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










Publication year range
1.
Melanoma Res ; 32(2): 88-97, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35254331

ABSTRACT

Heat shock proteins (hsp) are intracellular chaperones that possess extracellular immunostimulatory properties when complexed with antigens. A recombinant Hsp110-gp100 chaperone complex vaccine showed an antitumor response and prolonged survival in murine melanoma. A phase Ib dose-escalation study of a recombinant human Hsp110-gp100 vaccine in advanced-stage melanoma patients was performed to evaluate toxicity, immunostimulatory potential and clinical response. Patients with pretreated, unresectable stage IIIB/C/IV melanoma received the chaperone complex vaccine in a dose-escalation protocol; three vaccinations over a 43-day-period. Tumor response, clinical toxicity and immune response were measured. Ten patients (eight female, median age 70 years) were enrolled and two patients had grade 1 adverse events; minor skin rash, hyperhidrosis and fever (no grade 2 or higher adverse events). Median progression-free survival was longer for lower vaccine doses as compared to the maximum dose of 180 mcg (4.5 vs. 2.9 months; P = 0.018). The lowest dose patients (30 and 60 mcg) had clinical tumor responses (one partial response, one stable disease). CD8+ T cell interferon-γ responses to gp100 were greater in the clinically responding patients. A pattern of B cell responses to vaccination was not observed. Regulatory T cell populations and co-stimulatory molecules including cytotoxic T-lymphocyte-associated protein 4 and PD-1 appeared to differ in responders versus nonresponders. A fully recombinant human Hsp110-gp100 chaperone complex vaccine had minimal toxicity, measurable tumor responses at lower doses and produced peripheral CD8+ T cell activation in patients with advanced, pretreated melanoma. Combination with currently available immunotherapies may augment clinical responses.


Subject(s)
Cancer Vaccines , Melanoma , Skin Neoplasms , Aged , Animals , CD8-Positive T-Lymphocytes , Cancer Vaccines/adverse effects , Female , Humans , Male , Melanoma/drug therapy , Mice , Skin Neoplasms/chemically induced , Skin Neoplasms/drug therapy , gp100 Melanoma Antigen/metabolism
2.
J Immunother Cancer ; 9(1)2021 01.
Article in English | MEDLINE | ID: mdl-33468554

ABSTRACT

BACKGROUND: The efficacy of cancer immunotherapy can be limited by the poor immunogenicity of cancer and the immunosuppressive tumor microenvironment (TME). Immunologically programming the TME and creating an immune-inflamed tumor phenotype is critical for improving the immune-responsiveness of cancers. Here, we interrogate the immune modulator Flagrp170, engineered via incorporation of a pathogen-associated molecular pattern (ie, flagellin) into an immunostimulatory chaperone molecule, in transforming poorly immunogenic tumors and establishing a highly immunostimulatory milieu for immune augmentation. METHODS: Multiple murine cancer models were used to evaluate the immunostimulatory activity, antitumor potency, and potential side effects of Flagrp170 on administration into the tumors using a replication impaired adenovirus. Antibody neutralization and mice deficient in pattern recognition receptors, that is, toll-like receptor 5 (TLR5) and NOD like receptor (NLR) family caspase activation and recruitment domain (CARD) domain-containing protein 4 (NLRC4), both of which can recognize flagellin, were employed to understand the immunological mechanism of action of the Flagrp170. RESULTS: Intratumoral delivery of mouse or human version of Flagrp170 resulted in robust inhibition of multiple malignancies including head and neck squamous cell carcinoma and breast cancer, without tissue toxicities. This in situ Flagrp170 treatment induced a set of cytokines in the TME known to support Th1/Tc1-dominant antitumor immunity. Additionally, granulocyte macrophage colony-stimulating factor derived from mobilized CD8+ T cells was involved in the therapeutic activity of Flagrp170. We also made a striking finding that NLRC4, not TLR5, is required for Flagrp170-mediated antitumor immune responses. CONCLUSION: Our results elucidate a novel immune-potentiating activity of Flagrp170 via engaging the innate pattern recognition receptor NLRC4, and support its potential clinical use to reshape cancer immune phenotype for overcoming therapeutic resistance.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/therapy , Calcium-Binding Proteins/genetics , Flagellin/genetics , HSP70 Heat-Shock Proteins/genetics , Head and Neck Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/therapy , Adenoviridae/genetics , Animals , Antibodies, Neutralizing/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Flagellin/metabolism , HSP70 Heat-Shock Proteins/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Humans , Mice , Recombinant Proteins , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Toll-Like Receptor 5/genetics , Treatment Outcome , Tumor Microenvironment , Xenograft Model Antitumor Assays
3.
Nat Commun ; 11(1): 1821, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286326

