Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Hum Gene Ther ; 32(13-14): 744-760, 2021 07.
Article in English | MEDLINE | ID: mdl-33554732

ABSTRACT

Effectiveness of adoptively transferred chimeric antigen receptor (CAR) T cells strongly depends on the quality of CAR-mediated interaction of the effector cells with the target antigen on tumor cells. A major role in this interaction is played by the affinity of the single-chain variable fragment (scFv) for the antigen, and by the CAR design. In particular, the spacer domain may impact on the CAR T cell function by affecting the length and flexibility of the resulting CAR. This study addresses the need to improve the manufacturing process and the antitumor activity of CD44v6-specific CAR T cells by defining the optimal structure of a spacer region derived from the extracellular domain of the human low-affinity nerve growth factor receptor (LNGFR). We tailored the LNGFR spacer to modulate CAR length to efficiently recognize distal or proximal epitopes and to allow selection of transduced CAR T cells by the use of clinical-grade validated manufacturing systems. The different LNGFR spacers investigated in this study are responsible for the generation of CAR T cells with a different memory phenotype, which is mainly related to the level of CAR expression and the extent of the associated tonic signaling. In particular, the CD44v6-NWN2.CAR T cells are enriched in central memory cells and show improved in vitro functions in terms of killing capability, and in vivo antitumor activity against hematological and solid tumors. Clinical Trial Registration numbers: clinicaltrial.gov NCT04097301; ClinicalTrials.gov, NCT00423124.


Subject(s)
Receptors, Chimeric Antigen , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Receptor, Nerve Growth Factor , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Nerve Growth Factor , T-Lymphocytes , Xenograft Model Antitumor Assays
2.
Front Immunol ; 11: 99, 2020.
Article in English | MEDLINE | ID: mdl-32117253

ABSTRACT

The main challenge of adoptive therapy with Chimeric Antigen Receptor modified T cells (CAR T) is the application to the field of solid tumors, where the identification of a proper antigen has emerged as one of the major drawbacks to CAR T cell treatment success. CD44 is a glycoprotein involved in cell-cell and cell-matrix interactions. The isoform containing the variant domain 6 of CD44 gene (CD44v6) has been implicated in tumorigenesis, tumor cell invasion and metastasis and represents an attractive target for CAR T cell therapies. Targeting CD44v6 antigen has been shown to control tumor growth in acute myeloid leukemia and multiple myeloma mouse models. While CAR T approach for the treatment of B cell malignancies has shown great success, response rates among patients with solid cancer are less favorable. The purpose of our study was to test the efficacy of CD44v6.CAR T cells, produced in compliance with Good Manufacturing Practice (GMP), in adenocarcinoma tumor models. We generated a bicistronic retroviral vector containing the CD44v6 CAR and the HSV-TK Mut2 suicide gene to enhance the safety of the proposed CAR T cell therapy. CD44v6 transduced CAR T cells were homogeneously positive for ΔLNGFR selection marker, were enriched in T central memory (TCM) and T memory stem cells (TSCM) and displayed a highly activated phenotype. In vitro assays revealed antigen-specific activation and cytotoxicity of human CD44v6.CAR T cells against CD44v6 expressing tumor cell lines. When infused in immunodeficient tumor bearing mice, human CD44v6.CAR T cells were able to reach, infiltrate and proliferate at tumor sites, finally resulting in tumor growth control. Next, we checked if cells produced in compliance with GMP grade standards retained the same antitumor activity of those produced with research grade materials and protocols. Noteworthy, no differences in the potency of the CAR T obtained with the two manufacturing processes were observed. In conclusion, our preclinical results suggest that CD44v6.CAR T based adoptive therapy could be a promising strategy in solid cancer treatment.


