Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Clin Cancer Res ; 30(9): 1859-1877, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38393682

ABSTRACT

PURPOSE: Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN: Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS: We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS: CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.


Subject(s)
CD3 Complex , Endopeptidases , GPI-Linked Proteins , Immunotherapy, Adoptive , Mesothelin , Pancreatic Neoplasms , Receptors, Chimeric Antigen , Tumor Microenvironment , Xenograft Model Antitumor Assays , Humans , Animals , Mice , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Tumor Microenvironment/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , CD3 Complex/immunology , CD3 Complex/metabolism , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Cell Line, Tumor , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism , Adenocarcinoma/immunology , Adenocarcinoma/therapy , Adenocarcinoma/pathology
2.
Immunol Rev ; 320(1): 100-119, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37694970

ABSTRACT

Digestive tract cancers (DTC) belong to the most investigated family of tumors. The incidence, prevalence, and mortality rate of DTC remain high, especially for patients with pancreatic cancer. Even though immunotherapy such as immune checkpoint inhibitors (ICI) have revolutionized the treatment of solid cancer types, ICI are still restricted to a very small group of patients and seem to be more efficacious in combination with chemotherapy. Cellular immunotherapy such as CAR T-cell therapy has entered clinical routine in hematological malignancies with outstanding results. There is growing interest on translating this kind of immunotherapy and success into patients with solid malignancies, such as DTC. This review attempts to describe the major advances in preclinical and clinical research with CAR T cells in DTC, considering the most relevant hurdles in each subtype of DTC.


Subject(s)
Neoplasms , Pancreatic Neoplasms , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell , T-Lymphocytes , Gastrointestinal Tract
3.
Cancer Cell ; 40(5): 494-508.e5, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35452603

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy is effective in lymphoid malignancies, but there has been limited data in myeloid cancers. Here, we start with a CD27-based CAR to target CD70 ("native") in acute myeloid leukemia (AML), and we find modest efficacy in vivo, consistent with prior reports. We then use orthogonal approaches to increase binding on both the tumor and CAR-T cell sides of the immune synapse: a pharmacologic approach (azacitidine) to increase antigen density of CD70 in myeloid tumors, and an engineering approach to stabilize binding of the CAR to CD70. To accomplish the latter, we design a panel of hinge-modified regions to mitigate cleavage of the extracellular portion of CD27. Our CD8 hinge and transmembrane-modified CD70 CAR-T cells are less prone to cleavage, have enhanced binding avidity, and increased expansion, leading to more potent in vivo activity. This enhanced CD70-targeted CAR is a promising candidate for further clinical development.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive , Leukemia, Myeloid, Acute/therapy , T-Lymphocytes
4.
Blood Cancer Discov ; 3(2): 136-153, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35015685

ABSTRACT

Chimeric antigen receptor (CAR) T cells induce impressive responses in patients with hematologic malignancies but can also trigger cytokine release syndrome (CRS), a systemic toxicity caused by activated CAR T cells and innate immune cells. Although IFNγ production serves as a potency assay for CAR T cells, its biologic role in conferring responses in hematologic malignancies is not established. Here we show that pharmacologic blockade or genetic knockout of IFNγ reduced immune checkpoint protein expression with no detrimental effect on antitumor efficacy against hematologic malignancies in vitro or in vivo. Furthermore, IFNγ blockade reduced macrophage activation to a greater extent than currently used cytokine antagonists in immune cells from healthy donors and serum from patients with CAR T-cell-treated lymphoma who developed CRS. Collectively, these data show that IFNγ is not required for CAR T-cell efficacy against hematologic malignancies, and blocking IFNγ could simultaneously mitigate cytokine-related toxicities while preserving persistence and antitumor efficacy. SIGNIFICANCE: Blocking IFNγ in CAR T cells does not impair their cytotoxicity against hematologic tumor cells and paradoxically enhances their proliferation and reduces macrophage-mediated cytokines and chemokines associated with CRS. These findings suggest that IFNγ blockade may improve CAR T-cell function while reducing treatment-related toxicity in hematologic malignancies. See related content by McNerney et al., p. 90 (17). This article is highlighted in the In This Issue feature, p. 85.


