Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Immunol Cell Biol ; 102(1): 8-11, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37982351

ABSTRACT

The advent and clinical success of immune checkpoint inhibitors Ipilimumab, Nivolumab and Pembrolizumab has had a seismic impact on our drug discovery focus and rationale. Novel extrinsic targets that enhance immune responses to cancer are actively being pursued, while tumor intrinsic targets that render cancer cells more sensitive to the immune system have joined traditional intrinsic targets (e.g. directly cytotoxic) in the drug discovery pipeline. The phosphatase PTPN2 (TC-PTP) and its paralog PTPN1 (PTP-1B) are negative regulators of several cytokine signaling pathways and T cell receptor (TCR) signaling. In a recent publication, Baumgartner et al. demonstrate the pre-clinical efficacy of a first-in-class dual PTPN1/N2 active site inhibitor (ABBV-CLS-484/AC484) in cancer models.


Subject(s)
Antibodies, Monoclonal , Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Nivolumab/therapeutic use , Ipilimumab/therapeutic use , Neoplasms/drug therapy , Immunotherapy , Biology
2.
Cancer Cell ; 33(6): 1033-1047.e5, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894690

ABSTRACT

The co-engagement of fragment crystallizable (Fc) gamma receptors (FcγRs) with the Fc region of recombinant immunoglobulin monoclonal antibodies (mAbs) and its contribution to therapeutic activity has been extensively studied. For example, Fc-FcγR interactions have been shown to be important for mAb-directed effector cell activities, as well as mAb-dependent forward signaling into target cells via receptor clustering. Here we identify a function of mAbs targeting T cell-expressed antigens that involves FcγR co-engagement on antigen-presenting cells (APCs). In the case of mAbs targeting CTLA-4 and TIGIT, the interaction with FcγR on APCs enhanced antigen-specific T cell responses and tumoricidal activity. This mechanism extended to an anti-CD45RB mAb, which led to FcγR-dependent regulatory T cell expansion in mice.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antigen-Presenting Cells/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Protein Binding , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, IgG/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
3.
PLoS One ; 13(4): e0191926, 2018.
Article in English | MEDLINE | ID: mdl-29617360

ABSTRACT

CTLA-4 and CD28 exemplify a co-inhibitory and co-stimulatory signaling axis that dynamically sculpts the interaction of antigen-specific T cells with antigen-presenting cells. Anti-CTLA-4 antibodies enhance tumor-specific immunity through a variety of mechanisms including: blockade of CD80 or CD86 binding to CTLA-4, repressing regulatory T cell function and selective elimination of intratumoral regulatory T cells via an Fcγ receptor-dependent mechanism. AGEN1884 is a novel IgG1 antibody targeting CTLA-4. It potently enhanced antigen-specific T cell responsiveness that could be potentiated in combination with other immunomodulatory antibodies. AGEN1884 was well-tolerated in non-human primates and enhanced vaccine-mediated antigen-specific immunity. AGEN1884 combined effectively with PD-1 blockade to elicit a T cell proliferative response in the periphery. Interestingly, an IgG2 variant of AGEN1884 revealed distinct functional differences that may have implications for optimal dosing regimens in patients. Taken together, the pharmacological properties of AGEN1884 support its clinical investigation as a single therapeutic and combination agent.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CTLA-4 Antigen/immunology , Immunoglobulin G/pharmacology , Neoplasms/therapy , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/toxicity , Amino Acid Sequence , Animals , Antibody Formation/drug effects , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/toxicity , CHO Cells , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/pharmacology , Cells, Cultured , Cricetulus , Epitope Mapping , Humans , Immunity, Cellular/drug effects , Immunoglobulin G/chemistry , Immunoglobulin G/toxicity , Lymphocyte Activation/drug effects , Macaca fascicularis , Models, Molecular , Neoplasms/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
4.
Hum Antibodies ; 25(3-4): 87-109, 2017.
Article in English | MEDLINE | ID: mdl-28085016

ABSTRACT

Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy/methods , Neoplasms/drug therapy , Receptors, IgG/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Gene Expression Regulation , Humans , Immunity, Cellular/drug effects , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, IgG/agonists , Receptors, IgG/genetics , Receptors, Tumor Necrosis Factor/agonists , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
J Immunol ; 194(3): 878-82, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548231

ABSTRACT

CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.


