Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Immunotoxicol ; 11(1): 62-71, 2014.
Article in English | MEDLINE | ID: mdl-23738746

ABSTRACT

Immunogenicity is a major issue of concern for monoclonal antibodies used in human diseases and is by default mainly determined in non-human primates (NHP), as target molecules are considered most similar in NHP compared to human. In this manuscript the predictive value of immunogenicity testing in minipigs for human safety is evaluated, as the immune system of the pig is functionally similar to that in other mammalian species. Adalimumab and infliximab (both monoclonal antibodies blocking TNFα) were used as model substances. Female Göttingen minipigs (4/group) were treated every other week with low (0.1 mg/kg), mid (1.0 mg/kg), or high dose (5 mg/kg) adalimumab or 5 mg/kg infliximab subcutaneous (SC) over a period of 8 weeks. After first and last dosing, pharmacokinetic analysis was performed. Anti-drug antibodies (ADAs) were measured on several time points. Furthermore, hematology, clinical chemistry, body weight, clinical signs, and histopathology of several organs were evaluated. No signs of toxicity of the treatments were observed in the limited organs and tissues collected. Eleven out of 12 minipigs treated with adalimumab elicited a detectable ADA response. Induction of ADA was correlated with decreased plasma levels of adalimumab. Infliximab clearance was comparable after first and last dose. Therefore, the presence of ADA directed to infliximab was considered highly unlikely. It was concluded that the minipig and NHP showed comparable suitability for immunogenicity prediction in humans. More studies with other biopharmaceutical products are needed to strengthen the status of the minipig as an alternative model for immunotoxicity testing including immunogenicity.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal/administration & dosage , Adalimumab , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibody Formation/drug effects , Drug Evaluation, Preclinical , Feasibility Studies , Female , Humans , Infliximab , Injections, Subcutaneous , Metabolic Clearance Rate , Swine , Swine, Miniature , Tumor Necrosis Factor-alpha/metabolism
2.
J Immunotoxicol ; 10(1): 96-105, 2013.
Article in English | MEDLINE | ID: mdl-23134195

ABSTRACT

The use of recombinant human proteins for the treatment of several diseases has increased considerably during the last decades. A major safety and efficacy issue of biopharmaceuticals is their potential immunogenicity. To prevent immunogenicity, biotechnology-derived proteins are engineered to be as human-like as possible. Immunogenicity is mainly determined in non-human primates (NHP), as they are considered to be the best predictive animal species for human safety, based on their close relatedness to man. As minipigs are increasingly used in the safety evaluation of (bio)pharmaceuticals, the predictive value of the minipig in immunogenicity testing was evaluated in this study, using anakinra as a model compound. Animals were treated subcutaneously with either placebo, low- (0.5 mg/kg), or high-dose (5 mg/kg) anakinra daily on 29 consecutive days. After the first and last dose, the pharmacokinetic (PK) profile of anakinra was evaluated. Antibodies directed to anakinra were measured on several time points during the treatment period. Furthermore, hematology, clinical chemistry, body weight, clinical signs, and histopathology of several organs were evaluated. No signs of toxicity were observed upon treatment with anakinra. PK parameters were comparable with those found in human and NHP studies performed with anakinra. All animals developed anti-anakinra antibodies. The results obtained in minipigs were comparable to those observed in monkeys. For anakinra, the predictive value of the minipig for immunogenicity testing was found to be comparable to that seen in NHP. However, more studies evaluating additional biopharmaceutical products are needed to support the use of the minipig as an alternative model for (immuno)toxicity testing, including immunogenicity.


Subject(s)
Immunologic Tests , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Swine, Miniature/immunology , Animals , Antibodies/blood , Disease Models, Animal , Feasibility Studies , Humans , Injections, Subcutaneous , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Interleukin 1 Receptor Antagonist Protein/immunology , Male , Predictive Value of Tests , Receptors, Interleukin-1/antagonists & inhibitors , Recombinant Proteins/administration & dosage , Swine
3.
J Immunol ; 180(5): 2747-51, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292492

ABSTRACT

CD4+CD25+ regulatory T (Treg) cells play an essential role in maintaining tolerance to self and nonself. In several models of T cell-mediated (auto) immunity, Treg cells exert protective effects by the inhibition of pathogenic T cell responses. In addition, Treg cells can modulate T cell-independent inflammation. We now show that CD4+CD25+ Treg cells are able to shed large amounts of TNFRII. This is paralleled by their ability to inhibit the action of TNF-alpha both in vitro and in vivo. In vivo, Treg cells suppressed IL-6 production in response to LPS injection in mice. In contrast, Treg cells from TNFRII-deficient mice were unable to do so despite their unhampered capacity to suppress T cell proliferation in a conventional in vitro suppression assay. Thus, shedding of TNFRII represents a novel mechanism by which Treg cells can inhibit the action of TNF, a pivotal cytokine driving inflammation.


Subject(s)
Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Acute-Phase Proteins/antagonists & inhibitors , Acute-Phase Proteins/metabolism , Animals , Cells, Cultured , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/physiology , T-Lymphocytes, Regulatory/transplantation , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology
4.
Circulation ; 116(19): 2173-81, 2007 Nov 06.
Article in English | MEDLINE | ID: mdl-17967778

ABSTRACT

BACKGROUND: Lipopolysaccharide (LPS), which is released from gram-negative bacteria on multiplication or lysis, aggravates atherosclerosis in humans and rodents by inducing inflammation via toll-like receptors. Because apolipoprotein C-I (apoCI) enhances the LPS-induced inflammatory response in macrophages in vitro and in mice, we investigated the effect of endogenous apoCI expression on LPS-induced atherosclerosis in mice. METHODS AND RESULTS: Twelve-week-old apoe-/- apoc1-/- and apoe-/- apoc1+/+ mice received weekly intraperitoneal injections of LPS (50 microg) or vehicle for a period of 10 weeks, and atherosclerosis development was assessed in the aortic root. LPS administration did not affect atherosclerotic lesion area in apoe-/- apoc1-/- mice but increased it in apoe-/- apoc1+/+ mice. In fact, apoCI expression increased the LPS-induced atherosclerotic lesion area by 60% (P<0.05), concomitant with an increase in LPS-induced plasma levels of fibrinogen and E-selectin. This indicated that apoCI increased the LPS-induced inflammatory state, both systemically (ie, fibrinogen) and at the level of the vessel wall (ie, E-selectin). In addition, both macrophage-derived apoCI and HDL-associated apoCI increased the LPS-induced tumor necrosis factor-alpha response by macrophages in vitro. CONCLUSIONS: We conclude that apoCI is crucially involved in LPS-induced atherosclerosis in apoe-/- mice, which mainly relates to an increased inflammatory response toward LPS. We anticipate that apoCI plasma levels contribute to accelerated atherosclerosis development in individuals who have chronic infection.


Subject(s)
Apolipoproteins C/genetics , Apolipoproteins C/metabolism , Atherosclerosis/genetics , Atherosclerosis/immunology , Animals , Atherosclerosis/pathology , Biomarkers/blood , Cells, Cultured , Cholesterol, HDL/blood , E-Selectin/blood , Female , Fibrinogen/metabolism , Hypercholesterolemia/genetics , Hypercholesterolemia/immunology , Hypercholesterolemia/pathology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/pathology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/metabolism , Vasculitis/genetics , Vasculitis/immunology , Vasculitis/pathology
5.
Immunobiology ; 211(6-8): 627-32, 2006.
Article in English | MEDLINE | ID: mdl-16920501

ABSTRACT

Adaptive immune responses are orchestrated by specialized professional antigen-presenting cells (APCs), the dendritic cells (DCs), which play crucial roles as initiators and modulators of adaptive immune responses. A main feature of DCs is their phenotypic and functional plasticity. In the absence of any inflammation or pathogenic elements, most DCs in peripheral tissues and lymphoid organs have a resting, immature phenotype characterized by high endocytic capacity and low surface expression of MHC- and costimulatory molecules. However, upon interaction with microbial ligands, pro-inflammatory cytokines or CD40Ligand, DCs rapidly acquire an activated phenotype. These mature DCs have a very efficient T cell-priming ability as a consequence of upregulation of MHC- and costimulatory molecules on their cell surface. For this reason, DC-based vaccines have been used successfully to combat infections and malignancies. Nonetheless, evidence is accumulating that, especially immature, or semi-matured, DCs also have a potent ability to tolerize T cells or prevent undesired immune reactions. Therefore, current and prospective strategies to promote the inherent tolerogenic potential of DCs are a rational approach for the therapy of autoimmune diseases such as rheumatoid arthritis (RA). This review summarizes some aspects of the intriguing ability of DCs to steer the outcome of immunity and their potency to modulate the outcome of various pathological conditions.


