Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cancer Rep (Hoboken) ; 6(2): e1699, 2023 02.
Article in English | MEDLINE | ID: mdl-36806722

ABSTRACT

BACKGROUND: Elevated tissue factor (TF) expression, although restricted in normal tissue, has been reported in multiple solid cancers, and expression has been associated with poor prognosis. This manuscript compares TF expression across various solid tumor types via immunohistochemistry in a single study, which has not been performed previously. AIMS: To increase insight in the prevalence and cellular localization of TF expression across solid cancer types, we performed a detailed and systematic analysis of TF expression in tumor tissue obtained from patients with ovarian, esophageal, bladder, cervical, endometrial, pancreatic, prostate, colon, breast, non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), and glioblastoma. The spatial and temporal variation of TF expression was analyzed over time and upon disease progression in patient-matched biopsies taken at different timepoints. In addition, TF expression in patient-matched primary tumor and metastatic lesions was also analyzed. METHODS AND RESULTS: TF expression was detected via immunohistochemistry (IHC) using a validated TF-specific antibody. TF was expressed in all cancer types tested, with highest prevalence in pancreatic cancer, cervical cancer, colon cancer, glioblastoma, HNSCC, and NSCLC, and lowest in breast cancer. Staining was predominantly membranous in pancreatic, cervical, and HNSCC, and cytoplasmic in glioblastoma and bladder cancer. In general, expression was consistent between biopsies obtained from the same patient over time, although variability was observed for individual patients. NSCLC biopsies of primary tumor and matched lymph node metastases showed no clear difference in TF expression overall, although individual patient changes were observed. CONCLUSION: This study shows that TF is expressed across a broad range of solid cancer types, and expression is present upon tumor dissemination and over the course of treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Glioblastoma , Head and Neck Neoplasms , Lung Neoplasms , Male , Female , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Squamous Cell Carcinoma of Head and Neck , Thromboplastin/analysis , Lung Neoplasms/pathology
2.
EBioMedicine ; 52: 102625, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31981978

ABSTRACT

BACKGROUND: DuoBody®-CD3xCD20 (GEN3013) is a full-length human IgG1 bispecific antibody (bsAb) recognizing CD3 and CD20, generated by controlled Fab-arm exchange. Its Fc domain was silenced by introduction of mutations L234F L235E D265A. METHODS: T-cell activation and T-cell-mediated cytotoxicity were measured by flow cytometry following co-culture with tumour cells. Anti-tumour activity of DuoBody-CD3xCD20 was assessed in humanized mouse models in vivo. Non-clinical safety studies were performed in cynomolgus monkeys. FINDINGS: DuoBody-CD3xCD20 induced highly potent T-cell activation and T-cell-mediated cytotoxicity towards malignant B cells in vitro. Comparison of DuoBody-CD3xCD20 to CD3 bsAb targeting alternative B-cell antigens, or to CD3xCD20 bsAb generated using alternative CD20 Ab, emphasized its exceptional potency. In vitro comparison with other CD3xCD20 bsAb in clinical development showed that DuoBody-CD3xCD20 was significantly more potent than three other bsAb with single CD3 and CD20 binding regions and equally potent as a bsAb with a single CD3 and two CD20 binding regions. DuoBody-CD3xCD20 showed promising anti-tumour activity in vivo, also in the presence of excess levels of a CD20 Ab that competes for binding. In cynomolgus monkeys, DuoBody-CD3xCD20 demonstrated profound and long-lasting B-cell depletion from peripheral blood and lymphoid organs, which was comparable after subcutaneous and intravenous administration. Peak plasma levels of DuoBody-CD3xCD20 were lower and delayed after subcutaneous administration, which was associated with a reduction in plasma cytokine levels compared to intravenous administration, while bioavailability was comparable. INTERPRETATION: Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of subcutaneous DuoBody-CD3xCD20 in patients with B-cell malignancies. FUNDING: Genmab.


Subject(s)
Antibodies, Bispecific/immunology , Antigens, CD20/metabolism , CD3 Complex/metabolism , Cytotoxicity, Immunologic , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacology , Antibody Specificity/immunology , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/etiology , Leukemia, B-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/pathology , Macaca fascicularis , Mice , Mutation , Recombinant Proteins , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 18(2): 312-322, 2019 02.
Article in English | MEDLINE | ID: mdl-30381448

ABSTRACT

Immunotherapy of cancer with CD3-targeting bispecific antibodies (CD3 bsAb) is a fast developing field, and multiple tumor-associated antigens (TAA) are evaluated for hematologic and solid malignancies. The efficacy of these CD3 bsAb is usually examined in xenograft mouse tumor models with human T cells or in genetically engineered mouse models, where human TAA are introduced. These models often fail to fully recapitulate the natural tumor environment, especially for solid cancers, because of interspecies differences. Here, we investigated the systemic and intratumoral effects of a mouse CD3 bsAb in a fully immune-competent mouse melanoma model. Systemic administration of 0.5 mg/kg antibody induced a brief overall T-cell activation that was selectively sustained in the tumor microenvironment for several days. A fast subsequent influx of inflammatory macrophages into the tumor microenvironment was observed, followed by an increase in the number of CD4+ and CD8+ T cells. Although the capacity to directly kill melanoma cells in vitro was very modest, optimal tumor elimination was observed in vivo, even in the absence of CD8+ T cells, implying a redundancy in T-cell subsets for therapeutic efficacy. Finally, we took advantage of the full immune competence of our mouse model and tested immune memory induction. Despite a strong initial immunity against melanoma, treatment with the CD3 bsAb did not install protective memory responses. The observed mechanisms of action revealed in this immune-competent mouse model might form a rational basis for combinatorial approaches.


Subject(s)
Antibodies, Bispecific/administration & dosage , CD3 Complex/antagonists & inhibitors , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Female , Humans , Immunologic Memory , Lymphocyte Activation , Melanoma/immunology , Mice , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Res ; 74(4): 1214-26, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24371232

ABSTRACT

Tissue factor (TF) is aberrantly expressed in solid cancers and is thought to contribute to disease progression through its procoagulant activity and its capacity to induce intracellular signaling in complex with factor VIIa (FVIIa). To explore the possibility of using tissue factor as a target for an antibody-drug conjugate (ADC), a panel of human tissue factor-specific antibodies (TF HuMab) was generated. Three tissue factor HuMab, that induced efficient inhibition of TF:FVIIa-dependent intracellular signaling, antibody-dependent cell-mediated cytotoxicity, and rapid target internalization, but had minimal impact on tissue factor procoagulant activity in vitro, were conjugated with the cytotoxic agents monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF). Tissue factor-specific ADCs showed potent cytotoxicity in vitro and in vivo, which was dependent on tissue factor expression. TF-011-MMAE (HuMax-TF-ADC) was the most potent ADC, and the dominant mechanism of action in vivo was auristatin-mediated tumor cell killing. Importantly, TF-011-MMAE showed excellent antitumor activity in patient-derived xenograft (PDX) models with variable levels of tissue factor expression, derived from seven different solid cancers. Complete tumor regression was observed in all PDX models, including models that showed tissue factor expression in only 25% to 50% of the tumor cells. In conclusion, TF-011-MMAE is a promising novel antitumor agent with potent activity in xenograft models that represent the heterogeneity of human tumors, including heterogeneous target expression.


Subject(s)
Aminobenzoates/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Oligopeptides/therapeutic use , Thromboplastin/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Cells, Cultured , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Thromboplastin/antagonists & inhibitors , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 18(19): 5460-70, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22896657

ABSTRACT

PURPOSE: Electroporation of dendritic cells (DC) with mRNA encoding tumor-associated antigens (TAA) has multiple advantages compared to peptide loading. We investigated the immunologic and clinical responses to vaccination with mRNA-electroporated DC in stage III and IV melanoma patients. EXPERIMENTAL DESIGN: Twenty-six stage III HLA*02:01 melanoma patients scheduled for radical lymph node dissection (stage III) and 19 melanoma patients with irresectable locoregional or distant metastatic disease (referred to as stage IV) were included. Monocyte-derived DC, electroporated with mRNA encoding gp100 and tyrosinase, were pulsed with keyhole limpet hemocyanin and administered intranodally. TAA-specific T-cell responses were monitored in blood and skin-test infiltrating lymphocyte (SKIL) cultures. RESULTS: Comparable numbers of vaccine-induced CD8(+) and/or CD4(+) TAA-specific T-cell responses were detected in SKIL cultures; 17/26 stage III patients and 11/19 stage IV patients. Strikingly, in this population, TAA-specific CD8(+) T cells that recognize multiple epitopes and produce elevated levels of IFNγ upon antigenic challenge in vitro, were significantly more often observed in stage III patients; 15/17 versus 3/11 stage IV patients, P = 0.0033. In stage IV patients, one mixed and one partial response were documented. The presence or absence of IFNγ-producing TAA-specific CD8(+) T cells in stage IV patients was associated with marked difference in median overall survival of 24.1 months versus 11.0 months, respectively. CONCLUSION: Vaccination with mRNA-electroporated DC induces a broad repertoire of IFNγ producing TAA-specific CD8(+) and CD4(+) T-cell responses, particularly in stage III melanoma patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Immunotherapy , Melanoma , Adult , Aged , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Dendritic Cells/immunology , Electroporation , Female , Humans , Interferon-gamma/blood , Male , Melanoma/drug therapy , Melanoma/immunology , Melanoma/pathology , Middle Aged , Monophenol Monooxygenase/administration & dosage , Monophenol Monooxygenase/genetics , Neoplasm Metastasis , Neoplasm Staging , RNA, Messenger/administration & dosage , RNA, Messenger/immunology , gp100 Melanoma Antigen/administration & dosage , gp100 Melanoma Antigen/genetics
6.
PLoS One ; 7(6): e39706, 2012.
Article in English | MEDLINE | ID: mdl-22761877

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitors are widely used in the treatment of cancer. EGFR-targeted treatment is known to be associated with a high incidence of dermatological adverse reactions, including papulopustular rash, which can be dose-limiting and may affect compliance to treatment. Currently, the pathways involved in EGFR inhibitor-induced rash are poorly understood and few treatment options for this adverse event are available. Here, we developed a model for induction of papulopustular rash in healthy human volunteers by subcutaneous injection of the anti-EGFR monoclonal antibody zalutumumab. The injection sites and surrounding skin were evaluated by a dermatologist for the presence or absence of papulopustular rash and skin biopsies were taken to confirm the macroscopical findings by immunohistochemistry. Locally injected zalutumumab induced a papulopustular rash, characterized by acute follicular neutrophil-rich hair follicle inflammation, and thus mimicked adverse events induced by systemic administration of EGFR inhibitors. In this model, we tested the hypothesis that neutrophils, attracted by IL-8, play a central role in the observed rash. Indeed, concomitant local repeat dose treatment with HuMab-10F8, a neutralizing human antibody against IL-8, reduced the rash. Inhibition of IL-8 can therefore ameliorate dermatological adverse events induced by treatment with EGFR inhibitors.


Subject(s)
Antibodies, Monoclonal/adverse effects , ErbB Receptors/antagonists & inhibitors , Interleukin-8/immunology , Neutralization Tests , Skin Diseases/prevention & control , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Humans , Injections, Subcutaneous , Skin Diseases/chemically induced , Skin Diseases/pathology
7.
Anticancer Res ; 30(12): 5091-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21187495

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are the professional antigen-presenting cells of the immune system. We have demonstrated that vaccination of autologous ex vivo cultured DCs results in the induction of tumor-specific immune responses in cancer patients, which correlates with clinical response. Optimization of antigen loading is one of the possibilities for further improving the efficacy of DC vaccination. Theoretically, transfection of DCs with RNA encoding a tumor-specific antigen may induce a broader immune response as compared to the most widely used technique of peptide pulsing. PATIENTS AND METHODS: In this clinical study, RNA transfection was compared with peptide pulsing as an antigen loading strategy for DC vaccination. Patients with resectable liver metastases of colorectal cancer were vaccinated intravenously and intradermally 3 times weekly with either carcinoembryogenic antigen (CEA)-derived HLA-A2 binding peptide-loaded or CEA mRNA electroporated DCs prior to surgical resection of the metastases. All DCs were loaded with keyhole limpet hemocyanin (KLH) as a control protein. Evaluation of vaccine-induced immune reactivity consisted of T-cell proliferative responses and B-cell antibody responses against KLH in peripheral blood. CEA reactivity was determined in T-cell cultures of biopsies of post-treatment delayed type hypersensitivity skin tests. RESULTS: Sixteen patients were included. All patients showed T-cell responses against KLH upon vaccination. CEA peptide-specific T-cells were detected in 8 out of 11 patients in the peptide group, but in none of the 5 patients in the RNA group. CONCLUSION: In our study, DC CEA mRNA transfection was not superior to DC CEA peptide pulsing in the induction of a tumor-specific immune response in colorectal cancer patients.


