Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Cells ; 10(11)2021 11 13.
Article in English | MEDLINE | ID: mdl-34831378

ABSTRACT

Exosomes are a subset of extracellular vesicles (EVs) that are released by cells and play a variety of physiological roles including regulation of the immune system. Exosomes are heterogeneous and present in vast numbers in tumor microenvironments. A large subset of these vesicles has been demonstrated to be immunosuppressive. In this review, we focus on the suppression of T cell function by exosomes in human tumor microenvironments. We start with a brief introduction to exosomes, with emphasis on their biogenesis, isolation and characterization. Next, we discuss the immunosuppressive effect of exosomes on T cells, reviewing in vitro studies demonstrating the role of different proteins, nucleic acids and lipids known to be associated with exosome-mediated suppression of T cell function. Here, we also discuss initial proof-of-principle studies that established the potential for rescuing T cell function by blocking or targeting exosomes. In the final section, we review different in vivo models that were utilized to study as well as target exosome-mediated immunosuppression, highlighting the Xenomimetic mouse (X-mouse) model and the Omental Tumor Xenograft (OTX) model that were featured in a recent study to evaluate the efficacy of a novel phosphatidylserine-binding molecule for targeting immunosuppressive tumor-associated exosomes.


Subject(s)
Exosomes/metabolism , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Animals , Humans , Immunosuppression Therapy
2.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34599030

ABSTRACT

BACKGROUND: The human tumor microenvironment (TME) is a complex and dynamic milieu of diverse acellular and cellular components, creating an immunosuppressive environment, which contributes to tumor progression. We have previously shown that phosphatidylserine (PS) expressed on the surface of exosomes isolated from human TMEs is causally linked to T-cell immunosuppression, representing a potential immunotherapeutic target. In this study, we investigated the effect of ExoBlock, a novel PS-binding molecule, on T-cell responses in the TME. METHODS: We designed and synthesized a new compound, (ZnDPA)6-DP-15K, a multivalent PS binder named ExoBlock. The PS-binding avidity of ExoBlock was tested using an in vitro competition assay. The ability of this molecule to reverse exosome-mediated immunosuppression in vitro was tested using human T-cell activation assays. The in vivo therapeutic efficacy of ExoBlock was then tested in two different human tumor xenograft models, the melanoma-based xenomimetic (X-)mouse model, and the ovarian tumor-based omental tumor xenograft (OTX) model. RESULTS: ExoBlock was able to bind PS with high avidity and was found to consistently and significantly block the immunosuppressive activity of human ovarian tumor and melanoma-associated exosomes in vitro. ExoBlock was also able to significantly enhance T cell-mediated tumor suppression in vivo in both the X-mouse and the OTX model. In the X-mouse model, ExoBlock suppressed tumor recurrence in a T cell-dependent manner. In the OTX model, ExoBlock treatment resulted in an increase in the number as well as function of CD4 and CD8 T cells in the TME, which was associated with a reduction in tumor burden and metastasis, as well as in the number of circulating PS+ exosomes in tumor-bearing mice. CONCLUSION: Our results establish that targeting exosomal PS in TMEs with ExoBlock represents a promising strategy to enhance antitumor T-cell responses.


Subject(s)
Exosomes/metabolism , Neoplasms/immunology , Ovarian Neoplasms/genetics , Phosphatidylserines/metabolism , T-Lymphocytes/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred NOD , Ovarian Neoplasms/pathology , Tumor Microenvironment
3.
Sci Rep ; 11(1): 17853, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34497305

ABSTRACT

The safety and efficacy of several life-saving therapeutic proteins are compromised due to their immunogenicity. Once a sustained immune response against a protein-based therapy is established, clinical options that are safe and cost-effective become limited. Prevention of immunogenicity of therapeutic proteins prior to their initial use is critical as it is often difficult to reverse an established immune response. Here, we discuss a rational design and testing of a phosphatidylserine-containing nanoparticle platform for novel oral prophylactic reverse vaccination approach, i.e., pre-treatment of a therapeutic protein in the presence of nanoparticles to prevent immunogenicity of protein therapies.


