Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nat Biotechnol ; 41(2): 239-251, 2023 02.
Article in English | MEDLINE | ID: mdl-36203013

ABSTRACT

Post-translational modification (PTM) of antigens provides an additional source of specificities targeted by immune responses to tumors or pathogens, but identifying antigen PTMs and assessing their role in shaping the immunopeptidome is challenging. Here we describe the Protein Modification Integrated Search Engine (PROMISE), an antigen discovery pipeline that enables the analysis of 29 different PTM combinations from multiple clinical cohorts and cell lines. We expanded the antigen landscape, uncovering human leukocyte antigen class I binding motifs defined by specific PTMs with haplotype-specific binding preferences and revealing disease-specific modified targets, including thousands of new cancer-specific antigens that can be shared between patients and across cancer types. Furthermore, we uncovered a subset of modified peptides that are specific to cancer tissue and driven by post-translational changes that occurred in the tumor proteome. Our findings highlight principles of PTM-driven antigenicity, which may have broad implications for T cell-mediated therapies in cancer and beyond.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Humans , Protein Processing, Post-Translational/genetics , Peptides/genetics , Antigens , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Neoplasms/genetics
2.
Int J Cancer ; 151(1): 107-119, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35179790

ABSTRACT

Lynch Syndrome (LS) is an autosomal dominant genetic condition that causes a high risk of colorectal cancer. The hallmark of LS is genetic instability as a result of mismatch repair (MMR) deficiency, particularly in repetitive low complexity regions called microsatellites (MS). MLH1-/- mice deficient in MMR are prone to developing tumors in the colon, upon oral administration of dextran sodium sulfate (DSS), at a rate of more than 70%. Using this LS mouse model, we found a novel tumor neo-antigen from a deletion mutation of the coding MS in the SENP6 gene that prevented tumorigenesis or hindered tumor growth rate in immunized mice. This was accomplished via high throughput exome sequencing of DSS-induced colorectal tumors in the MLH1-/- mice and predicting the most highly immunogenic mutant gene products processed and presented as antigens in C57BL/6 MHC-I molecules. Throughout our study, we were able to prove the validity of the vaccine by analyzing the colorectal tumors in immunized DSS-treated mice using either our epitope, called Sp6D1, or an unrelated peptide as a negative control. Tumors developed in this context were found to be antigenic and Sp6D1-specific CD8+ tumor infiltrating lymphocytes were detected by flow cytometry and cytotoxic T lymphocytes (CTL) killing assays. Additionally, immunohistochemistry showed that tumor-adjacent tertiary lymphoid organs were a potentially significant source of CD8+ lymphocytes. Altogether, our results indicate that there may be a protective effect to patients carrying LS mutations through the induction of a peptide-specific CTL response from the use of neoepitope vaccination.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , Vaccines , Animals , Antigens, Neoplasm/genetics , Brain Neoplasms , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/prevention & control , Cysteine Endopeptidases/genetics , DNA Mismatch Repair/genetics , Humans , Mice , Mice, Inbred C57BL , Microsatellite Instability , MutL Protein Homolog 1/genetics , Neoplastic Syndromes, Hereditary
3.
Eur J Immunol ; 51(6): 1505-1518, 2021 06.
Article in English | MEDLINE | ID: mdl-33835499

ABSTRACT

A T-cell receptor (TCR) with optimal avidity to a tumor antigen can be used to redirect T cells to eradicate cancer cells via adoptive cell transfer. Cancer testis antigens (CTAs) are attractive targets because they are expressed in the testis, which is immune-privileged, and in the tumor. However, CTAs are self-antigens and natural TCRs to CTAs have low affinity/avidity due to central tolerance. We previously described a method of directed evolution of TCR avidity using somatic hypermutation. In this study, we made several improvements to this method and enhanced the avidity of the hT27 TCR, which is specific for the cancer testis antigen HLA-A2-MAGE-A1278-286 . We identified eight point mutations with varying degrees of improved avidity. Human T cells transduced with TCRs containing these mutations displayed enhanced tetramer binding, IFN-γ and IL2 production, and cytotoxicity. Most of the mutations have retained specificity, except for one mutant with extremely high avidity. We demonstrate that somatic hypermutation is capable of optimizing avidity of clinically relevant TCRs for immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Cells, Cultured , Central Tolerance , Cytotoxicity, Immunologic , HLA-A2 Antigen/metabolism , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Point Mutation/genetics , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Somatic Hypermutation, Immunoglobulin , T-Lymphocytes/transplantation
4.
Cell ; 179(1): 219-235.e21, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31522890

