by inflammatory lesions containing pathologic CD207+ dendritic cells with constitutively activated ERK. Mutually exclusive somatic mutations in MAPK pathway genes have been identified in ∼75% of LCH cases, including recurrent BRAF-V600E and MAP2K1 mutations. To elucidate mechanisms of ERK activation in the remaining 25% of patients, we performed whole-exome sequencing (WES, n = 6), targeted BRAF sequencing (n = 19), and/or whole-transcriptome sequencing (RNA-seq, n = 6) on 24 LCH patient samples lacking BRAF-V600E or MAP2K1 mutations. WES and BRAF sequencing identified in-frame BRAF deletions in the ß3-αC loop in 6 lesions. RNA-seq revealed one case with an in-frame FAM73A-BRAF fusion lacking the BRAF autoinhibitory regulatory domain but retaining an intact kinase domain. High levels of phospho-ERK were detected in vitro in cells overexpressing either BRAF fusion or deletion constructs and ex vivo in CD207+ cells from lesions. ERK activation was resistant to BRAF-V600E inhibition, but responsive to both a second-generation BRAF inhibitor and a MEK inhibitor. These results support an emerging model of universal ERK-activating genetic alterations driving pathogenesis in LCH. A personalized approach in which patient-specific alterations are identified may be necessary to maximize benefit from targeted therapies for patients with LCH.


Subject(s)
Histiocytosis, Langerhans-Cell/genetics , Mutation , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Enzyme Activation/genetics , Female , Histiocytosis, Langerhans-Cell/enzymology , Humans , Infant , Male , Oncogene Proteins, Fusion/metabolism , Protein Domains , Proto-Oncogene Proteins B-raf/metabolism
8.
PLoS One ; 9(12): e112925, 2014.
Article in English | MEDLINE | ID: mdl-25437209

ABSTRACT

Therapies that target leukocyte trafficking pathways can reduce disease activity and improve clinical outcomes in multiple sclerosis (MS). Experimental autoimmune encephalomyelitis (EAE) is a widely studied animal model that shares many clinical and histological features with MS. Chemokine-like receptor-1 (CMKLR1) is a chemoattractant receptor that is expressed by key effector cells in EAE and MS, including macrophages, subsets of dendritic cells, natural killer cells and microglia. We previously showed that CMKLR1-deficient (CMKLR1 KO) mice develop less severe clinical and histological EAE than wild-type mice. In this study, we sought to identify CMKLR1 inhibitors that would pharmaceutically recapitulate the CMKLR1 KO phenotype in EAE. We identified 2-(α-naphthoyl) ethyltrimethylammonium iodide (α-NETA) as a CMKLR1 small molecule antagonist that inhibits chemerin-stimulated ß-arrestin2 association with CMKLR1, as well as chemerin-triggered CMKLR1+ cell migration. α-NETA significantly delayed the onset of EAE induced in C57BL/6 mice by both active immunization with myelin oligodendrocyte glycoprotein peptide 35-55 and by adoptive transfer of encephalitogenic T cells. In addition, α-NETA treatment significantly reduced mononuclear cell infiltrates within the CNS. This study provides additional proof-of-concept data that targeting CMKLR1:chemerin interactions may be beneficial in preventing or treating MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Naphthalenes/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Arrestins/metabolism , Brain/drug effects , Brain/metabolism , Cell Movement/drug effects , Chemokines/metabolism , Drug Evaluation, Preclinical , Drug Stability , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Leukocytes/drug effects , Mice , Mice, Inbred C57BL , Naphthalenes/adverse effects , Naphthalenes/chemistry , Naphthalenes/therapeutic use , Quaternary Ammonium Compounds/adverse effects , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/therapeutic use , Receptors, Chemokine , Safety , Spinal Cord/drug effects , Spinal Cord/metabolism , Structure-Activity Relationship , beta-Arrestins
9.
J Immunol Methods ; 413: 69-73, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25066631

ABSTRACT

Due to low numbers of endogenous dendritic cells (DCs) in vivo, exogenous DC-poietin Fms-like tyrosine kinase 3-ligand (FLT3L) is routinely used to generate DC for subsequent studies. We engineered a novel FLT3L-FC DNA construct that, when combined with hydrodynamic gene transfer (HDT), induced robust DC expansion in mice. DC generated in vivo by FLT3L-FC HDT produced cytokines in response to stimulation by an array of TLR agonists and promoted T cell proliferation. The FLT3L-FC protein produced in vivo spontaneously homodimerized to enable effective FLT signaling and the FC-domain enhanced its plasma half-life, providing an improved reagent and method to boost DC numbers.


Subject(s)
Dendritic Cells/immunology , Gene Transfer Techniques , Membrane Proteins/genetics , Plasmids/metabolism , Recombinant Fusion Proteins/genetics , Animals , Cell Count , Cell Proliferation , Dendritic Cells/cytology , Female , Gene Expression , Half-Life , Hydrodynamics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Protein Multimerization , Recombinant Fusion Proteins/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL