Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 28(11): 2270-2277, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35344029

ABSTRACT

PURPOSE: FGFR genomic alterations (amplification, mutations, and/or fusions) occur in ∼8% of gliomas, particularly FGFR1 and FGFR3. We conducted a multicenter open-label, single-arm, phase II study of a selective FGFR1-3 inhibitor, infigratinib (BGJ398), in patients with FGFR-altered recurrent gliomas. PATIENTS AND METHODS: Adults with recurrent/progressive gliomas harboring FGFR alterations received oral infigratinib 125 mg on days 1 to 21 of 28-day cycles. The primary endpoint was investigator-assessed 6-month progression-free survival (PFS) rate by Response Assessment in Neuro-Oncology criteria. Comprehensive genomic profiling was performed on available pretreatment archival tissue to explore additional molecular correlations with efficacy. RESULTS: Among 26 patients, the 6-month PFS rate was 16.0% [95% confidence interval (CI), 5.0-32.5], median PFS was 1.7 months (95% CI, 1.1-2.8), and objective response rate was 3.8%. However, 4 patients had durable disease control lasting longer than 1 year. Among these, 3 had tumors harboring activating point mutations at analogous positions of FGFR1 (K656E; n = 2) or FGFR3 (K650E; n = 1) in pretreatment tissue; an FGFR3-TACC3 fusion was detected in the other. Hyperphosphatemia was the most frequently reported treatment-related adverse event (all-grade, 76.9%; grade 3, 3.8%) and is a known on-target toxicity of FGFR inhibitors. CONCLUSIONS: FGFR inhibitor monotherapy with infigratinib had limited efficacy in a population of patients with recurrent gliomas and different FGFR genetic alterations, but durable disease control lasting more than 1 year was observed in patients with tumors harboring FGFR1 or FGFR3 point mutations or FGFR3-TACC3 fusions. A follow-up study with refined biomarker inclusion criteria and centralized FGFR testing is warranted.


Subject(s)
Glioma , Neoplasm Recurrence, Local , Adult , Follow-Up Studies , Glioma/drug therapy , Glioma/genetics , Humans , Microtubule-Associated Proteins , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Phenylurea Compounds , Protein Kinase Inhibitors/adverse effects , Pyrimidines , Receptor, Fibroblast Growth Factor, Type 3/genetics
2.
Am J Dermatopathol ; 38(3): 201-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26894771

ABSTRACT

Melanocyte differentiation antigens, such as gp100, tyrosinase, and Melan-A and their corresponding antibodies HMB45, T311, and A103, are major diagnostic tools in surgical pathology. Little is known about tyrosinase-related protein 2 (TRP-2, or dopachrome tautomerase/DCT) another melanocyte differentiation antigen, which is an enzymatic component of melanogenesis. We identified a commercial reagent to TRP-2, monoclonal antibody (mAb) C-9 and undertook a comprehensive analysis to assess its specificity and usefulness for surgical pathology. Subsequently, we analyzed panels of normal tissues and tumors. We show that TRP-2 is regularly expressed in melanocytes of the normal skin. In cutaneous nevi, TRP-2 is present in junctional as well as in dermal nevocytes. In malignant tumors, C-9 reactivity is restricted to melanocytic and related lesions and present in 84% and 58% of primary and metastatic melanomas, respectively. Ten primary melanomas of the anorectal mucosa were all positive. Like the other melanocyte differentiation antigens, TRP-2 was absent in 6 desmoplastic melanomas. Also, only 2 of 9 angiomyolipomas were TRP-2 positive. We conclude that mAb C-9 is a valuable reagent for the analysis of TRP-2 expression in archival surgical pathology material. The expression pattern of TRP-2 in melanocytic and related lesions appears to parallel other melanocyte differentiation antigens, although the overall incidence is lower than other antigens, such as Melan-A or gp100.