Subject(s)
Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/therapy , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Carcinoembryonic Antigen/genetics , Colorectal Neoplasms/immunology , Dendritic Cells/physiology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Hemocyanins/immunology , Humans , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/immunology , T-Lymphocytes/immunology , Transfection
8.
Eur J Immunol ; 39(7): 1765-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19544486

ABSTRACT

CD4(+) T cells are important for CD8(+) T-cell priming by providing cognate signals for DC maturation. We analyzed the capacity of CD4(+) T cells to influence CD8(+) T-cell responses induced by activated DC. Surprisingly, mice depleted for CD4(+) cells were able to generate stronger antigen-specific CD8(+) T-cell responses after DC vaccination than non-depleted mice. The same observation was made when mice were vaccinated with MHC class II(-/-) DC, indicating the presence of a MHC class II-dependent CD4(+) T-cell population inhibiting CD8(+) T-cell responses. Recently we described the expansion of DX5(+)CD4(+) T cells, a T-cell population displaying immune regulatory properties, upon vaccination with DC. Intriguingly, we now observe an inverse correlation between CD8(+) T-cell induction and expansion of DX5(+)CD4(+) T cells as the latter cells did not expand after vaccination with MHC class II(-/-) DC. In vitro, DX5(+)CD4(+) T cells were able to limit proliferation, modulate cytokine production and induce Foxp3(+) expression in OVA-specific CD8(+) T cells. Together, our data show an inhibitory role of CD4(+) T cells on the induction of CD8(+) T-cell responses by activated DC and indicate the involvement of DX5(+)CD4(+), but not CD4(+)CD25(+), T cells in this process.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Integrin alpha2/immunology , Animals , Antibodies/administration & dosage , Antibodies/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/immunology , Female , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination/methods
9.
Cancer Res ; 69(7): 2927-34, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318559

ABSTRACT

Electroporation of dendritic cells (DC) with mRNA encoding tumor-associated antigens (TAA) for cancer immunotherapy has been proved efficient and clinically safe. It obviates prior knowledge of CTL and Th epitopes in the antigen and leads to the presentation of multiple epitopes for several HLA alleles. Here we studied the migration capacity and the antigen expression of mRNA-electroporated DC (mRNA-DC) in lymph nodes after vaccination in melanoma patients. DC were electroporated with mRNA encoding gp100 or tyrosinase, labeled with indium-111 and superparamagnetic iron oxide particles, and injected intranodally in melanoma patients 24 to 48 hours before scheduled dissection of regional lymph nodes. Immunohistochemical analysis of the lymph nodes after surgery revealed that mRNA-DC migrated from the injection site into the T-cell areas of the same and subsequent lymph nodes, where they expressed the antigen encoded by the electroporated mRNA. Furthermore, vaccine-related CD8(+) T-cell responses could be detected in 7 of 11 patients vaccinated with mRNA-DC. Together these data show that mature DC electroporated with mRNA encoding TAA migrate and express antigens in the lymph nodes and induce specific immune responses.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lymph Nodes/immunology , Melanoma/immunology , Melanoma/therapy , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/immunology , Electroporation/methods , HLA-A2 Antigen/immunology , Humans , Indium Radioisotopes , Lymph Nodes/pathology , Lymphocyte Activation , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Cancer Immunol Immunother ; 58(7): 1109-15, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19018531

ABSTRACT

Tumor-derived peptides are used frequently as antigen (Ag) source in dendritic cell (DC) therapy in cancer patients. An alternative is to load DC with tumor-associated Ag (TAA)-encoding RNA. RNA-loading obviates prior knowledge of CTL and Th epitopes in the Ag. Multiple epitopes for many HLA alleles (both MHC class I and class II) are encoded by the RNA and loading is independent of the patient's HLA make-up. Herein, we determined the optimal conditions for mRNA-electroporation of monocyte-derived DC for clinical application in relation to different maturation cocktails. The data demonstrate that TAA carcinoembryonic antigen, gp100 and tyrosinase are expressed already 30 min after electroporation with the encoding mRNA. Moreover, gp100-specific CTL are activated by gp100 mRNA-electroporated DC. Importantly, we show here that the presence of polyinosinic-polycytidylic acid [poly(I:C)] in the maturation cocktail prevents effective protein expression of the electroporated mRNA as well as subsequent CTL recognition. This effect of poly(I:C) correlates with the induction of IFN-induced genes and innate anti-viral effector molecules in DC. Together these data show that electroporation of mature DC with TAA-encoding mRNA is attractive for use in DC vaccination protocols in cancer patients, but protein expression should be tested for each maturation cocktail.