Subject(s)
Hematologic Neoplasms , Immunotherapy, Adoptive , Cytokine Release Syndrome , Cytokines/metabolism , Hematologic Neoplasms/therapy , Humans , Immunotherapy, Adoptive/adverse effects , Interferon-gamma/metabolism , Macrophage Activation , T-Lymphocytes/metabolism
5.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-34996813

ABSTRACT

In addition to remarkable antitumor activity, chimeric antigen receptor (CAR) T-cell therapy is associated with acute toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Current treatment guidelines for CRS and ICANS include use of tocilizumab, a monoclonal antibody that blocks the interleukin (IL)-6 receptor, and corticosteroids. In patients with refractory CRS, use of several other agents as third-line therapy (including siltuximab, ruxolitinib, anakinra, dasatinib, and cyclophosphamide) has been reported on an anecdotal basis. At our institution, anakinra has become the standard treatment for the management of steroid-refractory ICANS with or without CRS, based on recent animal data demonstrating the role of IL-1 in the pathogenesis of ICANS/CRS. Here, we retrospectively analyzed clinical and laboratory parameters, including serum cytokines, in 14 patients at our center treated with anakinra for steroid-refractory ICANS with or without CRS after standard treatment with tisagenlecleucel (Kymriah) or axicabtagene ciloleucel (Yescarta) CD19-targeting CAR T. We observed statistically significant and rapid reductions in fever, inflammatory cytokines, and biomarkers associated with ICANS/CRS after anakinra treatment. With three daily subcutaneous doses, anakinra did not have a clear, clinically dramatic effect on neurotoxicity, and its use did not result in rapid tapering of corticosteroids; although neutropenia and thrombocytopenia were common at the time of anakinra dosing, there were no clear delays in hematopoietic recovery or infections that were directly attributable to anakinra. Anakinra may be useful adjunct to steroids and tocilizumab in the management of CRS and/or steroid-refractory ICANs resulting from CAR T-cell therapies, but prospective studies are needed to determine its efficacy in these settings.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/therapeutic use , Neurotoxicity Syndromes/drug therapy , Receptors, Chimeric Antigen/therapeutic use , Adult , Aged , Female , Humans , Interleukin 1 Receptor Antagonist Protein/pharmacology , Male , Middle Aged
6.
Curr Opin Pharmacol ; 59: 116-126, 2021 08.
Article in English | MEDLINE | ID: mdl-34198161

ABSTRACT

Building on the tremendous success of chimeric antigen receptor T-cell therapy in hematological malignancies, there are efforts under way to overcome the challenges associated with this treatment and expand its application to solid tumors. In this fast-evolving field, new therapeutic options are constantly generated, tested in model systems, and further evaluated in clinical trials. In this review, we provide an overview of recent challenges and developments associated with engineered T cells and chimeric antigen receptor T-cell applications. We report on the most recent progress in hematological malignancies and highlight technical advances for applications in solid tumors.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
7.
Br J Clin Pharmacol ; 87(8): 3234-3243, 2021 08.
Article in English | MEDLINE | ID: mdl-33491253

ABSTRACT

AIMS: The aim of this study was to identify risk variants and haplotypes that impair dihydropyrimidine dehydrogenase (DPD) activity and are, therefore, candidate risk variants for severe toxicity to 5-fluorouracil (5-FU) chemotherapy. METHODS: Plasma dihydrouracil/uracil (UH2 /U) ratios were measured as a population marker for DPD activity in a total of 1382 subjects from 4 independent studies. Genotype and haplotype correlations with UH2 /U ratios were assessed. RESULTS: Significantly lower UH2 /U ratios (panova < 2 × 10-16 ) were observed in carriers of the 4 well-studied 5-FU toxicity risk variants with mean differences (MD) of -43.7% for DPYD c.1905 + 1G > A (rs3918290), -46.0% for DPYD c.1679T > G (rs55886062), -37.1%, for DPYD c.2846A > T (rs67376798), and -13.2% for DPYD c.1129-5923C > G (rs75017182). An additional variant, DPYD c.496A > G (rs2297595), was also associated with lower UH2 /U ratios (P < .0001, MD: -12.6%). A haplotype analysis was performed for variants in linkage disequilibrium with c.496A > G, which consisted of the common variant c.85T > C (rs1801265) and the risk variant c.1129-5923C > G. Both haplotypes carrying c.496A > G were associated with decreased UH2 /U ratios (H3, P = .003, MD: -9.6%; H5, P = .002, MD: -16.9%). A haplotype carrying only the variant c.85T > C (H2) was associated with elevated ratios (P = .004, MD: +8.6%). CONCLUSIONS: Based on our data, DPYD-c.496A > G is a strong candidate risk allele for 5-FU toxicity. Our data suggest that DPYD-c.85T > C might be protective; however, the deleterious impacts of the linked alleles c.496A > G and c.1129-5923C > G likely limit this effect in patients. The possible protective effect of c.85T > C and linkage disequilibrium with c.496A > G and c.1129-5923C > G may have hampered prior association studies and should be considered in future clinical studies.