Subject(s)
Biomarkers, Tumor/analysis , Intramolecular Oxidoreductases/biosynthesis , Melanocytes/metabolism , Skin/metabolism , Antibodies, Monoclonal , Antibody Specificity , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Melanoma/metabolism , Nevus/metabolism , Real-Time Polymerase Chain Reaction , Skin Neoplasms/metabolism , Transcriptome
4.
Cancer Immunol Res ; 2(5): 448-58, 2014 May.
Article in English | MEDLINE | ID: mdl-24795357

ABSTRACT

Induction of potent immune responses to self-antigens remains a major challenge in tumor immunology. We have shown that a vaccine based on alphavirus replicon particles (VRP) activates strong cellular and humoral immunity to tyrosinase-related protein-2 (TRP2) melanoma antigen, providing prophylactic and therapeutic effects in stringent mouse models. Here, we report that the immunogenicity and efficacy of this vaccine is increased in combination with either antagonist anti-CTL antigen-4 (CTLA-4) or agonist anti-glucocorticoid-induced TNF family-related gene (GITR) immunomodulatory monoclonal antibodies (mAb). In the challenging therapeutic setting, VRP-TRP2 plus anti-GITR or anti-CTLA-4 mAb induced complete tumor regression in 90% and 50% of mice, respectively. These mAbs had similar adjuvant effects in priming an adaptive immune response against the vaccine-encoded antigen, augmenting, respectively, approximately 4- and 2-fold the TRP2-specific CD8(+) T-cell response and circulating Abs, compared with the vaccine alone. Furthermore, while both mAbs increased the frequency of tumor-infiltrating CD8(+) T cells, anti-CTLA-4 mAb also increased the quantity of intratumor CD4(+)Foxp3(-) T cells expressing the negative costimulatory molecule programmed death-1 (PD-1). Concurrent GITR expression on these cells suggests that they might be controlled by anti-GITR mAbs, thus potentially explaining their differential accumulation under the two treatment conditions. These findings indicate that combining immunomodulatory mAbs with alphavirus-based anticancer vaccines can provide therapeutic antitumor immune responses in a stringent mouse model, suggesting potential utility in clinical trials. They also indicate that tumor-infiltrating CD4(+)Foxp3(-)PD-1(+) T cells may affect the outcome of immunomodulatory treatments.


Subject(s)
Alphavirus/genetics , Antibodies, Monoclonal/pharmacology , Immunologic Factors/pharmacology , Melanoma, Experimental/immunology , Vaccines, Virus-Like Particle/immunology , Animals , CTLA-4 Antigen/antagonists & inhibitors , Disease Models, Animal , Female , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Immunoglobulin G/immunology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/therapy , Mice , Phenotype , Replicon , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Vaccines, Virus-Like Particle/administration & dosage
5.
PLoS Pathog ; 10(4): e1003989, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743339

ABSTRACT

Modified vaccinia virus Ankara (MVA) is an attenuated poxvirus that has been engineered as a vaccine against infectious agents and cancers. Our goal is to understand how MVA modulates innate immunity in dendritic cells (DCs), which can provide insights to vaccine design. In this study, using murine bone marrow-derived dendritic cells, we assessed type I interferon (IFN) gene induction and protein secretion in response to MVA infection. We report that MVA infection elicits the production of type I IFN in murine conventional dendritic cells (cDCs), but not in plasmacytoid dendritic cells (pDCs). Transcription factors IRF3 (IFN regulatory factor 3) and IRF7, and the positive feedback loop mediated by IFNAR1 (IFN alpha/beta receptor 1), are required for the induction. MVA induction of type I IFN is fully dependent on STING (stimulator of IFN genes) and the newly discovered cytosolic DNA sensor cGAS (cyclic guanosine monophosphate-adenosine monophosphate synthase). MVA infection of cDCs triggers phosphorylation of TBK1 (Tank-binding kinase 1) and IRF3, which is abolished in the absence of cGAS and STING. Furthermore, intravenous delivery of MVA induces type I IFN in wild-type mice, but not in mice lacking STING or IRF3. Treatment of cDCs with inhibitors of endosomal and lysosomal acidification or the lysosomal enzyme Cathepsin B attenuated MVA-induced type I IFN production, indicating that lysosomal enzymatic processing of virions is important for MVA sensing. Taken together, our results demonstrate a critical role of the cGAS/STING-mediated cytosolic DNA-sensing pathway for type I IFN induction in cDCs by MVA. We present evidence that vaccinia virulence factors E3 and N1 inhibit the activation of IRF3 and the induction of IFNB gene in MVA-infected cDCs.