ABSTRACT

The abscopal effect following ionizing radiation therapy (RT) is considered to be a rare event. This effect does occur more frequently when combined with other therapies, including immunotherapy. Here we demonstrate that the frequency of abscopal events following RT alone is highly dependent upon the degree of adrenergic stress in the tumor-bearing host. Using a combination of physiologic, pharmacologic and genetic strategies, we observe improvements in the control of both irradiated and non-irradiated distant tumors, including metastatic tumors, when adrenergic stress or signaling through ß-adrenergic receptor is reduced. Further, we observe cellular and molecular evidence of improved, antigen-specific, anti-tumor immune responses which also depend upon T cell egress from draining lymph nodes. These data suggest that blockade of ß2 adrenergic stress signaling could be a useful, safe, and feasible strategy to improve efficacy in cancer patients undergoing radiation therapy.


Subject(s)
Adrenergic Agents/pharmacology , Immunity , Neoplasms/immunology , Neoplasms/radiotherapy , Radiation, Ionizing , Stress, Physiological , Adrenergic beta-Antagonists/pharmacology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Lymph Nodes/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects
4.
Methods Mol Biol ; 1709: 345-357, 2018.
Article in English | MEDLINE | ID: mdl-29177671

ABSTRACT

Large heat shock proteins (HSPs) or stress proteins, including Hsp110 and Grp170, are unique molecular chaperones with superior capability of shuttling tumor protein antigens into professional antigen-presenting cells, such as dendritic cells, for highly efficient cross-presentation and T cell priming. Reconstituted chaperone complexes of large HSP and tumor protein antigen have been demonstrated to generate a robust antigen-specific T lymphocyte response with therapeutic potency against multiple cancer types in preclinical models. Here, we describe the methods for preparing this recombinant chaperone complex vaccine and analyzing the vaccine-induced activation of antigen-specific T cells using in vitro and in vivo systems.


Subject(s)
Cancer Vaccines/immunology , Glycoproteins/immunology , HSP110 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/immunology , Vaccines, Synthetic/immunology , Humans , Immunotherapy , Neoplasm Proteins/immunology , Neoplasms/metabolism , Neoplasms/therapy
5.
Adv Cancer Res ; 128: 309-64, 2015.
Article in English | MEDLINE | ID: mdl-26216637

ABSTRACT

Scavenger receptors constitute a large family of evolutionally conserved protein molecules that are structurally and functionally diverse. Although scavenger receptors were originally identified based on their capacity to scavenge modified lipoproteins, these molecules have been shown to recognize and bind to a broad spectrum of ligands, including modified and unmodified host-derived molecules or microbial components. As a major subset of innate pattern recognition receptors, scavenger receptors are mainly expressed on myeloid cells and function in a wide range of biological processes, such as endocytosis, adhesion, lipid transport, antigen presentation, and pathogen clearance. In addition to playing a crucial role in maintenance of host homeostasis, scavenger receptors have been implicated in the pathogenesis of a number of diseases, e.g., atherosclerosis, neurodegeneration, or metabolic disorders. Emerging evidence has begun to reveal these receptor molecules as important regulators of tumor behavior and host immune responses to cancer. This review summarizes our current understanding on the newly identified, distinct functions of scavenger receptors in cancer biology and immunology. The potential of scavenger receptors as diagnostic biomarkers and novel targets for therapeutic interventions to treat malignancies is also highlighted.