Subject(s)
Adenocarcinoma/therapy , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/therapeutic use , Adenocarcinoma/immunology , Animals , Antigens, CD19 , Cell Line, Tumor , Cell Proliferation , Female , Genes, Transgenic, Suicide , Humans , Hyaluronan Receptors/genetics , Immunotherapy, Adoptive , Lung/pathology , Mice , Mice, Transgenic , Molecular Targeted Therapy , Ovary/metabolism , Ovary/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology
3.
Int J Mol Sci ; 20(18)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547231

ABSTRACT

NGR-hTNF is a therapeutic agent for a solid tumor that specifically targets angiogenic tumor blood vessels, through the NGR motif. Its activity has been assessed in several clinical studies encompassing tumors of different histological types. The drug's activity is based on an improved permeabilization of newly formed tumor vasculature, which favors intratumor penetration of chemotherapeutic agents and leukocyte trafficking. This work investigated the binding and the signaling properties of the NGR-hTNF, to elucidate its mechanism of action. The crystal structure of NGR-hTNF and modeling of its interaction with TNFR suggested that the NGR region is available for binding to a specific receptor. Using 2D TR-NOESY experiments, this study confirmed that the NGR-peptides binds to a specific CD13 isoform, whose expression is restricted to tumor vasculature cells, and to some tumor cell lines. The interaction between hTNF or NGR-hTNF with immobilized TNFRs showed similar kinetic parameters, whereas the competition experiments performed on the cells expressing both TNFR and CD13 showed that NGR-hTNF had a higher binding affinity than hTNF. The analysis of the NGR-hTNF-triggered signal transduction events showed a specific impairment in the activation of pro-survival pathways (Ras, Erk and Akt), compared to hTNF. Since a signaling pattern identical to NGR-hTNF was obtained with hTNF and NGR-sequence given as distinct molecules, the inhibition observed on the survival pathways was presumably due to a direct effect of the NGR-CD13 engagement on the TNFR signaling pathway. The reduced activation of the pro survival pathways induced by NGR-hTNF correlated with the increased caspases activation and reduced cell survival. This study demonstrates that the binding of the NGR-motif to CD13 determines not only the homing of NGR-hTNF to tumor vessels, but also the increase in its antiangiogenic activity.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Neoplasms/blood supply , Oligopeptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Angiogenesis Inhibitors/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Human Umbilical Vein Endothelial Cells , Humans , Models, Molecular , Oligopeptides/chemistry , Recombinant Fusion Proteins/chemistry , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/chemistry
4.
J Med Chem ; 61(17): 7474-7485, 2018 09 13.
Article in English | MEDLINE | ID: mdl-29883545

ABSTRACT

The isoDGR sequence is an integrin-binding motif that has been successfully employed as a tumor-vasculature-homing molecule or for the targeted delivery of drugs and diagnostic agents to tumors. In this context, we previously demonstrated that cyclopeptide 2, the product of the conjugation of c(CGisoDGRG) (1) to 4-( N-maleimidomethyl)cyclohexane-1-carboxamide, can be successfully used as a tumor-homing ligand for nanodrug delivery to neoplastic tissues. Here, combining NMR, computational, and biochemical methods, we show that the succinimide ring contained in 2 contributes to stabilizing interactions with αvß3, an integrin overexpressed in the tumor vasculature. Furthermore, we demonstrate that various cyclopeptides containing the isoDGR sequence embedded in different molecular scaffolds do not induce αvß3 allosteric activation and work as pure integrin antagonists. These results could be profitably exploited for the rational design of novel isoDGR-based ligands and tumor-targeting molecules with improved αvß3-binding properties and devoid of adverse integrin-activating effects.