Subject(s)
Bone Marrow Cells/metabolism , Dendritic Cells/metabolism , Interferon-beta/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Vaccinia virus/metabolism , Vaccinia/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/virology , Dendritic Cells/immunology , Dendritic Cells/virology , Endosomes/genetics , Endosomes/immunology , Endosomes/metabolism , Female , Immunity, Innate/genetics , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Lysosomes/genetics , Lysosomes/immunology , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Phosphorylation/genetics , Phosphorylation/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Vaccinia/genetics , Vaccinia/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/metabolism
6.
Blood ; 119(22): 5182-90, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22510877

ABSTRACT

Human CD34(+) progenitor-derived Langerhans-type dendritic cells (LCs) are more potent stimulators of T-cell immunity against tumor and viral antigens in vitro than are monocyte-derived DCs (moDCs). The exact mechanisms have remained elusive until now, however. LCs synthesize the highest amounts of IL-15R-α mRNA and protein, which binds IL-15 for presentation to responder lymphocytes, thereby signaling the phosphorylation of signal transducer and activator of transcription 5 (pSTAT5). LCs electroporated with Wilms tumor 1 (WT1) mRNA achieve sufficiently sustained presentation of antigenic peptides, which together with IL-15R-α/IL-15, break tolerance against WT1 by stimulating robust autologous, WT1-specific cytolytic T-lymphocytes (CTLs). These CTLs develop from healthy persons after only 7 days' stimulation without exogenous cytokines and lyse MHC-restricted tumor targets, which include primary WT1(+) leukemic blasts. In contrast, moDCs require exogenous rhuIL-15 to phosphorylate STAT5 and attain stimulatory capacity comparable to LCs. LCs therefore provide a more potent costimulatory cytokine milieu for T-cell activation than do moDCs, thus accounting for their superior stimulation of MHC-restricted Ag-specific CTLs without need for exogenous cytokines. These data support the use of mRNA-electroporated LCs, or moDCs supplemented with exogenous rhuIL-15, as vaccines for cancer immunotherapy to break tolerance against self-differentiation antigens shared by tumors.


Subject(s)
Antigen Presentation , Immune Tolerance , Interleukin-15/immunology , Langerhans Cells/immunology , Receptors, Interleukin-15/immunology , STAT5 Transcription Factor/immunology , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/immunology , Blast Crisis/genetics , Blast Crisis/immunology , Blast Crisis/pathology , Blast Crisis/therapy , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Female , Humans , Interleukin-15/pharmacology , Langerhans Cells/pathology , Leukemia/genetics , Leukemia/immunology , Leukemia/pathology , Leukemia/therapy , Lymphocyte Activation/drug effects , Male , Receptors, Interleukin-15/genetics , STAT5 Transcription Factor/genetics , T-Lymphocytes, Cytotoxic/pathology , WT1 Proteins/genetics
8.
Cancer Res ; 72(4): 876-86, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22174368

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that accumulate during tumor formation, facilitate immune escape, and enable tumor progression. MDSCs are important contributors to the development of an immunosuppressive tumor microenvironment that blocks the action of cytotoxic antitumor T effector cells. Heterogeneity in these cells poses a significant barrier to studying the in vivo contributions of individual MDSC subtypes. Herein, we show that granulocyte-macrophage colony stimulating factor, a cytokine critical for the numeric and functional development of MDSC populations, promotes expansion of a monocyte-derived MDSC population characterized by expression of CD11b and the chemokine receptor CCR2. Using a toxin-mediated ablation strategy to target CCR2-expressing cells, we show that these monocytic MDSCs regulate entry of activated CD8 T cells into the tumor site, thereby limiting the efficacy of immunotherapy. Our results argue that therapeutic targeting of monocytic MDSCs would enhance outcomes in immunotherapy.


Subject(s)
Immune Tolerance , Melanoma/immunology , Myeloid Cells/immunology , Receptors, CCR2/metabolism , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , CD11b Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Granulocyte-Macrophage Colony-Stimulating Factor , Immune Tolerance/immunology , Lymphocyte Activation , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism
9.
J Virol ; 85(20): 10814-25, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21835795