Subject(s)
Immunotherapy , Nanoparticles , Animals , Mice
4.
Clin Transl Immunology ; 10(2): e1246, 2021.
Article in English | MEDLINE | ID: mdl-33552509

ABSTRACT

OBJECTIVES: With a rapidly growing list of candidate immune-based cancer therapeutics, there is a critical need to generate highly reliable animal models to preclinically evaluate the efficacy of emerging immune-based therapies, facilitating successful clinical translation. Our aim was to design and validate a novel in vivo model (called Xenomimetic or 'X' mouse) that allows monitoring of the ability of human tumor-specific T cells to suppress tumor growth following their entry into the tumor. METHODS: Tumor xenografts are established rapidly in the greater omentum of globally immunodeficient NOD-scid IL2Rγnull (NSG) mice following an intraperitoneal injection of melanoma target cells expressing tumor neoantigen peptides, as well as green fluorescent protein and/or luciferase. Changes in tumor burden, as well as in the number and phenotype of adoptively transferred patient-derived tumor neoantigen-specific T cells in response to immunotherapy, are measured by imaging to detect fluorescence/luminescence and flow cytometry, respectively. RESULTS: The tumors progress rapidly and disseminate in the mice unless patient-derived tumor-specific T cells are introduced. An initial T cell-mediated tumor arrest is later followed by a tumor escape, which correlates with the upregulation of the checkpoint molecules programmed cell death-1 (PD-1) and lymphocyte-activation gene 3 (LAG3) on T cells. Treatment with immune-based therapies that target these checkpoints, such as anti-PD-1 antibody (nivolumab) or interleukin-12 (IL-12), prevented or delayed the tumor escape. Furthermore, IL-12 treatment suppressed PD-1 and LAG3 upregulation on T cells. CONCLUSION: Together, these results validate the X-mouse model and establish its potential to preclinically evaluate the therapeutic efficacy of immune-based therapies.

5.
Immunol Invest ; 49(7): 726-743, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32299258

ABSTRACT

Background: T cells present in chronic inflammatory tissues such as nasal polyps (from chronic rhinosinusitis patients) have been demonstrated to be hypo-responsive to activation via the TCR, similar to tumor-specific T cells in multiple different human tumor microenvironments. While immunosuppressive exosomes have been known to contribute to the failure of the tumor-associated T cells to respond optimally to activation stimuli, it is not known whether they play a similar role in chronic inflammatory microenvironments. In the current study, we investigate whether exosomes derived from chronic inflammatory microenvironments contribute to the immune suppression of T cells. Methods: Exosomes were isolated by ultracentrifugation and characterized by size and composition using nanoparticle tracking analysis, scanning electron microscopy, antibody arrays and flow exometry. Immunosuppressive ability of the exosomes was measured by quantifying its effect on activation of T cells, using nuclear translocation of NFκB as an activation endpoint. Results: Exosomes were isolated and characterized from two different types of chronic inflammatory tissues - nasal polyps from chronic rhinosinusitis patients and synovial fluid from rheumatoid arthritis patients. These exosomes arrest the activation of T cells stimulated via the TCR. This immune suppression, like that which is seen in tumor microenvironments, is dependent in part upon a lipid, ganglioside GD3, which is expressed on the exosomal surface. Conclusion: Immunosuppressive exosomes present in non-malignant chronic inflammatory tissues represent a new T cell checkpoint, and potentially represent a novel therapeutic target to enhance the response to current therapies and prevent disease recurrences.


Subject(s)
Cellular Microenvironment/immunology , Exosomes/metabolism , Immunomodulation , Inflammation/etiology , Inflammation/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Arthritis/etiology , Arthritis/metabolism , Biomarkers , Chronic Disease , Disease Susceptibility , Exosomes/ultrastructure , Extracellular Vesicles/metabolism , Extracellular Vesicles/ultrastructure , Humans , Immunohistochemistry , Immunophenotyping , Inflammation/pathology , Lipid Metabolism , Lymphocyte Activation/immunology , NF-kappa B/metabolism , Nasal Polyps/etiology , Nasal Polyps/metabolism , Nasal Polyps/pathology , Protein Transport , Signal Transduction , Synovial Fluid/metabolism
6.
Sci Rep ; 9(1): 4959, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30874569