Subject(s)
Neoplasms/immunology , Neoplasms/metabolism , Receptors, Scavenger/immunology , Receptors, Scavenger/metabolism , Animals , Humans , Neoplasms/diagnosis , Neoplasms/therapy
6.
Front Oncol ; 4: 377, 2014.
Article in English | MEDLINE | ID: mdl-25629003

ABSTRACT

Glucose-regulated protein 170 (GRP170) is the largest member of glucose-regulated protein family that resides in the endoplasmic reticulum (ER). As a component of the ER chaperone network, GRP170 assists in protein folding, assembly, and transportation of secretory or transmembrane proteins. The well documented cytoprotective activity of intracellular GRP170 due to its intrinsic chaperoning property has been shown to provide a survival benefit in cancer cells during tumor progression or metastasis. Accumulating evidence shows that extracellular GRP170 displays a superior capacity in delivering tumor antigens to specialized antigen-presenting cells for cross-presentation, resulting in generation of an anti-tumor immune response dependent on cytotoxic CD8(+) T cells. This unique feature of GRP170 provides a molecular basis for using GRP170 as an immunostimulatory adjuvant to develop a recombinant vaccine for therapeutic immunization against cancers. This review summarizes the latest findings in understanding the biological effects of GRP170 on cell functions and tumor progression. The immunomodulating activities of GRP170 during interactions with the innate and adaptive arms of the immune system as well as its therapeutic applications in cancer immunotherapy will be discussed.

7.
J Immunol ; 191(12): 6010-21, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24218449

ABSTRACT

Efficient cross-presentation of protein Ags to CTLs by dendritic cells (DCs) is essential for the success of prophylactic and therapeutic vaccines. In this study, we report a previously underappreciated pathway involving Ag entry into the endoplasmic reticulum (ER) critically needed for T cell cross-priming induced by a DC-targeted vaccine. Directing the clinically relevant, melanoma Ag gp100 to mouse-derived DCs by molecular adjuvant and chaperone Grp170 substantially facilitates Ag access to the ER. Grp170 also strengthens the interaction of internalized protein Ag with molecular components involved in ER-associated protein dislocation and/or degradation, which culminates in cytosolic translocation for proteasome-dependent degradation and processing. Targeted disruption of protein retrotranslocation causes exclusive ER retention of tumor Ag in mouse bone marrow-derived DCs and splenic CD8(+) DCs. This results in the blockade of Ag ubiquitination and processing, which abrogates the priming of Ag-specific CD8(+) T cells in vitro and in vivo. Therefore, the improved ER entry of tumor Ag serves as a molecular basis for the superior cross-presenting capacity of Grp170-based vaccine platform. The ER access and retrotranslocation represents a distinct pathway that operates within DCs for cross-presentation and is required for the activation of Ag-specific CTLs by certain vaccines. These results also reinforce the importance of the ER-associated protein quality control machinery and the mode of the Ag delivery in regulating DC-elicited immune outcomes.


Subject(s)
Adjuvants, Immunologic , Antigen Presentation/immunology , Cancer Vaccines/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Endoplasmic Reticulum/immunology , Glycoproteins/immunology , HSP70 Heat-Shock Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination/methods , gp100 Melanoma Antigen/immunology , ADP Ribose Transferases/pharmacology , Adoptive Transfer , Animals , Bacterial Toxins/pharmacology , Bone Marrow Cells/immunology , Cancer Vaccines/pharmacokinetics , Cell Lineage , Cells, Cultured , Cytosol/metabolism , Dendritic Cells/classification , Endocytosis/immunology , Endosomes/metabolism , Exotoxins/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational , Protein Transport , Proteolysis , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics , SEC Translocation Channels , Spleen/cytology , Spleen/immunology , Ubiquitination , Virulence Factors/pharmacology , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/pharmacokinetics , Pseudomonas aeruginosa Exotoxin A
8.
Int J Hyperthermia ; 29(5): 364-75, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23829534

