Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Commun Biol ; 4(1): 497, 2021 04 22.
Article En | MEDLINE | ID: mdl-33888863

Cyclic dinucleotide (CDN) agonists of the STimulator of InterferoN Genes (STING) pathway have shown immune activation and tumor clearance in pre-clinical models. However, CDNs administered intratumorally also promote STING activation leading to direct cytotoxicity of many cell types in the tumor microenvironment (TME), systemic inflammation due to rapid tumor extravasation of the CDN, and immune ablation in the TME. These result in a failure to establish immunological memory. ExoSTING, an engineered extracellular vesicle (EV) exogenously loaded with CDN, enhances the potency of CDN and preferentially activates antigen presenting cells in the TME. Following intratumoral injection, exoSTING was retained within the tumor, enhanced local Th1 responses and recruitment of CD8+ T cells, and generated systemic anti-tumor immunity to the tumor. ExoSTING at therapeutically active doses did not induce systemic inflammatory cytokines, resulting in an enhanced therapeutic window. ExoSTING is a novel, differentiated therapeutic candidate that leverages the natural biology of EVs to enhance the activity of CDNs.


Extracellular Vesicles/physiology , Immunologic Surveillance , Tumor Microenvironment/physiology , Animals , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
Mol Ther ; 29(5): 1729-1743, 2021 05 05.
Article En | MEDLINE | ID: mdl-33484965

Extracellular vesicles (EVs) are an important intercellular communication system facilitating the transfer of macromolecules between cells. Delivery of exogenous cargo tethered to the EV surface or packaged inside the lumen are key strategies for generating therapeutic EVs. We identified two "scaffold" proteins, PTGFRN and BASP1, that are preferentially sorted into EVs and enable high-density surface display and luminal loading of a wide range of molecules, including cytokines, antibody fragments, RNA binding proteins, vaccine antigens, Cas9, and members of the TNF superfamily. Molecules were loaded into EVs at high density and exhibited potent in vitro activity when fused to full-length or truncated forms of PTGFRN or BASP1. Furthermore, these engineered EVs retained pharmacodynamic activity in a variety of animal models. This engineering platform provides a simple approach to functionalize EVs with topologically diverse macromolecules and represents a significant advance toward unlocking the therapeutic potential of EVs.


Extracellular Vesicles/transplantation , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proteins/administration & dosage , Repressor Proteins/metabolism , Animals , Cell Communication , Drug Delivery Systems , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Female , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Repressor Proteins/genetics
3.
BMC Immunol ; 18(1): 6, 2017 02 02.
Article En | MEDLINE | ID: mdl-28148223

BACKGROUND: Comprehensive understanding of cellular immune subsets involved in regulation of tumor progression is central to the development of cancer immunotherapies. Single cell immunophenotyping has historically been accomplished by flow cytometry (FC) analysis, enabling the analysis of up to 18 markers. Recent advancements in mass cytometry (MC) have facilitated detection of over 50 markers, utilizing high resolving power of mass spectrometry (MS). This study examined an analytical and operational feasibility of MC for an in-depth immunophenotyping analysis of the tumor microenvironment, using the commercial CyTOF™ instrument, and further interrogated challenges in managing the integrity of tumor specimens. RESULTS: Initial longitudinal studies with frozen peripheral blood mononuclear cells (PBMCs) showed minimal MC inter-assay variability over nine independent runs. In addition, detection of common leukocyte lineage markers using MC and FC detection confirmed that these methodologies are comparable in cell subset identification. An advanced multiparametric MC analysis of 39 total markers enabled a comprehensive evaluation of cell surface marker expression in fresh and cryopreserved tumor samples. This comparative analysis revealed significant reduction of expression levels of multiple markers upon cryopreservation. Most notably myeloid derived suppressor cells (MDSC), defined by co-expression of CD66b+ and CD15+, HLA-DRdim and CD14- phenotype, were undetectable in frozen samples. CONCLUSION: These results suggest that optimization and evaluation of cryopreservation protocols is necessary for accurate biomarker discovery in frozen tumor specimens.


Carcinogenesis/immunology , Carcinoma, Renal Cell/pathology , Colorectal Neoplasms/pathology , Cryopreservation , Immunophenotyping/methods , Mass Spectrometry/methods , Myeloid-Derived Suppressor Cells/immunology , Cell Differentiation , Cell Lineage , Cell Separation , Feasibility Studies , Humans , Phenotype , Single-Cell Analysis , Tumor Microenvironment
4.
Mol Cancer ; 14: 119, 2015 Jun 11.
Article En | MEDLINE | ID: mdl-26062742