Subject(s)
Autoimmune Diseases/prevention & control , Dendritic Cells/immunology , Immune Tolerance/physiology , Animals , Arthritis, Rheumatoid/immunology , Humans
6.
J Immunol ; 175(7): 4247-54, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16177064

ABSTRACT

Ags expressed at immune privileged sites and other peripheral tissues are able to induce T cell tolerance. In this study, we analyzed whether tolerance toward an intraocular tumor expressing a highly immunogenic CTL epitope is maintained, broken, or reverted into immunity in the event the anatomical integrity of the eye is lost. Inoculation of tumor cells into the anterior chamber of the eye of naive B6 mice leads to progressive intraocular tumor growth, an abortive form of CTL activation in the tumor-draining submandibular lymph node, and systemic tolerance as evidenced by the inability of these mice to reject an otherwise benign tumor cell inoculum. Loss of anatomical integrity of the eye as a consequence of phthisis resulted in loss of systemic tolerance and the emergence of effective antitumor immunity against an otherwise lethal tumor challenge. Phthisis was accompanied by dendritic cell maturation and preceded the induction of systemic tumor-specific CTL immunity. Our data show that normal tissue homeostasis and anatomical integrity is required for the maintenance of ocular tolerance and prevention of CTL-mediated immunity. These data also indicate that tissue injury in the absence of viral or microbial infection can act as a switch for the induction of CTL immunity.


Subject(s)
Eye Neoplasms/immunology , Homeostasis/immunology , Immune Tolerance/physiology , Animals , Anterior Chamber/immunology , Antigen Presentation/immunology , Antigen-Presenting Cells/metabolism , Antigens, Neoplasm/immunology , CD11c Antigen/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cross-Priming/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Cytotoxic/immunology
7.
Cancer Res ; 65(15): 6984-9, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16061684

ABSTRACT

Modulation of the immune response by established tumors may contribute to the limited success of therapeutic vaccination for the treatment of cancer compared with vaccination in a preventive setting. We analyzed the contribution of the CD4+ T-cell population to the induction or suppression of tumor-specific CD8+ T cells in a tumor model in which eradication of tumors crucially depends on CD8+ T cell-mediated immunity. Vaccine-mediated induction of protective antitumor immunity in the preventive setting (i.e., before tumor challenge) was CD4+ T cell dependent because depletion of this T-cell subset prevented CD8+ T-cell induction. In contrast, depletion of CD4+ cells in mice bearing established E1A+ tumors empowered the mice to raise strong CD8+ T-cell immunity capable of tumor eradication without the need for tumor-specific vaccination. Spontaneous eradication of tumors, which had initially grown out, was similarly observed in MHC class II-deficient mice, supporting the notion that the tumor-bearing mice harbor a class II MHC-restricted CD4+ T-cell subset capable of suppressing a tumor-specific CD8+ T-cell immune response. The deleterious effects of the presence of CD4+ T cells in tumor-bearing hosts could be overcome by CD40-triggering or injection of CpG. Together these results show that CD4+ T cells with a suppressive activity are rapidly induced following tumor development and that their suppressive effect can be overcome by agents that activate professional antigen-presenting cells. These observations are important for the development of immune interventions aiming at treatment of cancer.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen-Presenting Cells/immunology , CD4 Lymphocyte Count , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Growth Processes/immunology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/prevention & control , Neoplasms, Experimental/therapy , Receptors, Interleukin-2/immunology
8.
J Immunol ; 175(2): 855-63, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16002683

ABSTRACT

Immature dendritic cells (DC), in contrast to their mature counterparts, are incapable of mobilizing a CD8+ CTL response, and, instead, have been reported to induce CTL tolerance. We directly addressed the impact of immature vs mature DC on CTL responses by infusing adenovirus peptide-loaded DC (of the D1 cell line) into mice that had received adenovirus-specific naive TCR-transgenic CD8+ T cells. Whereas i.v. injection of mature DC triggered vigorous CTL expansion, immature DC elicited little proliferation involving only a minority of the TCR-transgenic CTL. Even though the latter CTL developed effector functions, including cytolytic activity and proinflammatory cytokine secretion, these cells differed significantly from CTL primed by mature DC in that they did not exhibit down-regulation of CD62L and CCR7, receptors involved in trapping of T cells in the lymphoid organs. Interestingly, adoptive transfer of CTL effector cells harvested after priming by either mature or immature DC into naive recipient mice, followed by exposure to adenovirus, yielded quantitatively and qualitatively indistinguishable CTL memory responses. Therefore, in vivo priming of naive CD8+ T cells by immature DC, although failing to induce a full-blown, systemic CTL response, resulted in the formation of central memory-like T cells that were able to expand and produce IFN-gamma upon secondary antigenic stimulation.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Clonal Deletion/immunology , Dendritic Cells/immunology , Immunologic Memory , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/immunology , Animals , Antigen Presentation/genetics , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , Cell Movement/immunology , Clonal Deletion/genetics , Cytotoxicity, Immunologic/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Down-Regulation/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Immunologic Memory/genetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology
9.
J Immunol ; 173(11): 6753-9, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15557168