ABSTRACT

Although the large stress/heat shock proteins (HSPs), i.e. Hsp110 and Grp170, were identified over 30 years ago, these abundant and highly conserved molecules have received much less attention compared to other conventional HSPs. Large stress proteins act as molecular chaperones with exceptional protein-holding capability and prevent the aggregation of proteins induced by thermal stress. The chaperoning properties of Hsp110 and Grp170 are integral to the ability of these molecules to modulate immune functions and are essential for developing large chaperone complex vaccines for cancer immunotherapy. The potent anti-tumour activity of the Hsp110/Grp170-tumour protein antigen complexes demonstrated in preclinical studies has led to a phase I clinical trial through the National Cancer Institute's rapid access to intervention development (RAID) programme that is presently underway. Here we review aspects of the structure and function of these large stress proteins, their roles as molecular chaperones in the biology of cell stress, and prospects for their use in immune regulation and cancer immunotherapy. Lastly, we will discuss the recently revealed immunosuppressive activity of scavenger receptor A that binds to Hsp110 and Grp170, as well as the feasibility of targeting this receptor to promote T-cell activation and anti-tumour immunity induced by large HSP vaccines and other immunotherapies.


Subject(s)
HSP110 Heat-Shock Proteins , HSP70 Heat-Shock Proteins , Animals , Cancer Vaccines/therapeutic use , HSP110 Heat-Shock Proteins/chemistry , HSP110 Heat-Shock Proteins/physiology , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/physiology , Humans , Immunotherapy , Molecular Weight , Neoplasms/therapy
9.
Adv Cancer Res ; 119: 421-75, 2013.
Article in English | MEDLINE | ID: mdl-23870514

ABSTRACT

Therapeutic vaccines represent a viable option for active immunotherapy of cancers that aim to treat late stage disease by using a patient's own immune system. The promising results from clinical trials recently led to the approval of the first therapeutic cancer vaccine by the U.S. Food and Drug Administration. This major breakthrough not only provides a new treatment modality for cancer management but also paves the way for rationally designing and optimizing future vaccines with improved anticancer efficacy. Numerous vaccine strategies are currently being evaluated both preclinically and clinically. This review discusses therapeutic cancer vaccines from diverse platforms or targets as well as the preclinical and clinical studies employing these therapeutic vaccines. We also consider tumor-induced immune suppression that hinders the potency of therapeutic vaccines, and potential strategies to counteract these mechanisms for generating more robust and durable antitumor immune responses.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasms/immunology , Neoplasms/therapy , Antigens, Neoplasm/chemistry , Clinical Trials as Topic , Dendritic Cells/pathology , Humans , Immune System , Medical Oncology/trends , United States , United States Food and Drug Administration , Vaccines, DNA/therapeutic use
11.
Cancer Res ; 73(7): 2093-103, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23333935

ABSTRACT

Converting the immunosuppressive tumor environment into one that is favorable to the induction of antitumor immunity is indispensable for effective cancer immunotherapy. Here, we strategically incorporate a pathogen (i.e., flagellin)-derived, NF-κB-stimulating "danger" signal into the large stress protein or chaperone Grp170 (HYOU1/ORP150) that was previously shown to facilitate antigen crosspresentation. This engineered chimeric molecule (i.e., Flagrp170) is capable of transporting tumor antigens and concurrently inducing functional activation of dendritic cells (DC). Intratumoral administration of adenoviruses expressing Flagrp170 induces a superior antitumor response against B16 melanoma and its distant lung metastasis compared with unmodified Grp170 and flagellin. The enhanced tumor destruction is accompanied with significantly increased tumor infiltration by CD8(+) cells as well as elevation of IFN-γ and interleukin (IL)-12 levels in the tumor sites. In situ Ad.Flagrp170 therapy provokes systemic activation of CTLs that recognize several antigens naturally expressing in melanoma (e.g., gp100/PMEL and TRP2/DCT). The mechanistic studies using CD11c-DTR transgenic mice and Batf3-deficient mice reveal that CD8α(+) DCs are required for the improved T-cell crosspriming. Antibody neutralization assays show that IL-12 and IFN-γ are essential for the Flagrp170-elicited antitumor response, which also involves CD8(+) T cells and natural killer cells. The therapeutic efficacy of Flagrp170 and its immunostimulating activity are also confirmed in mouse prostate cancer and colon carcinoma. Together, targeting the tumor microenvironment with this chimeric chaperone is highly effective in mobilizing or restoring antitumor immunity, supporting the potential therapeutic use of this novel immunomodulator in the treatment of metastatic diseases.