BACKGROUND: Carbonic anhydrase (CA) IX is a surface-expressed protein that is upregulated by the hypoxia inducible factor (HIF) and represents a prototypic tumor-associated antigen that is overexpressed on renal cell carcinoma (RCC). Therapeutic approaches targeting CAIX have focused on the development of CAIX inhibitors and specific immunotherapies including monoclonal antibodies (mAbs). However, current in vivo mouse models used to characterize the anti-tumor properties of fully human anti-CAIX mAbs have significant limitations since the role of human effector cells in tumor cell killing in vivo is not directly evaluated. METHODS: The role of human anti-CAIX mAbs on CAIX(+) RCC tumor cell killing by immunocytes or complement was tested in vitro by antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) as well as on CAIX(+) RCC cellular motility, wound healing, migration and proliferation. The in vivo therapeutic activity mediated by anti-CAIX mAbs was determined by using a novel orthotopic RCC xenograft humanized animal model and analyzed by histology and FACS staining. RESULTS: Our studies demonstrate the capacity of human anti-CAIX mAbs that inhibit CA enzymatic activity to result in immune-mediated killing of RCC, including nature killer (NK) cell-mediated ADCC, CDC, and macrophage-mediated ADCP. The killing activity correlated positively with the level of CAIX expression on RCC tumor cell lines. In addition, Fc engineering of anti-CAIX mAbs was shown to enhance the ADCC activity against RCC. We also demonstrate that these anti-CAIX mAbs inhibit migration of RCC cells in vitro. Finally, through the implementation of a novel orthotopic RCC model utilizing allogeneic human peripheral blood mononuclear cells in NOD/SCID/IL2Rγ(-/-) mice, we show that anti-CAIX mAbs are capable of mediating human immune response in vivo including tumor infiltration of NK cells and activation of T cells, resulting in inhibition of CAIX(+) tumor growth. CONCLUSIONS: Our findings demonstrate that these novel human anti-CAIX mAbs have therapeutic potential in the unmet medical need of targeted killing of HIF-driven CAIX(+)RCC. The orthotopic tumor xenografted humanized mouse provides an improved model to evaluate the in vivo anti-tumor capabilities of fully human mAbs for RCC therapy.


Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Carbonic Anhydrases/immunology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lymphocytes/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Carbonic Anhydrase IX , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endocytosis/drug effects , Humans , Kidney Neoplasms/pathology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes/drug effects , Mice , Protein Engineering , Single-Chain Antibodies/immunology
5.
FEMS Immunol Med Microbiol ; 58(3): 397-404, 2010 Apr.
Article En | MEDLINE | ID: mdl-20180848

Given their immune-modulating capacity, regulatory T cells (Treg) cells may be important players in the induction of the protective T-cell response (Th1) to genital chlamydial infection. Recent work has demonstrated that plasmacytoid dendritic cells (pDC) respond to genital chlamydial infection, and that pDC may be uniquely positioned for the induction of Treg cells during this infection. Here, we present the first data demonstrating that Treg influx into the draining lymph node and the site of infection during genital chlamydial infection. We found that pDC depletion altered the numbers of Treg and nonprotective inflammatory cells [interferongamma-(IFNgamma)-producing CD8+ T and IFNgamma-producing natural killer T cells] in the spleens of mice genitally infected with Chlamydia muridarum. Furthermore, pDC depletion did not alter Th1 cell numbers, indicating that pDC modulate cells that could inhibit and promote nonprotective inflammation during genital chlamydial infection. Finally, we demonstrate that depletion of pDC results in less severe dilation and collagen deposition in the oviduct following resolution of infection, implicating pDC activity in the formation of sequelae following genital C. muridarum infection.


Chlamydia Infections/immunology , Chlamydia muridarum/pathogenicity , Dendritic Cells/immunology , Genital Diseases, Female/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Chlamydia Infections/microbiology , Chlamydia muridarum/immunology , Female , Forkhead Transcription Factors/metabolism , Genital Diseases, Female/microbiology , Humans , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology
6.
PLoS One ; 4(4): e5409, 2009.
Article En | MEDLINE | ID: mdl-19404403

BACKGROUND: Generation of robust cell-mediated immune responses at mucosal surfaces while reducing overall inflammation is a primary goal for vaccination. Here we report the use of a recombinant nanoparticle as a vaccine delivery platform against mucosal infections requiring T cell-mediated immunity for eradication. METHODOLOGY/PRINCIPAL FINDINGS: We encapsulated an immunogenic protein, the major outer membrane protein (MOMP) of Chlamydia muridarum, within hollow, vault nanocapsules (MOMP-vaults) that were engineered to bind IgG for enhanced immunity. Intranasal immunization (i.n) with MOMP-vaults induced anti-chlamydial immunity plus significantly attenuated bacterial burden following challenge infection. Vault immunization induced anti-chlamydial immune responses and inflammasome formation but did not activate toll-like receptors. Moreover, MOMP-vault immunization enhanced microbial eradication without the inflammation usually associated with adjuvants. CONCLUSIONS/SIGNIFICANCE: Vault nanoparticles containing immunogenic proteins delivered to the respiratory tract by the i.n. route can act as "smart adjuvants" for inducing protective immunity at distant mucosal surfaces while avoiding destructive inflammation.