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

7.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30730851

ABSTRACT

Epithelial ovarian cancer (EOC) often presents with metastases and ascites. Granulocytic myeloid-derived suppressor cells are an immature population that impairs antitumor immunity. Since suppressive granulocytes in the ascites of patients with newly diagnosed EOC were morphologically mature, we hypothesized that PMN were rendered suppressive in the tumor microenvironment (TME). Circulating PMN from patients were not suppressive but acquired a suppressor phenotype (defined as ≥1 log10 reduction of anti-CD3/CD28-stimulated T cell proliferation) after ascites supernatant exposure. Ascites supernatants (20 of 31 supernatants) recapitulated the suppressor phenotype in PMN from healthy donors. T cell proliferation was restored with ascites removal and restimulation. PMN suppressors also inhibited T cell activation and cytokine production. PMN suppressors completely suppressed proliferation in naive, central memory, and effector memory T cells and in engineered tumor antigen-specific cytotoxic T lymphocytes, while antigen-specific cell lysis was unaffected. Inhibition of complement C3 activation and PMN effector functions, including CR3 signaling, protein synthesis, and vesicular trafficking, abrogated the PMN suppressor phenotype. Moreover, malignant effusions from patients with various metastatic cancers also induced the C3-dependent PMN suppressor phenotype. These results point to PMN impairing T cell expansion and activation in the TME and the potential for complement inhibition to abrogate this barrier to antitumor immunity.


Subject(s)
Immunity , Neutrophils/immunology , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Aged , CD28 Antigens , Cell Proliferation , Complement C3 , Cytokines , Female , Granulocytes , Humans , Lymphocyte Activation/immunology , Muromonab-CD3 , Myeloid-Derived Suppressor Cells/immunology , Ovarian Neoplasms/pathology
8.
J Immunol ; 201(12): 3750-3758, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30446565

ABSTRACT

The tumor microenvironment is rendered immunosuppressive by a variety of cellular and acellular factors that represent potential cancer therapeutic targets. Although exosomes isolated from ovarian tumor ascites fluids have been previously reported to induce a rapid and reversible T cell arrest, the factors present on or within exosomes that contribute to immunosuppression have not been fully defined. In this study, we establish that GD3, a ganglioside expressed on the surface of exosomes isolated from human ovarian tumor ascites fluids, is causally linked to the functional arrest of T cells activated through their TCR. This arrest is inhibited by Ab blockade of exosomal GD3 or by the removal of GD3+ exosomes. Empty liposomes expressing GD3 on the surface also inhibit the activation of T cells, establishing that GD3 contributes to the functional arrest of T cells independent of factors present in exosomes. Finally, we demonstrate that the GD3-mediated arrest of the TCR activation is dependent upon sialic acid groups, because their enzymatic removal from exosomes or liposomes results in a loss of inhibitory capacity. Collectively, these data define GD3 as a potential immunotherapeutic target.


Subject(s)
Ascitic Fluid/metabolism , Exosomes/metabolism , Gangliosides/metabolism , Immunotherapy/methods , N-Acetylneuraminic Acid/metabolism , Ovarian Neoplasms/metabolism , T-Lymphocytes/immunology , Ascites , Cells, Cultured , Female , Humans , Immune Tolerance , NF-kappa B/metabolism , Neoplasm Staging , Ovarian Neoplasms/immunology , Tumor Microenvironment
9.
Sci Rep ; 8(1): 12905, 2018 08 27.
Article in English | MEDLINE | ID: mdl-30150674

ABSTRACT

Local acidification of stroma is proposed to favour pre-metastatic niche formation but the mechanism of initiation is unclear. We investigated whether Human Melanoma-derived exosomes (HMEX) could reprogram human adult dermal fibroblasts (HADF) and cause extracellular acidification. HMEX were isolated from supernatants of six melanoma cell lines (3 BRAF V600E mutant cell lines and 3 BRAF wild-type cell lines) using ultracentrifugation or Size Exclusion Chromatography (SEC). Rapid uptake of exosomes by HADF was demonstrated following 18 hours co-incubation. Exposure of HDAF to HMEX leads to an increase in aerobic glycolysis and decrease in oxidative phosphorylation (OXPHOS) in HADF, consequently increasing extracellular acidification. Using a novel immuno-biochip, exosomal miR-155 and miR-210 were detected in HMEX. These miRNAs were present in HMEX from all six melanoma cell lines and were instrumental in promoting glycolysis and inhibiting OXPHOS in tumour cells. Inhibition of miR-155 and miR-210 activity by transfection of miRNA inhibitors into HMEX reversed the exosome-induced metabolic reprogramming of HADF. The data indicate that melanoma-derived exosomes modulate stromal cell metabolism and may contribute to the creation of a pre-metastatic niche that promotes the development of metastasis.