Subject(s)
Integrin alphaVbeta3/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Succinimides/chemistry , Allosteric Regulation , Binding, Competitive , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , HEK293 Cells , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/chemistry , Magnetic Resonance Spectroscopy , Melanoma/pathology , Molecular Docking Simulation , Peptides, Cyclic/pharmacology , Protein Conformation , Snake Venoms/pharmacology , Structure-Activity Relationship , Tyrosine/metabolism
5.
Oncoimmunology ; 4(10): e1041700, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26451306

ABSTRACT

NGR-TNF is a vascular targeting agent in advanced clinical development, coupling tumor necrosis factor-α (TNF) with the CNGRCG peptide, which targets a CD13 isoform specifically expressed by angiogenic vessels. Antitumor efficacy of NGR-TNF has been described in different transplantation tumor models. Nevertheless, the mechanism underlying its activity is not fully understood. In the wild type and in the immunodeficient (RAG-/-) RIP1-Tag2 models of multistage pancreatic carcinogenesis, we demonstrate that CD13 is highly expressed on endothelial cells of hyperplastic and angiogenic islets, whereas its expression is down regulated in tumors where it partially colocalize with pericytes. In vivo CNGRCG peptides coupled to fluorescent nanoparticles (quantum dots) bind to CD13 and colocalize with anti-CD31, in pancreatic islets. At early stage, low doses of NGR-murine (m)TNF have a direct cytotoxic effect inducing endothelial cell apoptosis, reducing vessel density and eventually inhibiting the development of angiogenic islets. At a later stage, NGR-mTNF is able to reduce tumor growth inducing vascular normalization, exclusively when treatment is carried out in the immunocompetent mice. Interestingly, NGR-mTNF-treated tumors from these mice are characterized by CD8+ T cell infiltration. At molecular level, overexpression of genes involved in vessels normalization was detected only in NGR-mTNF-treated tumors from immunocompetent mice. These findings identified a new mechanism of action of NGR-mTNF, providing support for the development of new therapeutic strategies combining chemotherapy or active/adoptive immunotherapies to low dose NGR-TNF treatment.

6.
Clin Exp Metastasis ; 32(3): 289-300, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25648442

ABSTRACT

Tumor vessels are an attractive target for cancer therapy, including metastasis treatment. Angiogenesis inhibitors targeting the VEGF signalling pathway have proven to be efficacious in preclinical cancer models and in clinical trials. However, angiogenesis inhibition concomitantly elicits tumor adaptation and progression to stages of greater malignancy, with heightened invasiveness and in some cases increased distant metastasis. Here, we investigated whether NGR-TNF, a vascular targeting agent in phase III clinical development, coupling the CNGRCG angiogenic vessel-homing peptide with TNF-α, has an effect on metastasis in a model of murine breast cancer, which spontaneously metastasize to lungs, and on the growth of experimental melanoma lung metastasis. We report that NGR-TNF does not increase cancer invasiveness, as other antiangiogenics agents do, but controls metastatic growth in both models, both when administered as primary treatment and in adjuvant settings, improving the overall survival of metastasis-bearing mice.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Mammary Neoplasms, Animal/drug therapy , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/therapeutic use , Animals , Female , Flow Cytometry , Immunoenzyme Techniques , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/mortality , Mammary Neoplasms, Animal/pathology , Melanoma, Experimental/mortality , Melanoma, Experimental/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Rate , Tumor Cells, Cultured
7.
Angew Chem Int Ed Engl ; 51(31): 7702-5, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22718573

ABSTRACT

Ain't got that swing(-out): The cyclopeptide isoDGR is emerging as a new αvß3 integrin binding motif. Agreement between the results of computational and biochemical studies reveals that isoDGR-containing cyclopeptides are true αvß3 integrin antagonists that block αvß3 in its inactive conformation (see scheme). isoDGR-based ligands may give αvß3 antagonists without paradoxical effects.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Molecular Dynamics Simulation , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Allosteric Regulation , Integrin alphaVbeta3/metabolism , Models, Molecular , Oligopeptides/chemistry , Peptides, Cyclic/chemistry
8.
Nat Med ; 16(1): 98-105, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20037595