Subject(s)
Dihydrouracil Dehydrogenase (NADP) , Drug-Related Side Effects and Adverse Reactions , Dihydrouracil Dehydrogenase (NADP)/genetics , Fluorouracil/adverse effects , Genotype , Haplotypes , Humans
8.
Annu Rev Med ; 72: 365-382, 2021 01 27.
Article in English | MEDLINE | ID: mdl-32776808

ABSTRACT

Adoptive transfer of T cells modified with chimeric antigen receptors (CAR-T cells) has changed the therapeutic landscape of hematological malignancies, particularly for acute lymphoblastic leukemia and large B cell lymphoma, where two different CAR-T products are now considered standard of care. Furthermore, intense research efforts are under way to expand the clinical application of CAR-T cell therapy for the benefit of patients suffering from other types of cancers. Nevertheless, CAR-T cell treatment is associated with toxicities such as cytokine release syndrome, which can range in severity from mild flu-like symptoms to life-threatening vasodilatory shock, and a neurological syndrome termed ICANS (immune effector cell-associated neurotoxicity syndrome), which can also range in severity from a temporary cognitive deficit lasting only a few hours to lethal cerebral edema. In this review, we provide an in-depth discussion of different types of CAR-T cell-associated toxicities, including an overview of clinical presentation and grading, pathophysiology, and treatment options. We also address future perspectives and opportunities, with a special focus on hematological malignancies.


Subject(s)
Hematologic Neoplasms/therapy , Immunity, Cellular , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Hematologic Neoplasms/immunology , Humans
9.
J Immunol ; 205(10): 2640-2648, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33008951

ABSTRACT

IVIG preparations consisting of pooled IgG are increasingly used for the treatment of autoimmune diseases. IVIG is known to regulate the viability of immune cells, including neutrophils. We report that plasma-derived IgA efficiently triggers death of neutrophils primed by cytokines or TLR agonists. IgA-mediated programmed neutrophil death was PI3K-, p38 MAPK-, and JNK-dependent and evoked anti-inflammatory cytokines in macrophage cocultures. Neutrophils from patients with acute Crohn's disease, rheumatoid arthritis, or sepsis were susceptible to both IgA- and IVIG-mediated death. In contrast to IVIG, IgA did not promote cell death of quiescent neutrophils. Our findings suggest that plasma-derived IgA might provide a therapeutic option for the treatment of neutrophil-associated inflammatory disorders.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Crohn Disease/drug therapy , Immunoglobulin A/pharmacology , Neutrophils/drug effects , Sepsis/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Crohn Disease/blood , Crohn Disease/immunology , Humans , Immunoglobulin A/therapeutic use , Immunoglobulins, Intravenous/pharmacology , Immunoglobulins, Intravenous/therapeutic use , Macrophages , Mice , Neutrophils/immunology , Primary Cell Culture , Sepsis/blood , Sepsis/immunology
11.
Methods Mol Biol ; 1643: 23-31, 2017.
Article in English | MEDLINE | ID: mdl-28667527

ABSTRACT

Different protocols are required for the collection and isolation of antibodies from various body sites. For the sample collection factors to be considered include anatomic or physiological particularities. Secretory fluids such as saliva, gastrointestinal fluid, or breast milk may contain degrading enzymes that potentially affect the integrity of isolated antibodies. While the isolation of IgG from plasma is a common and often-described procedure, here we focus on methodological approaches to isolate antibodies immunoglobulin A (IgA) or IgM from plasma or secretory fluids. These protocols shall facilitate research on natural and induced antibodies.