ABSTRACT

Although clonal neo-antigen burden is associated with improved response to immune therapy, the functional basis for this remains unclear. Here we study this question in a novel controlled mouse melanoma model that enables us to explore the effects of intra-tumor heterogeneity (ITH) on tumor aggressiveness and immunity independent of tumor mutational burden. Induction of UVB-derived mutations yields highly aggressive tumors with decreased anti-tumor activity. However, single-cell-derived tumors with reduced ITH are swiftly rejected. Their rejection is accompanied by increased T cell reactivity and a less suppressive microenvironment. Using phylogenetic analyses and mixing experiments of single-cell clones, we dissect two characteristics of ITH: the number of clones forming the tumor and their clonal diversity. Our analysis of melanoma patient tumor data recapitulates our results in terms of overall survival and response to immune checkpoint therapy. These findings highlight the importance of clonal mutations in robust immune surveillance and the need to quantify patient ITH to determine the response to checkpoint blockade.


Subject(s)
Genetic Heterogeneity/radiation effects , Melanoma/genetics , Melanoma/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Ultraviolet Rays/adverse effects , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cohort Studies , Disease Models, Animal , Female , Humans , Lymphocytes, Tumor-Infiltrating , Melanoma/mortality , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mutation/radiation effects , Phylogeny , Skin Neoplasms/mortality , Survival Rate , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
5.
Int J Cancer ; 145(10): 2816-2826, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31381134

ABSTRACT

Adoptive transfer of T cells that have been genetically modified to express an antitumor T-cell receptor (TCR) is a potent immunotherapy, but only if TCR avidity is sufficiently high. Endogenous TCRs specific to shared (self) tumor-associated antigens (TAAs) have low affinity due to central tolerance. Therefore, for effective therapy, anti-TAA TCRs with higher and optimal avidity must be generated. Here, we describe a new in vitro system for directed evolution of TCR avidity using somatic hypermutation (SHM), a mechanism used in nature by B cells for antibody optimization. We identified 44 point mutations to the Pmel-1 TCR, specific for the H-2Db -gp10025-33 melanoma antigen. Primary T cells transduced with TCRs containing two or three of these mutations had enhanced activity in vitro. Furthermore, the triple-mutant TCR improved in vivo therapy of tumor-bearing mice, which exhibited improved survival, smaller tumors and delayed or no relapse. TCR avidity maturation by SHM may be an effective strategy to improve cancer immunotherapy.


Subject(s)
Directed Molecular Evolution/methods , Melanoma, Experimental/therapy , Receptors, Antigen, T-Cell/genetics , Skin Neoplasms/therapy , gp100 Melanoma Antigen/immunology , Animals , Cell Line, Tumor , HEK293 Cells , Histocompatibility Antigen H-2D , Humans , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed/methods , Peptides/immunology , Point Mutation , Proof of Concept Study , Receptors, Antigen, T-Cell/immunology , Skin Neoplasms/immunology
6.
Int J Cancer ; 144(4): 909-921, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30106470

ABSTRACT

For many years, clinicians and scientists attempt to develop methods to stimulate the immune system to target malignant cells. Recent data suggest that effective cancer vaccination requires combination immunotherapies to overcome tumor immune evasion. Through presentation of both MHC-I and II molecules, DCs-based vaccine platforms are effective in generating detectable CD4 and CD8 T cell responses against tumor-associated antigens. Several platforms include DC transfection with mRNA of the desired tumor antigen. These DCs are then delivered to the host and elicit an immune response against the antigen of interest. We have recently established an mRNA genetic platform which induced specific CD8+ cytotoxic T cell response by DC vaccination against melanoma. In our study, an MHC-II mRNA DCs vaccine platform was developed to activate CD4+ T cells and to enhance the anti-tumor response. The invariant chain (Ii) was modified and the semi-peptide CLIP was replaced with an MHC-II binding peptide sequences of melanoma antigens. These chimeric MHC-II constructs are presented by DCs and induce proliferation of tumor specific CD4+ T cells. When administered in combination with the MHC-I platform into tumor bearing mice, these constructs were able to inhibit tumor growth, and improve mouse survival. Deciphering the immunological mechanism of action, we observed an efficient CTLs killing in addition to higher levels of Th1 and Th2 subsets in the groups immunized with a combination of the MHC-I and MHC-II constructs. These universal constructs can be applied in multiple combinations and offer an attractive opportunity to improve cancer treatment.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Cancer Vaccines/immunology , Histocompatibility Antigens Class II/immunology , Melanoma, Experimental/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Progression , Immunotherapy/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
7.
Oncoimmunology ; 7(7): e1445457, 2018.
Article in English | MEDLINE | ID: mdl-29900058