ABSTRACT

Sterol metabolism has recently been linked to innate and adaptive immune responses through liver X receptor (LXR) signaling. Whether products of sterol metabolism interfere with antitumor responses is currently unknown. Dendritic cells (DCs) initiate immune responses, including antitumor activity after their CC chemokine receptor-7 (CCR7)-dependent migration to lymphoid organs. Here we report that human and mouse tumors produce LXR ligands that inhibit CCR7 expression on maturing DCs and, therefore, their migration to lymphoid organs. In agreement with this observation, we detected CD83(+)CCR7(-) DCs within human tumors. Mice injected with tumors expressing the LXR ligand-inactivating enzyme sulfotransferase 2B1b (SULT2B1b) successfully controlled tumor growth by regaining DC migration to tumor-draining lymph nodes and by developing overt inflammation within tumors. The control of tumor growth was also observed in chimeric mice transplanted with bone marrow from mice lacking the gene encoding LXR-alpha (Nr1h3(-/-) mice) Thus, we show a new mechanism of tumor immunoescape involving products of cholesterol metabolism. The manipulation of this pathway could restore antitumor immunity in individuals with cancer.


Subject(s)
Dendritic Cells/physiology , Neoplasms, Experimental/physiopathology , Orphan Nuclear Receptors/physiology , Receptors, CCR7/biosynthesis , Tumor Escape/physiology , Animals , Antigens, CD/immunology , Cell Line, Tumor , Cell Movement/physiology , Gene Expression Regulation/physiology , Humans , Immunoglobulins/immunology , Liver X Receptors , Lymph Nodes/immunology , Lymph Nodes/physiopathology , Melanoma/immunology , Melanoma/physiopathology , Membrane Glycoproteins/immunology , Mice , Neoplasms, Experimental/immunology , Signal Transduction/physiology , Tumor Escape/immunology , CD83 Antigen
9.
J Leukoc Biol ; 84(3): 871-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18515328

ABSTRACT

Dendritic cell (DC) migration to secondary lymphoid organs is a crucial step to initiate adaptive immune responses. This step requires the expression of a functional CCR7 chemokine receptor on DC undergoing maturation. Here, we show that the natural retinoid 9-cis retinoic acid (9cRA) and the synthetic retinoid fenretinide (4-HPR) specifically inhibit the functional up-regulation of CCR7 on maturing human DCs, without affecting early steps of DC maturation. As a consequence, mature DCs do not migrate in vitro toward the chemokine CCL19. Importantly, 4-HPR and 9cRA by inhibiting the expression of CCR7 on bone marrow-derived murine DCs dampen their in vivo migration to draining lymph nodes. 4-HPR also inhibits the expression of the chemokine receptors CXCR4, therefore, impairing in vitro migration of human DCs to CXCL12 and inhibiting in vivo the CXCR4-dependent migration of the posterior lateral line primordium (PLLp) in zebrafish embryos. Taken together, these data highlight a novel function of retinoids and suggest the possibility of using retinoids to treat inflammatory or autoimmune diseases.


Subject(s)
Cell Movement/drug effects , Dendritic Cells/drug effects , Embryo, Nonmammalian/drug effects , Fenretinide/pharmacology , Receptors, CCR7/physiology , Receptors, CXCR4/physiology , Tretinoin/pharmacology , Zebrafish Proteins/physiology , Alitretinoin , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Survival/drug effects , Cells, Cultured , Chemokine CXCL12/metabolism , Chemotaxis , Dendritic Cells/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , In Vitro Techniques , Interleukin-6/metabolism , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish/physiology
10.
J Leukoc Biol ; 84(1): 199-206, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18456818