Subject(s)
Antibodies/immunology , Antibodies/isolation & purification , Gastric Juice , Intestinal Secretions , Milk, Human , Plasma , Saliva , Chromatography, Affinity , Humans , Immunoglobulin A, Secretory/immunology , Immunoglobulin A, Secretory/isolation & purification , Immunoglobulin M/immunology , Immunoglobulin M/isolation & purification
12.
Oncologist ; 22(4): 480-483, 2017 04.
Article in English | MEDLINE | ID: mdl-28242791

ABSTRACT

This brief communication reports on a patient with an exceedingly rare "8p11 (eight-p-eleven) myeloproliferative syndrome" (EMS) with CEP110-FGFR1 rearrangement who responded to treatment with the multi-tyrosine kinase inhibitor (TKI) dasatinib. Dasatinib improved quality of life substantially by increasing blood counts and reducing the need for transfusions. This report demonstrates that the second-generation TKI may provide a therapeutic option for elderly and frail EMS patients who cannot be offered aggressive therapy, including allogeneic hematopoietic cell transplantation. The Oncologist 2017;22:480-483.


Subject(s)
Cell Cycle Proteins/genetics , Down Syndrome/drug therapy , Leukemoid Reaction/drug therapy , Neoplasms/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/genetics , Chromosomes, Human, Pair 8/genetics , Dasatinib/administration & dosage , Down Syndrome/genetics , Down Syndrome/pathology , Female , Humans , Leukemoid Reaction/genetics , Leukemoid Reaction/pathology , Middle Aged , Neoplasms/genetics , Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Protein Kinase Inhibitors/administration & dosage , Quality of Life , Translocation, Genetic/genetics
13.
J Immunol ; 193(11): 5649-59, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25339672

ABSTRACT

FcαRI (CD89), the human Fc receptor for IgA, is highly expressed on neutrophil granulocytes. In this study, we show that FcαRI induces different forms of neutrophil death, depending on the inflammatory microenvironment. The susceptibility of inflammatory neutrophils from sepsis or rheumatoid arthritis toward death induced by specific mAb, or soluble IgA at high concentrations, was enhanced. Although unstimulated cells experienced apoptosis following anti-FcαRI mAb stimulation, preactivation with cytokines or TLR agonists in vitro enhanced FcαRI-mediated death by additional recruitment of caspase-independent pathways, but this required PI3K class IA and MAPK signaling. Transmission electron microscopy of FcαRI-stimulated cells revealed cytoplasmic changes with vacuolization and mitochondrial swelling, nuclear condensation, and sustained plasma membrane. Coculture experiments with macrophages revealed anti-inflammatory effects of the partially caspase-independent death of primed cells following FcαRI engagement. Our data suggest that FcαRI has the ability to regulate neutrophil viability and to induce different forms of neutrophils depending on the inflammatory microenvironment and specific characteristics of the ligand-receptor interactions. Furthermore, these findings have potential implications for FcαRI-targeted strategies to treat neutrophil-associated inflammatory diseases.


Subject(s)
Antigens, CD/metabolism , Macrophages/immunology , Neutrophils/immunology , Receptors, Fc/metabolism , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Cell Death/drug effects , Cells, Cultured , Cellular Microenvironment , Coculture Techniques , Cytokines/immunology , Humans , Immunity, Innate , Inflammation Mediators/metabolism , MAP Kinase Signaling System/drug effects , Neutrophils/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Fc/immunology , Toll-Like Receptors/agonists
15.
J Clin Invest ; 124(4): 1810-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569453