ABSTRACT

Despite melanoma immunogenicity and remarkable therapeutic effects of negative immune checkpoint inhibitors, a significant fraction of patients does not respond to current treatments. This could be due to limitations in tumor immunogenicity and profound immunosuppression in the melanoma microenvironment. Moreover, insufficient tumor antigen processing and presentation by dendritic cells (DC) may hamper the development of tumor-specific T cells. Using two genetically engineered mouse melanoma models (RET and BRAFV600E transgenic mice), in which checkpoint inhibitor therapy alone is not efficacious, we performed proof-of-concept studies with an improved, multivalent DC vaccination strategy based on our recently developed genetic mRNA cancer vaccines. The in vivo expression of multiple chimeric MHC class I receptors allows a simultaneous presentation of several melanoma-associated shared antigens tyrosinase related protein (TRP)-1, tyrosinase, human glycoprotein 100 and TRP-2. The DC vaccine induced a significantly improved survival in both transgenic mouse models. Vaccinated melanoma-bearing mice displayed an increased CD8 T cell reactivity indicated by a higher IFN-γ production and an upregulation of activation marker expression along with an attenuated immunosuppressive pattern of myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg). The combination of DC vaccination with ultra-low doses of paclitaxel or anti-PD-1 antibodies resulted in further prolongation of mouse survival associated with a stronger reduction of MDSC and Treg immunosuppressive phenotype. Our data suggest that an improved multivalent DC vaccine based on shared tumor antigens induces potent anti-tumor effects and could be combined with checkpoint inhibitors or targeting immunosuppressive cells to further improve their therapeutic efficiency.

8.
Nanomedicine ; 14(3): 835-847, 2018 04.
Article in English | MEDLINE | ID: mdl-29306001

ABSTRACT

Nanoparticulate vaccines are promising tools to overcome cancer immune evasion. However, a deeper understanding on nanoparticle-immune cell interactions and treatments regime is required for optimal efficacy. We provide a comprehensive study of treatment schedules and mode of antigen-association to nanovaccines on the modulation of T cell immunity in vivo, under steady-state and tumor-bearing mice. The coordinated delivery of antigen and two adjuvants (Monophosphoryl lipid A, oligodeoxynucleotide cytosine-phosphate-guanine motifs (CpG)) by nanoparticles was crucial for dendritic cell activation. A single vaccination dictated a 3-fold increase on cytotoxic memory-T cells and raised antigen-specific immune responses against B16.M05 melanoma. It generated at least a 5-fold increase on IFN-γ cytokine production, and presented over 50% higher lymphocyte count in the tumor microenvironment, compared to the control. The number of lymphocytes at the tumor site doubled with triple immunization. This lymphocyte infiltration pattern was confirmed in mammary huHER2 carcinoma, with significant tumor reduction.