Subject(s)
Antigen Presentation/drug effects , Antigens, Neoplasm/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interferon Inducers/pharmacology , Poly I-C/pharmacology , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Dendritic Cells/transplantation , Electroporation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/immunology , RNA, Messenger/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , gp100 Melanoma Antigen
11.
Cancer Immunol Immunother ; 57(11): 1589-97, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18322684

ABSTRACT

Dendritic cells (DC) are professional antigen-presenting cells of the immune system that play a key role in regulating T cell-based immunity. In vivo, the capacity of DC to activate T cells depends on their ability to migrate to the T cell areas of lymph nodes as well as on their maturation state. Depending on their cytokine-secreting profile, DC are able to skew the immune response in a specific direction. In particular, IL-12p70 producing DC drive T cells towards a T helper 1 type response. A serious disadvantage of current clinical grade ex vivo generated monocyte-derived DC is the poor IL-12p70 production. We have investigated the effects of Toll-like receptor (TLR)-mediated maturation on ex vivo generated human monocyte-derived DC. We demonstrate that in contrast to cytokine-matured DC, DC matured with poly(I:C) (TLR3 ligand) and/or R848 (TLR7/8 ligand) are able to produce vast amounts of IL-12p70, but exhibit a reduced migratory capacity. The addition of prostaglandin E2 (PGE2) improved the migratory capacity of TLR-ligand matured DC while maintaining their IL-12p70 production upon T cell encounter. We propose a novel clinical grade maturation protocol in which TLR ligands poly(I:C) and R848 are combined with PGE2 to generate DC with both high migratory capacity and IL-12p70 production upon T cell encounter.


Subject(s)
Cell Movement/physiology , Dendritic Cells/physiology , Dinoprostone/metabolism , Interleukin-12/biosynthesis , Toll-Like Receptors/agonists , CD40 Ligand/metabolism , Cell Proliferation , Cells, Cultured , Humans , Imidazoles/pharmacology , Interferon-gamma/metabolism , Ligands , Monocytes/cytology , Poly I-C/pharmacology , Th1 Cells/immunology , Th1 Cells/metabolism , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/physiology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/physiology , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/physiology , Toll-Like Receptors/physiology
12.
J Immunol ; 178(12): 7581-6, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17548593

ABSTRACT

Ag-IgG immune complexes (IC) are efficiently taken up, and Ag-derived peptides are subsequently processed and presented by APC. In vitro experiments indicate that IgG Fc Receptors (FcgammaR) facilitate the efficient uptake of IC by dendritic cells. Previous experiments showed that the cross-presentation of Ag-derived peptides after s.c. administration of IC is FcgammaR-dependent. To study the role of different FcgammaR and complement in MHC class I Ag presentation after i.v. administration, we used mice deficient for FcgammaRs and complement components. These mice were injected with CFSE-labeled OVA-specific CD8+ T cells followed by administration of IC composed of OVA and rabbit anti-OVA IgG i.v. to measure MHC class I presentation of OVA-derived peptides. The Ag presentation was partly reduced in FcRgamma-chain-deficient mice, but not affected in FcgammaRI/II/III-deficient mice, complement factor C3-deficient mice, or FcgammaRI/II/III x C3-deficient mice. Importantly, CD8+ T cell proliferation was significantly reduced in mice deficient for C1q. This proliferation could be restored when IC were incubated with purified human C1q before injection. Likewise, purified C1q could strongly enhance the uptake and presentation of IC by dendritic cells in vitro. Heat inactivation abrogated the C1q-mediated uptake of IC. In addition, in vivo uptake of OVA-IC in the spleen was significantly reduced in C1q-deficient mice compared with wild-type mice. Together, these results indicate a novel function of C1q, which is present in high levels in the bloodstream, by directly enhancing the uptake and MHC class I presentation of Ag captured in IC by APC to CD8+ T cells.