Subject(s)
Cancer Vaccines/therapeutic use , Colonic Neoplasms/therapy , Immunotherapy , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Molecular Chaperones/immunology , Prostatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flagellin/genetics , Flagellin/immunology , Flagellin/metabolism , Glycoproteins/genetics , Glycoproteins/immunology , Glycoproteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunoenzyme Techniques , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , NF-kappa B , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured
12.
Hepatology ; 57(1): 228-38, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22821642

ABSTRACT

UNLABELLED: Negative feedback immune mechanisms are essential for maintenance of hepatic homeostasis and prevention of immune-mediated liver injury. We show here that scavenger receptor A (SRA/CD204), a pattern recognition molecule, is highly up-regulated in the livers of patients with autoimmune or viral hepatitis, and of mice during concanavalin A (Con A)-induced hepatitis (CIH). Strikingly, genetic SRA ablation strongly sensitizes mice to Con A-induced liver injury. SRA loss, increased mortality and liver pathology correlate with excessive production of IFN-γ and heightened activation of T cells. Increased liver expression of SRA primarily occurs in mobilized hepatic myeloid cells during CIH, including CD11b(+) Gr-1(+) cells. Mechanistic studies establish that SRA on these cells functions as a negative regulator limiting T-cell activity and cytokine production. SRA-mediated protection from CIH is further validated by adoptive transfer of SRA(+) hepatic mononuclear cells or administration of a lentivirus-expressing SRA, which effectively ameliorates Con A-induced hepatic injury. Also, CIH and clinical hepatitis are associated with increased levels of soluble SRA. This soluble SRA displays a direct T-cell inhibitory effect and is capable of mitigating Con A-induced liver pathology. CONCLUSION: Our findings demonstrate an unexpected role of SRA in attenuation of Con A-induced, T-cell-mediated hepatic injury. We propose that SRA serves as an important negative feedback mechanism in liver immune homeostasis, and may be exploited for therapeutic treatment of inflammatory liver diseases.


Subject(s)
Chemical and Drug Induced Liver Injury/immunology , Concanavalin A/toxicity , Hepatitis, Animal/immunology , Scavenger Receptors, Class A/metabolism , T-Lymphocytes/physiology , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Hepatitis, Animal/metabolism , Interferon-gamma/metabolism , Liver/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Mol Cancer Ther ; 11(11): 2331-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22896667

ABSTRACT

Given the complexity of prostate cancer progression and metastasis, multimodalities that target different aspects of tumor biology, for example, radiotherapy in conjunction with immunotherapy, may provide the best opportunities for promoting clinical benefits in patients with high-risk localized prostate cancer. Here, we show that intratumoral administration of unmodified dendritic cells (DC) failed to synergize with fractionated radiotherapy. However, ionizing radiation combined with in situ vaccination with DCs, in which the immunosuppressive scavenger receptor A (SRA/CD204) has been downregulated by lentivirus-mediated gene silencing, profoundly suppressed the growth of two mouse prostate cancers (e.g., RM1 and TRAMP-C2) and prolonged the lifespan of tumor-bearing animals. Treatment of subcutaneous tumors with this novel combinatorial radioimmunotherapeutic regimen resulted in a significant reduction in distant experimental metastases. SRA/CD204-silenced DCs were highly efficient in generating antigen or tumor-specific T cells with increased effector functions (e.g., cytokine production and tumoricidal activity). SRA/CD204 silencing-enhanced tumor cell death was associated with elevated IFN-γ levels in tumor tissue and increased tumor-infiltrating CD8(+) cells. IFN-γ neutralization or depletion of CD8(+) cells abrogated the SRA/CD204 downregulation-promoted antitumor efficacy, indicating a critical role of IFN-γ-producing CD8(+) T cells. Therefore, blocking SRA/CD204 activity significantly enhances the therapeutic potency of local radiotherapy combined with in situ DC vaccination by promoting a robust systemic antitumor immunity. Further studies are warranted to test this novel combinatorial approach for translating into improved clinical outcomes in patients with prostate cancer.