Subject(s)
Breast Neoplasms/prevention & control , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Carcinogenesis/drug effects , Nanoparticles/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer Vaccines/chemistry , Carcinogenesis/metabolism , Carcinogenesis/pathology , Female , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Tumor Cells, Cultured
9.
Immunol Cell Biol ; 96(3): 284-297, 2018 03.
Article in English | MEDLINE | ID: mdl-29356071

ABSTRACT

Inflammation plays pivotal roles in different stages of tumor development. Screening for predisposing genetic abnormalities and understanding the roles these genes play in the crosstalk between immune and cancer cells will provide new targets for cancer therapy and prevention. The interferon inducible transmembrane (IFITM) genes are involved in pathogenesis of the gastro-intestinal tract. We aimed at delineating the role of IFITM3 in colonic epithelial homeostasis, inflammation and colitis-associated tumorigenesis using IFITM3-deficient mice. Chemical induction of colitis in IFITM3-deficient mice results in significantly increased clinical signs of inflammation and induction of invasive tumorigenesis. Bone marrow transplantation showed that cells of the hematopoietic system are responsible for colitis deterioration. In these mice, impaired cytokine expression skewed inflammatory response toward pathogenic Th17 with reduced expression of the anti-inflammatory cytokine IL10 during the recovery phase. Intriguingly, mice lacking the entire IFITM locus developed spontaneous chronic colitis from the age of 14 weeks. Sequencing the 16S rRNA of naïve mice lacking IFITM3 gene, or the entire locus containing five IFITM genes, revealed these mice had significant bacterial differences from their wild-type littermates. Our novel results provide strong evidence for the essential role of IFITM genes in ameliorating colitis and colitis-associated tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Colitis/immunology , Colitis/microbiology , Immunity , Inflammation/genetics , Membrane Proteins/genetics , Microbiota , Animals , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Disease Progression , Disease Susceptibility , Hematopoiesis , Immunity/genetics , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Microbiota/genetics , Myeloid Cells/pathology
10.
Oncoimmunology ; 5(6): e1160183, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471629

ABSTRACT

Malignant melanoma is characterized by a rapid progression, metastasis to distant organs and resistance to chemo and radiotherapy. Although melanoma is capable of eliciting an immune response, the disease progresses and the overall results of immunotherapeutic clinical studies are not satisfactory. Recently, we have developed a novel genetic platform for improving an induction of peptide-specific CD8(+) T cells by dendritic cell (DC) based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at the N-terminus and to the cytosolic domain of TLR4 at the C-terminus. In vitro transcribed mRNA transfection of antigen-presenting cells (APCs) resulted in an efficient coupling of peptide presentation and cell activation. In this research, we utilize the chimeric platform to induce an immune response in ret transgenic mice that spontaneously develop malignant skin melanoma and to examine its effect on the overall survival of tumor-bearing mice. Following immunization with chimeric construct system, we observe a significantly prolonged survival of tumor-bearing mice as compared to the control group. Moreover, we see elevations in the frequency of CD62L(hi)CD44(hi) central and CD62L(lo)CD44(hi) effector memory CD8(+) T-cell subsets. Importantly, we do not observe any changes in frequencies of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the vaccinated groups. Our data suggest that this novel vaccination approach could be efficiently applied for the immunotherapy of malignant melanoma.

11.
Mol Ther ; 23(8): 1391-1400, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25997427

ABSTRACT

Recently, we have developed a novel genetic platform for improving dendritic cell (DC) induction of peptide-specific CD8 T cells, based on membrane-anchored ß2-microglobulin (ß2m) linked to a selected antigenic peptide at its N-terminus and to the cytosolic domain of toll-like receptor (TLR)4 C-terminally. In vitro transcribed mRNA transfection of antigen presenting cells resulted in polypeptides that efficiently coupled peptide presentation to cellular activation. In the present study, we evaluated the immunogenicity of such constructs in mRNA-transfected immature murine bone marrow-derived DCs. We show that the encoded peptide ß2m-TLR4 products were expressed at the cell surface up to 72 hours and stimulated the maturation of DCs. In vivo, these DCs prompted efficient peptide-specific T-cell activation and target cell killing which were superior to those induced by peptide-loaded, LPS-stimulated DCs. This superiority was also evident in the ability to protect mice from tumor progression following the administration of B16F10.9 melanoma cells and to inhibit the development of pre-established B16F10.9 tumors. Our results provide evidence that the products of two recombinant genes encoding for peptide-hß2m-TLR4 and peptide-hß2m-K(b) expressed from exogenous mRNA can cooperate to couple Major Histocompatibility Complex (MHC-I) peptide presentation to TLR-mediated signaling, offering a safe, economical and highly versatile genetic platform for a novel category of CTL-inducing vaccines.