ABSTRACT

Poxviruses are large DNA viruses that replicate in the cytoplasm of infected cells. Myxoma virus is a rabbit poxvirus that belongs to the Leporipoxvirus genus. It causes a lethal disease called myxomatosis in European rabbits but cannot sustain any detectable infection in nonlagomorphs. Vaccinia virus is a prototypal orthopoxvirus that was used as a vaccine to eradicate smallpox. Myxoma virus is nonpathogenic in mice, whereas systemic infection with vaccinia virus can be lethal even in immunocompetent mice. Plasmacytoid dendritic cells (pDCs) are potent type I interferon (IFN)-producing cells that play important roles in antiviral innate immunity. How poxviruses are sensed by pDCs to induce type I IFN production is not well understood. Here we report that infection of primary murine pDCs with myxoma virus, but not with vaccinia virus, induces IFN-α, IFN-ß, tumor necrosis factor (TNF), and interleukin-12p70 (IL-12p70) production. Using pDCs derived from genetic knockout mice, we show that the myxoma virus-induced innate immune response requires the endosomal DNA sensor TLR9 and its adaptor MyD88, transcription factors IRF5 and IRF7, and the type I IFN positive-feedback loop mediated by IFNAR1. It is independent of the cytoplasmic RNA sensing pathway mediated by the mitochondrial adaptor molecule MAVS, the TLR3 adaptor TRIF, or the transcription factor IRF3. Using pharmacological inhibitors, we demonstrate that myxoma virus-induced type I IFN and IL-12p70 production in murine pDCs is also dependent on phosphatidylinositol 3-kinase (PI3K) and Akt. Furthermore, our results reveal that the N-terminal Z-DNA/RNA binding domain of vaccinia virulence factor E3, which is missing in the orthologous M029 protein expressed by myxoma virus, plays an inhibitory role in poxvirus sensing and innate cytokine production by murine pDCs.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factors/immunology , Interferon Type I/metabolism , Myeloid Differentiation Factor 88/immunology , Myxoma virus/immunology , Toll-Like Receptor 9/immunology , Animals , Cells, Cultured , Female , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Toll-Like Receptor 9/metabolism , Vaccinia virus/immunology
10.
PLoS One ; 6(6): e21214, 2011.
Article in English | MEDLINE | ID: mdl-21731676

ABSTRACT

Determining how tumor immunity is regulated requires understanding the extent to which the anti-tumor immune response "functions" in vivo without therapeutic intervention. To better understand this question, we developed advanced multimodal reflectance confocal/two photon fluorescence intra-vital imaging techniques to use in combination with traditional ex vivo analysis of tumor specific T cells. By transferring small numbers of melanoma-specific CD8+ T cells (Pmel-1), in an attempt to mimic physiologic conditions, we found that B16 tumor growth alone was sufficient to induce naive Pmel-1 T cell proliferation and acquisition of effector phenotype. Tumor -primed Pmel-1 T cells, are capable of killing target cells in the periphery and secrete IFNγ, but are unable to mediate tumor regression. Within the tumor, Pmel-1 T cells have highly confined mobility, displaying long term interactions with tumor cells. In contrast, adoptively transferred non tumor-specific OT-I T cells show neither confined mobility, nor long term interaction with B16 tumor cells, suggesting that intra-tumor recognition of cognate self antigen by Pmel-1 T cells occurs during tumor growth. Together, these data indicate that lack of anti-tumor efficacy is not solely due to ignorance of self antigen in the tumor microenvironment but rather to active immunosuppressive influences preventing a protective immune response.


Subject(s)
Antigens, Neoplasm/immunology , Autoantigens/immunology , Disease Progression , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Microscopy, Confocal/methods , Adoptive Transfer , Animals , Cell Communication , Cell Proliferation , Epitopes/immunology , Kinetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology , Time Factors
11.
Curr Top Microbiol Immunol ; 344: 211-44, 2011.
Article in English | MEDLINE | ID: mdl-20563707

ABSTRACT

The rational manipulation of antigen-specific T cells to reignite a tumor-specific immune response in cancer patients is a challenge for cancer immunotherapy. Targeting coinhibitory and costimulatory T cell receptors with specific antibodies in cancer patients is an emerging approach to T cell manipulation, namely "immune modulation." Cytotoxic T-lymphocyte antigen-4 (CTLA-4) and glucocorticoid-induced tumor necrosis factor family receptor (GITR) are potential targets for immune modulation through anti-CTLA-4 blocking antibodies and anti-GITR agonistic antibodies, respectively. In this review, we first discuss preclinical findings key to the understanding of the mechanisms of action of these immunomodulatory antibodies and the preclinical evidence of antitumor activity which preceded translation into the clinic. We next describe the outcomes and immune related adverse effects associated with anti-CTLA-4 based clinical trials with particular emphasis on specific biomarkers used to elucidate the mechanisms of tumor immunity in patients. The experience with anti-CTLA-4 therapy and the durable clinical benefit observed provide proof of principle to effective antitumor immune modulation and the promise of future clinical immune modulatory antibodies.