Subject(s)
Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigens, Surface/metabolism , Cell Line, Tumor , CpG Islands , DNA Methylation , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Humans , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Transgenic , Neoplasms/metabolism , Transforming Growth Factor beta/metabolism
7.
Immunol Cell Biol ; 92(6): 475-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24732076

ABSTRACT

Antibodies targeting checkpoint inhibitors or co-stimulatory receptors on T cells have shown significant antitumor efficacy in preclinical and clinical studies. In mouse tumor models, engagement of activating Fcγ receptor (FcγR)-expressing immune cells was recently shown to be required for the tumoricidal activity of antibodies recognizing the tumor necrosis factor superfamily receptor (TNFR) GITR (CD357) and CTLA-4 (CD152). In particular, activating FcγRs facilitated the selective elimination of intratumoral T-cell populations. However, it remains unclear whether FcγRs contribute to the antitumor efficacy of other immunomodulatory antibodies. Here, we explored the mechanism of antitumor activity mediated by an agonistic antibody (clone OX86) to the co-stimulatory TNFR OX40 (CD134). OX40 was highly expressed by intratumoral T cells, particularly those of the FoxP3(+) regulatory T-cell (Treg) lineage. OX86 administration resulted in the depletion of intratumoral regulatory T cells in an activating FcγR-dependent manner, which correlated with tumor regression. Together with previous data from our group and others, these findings support a mechanism whereby antibodies targeting antigens highly expressed by intratumoral T cells can mediate their elimination by FcγR-expressing immune cells, and facilitate subsequent antitumor immunity.


Subject(s)
Neoplasms, Experimental/immunology , Receptors, IgG/immunology , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , Lymphocyte Depletion , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Receptors, IgG/genetics , Receptors, OX40/genetics , T-Lymphocytes, Regulatory/pathology
8.
J Immunol ; 192(7): 3259-68, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24610009

ABSTRACT

Adjuvants are an essential component of modern vaccines and used for their ability to elicit immunity to coadministered Ags. Many adjuvants in clinical development are particulates, but how they drive innate and adaptive immune responses remains poorly understood. Studies have shown that a number of vaccine adjuvants activate inflammasome pathways in isolated APCs. However, the contribution of inflammasome activation to vaccine-mediated immunity in vivo remains controversial. In this study, we evaluated immune cell responses to the ISCOMATRIX adjuvant (IMX) in mice. Like other particulate vaccine adjuvants, IMX potently activated the NALP-3-ASC-Caspase-1 inflammasome in APCs, leading to IL-1ß and IL-18 production. The IL-18R pathway, but not IL-1R, was required for early innate and subsequent cellular immune responses to a model IMX vaccine. APCs directly exposed to IMX underwent an endosome-mediated cell-death response, which we propose initiates inflammatory events locally at the injection site. Importantly, both inflammasome-related and -unrelated pathways contributed to IL-18 dependence in vivo following IMX administration. TNF-α provided a physiological priming signal for inflammasome-dependent IL-18 production by APCs, which correlated with reduced vaccine-mediated immune cell responses in TNF-α- or TNFR-deficient mice. Taken together, our findings highlight an important disconnect between the mechanisms of vaccine adjuvant action in vitro versus in vivo.


Subject(s)
Cholesterol/immunology , Immunity/immunology , Inflammasomes/immunology , Interleukin-18/immunology , Phospholipids/immunology , Saponins/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Blotting, Western , Cell Survival/drug effects , Cell Survival/immunology , Cholesterol/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Drug Combinations , Humans , Immunity/drug effects , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lysosomes/drug effects , Lysosomes/immunology , Lysosomes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phospholipids/pharmacology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Saponins/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Immunol Cell Biol ; 91(7): 451-60, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817579

ABSTRACT

Toll-like receptors (TLRs) enable metazoans to mount effective innate immune responses to microbial and viral pathogens, as well as to endogenous host-derived ligands. It is understood that genetic background of the host can influence TLR responsiveness, altering susceptibility to pathogen infection, autoimmunity and cancer. Macrophage stimulatory protein (MSP), which activates the receptor tyrosine kinase recepteur d'origine nantais (RON), promotes key macrophage functions such as motility and phagocytic activity. MSP also acts via RON to modulate signaling by TLR4, which recognizes a range of pathogen or endogenous host-derived molecules. Here, we show that RON exerts divergent control over TLR4 activity in macrophages from different mouse genetic backgrounds. RON potently modulated the TLR4 response in macrophages from M2-prone FVB mice, as compared with M1-skewed C57Bl6 mice. Moreover, global expression analysis revealed that RON suppresses the TLR4-dependent type-I interferon gene signature only in FVB macrophages. This leads to attenuated production of the potent inflammatory mediator, tumor necrosis factor-α. Eliminating RON kinase activity markedly decreased carcinogen-mediated tumorigenesis in M2/Th2-biased FVB mice. We propose that host genetic background influences RON function, thereby contributing to the variability in TLR4 responsiveness in rodents and, potentially, in humans. These findings provide novel insight into the complex interplay between genetic context and immune function.