Subject(s)
Dendritic Cells/cytology , Histocompatibility Antigens Class I/metabolism , Peptides/chemistry , RNA, Messenger/metabolism , Toll-Like Receptor 4/metabolism , beta 2-Microglobulin/metabolism , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/cytology , Antigens, Neoplasm/chemistry , Antineoplastic Agents/chemistry , Bone Marrow Cells/cytology , Cancer Vaccines/immunology , Cell Membrane/metabolism , Cloning, Molecular , Culture Media , Cytosol/metabolism , Female , Humans , Lipopolysaccharides/chemistry , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Signal Transduction
12.
Cancer Immunol Immunother ; 63(4): 369-80, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24452202

ABSTRACT

Cryoablation is a low-invasive surgical procedure for management of malignant tumors. Tissue destruction is obtained by repeated deep freezing and thawing and results in coagulative necrosis and in apoptosis. This procedure induces the release of tumor-associated antigens and proinflammatory factors into the microenvironment. Local administration of immature dendritic cells (DCs) potentiates the immune response induced by cryoablation. To further augment the induction of long-lasting antitumor immunity, we investigated the clinical value of combining cryoimmunotherapy consisting of cryoablation and inoculation of immature DCs with administration of the immune adjuvant, CpG oligodeoxynucleotides. Injection of the murine Lewis lung carcinoma, D122-luc-5.5 that expresses the luciferase gene, results in spontaneous metastases, which can be easily monitored in vivo. The local tumor was treated by the combined treatment. The clinical outcome was assessed by monitoring tumor growth, metastasis in distant organs, overall survival, and protection from tumor recurrence. The nature of the induced T cell responses was analyzed. Combined cryoimmunotherapy results in reduced tumor growth, low metastasis and significantly prolonged survival. Moreover, this treatment induces antitumor memory that protected mice from rechallenge. The underlying suggested mechanisms are the generation of tumor-specific type 1 T cell responses, subsequent induction of cytotoxic T lymphocytes, and generation of systemic memory. Our data highlight the combined cryoimmunotherapy as a novel antitumor vaccine with promising preclinical results. Adjustment of this technique into practice will provide the therapeutic benefits of both, ablation of the primary tumor and induction of robust antitumor and antimetastatic immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Carcinoma, Lewis Lung/therapy , CpG Islands , Cryosurgery/methods , Dendritic Cells/immunology , Immunotherapy/methods , Lung Neoplasms/therapy , Oligodeoxyribonucleotides/therapeutic use , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/secondary , Carcinoma, Lewis Lung/surgery , Cells, Cultured , Combined Modality Therapy , Dendritic Cells/transplantation , Foot , Immunologic Memory , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/surgery , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Oligodeoxyribonucleotides/administration & dosage , Recurrence , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/immunology
13.
Ann N Y Acad Sci ; 1283: 87-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23387369

ABSTRACT

Cytotoxic T lymphocytes (CTLs) are the major effector arm of the immune system against tumors. Many tumor-associated antigens (TAAs), known today as potential rejection antigens, were identified by their ability to induce CTL responses. CTLs utilize their clonotypic T cell receptor (TCR) to recognize short antigenic peptides presented on major histocompatibility complex (MHC)-I proteins. These consist of a membrane-attached α heavy chain, which forms the peptide binding pocket, and a noncovalently associated ß2m light chain, not anchored to the cell membrane. CTL activation requires that antigenic peptides be presented initially on professional antigen presenting cells (APCs), primarily dendritic cells (DCs). Autologous DCs are a powerful tool for the induction of antitumor responses and are thus widely explored as vehicles for cancer vaccines. Although encouraging evidence for the induction of tumor-specific CTLs by ex vivo-manipulated DCs came from numerous animal studies, reproducible objective clinical response in human trials is yet to be demonstrated.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Animals , Cancer Vaccines/therapeutic use , Drug Design , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/therapeutic use , Mice , Protein Engineering , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
14.
PLoS One ; 8(2): e55583, 2013.
Article in English | MEDLINE | ID: mdl-23383339