Subject(s)
Antigens, CD/physiology , Immunotherapy , Neoplasms/therapy , Receptors, Nerve Growth Factor/agonists , Receptors, Tumor Necrosis Factor/agonists , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , CTLA-4 Antigen , Glucocorticoid-Induced TNFR-Related Protein , Humans , Ipilimumab , Lymphocyte Activation , Neoplasms/immunology
12.
PLoS One ; 5(9)2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20844763

ABSTRACT

BACKGROUND: Malignant melanoma is the deadliest form of skin cancer and is refractory to conventional chemotherapy and radiotherapy. Therefore alternative approaches to treat this disease, such as immunotherapy, are needed. Melanoma vaccine design has mainly focused on targeting CD8+ T cells. Activation of effector CD8+ T cells has been achieved in patients, but provided limited clinical benefit, due to immune-escape mechanisms established by advanced tumors. We have previously shown that alphavirus-based virus-like replicon particles (VRP) simultaneously activate strong cellular and humoral immunity against the weakly immunogenic melanoma differentiation antigen (MDA) tyrosinase. Here we further investigate the antitumor effect and the immune mechanisms of VRP encoding different MDAs. METHODOLOGY/PRINCIPAL FINDINGS: VRP encoding different MDAs were screened for their ability to prevent the growth of the B16 mouse transplantable melanoma. The immunologic mechanisms of efficacy were investigated for the most effective vaccine identified, focusing on CD8+ T cells and humoral responses. To this end, ex vivo immune assays and transgenic mice lacking specific immune effector functions were used. The studies identified a potent therapeutic VRP vaccine, encoding tyrosinase related protein 2 (TRP-2), which provided a durable anti-tumor effect. The efficacy of VRP-TRP2 relies on a novel immune mechanism of action requiring the activation of both IgG and CD8+ T cell effector responses, and depends on signaling through activating Fcγ receptors. CONCLUSIONS/SIGNIFICANCE: This study identifies a VRP-based vaccine able to elicit humoral immunity against TRP-2, which plays a role in melanoma immunotherapy and synergizes with tumor-specific CD8+ T cell responses. These findings will aid in the rational design of future immunotherapy clinical trials.


Subject(s)
Alphavirus/genetics , Immunity, Cellular , Immunity, Humoral , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/therapeutic use , Melanoma/therapy , Replicon , Alphavirus/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Female , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Immunotherapy , Intramolecular Oxidoreductases/immunology , Melanoma/immunology , Melanoma/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Sci Transl Med ; 2(44): 44ra57, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20702856

ABSTRACT

Antigen-presenting dendritic cells (DCs) trigger the activation of cytotoxic CD8 T cells that target and eliminate cells with the antigen on their surface. Although DCs usually pick up and process antigens themselves, they can also receive peptide antigens from other cells via gap junctions. We demonstrate here that infection with Salmonella can induce, in both human and murine melanoma cells, the up-regulation of connexin 43 (Cx43), a ubiquitous protein that forms gap junctions and that is normally lost during melanoma progression. Bacteria-treated melanoma cells can establish functional gap junctions with adjacent DCs. After bacterial infection, these gap junctions transferred preprocessed antigenic peptides from the tumor cells to the DCs, which then presented those peptides on their surface. These peptides activated cytotoxic T cells against the tumor antigen, which could control the growth of distant uninfected tumors. Melanoma cells in which Cx43 had been silenced, when infected in vivo with bacteria, failed to elicit a cytotoxic antitumor response, indicating that this Cx43 mechanism is the principal one used in vivo for the generation of antitumor responses. The Cx43-dependent cross-presentation pathway is more effective than standard protocols of DC loading (peptide, tumor lysates, or apoptotic bodies) for generating DC-based tumor vaccines that both inhibit existing tumors and prevent tumor establishment. In conclusion, we exploited an antimicrobial response present in tumor cells to activate cytotoxic CD8 T cells specific for tumor-generated peptides that could directly recognize and kill tumor cells.