Subject(s)
Fibrosarcoma/immunology , Macrophages, Peritoneal/immunology , Papilloma/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/immunology , Toll-Like Receptor 4/immunology , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , Animals , Carcinogenesis , Cell Movement/drug effects , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Genotype , Hepatocyte Growth Factor/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Methylcholanthrene/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Papilloma/chemically induced , Papilloma/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Th1-Th2 Balance , Transcriptional Activation/genetics , Transcriptome
10.
J Exp Med ; 210(9): 1685-93, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23897982

ABSTRACT

Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Immunologic Factors/pharmacology , Neoplasms/immunology , Receptors, IgG/metabolism , Animals , CTLA-4 Antigen/metabolism , Female , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Neoplasms/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
11.
Cancer Cell ; 22(1): 80-90, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22789540

ABSTRACT

The proapoptotic death receptor DR5 has been studied extensively in cancer cells, but its action in the tumor microenvironment is not well defined. Here, we uncover a role for DR5 signaling in tumor endothelial cells (ECs). We detected DR5 expression in ECs within tumors but not normal tissues. Treatment of tumor-bearing mice with an oligomeric form of the DR5 ligand Apo2L/TRAIL induced apoptosis in tumor ECs, collapsing blood vessels and reducing tumor growth: Vascular disruption and antitumor activity required DR5 expression on tumor ECs but not malignant cells. These results establish a therapeutic paradigm for proapoptotic receptor agonists as selective tumor vascular disruption agents, providing an alternative, perhaps complementary, strategy to their use as activators of apoptosis in malignant cells.


Subject(s)
Apoptosis , Cell Division , Endothelium, Vascular/metabolism , Neoplasms/blood supply , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Humans , Mice , Neoplasms/pathology
12.
Immunol Cell Biol ; 90(5): 540-52, 2012 May.
Article in English | MEDLINE | ID: mdl-21894173

ABSTRACT

Generating a cytotoxic CD8(+) T-cell response that can eradicate malignant cells is the primary objective of cancer vaccine strategies. In this study we have characterized the innate and adaptive immune response to the ISCOMATRIX adjuvant, and the ability of vaccine antigens formulated with this adjuvant to promote antitumor immunity. ISCOMATRIX adjuvant led to a rapid innate immune cell response at the injection site, followed by the activation of natural killer and dendritic cells (DC) in regional draining lymph nodes. Strikingly, major histocompatibility complex (MHC) class I cross-presentation by CD8α(+) and CD8α(-) DCs was enhanced by up to 100-fold when antigen was formulated with ISCOMATRIX adjuvant. These coordinated features enabled efficient CD8(+) T-cell cross-priming, which exhibited prophylactic and therapeutic tumoricidal activity. The therapeutic efficacy of an ISCOMATRIX vaccine was further improved when co-administered with an anti-CD40 agonist antibody, suggesting that ISCOMATRIX-based vaccines may combine favorably with other immune modifiers in clinical development to treat cancer. Finally, we identified a requirement for the myeloid differentiation primary response gene 88 (MyD88) adapter protein for both innate and adaptive immune responses to ISCOMATRIX vaccines in vivo. Taken together, our findings support the utility of the ISCOMATRIX adjuvant for use in the development of novel vaccines, particularly those requiring strong CD8(+) T-cell immune responses, such as therapeutic cancer vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cholesterol/immunology , Phospholipids/immunology , Saponins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Neoplasm/immunology , CD40 Antigens/immunology , CD8-Positive T-Lymphocytes/drug effects , Cancer Vaccines/administration & dosage , Cholesterol/administration & dosage , Cross-Priming/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Combinations , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Ovalbumin/immunology , Phospholipids/administration & dosage , Receptor Cross-Talk/drug effects , Saponins/administration & dosage , Signal Transduction/drug effects
13.
Cancer Cell ; 19(1): 101-13, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21251615

ABSTRACT

Antibodies to cell-surface antigens trigger activatory Fcγ receptor (FcγR)-mediated retrograde signals in leukocytes to control immune effector functions. Here, we uncover an FcγR mechanism that drives antibody-dependent forward signaling in target cells. Agonistic antibodies to death receptor 5 (DR5) induce cancer-cell apoptosis and are in clinical trials; however, their mechanism of action in vivo is not fully defined. Interaction of the DR5-agonistic antibody drozitumab with leukocyte FcγRs promoted DR5-mediated tumor-cell apoptosis. Whereas the anti-CD20 antibody rituximab required activatory FcγRs for tumoricidal function, drozitumab was effective in the context of either activatory or inhibitory FcγRs. A CD40-agonistic antibody required similar FcγR interactions to stimulate nuclear factor-κB activity in B cells. Thus, FcγRs can drive antibody-mediated receptor signaling in target cells.