ABSTRACT

Identification and quantification of immunogenic peptides and tumor-derived epitopes presented on MHC-I molecules are essential for basic studies and vaccines generation. Although lymphocytes derived from transgenic mice can serve as sensitive detectors of processes of antigen presentation and recognition, they are not always available. The use of cell lines might be extremely useful. In this study, we generated a lacZ inducible CD8⁺ hybridoma (BUSA14) capable of recognizing both human and mouse gp10025₋33 melanoma antigens presented on dendritic and tumor cell lines. This hybridoma expresses a variety of membranal T cell markers and secretes IL-2 and TNFα. Thus, BUSA14 offers a quantifiable, cheap and straightforward tool for studying peptide presentation by MHC-I molecules on the cell surface.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Histocompatibility Antigens Class I/metabolism , Hybridomas/metabolism , gp100 Melanoma Antigen/metabolism , Animals , Cell Line, Tumor , Dendritic Cells/metabolism , Flow Cytometry , Humans , Hybridomas/cytology , Interleukin-2/metabolism , Lac Operon , Mice , Tumor Necrosis Factor-alpha/metabolism
15.
PLoS One ; 7(6): e39193, 2012.
Article in English | MEDLINE | ID: mdl-22723962

ABSTRACT

Mice are exceedingly sensitive to intra-peritoneal (IP) challenge with some virulent pneumococci (LD50 = 1 bacterium). To investigate how peripheral contact with bacterial capsular polysaccharide (PS) antigen can induce resistance, we pulsed bone marrow dendritic cells (BMDC) of C57BL/6 mice with type 4 or type 3 PS, injected the BMDC intra-foot pad (IFP) and challenged the mice IP with supra-lethal doses of pneumococci. We examined the responses of T cells and B cells in the draining popliteal lymph node and measured the effects on the bacteria in the peritoneum and blood. We now report that: 1) The PS co-localized with MHC molecules on the BMDC surface; 2) PS-specific T and B cell proliferation and IFNγ secretion was detected in the draining popliteal lymph nodes on day 4; 3) Type-specific resistance to lethal IP challenge was manifested only after day 5; 4) Type-specific IgM and IgG antibodies were detected in the sera of only some of the mice, but B cells were essential for resistance; 5) Control mice vaccinated with a single injection of soluble PS did not develop a response in the draining popliteal lymph node and were not protected; 6) Mice injected with unpulsed BMDC also did not resist challenge: In unprotected mice, pneumococci entered the blood shortly after IP inoculation and multiplied exponentially in both blood and peritoneum killing the mice within 20 hours. Mice vaccinated with PS-pulsed BMDC trapped the bacteria in the peritoneum. The trapped bacteria proliferated exponentially IP, but died suddenly at 18-20 hours. Thus, a single injection of PS antigen associated with intact BMDC is a more effective vaccine than the soluble PS alone. This model system provides a platform for studying novel aspects of PS-targeted vaccination.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Polysaccharides, Bacterial/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes/immunology , Animals , Antibody Specificity/immunology , Antigen Presentation/immunology , Antigens, CD1d/immunology , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Interferon-gamma/biosynthesis , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Peritoneum/immunology , Peritoneum/microbiology , Pneumococcal Vaccines/immunology , Polysaccharides, Bacterial/administration & dosage , Receptors, Antigen, T-Cell/deficiency , Receptors, Antigen, T-Cell/immunology , Time Factors
16.
J Immunol ; 187(10): 5452-62, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998458

ABSTRACT

Gliomas that grow uninhibited in the brain almost never metastasize outside the CNS. The rare occurrences of extracranial metastasis are usually associated with a suppressed immune system. This observation raises the possibility that some gliomas might not grow outside the CNS due to an inherent immune response, We report in this study that the highly malignant F98 Fischer rat undifferentiated glioma, which grows aggressively in the brain, spontaneously regresses when injected live s.c. We found that this regression is immune-mediated and that it markedly enhances the survival or cures rats challenged with the same tumor intracranially either before or after the s.c. live-cell treatment. Adoptive transfer experiments showed the effect was immune-mediated and that the CD8 T cell fraction, which exhibited direct tumor cytotoxicity, was more effective than the CD4 T cell fraction in mediating resistance to intracranial challenge of naive rats. Brain tumors from treated rats exhibited enhanced CD3(+)CD8(+)CD4(-) and CD3(+)CD4(+)CD8(-) T cell infiltration and IFN-γ secretion. The results in the F98 glioma were corroborated in the Lewis rat CNS-1 astrocytoma. In both tumor models, s.c. treatment with live cells was significantly better than immunization with irradiated cells. We propose in this study a location-based immunotherapeutic phenomenon we term "split immunity": a tumor that thrives in an immune-privileged site may be inhibited by injecting live, unmodified tumor cells into a site that is not privileged, generating protective immunity that spreads back to the privileged site. Split immunity could explain several long-standing paradoxes regarding the lack of overt extracranial metastasis in patients with primary brain tumors.