Subject(s)
Antigens, Neoplasm/immunology , Cross-Priming/immunology , Gap Junctions/metabolism , Melanoma/immunology , Melanoma/pathology , Salmonella/immunology , Animals , Antigen Presentation/immunology , Connexin 43/metabolism , Dendritic Cells/immunology , Female , Fluorescent Dyes/metabolism , Humans , Interferon-gamma/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Phagocytosis , RNA, Small Interfering/metabolism , Salmonella Infections/immunology
14.
Blood ; 115(22): 4384-92, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20154220

ABSTRACT

Cyclophosphamide (CTX), a commonly used chemotherapeutic agent can enhance immune responses. The ability of CTX to promote the proliferation of effector T cells and abrogate the function of regulatory T cells (Tregs) has been described. In this study, we examined the effects of CTX treatment on dendritic cell (DC) subsets and the subsequent outcome on the effector and suppressive arms of adaptive immunity. In secondary lymphoid tissues, tissue-derived migratory DCs (migratory DCs), lymphoid tissue-resident DCs (resident DCs), and plasmacytoid DCs (pDCs) are well described. CTX has profound and selective cytotoxic effects on CD8(+) resident DCs, but not skin-derived migratory DCs or pDCs in lymph nodes (LNs) and spleen, causing an imbalance among these DC subsets. CTX treatment increases the potency of DCs in antigen presentation and cytokine secretion, and partially inhibits the suppressor activity of Tregs. Adoptive transfer of CD8(+) DCs can reconstitute this population in regional draining LNs and abrogate the immune-enhancing effects of CTX in vivo. These findings demonstrate that CTX may improve immune responses by preferentially depleting CD8(+) lymphoid-resident DCs, which leads to diminished Treg suppression and enhanced effector T-cell function in vivo.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cyclophosphamide/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Lymphoid Tissue/drug effects , Lymphoid Tissue/immunology , Adoptive Transfer , Animals , Antigen Presentation/drug effects , CD8 Antigens/metabolism , Dendritic Cells/classification , Female , In Vitro Techniques , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Lymphoid Tissue/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
15.
J Exp Med ; 206(5): 1103-16, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19414558

ABSTRACT

Expansion and recruitment of CD4(+) Foxp3(+) regulatory T (T reg) cells are mechanisms used by growing tumors to evade immune elimination. In addition to expansion of effector T cells, successful therapeutic interventions may require reduction of T reg cells within the tumor microenvironment. We report that the combined use of the alkylating agent cyclophosphamide (CTX) and an agonist antibody targeting the co-stimulatory receptor OX40 (OX86) provides potent antitumor immunity capable of regressing established, poorly immunogenic B16 melanoma tumors. CTX administration resulted in tumor antigen release, which after OX86 treatment significantly enhanced the antitumor T cell response. We demonstrated that T reg cells are an important cellular target of the combination therapy. Paradoxically, the combination therapy led to an expansion of T reg cells in the periphery. In the tumor, however, the combination therapy induced a profound T reg cell depletion that was accompanied by an influx of effector CD8(+) T cells leading to a favorable T effector/T reg cell ratio. Closer examination revealed that diminished intratumoral T reg cell levels resulted from hyperactivation and T reg cell-specific apoptosis. Thus, we propose that CTX and OX40 engagement represents a novel and rational chemoimmunotherapy.


Subject(s)
Melanoma, Experimental/drug therapy , Receptors, OX40/genetics , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/therapeutic use , Drug Therapy, Combination , Immunotherapy/methods , Lymphocyte Depletion , Melanoma, Experimental/immunology , Mice , T-Lymphocytes, Regulatory/drug effects
16.
Eur J Immunol ; 38(7): 1937-47, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18581324

ABSTRACT

Salmonella typhimurium is a facultative anaerobic bacterium able to multiply preferentially in tumors and inhibit their growth. The mechanisms through which Salmonella exerts its anti-cancer properties are not fully understood. We recently showed that intra-tumoral Salmonella injection results not only in the regression of even bulky tumor masses, but also impacts on the growth of distant untreated lesions. Here we describe how Salmonella exerts its systemic anti-cancer effects and means to potentiate them. The outburst of an early inflammatory reaction in the treated tumor promotes the development of an immunostimulatory cytokine environment both locally and in the draining lymph node. Within the next 10 days, an efficient cross-presentation of endogenous tumor antigens by dendritic cells at the tumor-draining lymph node leads to the priming of effective anti-tumor CD8+ T cell responses. This potentially broadly reactive T cell repertoire can be directed to other pre-established melanomas by low-dose radiotherapy enhancing the Salmonella anti-cancer effect. We demonstrate that Salmonella-based therapy coupled to low-dose radiotherapy dampens tumor immune escape mechanisms at different levels and allows controlling systemic disease in a CD8+ T cell-dependent manner.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Melanoma, Experimental/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Combined Modality Therapy , Cross-Priming , Cytokines/immunology , Dendritic Cells/metabolism , Female , Lymph Nodes/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/microbiology , Melanoma, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/microbiology
17.
Biochim Biophys Acta ; 1776(1): 108-23, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17720322