Subject(s)
Dendritic Cells/immunology , Gene Silencing , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , Scavenger Receptors, Class A/genetics , Vaccination , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Death , Cross-Priming/immunology , Humans , Interferon-gamma/biosynthesis , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , RNA, Small Interfering/metabolism , T-Lymphocytes, Cytotoxic/immunology , Treatment Outcome
14.
Immunol Cell Biol ; 90(1): 101-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21383767

ABSTRACT

Given the primary expression of scavenger receptor A (SRA) or CD204 on antigen-presenting cells, we investigate the immunoregulatory activities of SRA/CD204 in the context of cross-presentation of cell-associated antigen and the immunogenicity of dying tumor cells. Immunization with dying prostate cancer cells results in profoundly increased control of subsequently inoculated tumors in SRA/CD204 knockout mice. Using OVA-expressing RM1 prostate tumor line (RM1-OVA), we show for the first time that SRA absence greatly enhances dendritic cells (DCs)-mediated cross-presentation of OVA antigen derived from dying RM1 cells. While the phagocytic ability of DCs is not significantly impacted by the lack of SRA/CD204, DCs deficient in SRA/CD204 display increased expression of inflammatory cytokines and chemokines, as well as co-stimulatory molecules upon interaction with dying RM1 cells, implicating a suppressive regulation of the functional activation of DCs by SRA/CD204. Further, SRA/CD204-deficient DCs pulsed with dying RM1-OVA cells are more effective than wild-type counterparts in priming antigen-specific T-cell responses, resulting in improved control of RM1 tumor growth in both prophylactic and therapeutic settings. Our findings suggest that the increased immunogenicity of dying tumor cells in SRA/CD204 knockout mice is attributed to the altered functions of DCs in the absence of SRA/CD204, which underscores the important role of SRA/CD204 in host immune homeostasis. Selective downregulation or blockade of this immunoregulatory molecule may lead to enhanced potency of DC-based vaccines capable of breaking immune tolerance against cancer.


Subject(s)
Antigens/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Neoplasms, Experimental/immunology , Scavenger Receptors, Class A/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens/genetics , Antigens/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Flow Cytometry , Immunotherapy, Adoptive/methods , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Phagocytosis/immunology , Scavenger Receptors, Class A/deficiency , Scavenger Receptors, Class A/genetics , Tumor Burden/genetics , Tumor Burden/immunology
15.
FASEB J ; 26(4): 1493-505, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22207611

ABSTRACT

Recognition of pathogen-associated molecular patterns by innate immune receptors is essential for host defense responses. Although extracellular stress proteins are considered as indicators of the stressful conditions (e.g., infection or cell injury), the exact roles of these molecules in the extracellular milieu remain less defined. We found that glucose-regulated protein 170 (Grp170), the largest stress protein and molecular chaperone, is highly efficient in binding CpG oligodeoxynucleotides (CpG-ODN), the microbial DNA mimetic sensed by toll-like receptor 9 (TLR9). Extracellular Grp170 markedly potentiates the endocytosis and internalization of CpG-ODN by mouse bone marrow-derived macrophages and directly interacts with endosomal TLR9 on cell entry. These molecular collaborations result in the synergistic activation of the MyD88-dependent signaling and enhanced production of proinflammatory cytokines and nitric oxide in mouse primary macrophages as well as human THP-1 monocyte-derived macrophages, suggesting that Grp170 released from injured cells facilitates the sensing of pathogen-associated "danger" signals by intracellular receptors. This CpG-ODN chaperone complex-promoted innate immunity confers increased resistance in mice to infection of Listeria monocytogenes compared with CpG-ODN treatment alone. Our studies reveal a previously unrecognized attribute of Grp170 as a superior DNA-binding chaperone capable of amplifying TLR9 activation on pathogen recognition, which provides a conceptual advance in understanding the dynamics of ancient chaperoning functions inside and outside the cell.