Subject(s)
Astrocytoma/immunology , Astrocytoma/prevention & control , Cell Differentiation/immunology , Glioma/immunology , Glioma/prevention & control , Graft Rejection/immunology , Skin Neoplasms/immunology , Skin Neoplasms/prevention & control , Animals , Astrocytoma/pathology , Cell Survival/immunology , Cells, Cultured , Clone Cells , Coculture Techniques , Dose-Response Relationship, Immunologic , Female , Glioma/pathology , Graft Rejection/pathology , Injections, Intraventricular , Injections, Subcutaneous , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Skin Neoplasms/pathology , Tumor Cells, Cultured
17.
Int Immunol ; 23(7): 453-61, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21652516

ABSTRACT

Priming of naive CD8 T cells by dendritic cells (DCs) entails both effective antigen presentation on MHC class I products and co-stimulatory signaling. Their optimal coupling is a major goal in the development of CTL-inducing vaccines. We recently reported that a membranal derivative of the invariant MHC-I light chain, ß(2)-microglobulin (ß(2)m), markedly stabilizes MHC-I molecules and can serve as a universal platform for exceptional presentation of genetically linked peptides. To test whether it is possible to equip the resulting MHC-I complexes with an inherent ability to activate antigen-presenting cells, we engrafted the intracellular Toll/IL-1 receptor domain of mouse Toll-like receptor (TLR) 4 or TLR2 onto the peptide-ß(2)m scaffold. We evaluated the level of peptide presentation and status of cell activation conferred by such constructs in stably transfected mouse RAW264.7 macrophages and mRNA-transfected mouse DC2.4 DCs. We show that the encoded peptide-ß(2)m-TLR polypeptides are expressed at the cell surface, pair with endogenous heavy chains, stabilize MHC-I products, prompt efficient peptide-specific T-cell recognition and confer a constitutively activated phenotype on the transfected cells, as judged by the up-regulation of pro-inflammatory genes and surface co-stimulatory molecules. Our results provide evidence that the product of a single recombinant gene can couple MHC peptide presentation to TLR-mediated signaling and offer a safe, economical and highly versatile modality for a novel category of genetic CTL-inducing vaccines.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , H-2 Antigens/immunology , Peptides/immunology , Animals , Genetic Vectors/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Transfection , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
18.
J Autoimmun ; 35(2): 135-44, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20580204

ABSTRACT

T cell vaccination (TCV) with irradiated encephalitogenic T cells induces resistance to EAE. However, the fate of the encephalitogenic T cells in vivo following TCV has yet to be studied. Here we used anti-MBP encephalitogenic T cells that were transduced to express GFP to study the effects of TCV on these cells. In naïve rats or in control-vaccinated (Ova-GFP) rats injected i.v. with GFP-labeled effector cells, high numbers of effector T cells were found along with macrophages, CD8 T cells and Non-GFP CD4 cells in the spleens, parathymic lymph nodes (PTLN) and spinal cords. In contrast, the recipients that had been treated with TCV (anti-MBP T-cell lines) showed few if any GFP-labeled effector T cells throughout the disease (day 1-8) and their spinal cords were almost clear of macrophages, CD4 and CD8 cells. Splenocytes in the control groups secreted IFNgamma in response to MBP and showed high numbers of IFNgamma secreting CD4 and CD8 cells in their spinal cords at the disease peak. In the TCV-protected groups, splenocytes showed no reactivity to MBP but secreted IFNgamma in response to irradiated encephalitogenic T cells--an anti-idiotypic response. Thus, TCV leads to a marked decrease in the numbers of effector T cells in the CNS and lymphoid organs, to a marked reduction in the Th1 cytokine producing cells in the CNS, and to the appearance of T cells responsive to the anti-MBP effector T cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Myelin Basic Protein/metabolism , T-Lymphocytes/metabolism , Animals , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Line , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Green Fluorescent Proteins/genetics , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Macrophages/pathology , Myelin Basic Protein/immunology , Rats , Rats, Inbred Lew , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Vaccination
19.
Immunology ; 128(1): 92-102, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19689739