ABSTRACT

The concept of cancer immunotherapy stems from the proposed function of the immune system, called immunosurveillance, to protect against growing tumors. Due to genetic aberrations, tumor cells display an altered repertoire of MHC-associated peptides that can lead to the activation of immune cells able to eliminate the transformed cells. In some instances, under the pressure of the immune system, both the tumor and its microenvironment are shaped and immune-resistant tumor variants are selected initiating the process of cancer immunoediting. This can impair not only host-generated immunosurveillance, but also attempts to harness the immune response for therapeutic purposes, namely immunotherapy. Rather than being an exhaustive review of the different approaches of cancer immunotherapy, the focus of this review is to provide the reader with future challenges of the field by proposing 'second generation' immunotherapy approaches that take into account immunosubversive mechanisms adopted by tumor cells. After an introduction on the process of immunosurveillance and immunoescape we will analyze why current immunotherapy approaches have not fulfilled their promise and will finish by summarizing what are the challenges for future approaches.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy , Neoplasms/therapy , Tumor Escape , Animals , Antigen-Presenting Cells , Antigens, Neoplasm/immunology , Humans , Immunologic Surveillance , Neoplasms/immunology
18.
Cancer Res ; 65(9): 3920-7, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867392

ABSTRACT

A major obstacle for the development of effective immunotherapy is the ability of tumors to escape the immune system. The possibility to kill tumor cells because they are recognized as infected rather than as malignant could help overcome immune escape mechanisms. Here we report a conceptually new approach of cancer immunotherapy based on in vivo infection of tumors and killing of infected tumor cells. Attenuated but still invasive, Salmonella typhimurium can be successfully exploited to invade melanoma cells that can present antigenic determinants of bacterial origin and become targets for anti-Salmonella-specific T cells. However, to fully appreciate the anticancer therapeutic properties of S. typhimurium, tumor-bearing mice need to be vaccinated against S. typhimurium before intratumoral Salmonella injection. Tumor infection when coupled to anti-Salmonella vaccination leads to 50% to 100% tumor-free mice with a better outcome on larger tumors. Invasive Salmonella also exert an indirect toxic effect on tumor cells through the recruitment of inflammatory cells and the cross-presentation of tumor antigens, which allow induction of tumor-specific immune response. This is effective in retarding the growth of untreated established distant tumors and in protecting the mice from subsequent tumor challenges.


Subject(s)
Immunotherapy/methods , Melanoma/microbiology , Melanoma/therapy , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Antigens, Bacterial/immunology , Epitopes, T-Lymphocyte/immunology , Female , Humans , Melanoma/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Salmonella Vaccines/immunology , Salmonella Vaccines/pharmacology , T-Lymphocytes, Cytotoxic/immunology
19.
Nat Immunol ; 6(5): 507-14, 2005 May.
Article in English | MEDLINE | ID: mdl-15821737

ABSTRACT

The control of damaging inflammation by the mucosal immune system in response to commensal and harmful ingested bacteria is unknown. Here we show epithelial cells conditioned mucosal dendritic cells through the constitutive release of thymic stromal lymphopoietin and other mediators, resulting in the induction of 'noninflammatory' dendritic cells. Epithelial cell-conditioned dendritic cells released interleukins 10 and 6 but not interleukin 12, and they promoted the polarization of T cells toward a 'classical' noninflammatory T helper type 2 response, even after exposure to a T helper type 1-inducing pathogen. This control of immune responses seemed to be lost in patients with Crohn disease. Thus, the intimate interplay between intestinal epithelial cells and dendritic cells may help to maintain gut immune homeostasis.


Subject(s)
Dendritic Cells/immunology , Epithelial Cells/immunology , Homeostasis/immunology , Intestines/immunology , Cell Differentiation , Cells, Cultured , Colon/cytology , Colon/immunology , Cytokines/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-12/metabolism , Intestines/cytology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Thymic Stromal Lymphopoietin
SELECTION OF CITATIONS
SEARCH DETAIL
...