Subject(s)
Antigen Presentation , Antigen-Antibody Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Complement C1q/immunology , Histocompatibility Antigens Class I/immunology , Receptors, IgG/immunology , Animals , Complement C1q/genetics , Cross-Priming , Immunoglobulin G/immunology , Mice , Mice, Mutant Strains , Ovalbumin/immunology , Rabbits , Receptors, IgG/genetics
13.
Immunology ; 119(4): 499-506, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16995881

ABSTRACT

Professional antigen-presenting cells (APC) are able to process and present exogenous antigen leading to the activation of T cells. Antigen-immunoglobulin (Ig)G complexes (IC) are much more efficiently processed and presented than soluble antigen. Dendritic cells (DC) are known for their ability to take up and process immune complex (IC) via FcgammaR, and they have been shown to play a crucial role in IC-processing onto major histocompatibility complex (MHC) class I as they contain a specialized cross-presenting transport system required for MHC class I antigen-processing. However, the MHC class II-antigen-processing pathway is distinct. Therefore various other professional APC, like macrophages and B cells, all displaying FcgammaR, are thought to present IC-delivered antigen in MHC class II. Nonetheless, the relative contribution of these APC in IC-facilitated antigen-presentation for MHC class II in vivo is not known. Here we show that, in mice, both macrophages and DC, but not B cells, efficiently capture IC. However, only DC, but not macrophages, efficiently activate antigen-specific MHC class II restricted CD4(+) T cells. These results indicate that mainly DC and not other professional APC, despite expressing FcgammaR and MHC class II, contribute significantly to IC-facilitated T cell activation in vivo under steady-state conditions.


Subject(s)
Antigen-Antibody Complex/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class II/metabolism , Animals , Antigen Presentation/immunology , B-Lymphocytes/immunology , Cell Proliferation , Immunoglobulin G/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred BALB C
14.
J Immunol ; 176(8): 4573-80, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16585547

ABSTRACT

Dendritic cells (DCs) play an important role in the induction of T cell responses. Fc gammaRs, expressed on DCs, facilitate the uptake of complexed Ag, resulting in efficient MHC class I and MHC class II Ag presentation and DC maturation. In the present study, we show that prophylactic immunization with DCs loaded with Ag-IgG immune complexes (ICs) leads to efficient induction of tumor protection in mice. Therapeutic vaccinations strongly delay tumor growth or even prevent tumors from growing out. By depleting CD4+ and CD8+ cell populations before tumor challenge, we identify CD8+ cells as the main effector cells involved in tumor eradication. Importantly, we show that DCs that are preloaded in vitro with ICs are at least 1000-fold more potent than ICs injected directly into mice or DCs loaded with the same amount of noncomplexed protein. The contribution of individual Fc gammaRs to Ag presentation, T cell response induction, and induction of tumor protection was assessed. We show that Fc gammaRI and Fc gammaRIII are capable of enhancing MHC class I-restricted Ag presentation to CD8+ T cells in vitro and that these activating Fc gammaRs on DCs are required for efficient priming of Ag-specific CD8+ cells in vivo and induction of tumor protection. These findings show that targeting ICs via the activating Fc gammaRs to DCs in vitro is superior to direct IC vaccination to induce protective tumor immunity in vivo.


Subject(s)
Antigen-Antibody Complex/administration & dosage , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , In Vitro Techniques , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Receptors, IgG/deficiency , Receptors, IgG/genetics , Receptors, IgG/metabolism , Solubility
15.
Int Arch Allergy Immunol ; 140(1): 53-72, 2006.
Article in English | MEDLINE | ID: mdl-16534219