ABSTRACT

Fischer strain rats resist active induction of experimental autoimmune encephalomyelitis (EAE) following immunization with guinea-pig myelin basic protein (MBP) in complete Freund's adjuvant (CFA). Nevertheless, we now report that an encephalitogenic CD4(+) anti-MBP T-cell line could be developed from actively immunized Fischer rats. Adoptive transfer of the activated line mediated acute EAE in adult Fischer rats, but not in 1-day-old rats. Moreover, we found that both resting and activated anti-MBP T cells injected 1 day post-natally rendered these rats susceptible later in life to the active induction of EAE by immunization with MBP/CFA. The actively induced EAE manifested the accelerated onset of a secondary, memory-type response. Resting anti-MBP T cells injected even up to 2 weeks post-natally produced no clinical signs but seeded 50-100% of the recipients for an active encephalitogenic immune response to MBP. An earlier T-cell injection (1-2 days) produced a higher incidence and stronger response. The transferred resting T cells entered the neonatal spleen and thymus and proliferated there but did not change the total anti-MBP precursor number in adults. Splenocytes harvested from rats that were injected neonatally but not exposed to MBP in vivo proliferated strongly and produced significant amounts of interferon-gamma to MBP in vitro. Similar results were observed in rats injected with resting T-cell lines reactive to ovalbumin, suggesting that the neonatal injection of resting T cells specific for a self or for a foreign antigen can seed the immune system with the potential for an enhanced effector response to that antigen later in life.


Subject(s)
CD4-Positive T-Lymphocytes/transplantation , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Basic Protein/immunology , Adoptive Transfer , Animals , Animals, Newborn , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation , Disease Susceptibility , Lymphocyte Activation/immunology , Ovalbumin/immunology , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Spleen/immunology , Thymus Gland/immunology
20.
PLoS One ; 4(6): e6053, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19557135

ABSTRACT

Traditionally, immunology has considered a meaningful antibody response to be marked by large amounts of high-affinity antibodies reactive with the specific inciting antigen; the detection of small amounts of low-affinity antibodies binding to seemingly unrelated antigens has been considered to be beneath the threshold of immunological meaning. A systems-biology approach to immunology, however, suggests that large-scale patterns in the antibody repertoire might also reflect the functional state of the immune system. To investigate such global patterns of antibodies, we have used an antigen-microarray device combined with informatic analysis. Here we asked whether antibody-repertoire patterns might reflect the state of an implanted tumor. We studied the serum antibodies of inbred C57BL/6 mice before and after implantation of syngeneic 3LL tumor cells of either metastatic or non-metastatic clones. We analyzed patterns of IgG and IgM autoantibodies binding to over 300 self-antigens arrayed on slides using support vector machines and genetic algorithm techniques. We now report that antibody patterns, but not single antibodies, were informative: 1) mice, even before tumor implantation, manifest both individual and common patterns of low-titer natural autoantibodies; 2) the patterns of these autoantibodies respond to the growth of the tumor cells, and can distinguish between metastatic and non-metastatic tumor clones; and 3) curative tumor resection induces dynamic changes in these low-titer autoantibody patterns. The informative patterns included autoantibodies binding to self-molecules not known to be tumor-associated antigens (including insulin, DNA, myosin, fibrinogen) as well as to known tumor-associated antigens (including p53, cytokeratin, carbonic anhydrases, tyrosinase). Thus, low-titer autoantibodies that are not the direct products of tumor-specific immunization can still generate an immune biomarker of the body-tumor interaction. System-wide profiling of autoantibody repertoires can be informative.


Subject(s)
Autoantibodies/chemistry , Neoplasms, Experimental/immunology , Algorithms , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cluster Analysis , Immunoglobulin G/chemistry , Immunoglobulin M/chemistry , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Systems Biology
SELECTION OF CITATIONS
SEARCH DETAIL
...