ABSTRACT

Heat shock proteins (HSPs) are potent inducers of an antigen-specific immunological response. A role of chaperon of immunogenic peptides and a direct effect on APC activation and function have been described. However, the signal transduction events involved in the activation of human APCs are poorly characterized. We investigated, using human monocyte-derived dendritic cells (DCs), the signal transduction pathways activated by a human recombinant HSP70 (r)HSP70 purified from eukaryotic cells. rHSP70 effectively induced a partial maturation of DCs in vitro and a significant increase in the titers of antigen-specific IgG when used as a vaccine adjuvant in vivo. rHSP70 did not desensitize human DCs to LPS stimulation and retained its adjuvant properties in C3H/HeJ mice, which are LPS-resistant as a result of a mutation in TLR-4, ruling out the potential interference of LPS contamination. Effects on DC maturation and in vivo functions correlate to the ability of rHSP70 to activate IkappaB-alpha/NF-kappaB and ERK1/2 pathways in human DCs. No activation of p38 was induced in the same experimental conditions. Our data suggest that the IkappaB-alpha/NF-kappaB pathway has a critical role in the partial maturation of DCs induced by rHSP70.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , HSP70 Heat-Shock Proteins/pharmacology , Recombinant Proteins/pharmacology , Animals , Antigens , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/isolation & purification , Humans , Mice , NIH 3T3 Cells , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Signal Transduction/drug effects , Solubility/drug effects
11.
J Immunol ; 174(12): 7506-15, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15944249

ABSTRACT

High mobility group box 1 (HMGB1) is an abundant and conserved nuclear protein that is released by necrotic cells and acts in the extracellular environment as a primary proinflammatory signal. In this study we show that human dendritic cells, which are specialized in Ag presentation to T cells, actively release their own HMGB1 into the extracellular milieu upon activation. This secreted HMGB1 is necessary for the up-regulation of CD80, CD83, and CD86 surface markers of human dendritic cells and for IL-12 production. The HMGB1 secreted by dendritic cells is also required for the clonal expansion, survival, and functional polarization of naive T cells. Using neutralizing Abs and receptor for advanced glycation end product-deficient (RAGE(-/-)) cells, we demonstrate that RAGE is required for the effect of HMGB1 on dendritic cells. HMGB1/RAGE interaction results in downstream activation of MAPKs and NF-kappaB. The use of an ancient signal of necrosis, HMGB1, by dendritic cells to sustain their own maturation and for activation of T lymphocytes represents a profitable evolutionary mechanism.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , HMGB1 Protein/metabolism , Lymphocyte Activation/physiology , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/physiology , Active Transport, Cell Nucleus/physiology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/physiology , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Clone Cells , Coculture Techniques , Cytosol/metabolism , Dendritic Cells/cytology , Extracellular Signal-Regulated MAP Kinases/physiology , Extracellular Space/metabolism , Growth Inhibitors/pharmacology , HMGB1 Protein/physiology , Humans , Immune Sera/pharmacology , Interleukin-12/biosynthesis , NF-kappa B/physiology , Receptor for Advanced Glycation End Products , Resting Phase, Cell Cycle/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Th1 Cells/cytology , Th1 Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/physiology
12.
J Biol Chem ; 280(14): 14264-71, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15671028