ABSTRACT

Dendritic cells (DC) are professional antigen-presenting cells which are strategically positioned at the boundaries between the inner and the outside world, in this way bridging innate and adaptive immunity. DC can initiate T cell responses against microbial pathogens and tumors due to their capacity to stimulate naïve T cells. The development of DC occurs in distinct stages. DC precursors develop in the bone marrow and home to a large variety of tissues. Immature DC capture antigen (Ag) and, following proinflammatory signals, migrate to the lymphoid organs where, after maturation, they present captured Ag to naïve T cells, thereby inducing differentiation of naïve T cells into effector T cells. An important cognate event in the development of cell-mediated immunity is the interaction between CD40 and CD40 ligand. Ligation of CD40 on DC by its ligand results in maturation of the DC. In addition to CD40 ligand (expressed by activated Th cells), inflammatory cytokines, bacterial components or Ag-Ab immune complexes can induce maturation of DC. Maturation of DC is crucial for the priming of efficient T cell responses and is characterized by a decreased Ag processing capacity, an increased cell surface expression of MHC and costimulatory molecules, and rearrangement of cytoskeleton, adhesion molecules, and cytokine receptors. Mature DC migrate from peripheral tissues to secondary lymphoid organs, where T cell priming occurs. DC are not only critical in initiating T cell immunity, they also play a role in the induction of T cell tolerance and the regulation of the type of T cell response that is induced. Here we give an overview of the dendritic cell system.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Animals , Antigen-Presenting Cells/classification , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Dendritic Cells/classification , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans
16.
Mol Immunol ; 43(13): 2045-50, 2006 May.
Article in English | MEDLINE | ID: mdl-16513171

ABSTRACT

Antigen(Ag)-immunoglobulin (Ig)G complexes (IC) are more efficiently processed and presented than soluble Ag. IC can bind to various cell types via different types of Fc-Receptors or, upon binding to complement factors, by complement receptors. Murine professional antigen-presenting cells (APC) express four types of FcgammaReceptors (FcgammaR) via which they are able to capture IC; three activating receptors (FcgammaRI, III and IV) and one inhibitory receptor (FcgammaRII). It has been demonstrated that FcgammaR play a pivotal role in facilitating the presentation of Ag derived from IC. Nonetheless, relative little information is available on the relative contribution of the activating or inhibitory FcgammaR or complement to the presentation of immune-complexed Ag to CD8+ T cells. To study the contribution of the different FcgammaR and complement receptors in IC-facilitated Ag-presentation, we analyzed the ovalbumin(OVA)-specific CD8+ T cell proliferation in FcgammaR- and complement component 3 (C3)-deficient mice after subcutaneous injection of OVA-IC. Here we show that the efficient Ag-presentation was FcgammaR-, but not C3-mediated, as it was inhibited in FcgammaRI/II/III-deficient mice but unaffected in the C3-depleted mice. Moreover, FcgammaRIV does not play a role under these conditions. However, no difference was found between wild-type and FcgammaRI/III-deficient or wild-type and FcgammaRII-deficient mice. These results indicate that Ag-presentation via the activating FcgammaR is not enhanced in the absence of FcgammaRII, and point to redundancy of the FcgammaR, including FcgammaRII, in the uptake and presentation of s.c. injected soluble IC to CD8+ T cells.


Subject(s)
Antigen Presentation/immunology , Antigen-Antibody Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, IgG/immunology , Animals , Antigen Presentation/drug effects , Antigen-Antibody Complex/pharmacology , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation/drug effects , Cells, Cultured , Complement C3/deficiency , Complement C3/immunology , Mice , Mice, Knockout , Receptors, Complement/immunology , Receptors, IgG/deficiency
17.
Transplantation ; 76(11): 1608-15, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14702533