Subject(s)
Cellular Reprogramming/physiology , Exosomes/metabolism , Melanoma/metabolism , MicroRNAs/metabolism , Aerobiosis/genetics , Aerobiosis/physiology , Cell Line, Tumor , Cellular Reprogramming/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Glycolysis/genetics , Glycolysis/physiology , Humans , Melanoma/genetics , MicroRNAs/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
10.
J Pharm Sci ; 107(8): 2048-2054, 2018 08.
Article in English | MEDLINE | ID: mdl-29649469

ABSTRACT

Phosphatidylserine (PS) exposure during apoptosis leads to silent clearance of cells without adverse immune reactions to self-proteins. Given the biological functions of PS in cellular cleanup and global immunosuppression, we hypothesized that administration of PS-protein complexes would reduce immunogenicity. Here, we report that exposing Pompe disease mice to acid alpha glucosidase (rhGAA) with PS or immunosuppressant dexamethasone resulted in lower anti-rhGAA antibodies than in animals receiving rhGAA alone. However, upon rechallenge with rhGAA, only PS-rhGAA pre-exposed mice displayed a durable hyporesponsiveness even after PS administration was ceased. Thus, pre-exposure of antigens administered together with PS were not silently cleared, but the immune system acquired memory about the antigen that averted mounting of a response during rechallenge. In hemophilia A mice, PS hyporesponsiveness toward Factor VIII was reversed by administration of function-blocking antibody against the PS receptor T-cell immunoglobulin and mucin 4, implicating this receptor in PS's effect. Moreover, pre-exposure of myelin oligodendrocyte glycoprotein peptide with PS delayed the onset and reduced the severity of experimental autoimmune encephalomyelitis. These observations suggest that PS's function in apoptosis is not limited to silent antigen clearance without immune responses toward self-proteins but shows that PS reduces immune response during rechallenge to several antigens that also involves initiation of antigen tolerance.


Subject(s)
Dexamethasone/immunology , Glycogen Storage Disease Type II/immunology , Immunosuppressive Agents/immunology , Phosphatidylserines/immunology , alpha-Glucosidases/immunology , Animals , Antibody Formation , Apoptosis , Dexamethasone/administration & dosage , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Factor VIII/administration & dosage , Factor VIII/immunology , Female , Hemophilia A/immunology , Humans , Immune Tolerance , Immunosuppressive Agents/administration & dosage , Liposomes/administration & dosage , Liposomes/immunology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Phosphatidylserines/administration & dosage , alpha-Glucosidases/administration & dosage
11.
Cancer Immunol Res ; 6(2): 236-247, 2018 02.
Article in English | MEDLINE | ID: mdl-29301753

ABSTRACT

Nano-sized membrane-encapsulated extracellular vesicles isolated from the ascites fluids of ovarian cancer patients are identified as exosomes based on their biophysical and compositional characteristics. We report here that T cells pulsed with these tumor-associated exosomes during TCR-dependent activation inhibit various activation endpoints including translocation of NFκB and NFAT into the nucleus, upregulation of CD69 and CD107a, production of cytokines, and cell proliferation. In addition, the activation of virus-specific CD8+ T cells that are stimulated with the cognate viral peptides presented in the context of class I MHC is also suppressed by the exosomes. The inhibition occurs without loss of cell viability and coincidentally with the binding and internalization of these exosomes. This exosome-mediated inhibition of T cells was transient and reversible: T cells exposed to exosomes can be reactivated once exosomes are removed. We conclude that tumor-associated exosomes are immunosuppressive and represent a therapeutic target, blockade of which would enhance the antitumor response of quiescent tumor-associated T cells and prevent the functional arrest of adoptively transferred tumor-specific T cells or chimeric antigen receptor T cells. Cancer Immunol Res; 6(2); 236-47. ©2018 AACR.