ABSTRACT

The fate of naive CD8(+) T cells is determined by the environment in which they encounter MHC class I presented peptide Ags. The manner in which tumor Ags are presented is a longstanding matter of debate. Ag presentation might be mediated by tumor cells in tumor draining lymph nodes or via cross-presentation by professional APC. Either pathway is insufficient to elicit protective antitumor immunity. We now demonstrate using a syngeneic mouse tumor model, expressing an Ag derived from the early region 1A of human adenovirus type 5, that the inadequate nature of the antitumor CTL response is not due to direct Ag presentation by the tumor cells, but results from presentation of tumor-derived Ag by nonactivated CD11c(+) APC. Although this event results in division of naive CTL in tumor draining lymph nodes, it does not establish a productive immune response. Treatment of tumor-bearing mice with dendritic cell-stimulating agonistic anti-CD40 mAb resulted in systemic efflux of CTL with robust effector function capable to eradicate established tumors. For efficacy of anti-CD40 treatment, CD40 ligation of host APC is required because adoptive transfer of CD40-proficient tumor-specific TCR transgenic CTL into CD40-deficient tumor-bearing mice did not lead to productive antitumor immunity after CD40 triggering in vivo. CpG and detoxified LPS (MPL) acted similarly as agonistic anti-CD40 mAb with respect to CD8(+) CTL efflux and tumor eradication. Together these results indicate that dendritic cells, depending on their activation state, orchestrate the outcome of CTL-mediated immunity against tumors, leading either to an ineffective immune response or potent antitumor immunity.


Subject(s)
Antigens, Viral, Tumor/metabolism , Cross-Priming/immunology , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Adenovirus E1A Proteins/immunology , Adenovirus E1A Proteins/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Viral, Tumor/immunology , CD11c Antigen/biosynthesis , CD40 Antigens/biosynthesis , CD40 Antigens/immunology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Injections, Intralesional , Injections, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured
10.
J Immunol ; 172(10): 6074-9, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15128791

ABSTRACT

Naive T cells can be tolerized in the periphery by diverse mechanisms. However, the extent to which memory T cells are susceptible to tolerance induction is less well defined. Vaccination of mice with a minimal CTL epitope derived from human adenovirus type 5 E1A in IFA s.c. readily tolerizes naive as well as recently activated CD8(+) T cells due to the overwhelming systemic and persistent presence of the peptide. We have now studied the effect of this peptide on established memory cells, which were induced at least 50 days before by virus vaccination. Memory cells did not undergo peripheral deletion and kept their ability to produce IFN-gamma as well as their cytolytic activity in response to Ag directly ex vivo. However, memory CTL responses in virus vaccinated mice injected with peptide ceased to control tumor outgrowth. Interestingly, functional capacities were regained when T cells were transferred to an Ag-free environment in vivo as determined by their ability to reject an otherwise lethal tumor challenge. Together, these findings indicate that memory CTL responses can be functionally incapacitated, but are not, in contrast to naive or recently activated T cells, irreversibly tolerized by persistent systemic Ag, as memory T cells quickly regain effector function upon disappearance of the Ag.


Subject(s)
Adenovirus E1A Proteins/immunology , Adenovirus E1A Proteins/metabolism , Antigen Presentation , Cytotoxicity, Immunologic , Immunologic Memory , T-Lymphocytes, Cytotoxic/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/therapy , Adenovirus E1A Proteins/administration & dosage , Adoptive Transfer , Animals , Antigen Presentation/immunology , Cell Division/immunology , Cells, Cultured , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Immune Tolerance/immunology , Injections, Subcutaneous , Interphase/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/transplantation , Tumor Virus Infections/pathology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
11.
J Immunol ; 172(3): 1567-74, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734736

ABSTRACT

Ocular immune privilege is considered essential in the protection against sight-threatening immune responses, as illustrated by the ability of the ocular environment to permit the growth of tumors that are rejected when implanted at other sites. Although several studies indicate that soluble Ag can drain directly into the spleen when injected into the anterior chamber, the primary site of intraocular tumor Ag presentation to tumor-specific CTLs has not been studied. To gain a better understanding of the mechanism involved in ocular immune privilege, we examined to which lymphoid organs anterior chamber tumor Ags primarily drain. Our data show that intraocular tumor Ag drains exclusively to the submandibular lymph nodes, resulting in activation of tumor-specific CTLs, whereas no Ag drainage was found in spleen. However, these tumor-specific CTLs do not distribute systemically and, as a consequence, intraocular tumor growth is unhampered. A similar lack of CTL efficacy has been observed in mice bearing s.c. tumors, which is converted to a systemic tumoricidal CTL response by administration of agonistic anti-CD40 mAb. In contrast, systemic anti-CD40 treatment of eye tumor-bearing mice did not result in mobilizing tumor-specific CTLs or tumor eradication. Together, these results show that intraocular tumor Ag drains to regional lymph nodes for activation of tumor-specific CTLs. However, the induced tumor-specific immunity is insufficient for tumor clearance, even combined with otherwise highly effective immune intervention protocols.