ABSTRACT

BACKGROUND: Activation of immature dendritic cells (DC) in the presence of the glucocorticoid hormone dexamethasone (DEX) results in alternatively matured DC that present antigen in the absence of a proper co-stimulatory context. This maturation process is irreversible, making these cells an attractive potential tool for the induction of antigen-specific T-cell tolerance in vivo. The authors explored the possibility of using these DC for the induction of transplantation tolerance in a fully allogeneic setting in mice. METHODS: Immature dendritic cells (D1, an immature splenic DC line derived from B6 mice) were pretreated with DEX for 24 hr, after which lipopolysaccharide or nothing was added to the culture for another 48 hr. These cells were analyzed for their in vitro and in vivo stimulating or tolerizing capacities. RESULTS: In line with their phenotype, including decreased interleukin (IL)-12 production, in vitro co-culture of alternatively matured D1 (B6 origin; H-2b) with completely allogeneic T cells of BALB/c origin led to a significant decrease in the alloreactive T-cell response. A single injection of 1 x 10(6) alternatively matured H-2b DC into BALB/c mice induced a different alloimmune response compared with mature DC. The responding T cells showed a lower proliferation rate and a lower interferon-gamma production, whereas a significantly higher proportion of the cells produced IL-10 as measured ex vivo by enzyme-linked immunospot assay. Furthermore, injection with alternatively matured DC, followed by transplantation of fully mismatched skin grafts (C57BL/6), led to a significantly prolonged survival compared with that of mature DC-pretreated mice or untreated mice. The immunomodulatory effect was antigen specific, as third-party reactive alloresponses were not affected. CONCLUSIONS: The authors' data constitute the first direct demonstration that DC alternatively matured in the presence of glucocorticoid hormones can be exploited for the specific suppression of the alloreactive Th1 response, resulting in a delayed skin graft rejection in a complete major histocompatibility complex-incompatible strain combination.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft Survival/physiology , Skin Transplantation/physiology , Animals , Cells, Cultured , Dendritic Cells/cytology , Dexamethasone/pharmacology , Female , Flow Cytometry , Immunosuppression Therapy/methods , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Skin Transplantation/immunology , Transplantation, Homologous/immunology
18.
J Immunol ; 170(1): 33-40, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12496380

ABSTRACT

The interaction between TNFR family member CD27 and its ligand CD70 promotes lymphocyte expansion and effector cell formation. In humans, control of CD27 function is partly regulated by the restricted expression of CD70. We used newly developed mAbs to characterize murine (m) CD70 expression in vitro and in vivo. On resting lymphocytes and immature dendritic cells (DC), mCD70 is absent. In vitro, Ag receptor triggering induced mCD70 mRNA in T cells, but cell surface protein expression was very low. Activated B cells synthesized much higher levels of mCD70 mRNA than activated T cells and clearly expressed mCD70 at the cell surface. mCD70 cell surface expression could also be induced on the DC line D1 and on in vitro-generated murine DC upon maturation. In lymphoid organs of naive mice, virtually no mCD70-expressing cells were found, with exception of cells in the thymic medulla, which may be epithelial in origin. However, after intranasal infection with influenza virus, lung-infiltrating T cells and T and B cells in draining lymph nodes expressed mCD70 according to immunohistology. In such activated lymphocytes, mCD70 protein is largely retained intracellularly. Plasma membrane expression of mCD70 was only detectable by flow cytometry on a small proportion of lung-infiltrating T cells and peaked at the height of the primary response. Thus, expression of CD70 in the mouse is highly regulated at the transcriptional and posttranslational level. This most likely serves to limit excessive effector cell formation after antigenic stimulation.


Subject(s)
Antigens, CD , Membrane Proteins/biosynthesis , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , B-Lymphocytes/chemistry , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD27 Ligand , Cricetinae , Cricetulus , Humans , Hybridomas , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/pathology , Ligands , Lung/chemistry , Lung/immunology , Lung/pathology , Lymph Nodes/chemistry , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity/immunology , Spleen/chemistry , Spleen/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured
19.
J Immunol ; 168(5): 2240-6, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11859111

ABSTRACT

Dendritic cells (DCs) require a maturation signal to acquire efficient CTL-priming capacity. In vitro FcgammaR-mediated internalization of Ag-Ab immune complexes (ICs) can induce maturation of DCs. In this study, we show that IC-induced DC maturation in vitro enables DCs to prime peptide-specific CD8+ CTLs in vivo, independently of CD4+ Th cells. Importantly, OVA/anti-OVA IC-treated DCs not only primed CD8+ CTLs to an exogenously loaded peptide nonrelated to OVA, but also efficiently primed CTLs against the dominant CTL epitope derived from the OVA Ag present in the ICs. Our studies show that ICs fulfill a dual role in priming of CD8+ CTL responses to exogenous Ags: enhancement of Ag uptake by DCs and activation of DCs, resulting in "license to kill." These findings indicate that the presence of specific Abs can crucially affect the induction of cytotoxic cellular responses.


Subject(s)
Antigen Presentation , Antigen-Antibody Complex/immunology , Dendritic Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Differentiation , Cell Line, Transformed , Cytotoxicity Tests, Immunologic , Endocytosis , Female , Histocompatibility Antigens Class I/physiology , Hybridomas , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Ovalbumin/immunology , Ovalbumin/metabolism , Peptides/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...