Subject(s)
Antibodies, Monoclonal/metabolism , Neoplasms/metabolism , Receptors, IgG/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Apoptosis/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , CD40 Antigens/agonists , CD40 Antigens/immunology , Cell Line, Tumor , Female , HCT116 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Killer Cells, Natural/immunology , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Mutation/immunology , Myeloid Cells/immunology , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/immunology , Protein Binding/genetics , Protein Binding/immunology , Receptor Aggregation/immunology , Receptors, IgG/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Xenograft Model Antitumor Assays
14.
Curr Opin Cell Biol ; 22(6): 837-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20813513

ABSTRACT

Proapoptotic receptor agonists (PARAs) targeting death receptors (DRs) 4 and 5 hold promise for cancer therapy based on their selective ability to kill malignant versus healthy cells. Emerging clinical results have confirmed that DR4/5 PARAs are relatively well-tolerated and suitable for further investigation. Given that some cancer cell lines and models are not sensitive to PARAs, it is important to develop strategies to identify what specific types of tumor cells may be most responsive to PARA-based therapy and how to overcome apoptosis resistance mechanisms in tumors. Here we review the molecular and biological determinants of responsiveness to PARAs in cancer cells, and discuss the potential for predictive biomarkers and drug combination strategies to maximize the anti-tumor activity of these agents.


Subject(s)
Apoptosis/physiology , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/physiology , Animals , Cell Membrane/metabolism , Humans , Mitochondria/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism
15.
Nat Immunol ; 10(4): 348-55, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295631

ABSTRACT

Death receptors (DRs) are members of the tumor necrosis factor receptor superfamily that possess a cytoplasmic death domain (DD). DRs regulate important operational and homeostatic aspects of the immune system. They transmit signals through apical protein complexes, which are nucleated by the DD adaptors FADD and TRADD, to control cellular outcomes that range from apoptosis to gene activation. FADD and TRADD also nucleate several distal signaling complexes, which mediate cross-talk between distinct DR signaling pathways. Moreover, together with other DR signal transducers, FADD and TRADD participate in functional complexes assembled by certain non-DR immune cell receptors, such as pattern-recognition receptors. Thus, DR signal transducers may provide important nodes of coordination in immune signaling networks.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Fas-Associated Death Domain Protein/physiology , Receptors, Death Domain/physiology , Signal Transduction/physiology , TNF Receptor-Associated Death Domain Protein/physiology , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis/physiology , Fas-Associated Death Domain Protein/immunology , Humans , Immunity, Active , Immunity, Innate , Receptors, Death Domain/immunology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , TNF Receptor-Associated Death Domain Protein/immunology
16.
Nat Immunol ; 9(11): 1244-52, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849989

ABSTRACT

The importance of conventional dendritic cells (cDCs) in the processing and presentation of antigen is well established, but the contribution of plasmacytoid dendritic cells (pDCs) to these processes, and hence to T cell immunity, remains unclear. Here we showed that unlike cDCs, pDCs continued to synthesize major histocompatibility complex (MHC) class II molecules and the MHC class II ubiquitin ligase MARCH1 long after activation. Sustained MHC class II-peptide complex formation, ubiquitination and turnover rendered pDCs inefficient in the presentation of exogenous antigens but enabled pDCs to continuously present endogenous viral antigens in their activated state. As the antigen-presenting abilities of cDCs and pDCs are fundamentally distinct, these two cell types may activate largely nonoverlapping repertoires of CD4(+) T cells.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Histocompatibility Antigens Class II/metabolism , Ubiquitination , Animals , Antigens, Viral/immunology , CD11 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Histocompatibility Antigens Class II/biosynthesis , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , Mice , Mice, Inbred Strains , Mice, Knockout , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
17.
Immunol Cell Biol ; 86(4): 333-42, 2008.
Article in English | MEDLINE | ID: mdl-18347609