Subject(s)
Glycoproteins/immunology , HSP70 Heat-Shock Proteins/immunology , Immunity, Innate/immunology , Macrophages/immunology , Molecular Chaperones/immunology , Animals , Endocytosis/immunology , Endosomes/metabolism , Glycoproteins/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Listeria monocytogenes/immunology , Macrophages/cytology , Mice , Mice, Inbred Strains , Molecular Chaperones/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Oligodeoxyribonucleotides/immunology , Toll-Like Receptor 9/immunology
16.
J Mol Med (Berl) ; 90(4): 413-26, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22083206

ABSTRACT

Pattern recognition scavenger receptor SRA/CD204, primarily expressed on specialized antigen-presenting cells (APCs), including dendritic cells (DCs) and macrophages, has been implicated in multiple physiological and pathological processes, including atherosclerosis, Alzheimer's disease, endotoxic shock, host defense, and cancer development. SRA/CD204 was also recently shown to function as an attenuator of vaccine response and antitumor immunity. Here, we, for the first time, report that SRA/CD204 knockout (SRA(-/-)) mice developed a more robust CD4(+) T cell response than wild-type mice after ovalbumin immunization. Splenic DCs from the immunized SRA(-/-) mice were much more efficient than those from WT mice in stimulating naïve OT-II cells, indicating that the suppressive activity of SRA/CD204 is mediated by DCs. Strikingly, antigen-exposed SRA(-/-) DCs with or without lipopolysaccharide treatment exhibited increased T-cell-stimulating activity in vitro, which was independent of the classical endocytic property of the SRA/CD204. Additionally, absence of SRA/CD204 resulted in significantly elevated IL12p35 expression in DCs upon CD40 ligation plus interferon gamma (IFN-γ) stimulation. Molecular studies reveal that SRA/CD204 inhibited the activation of STAT1, mitogen activated protein kinase p38, and nuclear factor-kappa B signaling activation in DCs treated with anti-CD40 antibodies and IFN-γ. Furthermore, splenocytes from the generated SRA(-/-) OT-II mice showed heightened proliferation upon stimulation with OVA protein or MHC-II-restricted OVA(323-339) peptide compared with cells from the SRA(+/+) OT-II mice. These results not only establish a new role of SRA/CD204 in limiting the intrinsic immunogenicity of APCs and CD4(+) T cell activation but also provide additional insights into the molecular mechanisms involved in the immune suppression by this molecule.


Subject(s)
Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Ovalbumin/immunology , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/cytology , Forkhead Transcription Factors/immunology , Gene Knockout Techniques , Immunization , Interferon-gamma/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
17.
Methods Mol Biol ; 787: 277-87, 2011.
Article in English | MEDLINE | ID: mdl-21898243

ABSTRACT

Large heat-shock proteins (HSPs), including hsp110 and grp170, are unique immunochaperones capable of carrying and introducing antigens into professional antigen-presenting cells for efficient cross-presentation. Therefore, reconstituted chaperone complexes of large HSPs and protein antigen may be exploited for augmentation of an antigen-specific immune response. The methods for the preparation of the recombinant protein antigen chaperone complex and characterization of its T-cell priming capability in both in vitro and in vivo settings are described.


Subject(s)
Antigen Presentation/immunology , Glycoproteins/immunology , HSP110 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Baculoviridae/genetics , Baculoviridae/metabolism , Cancer Vaccines/immunology , Cross-Priming/immunology , Glycoproteins/metabolism , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Transgenic , Molecular Chaperones/immunology , Protein Folding , Recombinant Proteins/immunology
18.
Cancer Res ; 71(21): 6611-20, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21914786