ABSTRACT

Polymyxin B is a lipopolysaccharide binding antibiotic used to inactivate potential lipopolysaccharide contaminations when evaluating the activity of different agents on innate immune cells. We report that polymyxin B is able to induce directly in monocyte-derived human dendritic cells (DCs) several functional and molecular modifications characteristic of DCs undergoing a maturation process. DCs incubated with polymyxin B up-regulate the expression of HLA class I and II, the co-stimulatory CD86 molecule, and show an increase in the fraction of adherent cells at short time, which persist at 48 h of incubation. Adhesion to the plate was required for the polymyxin B-induced DCs maturation. A transient activation of IkappaB-alpha/NF-kappaB and ERK1/2 pathways at short time and a further ERK1/2 activation at long term were also detected. Neither up-regulation of the maturation marker CD83 nor activation of p38 nor induction of cytokines secretion was observed in DCs treated with polymyxin B. We demonstrated that inhibition of IkappaB-alpha/NF-kappaB pathway abolishes polymyxin B effects. ERK1/2 inhibition instead allowed DCs treated with polymyxin B to progress in their maturation process as revealed by the increased up-regulation of the CD83 co-stimulatory molecules, the activation of p38, and the reduced adhesion to culture plates at 48 h of incubation. Our results indicate that polymyxin B induces a partial maturation of human DCs through increased adhesion to a substrate and activation of the IkappaB-alpha/NF-kappaB pathway. The increased ERK1/2 activation observed, even though correlating with the initial phases of the maturation process, actually inhibits the occurrence of full maturation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/physiology , Polymyxin B/pharmacology , Signal Transduction/physiology , Antigens, CD/genetics , Antigens, CD/metabolism , B7-2 Antigen , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Enzyme Activation , Enzyme Inhibitors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/metabolism , Genes, MHC Class I , Genes, MHC Class II , HLA Antigens/genetics , HLA Antigens/metabolism , Humans , I-kappa B Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Tosylphenylalanyl Chloromethyl Ketone/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
EMBO Rep ; 5(8): 825-30, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15272298

ABSTRACT

Immune responses against pathogens require that microbial components promote the activation of antigen-presenting cells (APCs). Autoimmune diseases and graft rejections occur in the absence of pathogens; in these conditions, endogenous molecules, the so-called 'innate adjuvants', activate APCs. Necrotic cells contain and release innate adjuvants; necrotic cells also release high-mobility group B1 protein (HMGB1), an abundant and conserved constituent of vertebrate nuclei. Here, we show that necrotic HMGB1(-/-) cells have a reduced ability to activate APCs, and HMGB1 blockade reduces the activation induced by necrotic wild-type cell supernatants. In vivo, HMGB1 enhances the primary antibody responses to soluble antigens and transforms poorly immunogenic apoptotic lymphoma cells into efficient vaccines.


Subject(s)
Adjuvants, Immunologic , HMGB1 Protein/immunology , Necrosis , Animals , Antigens, CD/immunology , Cell Line , Dendritic Cells/immunology , Fibroblasts/cytology , Fibroblasts/physiology , HMGB1 Protein/genetics , HMGB1 Protein/isolation & purification , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Cancer Immunol Immunother ; 53(6): 519-24, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14727086

ABSTRACT

The search for alternative strategies of therapy remains a major issue for most neoplastic diseases. The expression of several tumor antigens makes human rhabdomyosarcomas, which are the most frequent form of soft tissue tumor in children, a good candidate for tumor-specific immunotherapy. To assess the feasibility of this approach, we evaluated the ability of rhabdomyosarcoma cell lines to process and present the MAGE-A tumor antigens to effectors of the immune system. To this end, we investigated recognition of MAGE-A-positive rhabdomyosarcoma cells by HLA-B*3701-restricted T cells specific for a MAGE-A-derived peptide. Low level of HLA expression impaired recognition of the tumor cells. Therefore, to obtain HLA expression avoiding the use of IFN-gamma and TNF-alpha, which could affect the proteasome activity, a rhabdomyosarcoma line was transduced by a retroviral vector encoding the HLA-B*3701 allele. Recognition of the infected cells was then observed also in the absence of IFN-gamma and TNF-alpha treatment, thus demonstrating that rhabdomyosarcoma cells were indeed able to naturally process and present the MAGE-A antigens. These results demonstrate that rhabdomyosarcoma cells expressing MAGE-A can be targets of tumor-specific effectors, suggesting the feasibility of clinical protocols of specific immunotherapy also for the treatment of rhabdomyosarcoma.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy , Neoplasm Proteins/immunology , Rhabdomyosarcoma/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/genetics , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Feasibility Studies , HLA-B Antigens/genetics , HLA-B Antigens/pharmacology , Humans , Interferon-gamma/immunology , Melanoma/immunology , Melanoma/pathology , Melanoma-Specific Antigens , Neoplasm Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/pathology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...