ABSTRACT

Protective immunity against viral pathogens depends on the generation and maintenance of a small population of memory CD8(+) T cells. Successful memory cell generation begins with early interactions between naïve T cell and dendritic cells (DCs) within the inflammatory milieu of the secondary lymphoid tissues. Recent insights into the role of different populations of DCs, and kinetics of antigen presentation, during viral infections have helped to understand how DCs can shape the immune response. Here, we review the recent progress that has been made towards defining how specific DC subsets drive effector CD8(+) T-cell expansion and differentiation into memory cells. Further, we endeavour to examine how the molecular signals imparted by DCs coordinate to generate protective CD8(+) T-cell immunity.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Inflammation/virology
18.
Immunol Cell Biol ; 86(2): 200-5, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18026177

ABSTRACT

Dendritic cells (DCs) play major roles in immunosurveillance. In peripheral tissues, 'immature' DCs are dedicated to capturing antigens. Detection of pathogens through Toll-like receptors (TLRs) triggers DC migration to the lymph nodes (LNs), where they acquire a 'mature' phenotype specialized at presenting antigens. However, DCs migrate from tissues and mature even in the absence of overt infections. This has been attributed to detection of commensal flora in the skin, the gut or other peripheral tissues in the steady state. To test this assumption, we have analyzed the DCs contained in the lymphoid organs of germ-free mice and of mice lacking the TLR adapter molecules, MyD88 and TRIF. We show that the proportion and expression of maturation markers in DC immigrants in the LNs of these mice are similar to those in normal mice. These results suggest that DC migration from tissues, followed by their phenotypic maturation, is regulated in the steady state by an inherent program of DC differentiation or by the release of low levels of inflammatory signals from normal tissues.


Subject(s)
Dendritic Cells/physiology , Toll-Like Receptors/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Bacteria/immunology , Cell Movement , Dendritic Cells/immunology , Dendritic Cells/microbiology , Germ-Free Life , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Toll-Like Receptors/metabolism
19.
Proc Natl Acad Sci U S A ; 104(45): 17753-8, 2007 Nov 06.
Article in English | MEDLINE | ID: mdl-17978177

ABSTRACT

When dendritic cells (DCs) encounter signals associated with infection or inflammation, they become activated and undergo maturation. Mature DCs are very efficient at presenting antigens captured in association with their activating signal but fail to present subsequently encountered antigens, at least in vitro. Such impairment of MHC class II (MHC II) antigen presentation has generally been thought to be a consequence of down-regulation of endocytosis, so it might be expected that antigens synthesized by the DCs themselves (for instance, viral antigens) would still be presented by mature DCs. Here, we show that DCs matured in vivo could still capture and process soluble antigens, but were unable to present peptides derived from these antigens. Furthermore, presentation of viral antigens synthesized by the DCs themselves was also severely impaired. Indeed, i.v. injection of pathogen mimics, which caused systemic DC activation in vivo, impaired the induction of CD4 T cell responses against subsequently encountered protein antigens. This immunosuppressed state could be reversed by adoptive transfer of DCs loaded exogenously with antigens, demonstrating that impairment of CD4 T cell responses was due to lack of antigen presentation rather than to overt suppression of T cell activation. The biochemical mechanism underlying this phenomenon was the down-regulation of MHC II-peptide complex formation that accompanied DC maturation. These observations have important implications for the design of prophylactic and therapeutic DC vaccines and contribute to the understanding of the mechanisms causing immunosuppression during systemic blood infections.


Subject(s)
Antigens, Viral/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Viral Vaccines/immunology , Animals , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/virology , Flow Cytometry , Mice , Muramidase/immunology , Ovalbumin/immunology
20.
J Immunol ; 178(4): 2094-103, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277113

ABSTRACT

Peripheral tolerance is required to prevent autoimmune tissue destruction by self-reactive T cells that escape negative selection in the thymus. One mechanism of peripheral tolerance in CD8(+) T cells is their activation by resting dendritic cells (DC). In contrast, DC can be "licensed" by CD4(+) T cells to induce cytotoxic function in CD8(+) T cells. The question that then arises, whether CD4(+) T cell help could impair peripheral tolerance induction in self-reactive CD8(+) T cells, has not been addressed. In this study we show that CD4(+) T cell activation by resting DC results in helper function that transiently promotes the expansion and differentiation of cognate CD8(+) T cells. However, both the CD4(+) and CD8(+) T cell populations ultimately undergo partial deletion and acquire Ag unresponsiveness, disabling their ability to destroy OVA-expressing pancreatic beta cells and cause diabetes. Thus, effective peripheral tolerance can be induced by resting DC in the presence of CD4(+) and CD8(+) T cells with specificity for the same Ag.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Deletion/immunology , Immune Tolerance , Insulin-Secreting Cells/immunology , Lymphocyte Activation/immunology , Animals , Antigens/genetics , Antigens/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Gene Expression Regulation/immunology , Mice , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...