Subject(s)
Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic , Eye Neoplasms/immunology , Graft Rejection/immunology , Lymph Nodes/immunology , Submandibular Gland/immunology , T-Lymphocytes, Cytotoxic/immunology , Adenovirus E1A Proteins/biosynthesis , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/immunology , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , Anterior Chamber/immunology , Anterior Chamber/pathology , Anterior Chamber/virology , Antigens, Neoplasm/genetics , CD40 Antigens/physiology , Cell Division/genetics , Cell Division/immunology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/immunology , Eye Neoplasms/pathology , Eye Neoplasms/virology , Graft Rejection/pathology , Graft Rejection/virology , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/virology , Submandibular Gland/pathology , Submandibular Gland/virology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology
12.
Proc Natl Acad Sci U S A ; 99(8): 5561-6, 2002 Apr 16.
Article in English | MEDLINE | ID: mdl-11929985

ABSTRACT

Adequate spontaneous activation of tumor-specific T lymphocytes in tumor-bearing hosts is rare, despite the expression of tumor antigens that are potentially highly immunogenic. For example, failure of the immune system to raise competent responses against established tumors expressing the human adenovirus E1A-antigen allows this tumor to grow in immunocompetent mice. We show that systemic in vivo administration of agonistic anti-CD40 antibodies into tumor-bearing mice results in tumor eradication mediated by CD8(+) T cells. Treatment resulted in a strong expansion and systemic accumulation of E1A-specific CTL and depended on CD40 expression on host cells, as the tumor was CD40(-), and therapy failed in CD40-deficient mice. Local intratumoral administration of anti-CD40 mAb is equally effective in licensing strong, systemic CTL immunity, resulting in the clearance of distant tumor nodules. Our data indicate that the immune response after cancer-host interactions can be directed toward competence, leading to the cure of established tumors merely by delivery of a CD40-dependent "license to kill" signal.


Subject(s)
CD40 Antigens/biosynthesis , CD40 Antigens/pharmacology , Neoplasms/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , CD40 Antigens/metabolism , Coloring Agents/pharmacology , Epitopes , Flow Cytometry , Fluoresceins/pharmacology , Lymph Nodes/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Spleen/metabolism , Succinimides/pharmacology , Time Factors
13.
J Immunol ; 168(8): 3755-62, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11937526

ABSTRACT

Triggering of 4-1BB, a member of the TNFR family, through in vivo administration of agonistic anti-4-1BB Ab delivers a powerful costimulatory signal to CTL. We found this signal to effectively replace the need for CD4(+) T cell help in the cross-priming of tumor-specific CTL immunity. Furthermore, 4-1BB Ab can convert an otherwise tolerogenic peptide vaccine into a formulation capable of efficient CTL priming. Initial activation of naive CTL can occur in the absence of 4-1BB costimulation, but this signal permits increased survival of Ag-stimulated CTL. Because naive CTL do not express 4-1BB at their surface, susceptibility to 4-1BB triggering depends on prior up-regulation of this receptor. We show that this requires both stimulation of the TCR and CD28-dependent costimulation. Accordingly, blockade of the CD28-costimulatory pathway abrogates the capacity of agonistic anti-4-1BB Ab to trigger Th-independent CTL immunity. In conclusion, our data reveal that the 4-1BB-mediated survival signal is positioned downstream of Ag-specific TCR triggering and CD28-dependent costimulation of naive CTL. The powerful effects of 4-1BB triggering on the induction, amplification, and persistence of CTL responses provide a novel strategy for increasing the potency of vaccines against cancers.


Subject(s)
CD28 Antigens/physiology , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Lymphocyte Activation/immunology , Receptors, Nerve Growth Factor/physiology , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes, Cytotoxic/immunology , 4-1BB Ligand , Animals , Antigens, CD , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immune Sera/administration & dosage , Injections, Intraperitoneal , Injections, Intravenous , Interphase/immunology , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Tumor Necrosis Factor-alpha/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...