Subject(s)
Microscopy, Electron, Transmission/methods , T-Lymphocytes/metabolism , Cell Proliferation , Exosomes/immunology , Female , Humans , Ovarian Neoplasms/immunology
12.
Blood Adv ; 1(16): 1263-1273, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-29296768

ABSTRACT

To discern features of non-Hodgkin lymphomas (NHL) that are autonomous from those that are shaped by the tumor environment (TE), we used patient-derived xenografts (PDX) to probe the effects on neoplastic cells of manipulating the TE. Properties of neoplastic cells that are often considered to be autonomous include their relative independence from stromal support, their relative survival and/or proliferation advantages compared with nonneoplastic cells, and their state of differentiation. Prior approaches to creation of PDX models likely select for neoplasms, which are the most capable of engraftment, potentially masking the effects of the TE. To overcome this bias, we developed a robust protocol that rapidly produced xenografts with more than 85% of unselected, cryo-preserved, B-cell NHL specimens, including low-grade tumors such as follicular and marginal zone lymphoma. To discern features that are shaped by the TE, we extensively studied 4 low-grade lymphoma specimens. B-cell engraftment required components of the native TE; specifically, CD4+ cells. The relative survival of neoplastic compared with nonneoplastic B cells was not autonomous in 2 specimens; specifically, neoplastic B cells from 2 specimens showed a greater dependence on the TE than normal B cells for engraftment. Furthermore, the differentiation of neoplastic B cells was dependent on the TE; mature B-cell neoplasms converted to plasmacytoma-like lesions in the grafts. These results highlight the central and patient-specific roles of the TE in maintaining the relative survival of neoplastic cells compared with normal cells and in controlling the differentiation of neoplastic cells.

13.
Cancer Immunol Res ; 3(11): 1269-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26112921

ABSTRACT

The identification of immunosuppressive factors within human tumor microenvironments, and the ability to block these factors, would be expected to enhance patients' antitumor immune responses. We previously established that an unidentified factor, or factors, present in ovarian tumor ascites fluids reversibly inhibited the activation of T cells by arresting the T-cell signaling cascade. Ultracentrifugation of the tumor ascites fluid has now revealed a pellet that contains small extracellular vesicles (EV) with an average diameter of 80 nm. The T-cell arrest was determined to be causally linked to phosphatidylserine (PS) that is present on the outer leaflet of the vesicle bilayer, as a depletion of PS-expressing EV or a blockade of PS with anti-PS antibody significantly inhibits the vesicle-induced signaling arrest. The inhibitory EV were also isolated from solid tumor tissues. The presence of immunosuppressive vesicles in the microenvironments of ovarian tumors and our ability to block their inhibition of T-cell function represent a potential therapeutic target for patients with ovarian cancer.


Subject(s)
Extracellular Vesicles/immunology , Ovarian Neoplasms/immunology , Phosphatidylserines/physiology , T-Lymphocyte Subsets/immunology , Tumor Microenvironment/immunology , Ascites/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immune Tolerance , Immunophenotyping , Lipids/immunology , Lymphocyte Activation/immunology , Phosphatidylserines/metabolism , Signal Transduction/immunology
14.
AAPS J ; 15(4): 897-900, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856740

ABSTRACT

The administration of therapeutic proteins via the subcutaneous route (sc) is desired for compliance and convenience, but could be challenging due to perceived immunogenic potential or unwanted immune responses. There are clinical and preclinical data supporting as well as refuting the generalized notion that sc is more immunogenic. We provide a mechanistic perspective of immunogenicity of therapeutic proteins administered via the sc route and discuss strategies and opportunities for novel therapeutic approaches to mitigate immunogenicity.


Subject(s)
Antigen Presentation/physiology , Immunogenetic Phenomena/physiology , Skin/immunology , Skin/metabolism , Animals , Antigen Presentation/drug effects , Humans , Immunogenetic Phenomena/drug effects , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/immunology , Immunosuppressive Agents/metabolism , Injections, Subcutaneous , Proto-Oncogene Proteins/administration & dosage , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Skin/drug effects
15.
Cancer Immun ; 13: 14, 2013.
Article in English | MEDLINE | ID: mdl-23882159

ABSTRACT

Human memory T cells present in ovarian tumor ascites fluids fail to respond normally to stimulation via the T cell receptor (TCR). This immunosuppression is manifested by decreases in NF-κB and NFAT activation, IFN-γ production, and cell proliferation in response to TCR stimulation with immobilized antibodies to CD3 and CD28. The anergy of the tumor-associated T cells (TATs) is mediated by soluble factors present in ovarian tumor ascites fluids. The non-responsiveness of the T cells is quickly reversed when the cells are assayed in the absence of the ascites fluid, and is rapidly reestablished when a cell-free ascites fluid is added back to the T cells. Based upon the observed normal phosphorylation patterns of the TCR proximal signaling molecules, the inhibition of NF-κB, and NFAT activation in response to TCR stimulation, as well as the ability of the diacylglycerol analog PMA and the ionophore ionomycin to bypass the ascites fluid-induced TCR signaling arrest, the site of the arrest in the activation cascade appears to be at or just upstream of PLC-γ. An identical TCR signaling arrest pattern was observed when T cells derived from normal donor peripheral blood were incubated with either malignant or nonmalignant (cirrhotic) ascites fluids. The immunosuppressive activity of ascites fluids reported here suggests that soluble factors acting directly or indirectly upon T cells present within tumors contribute to the anergy that has previously been observed in T cells derived from malignant and nonmalignant inflammatory microenvironments. The soluble immunosuppressive factors represent potential therapeutic targets for ovarian cancer.