ABSTRACT

Alteration of the surface glycosylation pattern on malignant cells potentially affects tumor immunity by directly influencing interactions with glycan-binding proteins (lectins) on the surface of immunomodulatory cells. The sialic acid-binding Ig-like lectins Siglec-7 and -9 are MHC class I-independent inhibitory receptors on human NK cells that recognize sialic acid-containing carbohydrates. Here, we found that the presence of Siglec-9 defined a subset of cytotoxic NK cells with a mature phenotype and enhanced chemotactic potential. Interestingly, this Siglec-9+ NK cell population was reduced in the peripheral blood of cancer patients. Broad analysis of primary tumor samples revealed that ligands of Siglec-7 and -9 were expressed on human cancer cells of different histological types. Expression of Siglec-7 and -9 ligands was associated with susceptibility of NK cell-sensitive tumor cells and, unexpectedly, of presumably NK cell-resistant tumor cells to NK cell-mediated cytotoxicity. Together, these observations have direct implications for NK cell-based therapies and highlight the requirement to consider both MHC class I haplotype and tumor-specific glycosylation.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Killer Cells, Natural/immunology , Lectins/metabolism , Monitoring, Immunologic , Neoplasms/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Glycosylation , HeLa Cells , Humans , Immunity, Innate , K562 Cells , Killer Cells, Natural/classification , Ligands , Male , Mice , Mice, Inbred NOD , Mice, SCID
16.
Semin Hematol ; 50 Suppl 1: S109-15, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23664507

ABSTRACT

Immune thrombocytopenia (ITP) is a complex disease. The pathogenic and clinical heterogeneity of ITP is reflected by reports on variability in patient history and treatment response, in concert with recent evidence from mechanistic studies. Programmed cell death (PCD) pathways are thought to play a peculiar role in the megakaryocyte lineage in terms of hemostasis and the generation and function of megakaryocytes and platelets; unbalanced genetic or environmental disturbances of these tightly regulated pathways may cause thrombocytopenia. Dysregulated PCD has also been linked to peripheral platelet destruction, intramedullary apoptosis, and inefficient thrombopoiesis in ITP. In this article, we discuss novel and controversial findings on the role of PCD in the megakaryocyte lineage and their potential implications in terms of pathogenesis, diagnosis, and treatment of ITP.


Subject(s)
Thrombocytopenia/immunology , Thrombocytopenia/pathology , Animals , Apoptosis/immunology , Autoantibodies/immunology , Blood Platelets/immunology , Cell Death/immunology , Humans , Megakaryocytes/immunology , Thrombocytopenia/diagnosis , Thrombocytopenia/therapy
17.
Immunobiology ; 216(1-2): 86-95, 2011.
Article in English | MEDLINE | ID: mdl-20362352

ABSTRACT

The human airway epithelium serves as structural and functional barrier against inhaled particulate antigen. Previously, we demonstrated in an in vitro epithelial barrier model that monocyte derived dendritic cells (MDDC) and monocyte derived macrophages (MDM) take up particulate antigen by building a trans-epithelial interacting network. Although the epithelial tight junction (TJ) belt was penetrated by processes of MDDC and MDM, the integrity of the epithelium was not affected. These results brought up two main questions: (1) Do MDM and MDDC exchange particles? (2) Are those cells expressing TJ proteins, which are believed to interact with the TJ belt of the epithelium to preserve the epithelial integrity? The expression of TJ and adherens junction (AJ) mRNA and proteins in MDM and MDDC monocultures was determined by RT-PCR, and immunofluorescence, respectively. Particle uptake and exchange was quantified by flow cytometry and laser scanning microscopy in co-cultures of MDM and MDDC exposed to polystyrene particles (1 µm in diameter). MDM and MDDC constantly expressed TJ and AJ mRNA and proteins. Flow cytometry analysis of MDM and MDDC co-cultures showed increased particle uptake in MDDC while MDM lost particles over time. Quantitative analysis revealed significantly higher particle uptake by MDDC in co-cultures of epithelial cells with MDM and MDDC present, compared to co-cultures containing only epithelial cells and MDDC. We conclude from these findings that MDM and MDDC express TJ and AJ proteins which could help to preserve the epithelial integrity during particle uptake and exchange across the lung epithelium.


Subject(s)
Cell-Derived Microparticles/metabolism , Dendritic Cells/metabolism , Macrophages/metabolism , Respiratory Mucosa/pathology , Tight Junctions/metabolism , Adherens Junctions/genetics , Adherens Junctions/immunology , Adherens Junctions/metabolism , Airway Remodeling/immunology , Antigens/immunology , Cell Communication/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Cell-Derived Microparticles/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Endocytosis/immunology , Environmental Exposure/adverse effects , Humans , Macrophages/immunology , Macrophages/pathology , Monocytes/pathology , Particulate Matter/adverse effects , Particulate Matter/immunology , Tight Junctions/genetics , Tight Junctions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...