ABSTRACT

Although dendritic cell (DC) vaccines offer promise as cancer immunotherapy, further improvements are needed to amplify their clinical therapeutic efficacy. The pattern recognition scavenger receptor SRA/CD204 attenuates the ability of DCs to activate CD8(+) T-cell responses. Therefore, we examined the impact of SRA/CD204 on antitumor responses generated by DC vaccines and we also evaluated the feasibility of enhancing DC vaccine potency by SRA/CD204 blockade. DCs from SRA/CD204-deficient mice were more immunogenic in generating antitumor responses to B16 melanoma, compared with DCs from wild-type mice. Similarly, siRNA-mediated knockdown of SRA/CD204 by lentiviral vectors improved the ability of wild-type DCs to stimulate the expansion and activation of CD8(+) T cells specific for idealized or established melanoma antigens in mice. Using SRA/CD204-silenced DCs to generate antigen-targeted vaccines, we documented a marked increase in the level of antitumor immunity achieved against established B16 tumors and metastases. This increase was associated with enhanced activation of antigen specific CTLs, greater tumor infiltration by CD8(+) T cells and NK cells, and increased intratumoral ratios of both CD4(+) and CD8(+) T-effector cells to CD4(+)CD25(+) T-regulatory cells. Our studies establish that downregulating SRA/CD204 strongly enhances DC-mediated antitumor immunity. In addition, they provide a rationale to enhance DC vaccine potency through SRA/CD204-targeting approaches that can improve clinical outcomes in cancer treatment.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Genetic Therapy , Genetic Vectors/therapeutic use , Immunotherapy, Active , Lung Neoplasms/secondary , Melanoma, Experimental/secondary , Neoplasm Proteins/antagonists & inhibitors , RNA, Small Interfering/therapeutic use , Scavenger Receptors, Class A/antagonists & inhibitors , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/immunology , Combined Modality Therapy , Cytotoxicity, Immunologic , Drug Screening Assays, Antitumor , Genetic Vectors/pharmacology , Immunotherapy, Adoptive , Lentivirus/genetics , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , RNA, Small Interfering/pharmacology , Scavenger Receptors, Class A/deficiency , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/immunology , T-Lymphocytes, Cytotoxic/transplantation , Vaccination
19.
J Immunol ; 187(6): 2905-14, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21832164

ABSTRACT

We previously reported that scavenger receptor A (SRA/CD204), a binding structure on dendritic cells (DCs) for large stress/heat shock proteins (HSPs; e.g., hsp110 and grp170), attenuated an antitumor response elicited by large HSP-based vaccines. In this study, we show that SRA/CD204 interacts directly with exogenous hsp110, and lack of SRA/CD204 results in a reduction in the hsp110 binding and internalization by DCs. However, SRA(-/-) DCs pulsed with hsp110 or grp170-reconstituted gp100 chaperone complexes exhibit a profoundly increased capability of stimulating melanoma Ag gp100-specific naive T cells compared with wild-type (WT) DCs. Similar results were obtained when SRA/CD204 was silenced in DCs using short hairpin RNA-encoding lentiviruses. In addition, hsp110-stimulated SRA(-/-) DCs produced more inflammatory cytokines associated with increased NF-κB activation, implicating an immunosuppressive role for SRA/CD204. Immunization with the hsp110-gp100 vaccine resulted in a more robust gp100-specific CD8(+) T cell response in SRA(-/-) mice than in WT mice. Lastly, SRA/CD204 absence markedly improved the therapeutic efficacy of the hsp110-gp100 vaccine in mice established with B16 melanoma, which was accompanied by enhanced activation and tumor infiltration of CD8(+) T cells. Given the presence of multiple HSP-binding scavenger receptors on APCs, we propose that selective scavenger receptor interactions with HSPs may lead to highly distinct immunological consequences. Our findings provide new insights into the immune regulatory functions of SRA/CD204 and have important implications in the rational design of protein Ag-targeted recombinant chaperone vaccines for the treatment of cancer.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , HSP110 Heat-Shock Proteins/immunology , Melanoma, Experimental/immunology , Scavenger Receptors, Class A/immunology , gp100 Melanoma Antigen/immunology , Adoptive Transfer , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/metabolism , Cell Separation , Dendritic Cells/metabolism , Flow Cytometry , Gene Silencing , HSP110 Heat-Shock Proteins/metabolism , Immunoprecipitation , Lymphocyte Activation/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Scavenger Receptors, Class A/metabolism , gp100 Melanoma Antigen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...