Subject(s)
NF-kappa B/immunology , NFATC Transcription Factors/immunology , Ovarian Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , Ascites/immunology , Ascites/pathology , Female , Humans , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
16.
Cancer Immun ; 13: 11, 2013.
Article in English | MEDLINE | ID: mdl-23885217

ABSTRACT

Despite an initial response to chemotherapy, most patients with ovarian cancer eventually progress and succumb to their disease. Understanding why effector T cells that are known to infiltrate the tumor do not eradicate the disease after cytoreduction is critically important to the development of novel therapeutic strategies to augment tumor immunity and improve patient outcomes. Such studies have been hampered by the lack of a suitable in vivo model. We report here a simple and reliable model system in which ovarian tumor cell aggregates implanted intraperitoneally into severely immunodeficient NSG mice establish tumor microenvironments within the omentum. The rapid establishment of tumor xenografts within this small anatomically well-defined site enables the recovery, characterization, and quantification of tumor and tumor-associated T cells. We validate here the ability of the omental tumor xenograft (OTX) model to quantify changes in tumor cell number in response to therapy, to quantify changes in the tumor vasculature, and to demonstrate and study the immunosuppressive effects of the tumor microenvironment. Using the OTX model, we show that the tumor-associated T cells originally present within the tumor tissues are anergic and that fully functional autologous T cells injected into tumor-bearing mice localize within the tumor xenograft. The transferred T cells remain functional for up to 3 days within the tumor microenvironment but become unresponsive to activation after 7 days. The OTX model provides for the first time the opportunity to study in vivo the cellular and molecular events contributing to the arrest in T cell function in human ovarian tumors.


Subject(s)
Ovarian Neoplasms/blood supply , Ovarian Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Female , Heterografts , Humans , Immunochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , T-Lymphocytes/pathology , Tumor Microenvironment
17.
J Biol Chem ; 288(24): 17051-6, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23649621

ABSTRACT

Administration of recombinant factor VIII (FVIII), an important co-factor in blood clotting cascade, elicits unwanted anti-FVIII antibodies in hemophilia A (HA) patients. Previously, FVIII associated with phosphatidylserine (PS) showed significant reduction in the anti-FVIII antibody response in HA mice. The reduction in the immune response to FVIII-PS could be due either to a failure of the immune system to recognize the antigen (i.e. immunological ignorance) or to an active induction of an antigen-specific nonresponsiveness (i.e. immunological tolerance). If it were a result of tolerance, one would predict that pre-exposure to FVIII-PS would render the mice hypo-responsive to a subsequent FVIII challenge. Here, we have demonstrated that naive HA mice that were pretreated with FVIII-PS showed a significantly reduced FVIII immune response to further challenge with native FVIII and that this decreased responsiveness could be adoptively transferred to other mice. An increase in number of FoxP3-expressing CD4(+) regulatory T-cells (Treg) was observed for the FVIII-PS-immunized group as compared with animals that received FVIII alone, suggesting the involvement of Treg in PS-mediated hypo-responsiveness. The PS-mediated reduction in antibody response was reversed by the co-administration of function-blocking anti-TGF-ß antibody with FVIII-PS. The decreased response to FVIII induced by FVIII-PS was determined to be antigen-specific because the immune response to another non-cross-reactive antigen (ovalbumin) was not altered. These results are consistent with the notion that FVIII-PS is tolerogenic and suggest that immunization with this tolerogenic form of the protein could be a useful treatment option to minimize immunogenicity of FVIII and other protein-based therapeutics.