Bacterial Vaccines/administration & dosage , Immunity, Mucosal/drug effects , Nanoparticles/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/therapeutic use , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/therapeutic use , Chlamydia muridarum/immunology , Drug Compounding/methods , Immunity, Mucosal/immunology , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Treatment Outcome
7.
ACS Nano ; 3(1): 27-36, 2009 Jan 27.
Article En | MEDLINE | ID: mdl-19206245

As a naturally occurring nanocapsule abundantly expressed in nearly all-eukaryotic cells, the barrel-shaped vault particle is perhaps an ideal structure to engineer for targeting to specific cell types. Recombinant vault particles self-assemble from 96 copies of the major vault protein (MVP), have dimensions of 72.5 x 41 nm, and have a hollow interior large enough to encapsulate hundreds of proteins. In this study, three different tags were engineered onto the C-terminus of MVP: an 11 amino acid epitope tag, a 33 amino acid IgG-binding peptide, and the 55 amino acid epidermal growth factor (EGF). These modified vaults were produced using a baculovirus expression system. Our studies demonstrate that recombinant vaults assembled from MVPs containing C-terminal peptide extensions display these tags at the top and bottom of the vault on the outside of the particle and can be used to specifically bind the modified vaults to epithelial cancer cells (A431) via the epidermal growth factor receptor (EGFR), either directly (EGF modified vaults) or as mediated by a monoclonal antibody (anti-EGFR) bound to recombinant vaults containing the IgG-binding peptide. The ability to target vaults to specific cells represents an essential advance toward using recombinant vaults as delivery vehicles.


Cell Membrane/metabolism , Nanoparticles/chemistry , Receptors, Cell Surface/chemistry , Animals , Binding Sites , Cell Line, Tumor , Dendritic Cells/metabolism , Epitopes/chemistry , ErbB Receptors/chemistry , HeLa Cells , Humans , Immunoglobulin G/chemistry , Mice , Protein Binding , Protein Structure, Tertiary , Receptors, Cell Surface/metabolism
8.
FEMS Immunol Med Microbiol ; 55(2): 240-9, 2009 Mar.
Article En | MEDLINE | ID: mdl-19159428

New methods are needed to eradicate or prevent Chlamydia trachomatis infections. Blockade of epithelial membrane protein 2 (EMP2) by genetic silencing or neutralizing polyclonal antibody reduced chlamydial infectivity in vitro. This study tests the prediction that recombinant anti-EMP2 diabody could reduce early chlamydial infection of the genital tract in vivo. In a murine infection model, pretreatment with anti-EMP2 diabody, as compared with control diabody, significantly reduced bacterial load, tissue production of inflammatory cytokines, recruitment of polymorphonuclear leukocytes, and local tissue inflammation. These findings support EMP2 as a potential preventative and therapeutic target for genital chlamydial infection.


Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Genital Diseases, Female/prevention & control , Immunoglobulins/therapeutic use , Membrane Glycoproteins/antagonists & inhibitors , Animals , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Colony Count, Microbial , Cytokines/immunology , Cytokines/metabolism , Female , Genital Diseases, Female/immunology , Genital Diseases, Female/microbiology , Genital Diseases, Female/pathology , Immunoglobulins/genetics , Inflammation/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C
9.
FEMS Immunol Med Microbiol ; 55(2): 226-36, 2009 Mar.
Article En | MEDLINE | ID: mdl-19159430

Dendritic cells (DCs) are central for the induction of T-cell responses needed for chlamydial eradication. Here, we report the activation of two DC subsets: a classical CD11b+ (cDC) and plasmacytoid (pDC) during genital infection with Chlamydia muridarum. Genital infection induced an influx of cDC and pDC into the genital tract and its draining lymph node (iliac lymph nodes, ILN) as well as colocalization with T cells in the ILN. Genital infection with C. muridarum also stimulated high levels of costimulatory molecules on cDC central for the activation of naïve T cells in vivo. In contrast, pDC expressed low levels of most costimulatory molecules in vivo and did not secrete cytokines associated with the production of T helper (Th)1 cells in vitro. However, pDC upregulated inducible costimulatory ligand expression and produced IL-6 and IL-10 in response to chlamydial exposure in vitro. Our findings show that these two DC subsets likely have different functions in vivo. cDCs are prepared for induction of antichlamydial T-cell responses, whereas pDCs have characteristics associated with the differentiation of non-Th1 cell subsets.


Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia muridarum/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Genital Diseases, Female/immunology , Genital Diseases, Female/microbiology , Animals , Cytokines/biosynthesis , Dendritic Cells/classification , Female , Mice , Mice, Inbred BALB C
...