Subject(s)
Factor VIII/immunology , Hemophilia A/therapy , Phosphatidylserines/immunology , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , Antibodies, Neutralizing/blood , Factor VIII/administration & dosage , Forkhead Transcription Factors/metabolism , Hemophilia A/immunology , Humans , Immune Tolerance , Mice , Mice, Knockout , Phosphatidylserines/administration & dosage , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation
18.
J Pharm Sci ; 101(6): 2055-65, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22388918

ABSTRACT

The administration of recombinant factor VIII (FVIII) is the first-line therapy for hemophilia A (HA), but 25%-35% of patients develop an inhibitory antibody response. In general, the presence of aggregates contributes to unwanted immunogenic responses against therapeutic proteins. FVIII has been shown to form both native-like and nonnative aggregates. Previously, we showed that nonnative aggregates of FVIII are less immunogenic than the native protein. Here, we investigated the effect of native-like aggregates of FVIII on immunogenicity in HA and von Willebrand factor knockout (vWF(-/-)) mice. Mice immunized with native-like aggregates showed significantly higher inhibitory antibody titers than animals that received native FVIII. Following restimulation in vitro with native FVIII, the activation of CD4+ T-cells isolated from mice immunized with native-like aggregates is approximately fourfold higher than mice immunized with the native protein. Furthermore, this is associated with increases in the secretion of proinflammatory cytokines IL-6 and IL-17 in the native-like aggregate treatment group. The results indicate that the native-like aggregates of FVIII are more immunogenic than native FVIII for both the B-cell and the T-cell responses.


Subject(s)
Factor VIII/immunology , Hemophilia A/genetics , von Willebrand Factor/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Factor VIII/genetics , Mice
19.
J Assoc Res Otolaryngol ; 13(3): 423-35, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22310933

ABSTRACT

A majority of T cells from chronic inflammatory tissues derived from patients with nasal polyposis were found to express an effector memory phenotype. We report here that these memory T cells failed to activate NF-κB in response to TCR stimulation but responded normally when the proximal TCR signaling molecules were bypassed with PMA and ionomycin. The dysfunction of these cells was associated with a decrease in the phosphorylation of several TCR proximal signaling molecules including ZAP70, Lck and SLP-76. In addition to the disruption in the TCR signaling pathway, the nasal polyp-associated T cells were shown to have a defect in their ability to translocate LAMP-1 to the cell surface. The results presented here establish that the phenotype and anergy of the T cells in the nasal polyp are similar to those which is seen in memory T cells derived from human tumors and other sites of chronic inflammation.


Subject(s)
CD28 Antigens/metabolism , CD3 Complex/metabolism , Nasal Polyps/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Calcium Ionophores , Case-Control Studies , Flow Cytometry , Humans , Immunologic Memory , Immunophenotyping , Ionomycin , Lymphocyte Activation , Lysosomal Membrane Proteins/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Phenotype , Phosphorylation , Signal Transduction , T-Lymphocytes/cytology
20.
J Pharm Sci ; 101(1): 48-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21953409

ABSTRACT

Factor VIII (FVIII) is an important coagulation cofactor and its deficiency causes Hemophilia A, a bleeding disorder. Replacement therapy using recombinant FVIII is currently the first line of therapy for Hemophilia A, but the development of neutralizing antibody is a major clinical complication for this therapy. Recently, it has been shown that FVIII associated with phosphatidylinositol (PI)-containing lipidic nanoparticles reduced development of neutralizing antibodies in Hemophilia A mice (Peng A, Straubinger RM, Balu-Iyer SV. 2010. AAPS J 12(3):473-481). Here, we investigated the underlying mechanism of this reduction in antibody response in culturing conditions. In vitro, PI interfered with the processing of FVIII by cultured dendritic cells (DC), resulting in a reduction in the upregulation of phenotypic costimulatory signal CD40. Furthermore, PI increased secretion of regulatory cytokines Transforming Growth Factor ß1 and Interleukin 10 (IL-10) but reduced the secretion of proinflammatory cytokines IL-6 and IL-17. The data suggest that PI reduces immunogenicity of FVIII by modulating DC maturation and inducing secretion of regulatory cytokines.


Subject(s)
CD40 Antigens/metabolism , Dendritic Cells/immunology , Factor VIII/immunology , Factor VIII/pharmacology , Phosphatidylinositols/immunology , Phosphatidylinositols/pharmacology , Transforming Growth Factor beta1/metabolism , Animals , Antibodies, Neutralizing/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , CD40 Antigens/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Down-Regulation/drug effects , Down-Regulation/immunology , Factor VIII/metabolism , Humans , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...