Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Mol Immunol ; 135: 365-372, 2021 07.
Article in English | MEDLINE | ID: mdl-33990005

ABSTRACT

Although there are exceptions and outliers, T cell functional responses generally correlate with the affinity of a TCR for a peptide/MHC complex. In one recently described outlier case, the most promising clinical candidate in a series of TCRs specific for the gp100209 melanoma antigen bound with the weakest solution affinity and produced the least amount of cytokine in vitro. Hypotheses for this outlier behavior included unusual cytokine expression patterns arising from an atypical TCR binding geometry. Studying this instance in more detail, we found here that outlier behavior is attributable not to unusual cytokine patterns or TCR binding, but the use of a position 2 anchor-modified peptide variant in in vitro experiments instead of the wild type antigen that is present in vivo. Although the anchor-modified variant has been widely used in basic and clinical immunology as a surrogate for the wild type peptide, prior work has shown that TCRs can clearly distinguish between the two. We show that when this differential recognition is accounted for, the functional properties of gp100209-specific TCRs track with their affinity towards the peptide/MHC complex. Beyond demonstrating the correlates with T cell function for a clinically relevant TCR, our results provide important considerations for selection of TCRs for immunotherapy and the use of modified peptides in immunology.


Subject(s)
HLA-A2 Antigen/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , gp100 Melanoma Antigen/immunology , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Humans , Jurkat Cells , Protein Binding/immunology , Surface Plasmon Resonance
2.
Prostate ; 80(5): 424-431, 2020 04.
Article in English | MEDLINE | ID: mdl-32017163

ABSTRACT

BACKGROUND: Prostate cancer (PC) is the most commonly diagnosed solid tumor in men. A major challenge in PC immunotherapy is the lack of an animal model that resembles human adenocarcinoma and allows for manipulation or monitoring of the immune response. Mouse models are needed for preclinical testing of new immunotherapies, whether used alone or in combination with established drugs, and to develop companion biomarkers that can be validated in clinical trials. METHODS: To develop a syngeneic prostate adenocarcinoma model with a well-defined tumor antigen, murine RM1 PC cells were transfected with the endogenous mouse melanoma protein, gp100 (RM1-gp100). Gp100 was attractive because it is a self-protein and it instantly allowed us to use the large trove of immune research tools developed for melanoma research. A dendritic cell (DC) vaccine was used as model immunotherapy to demonstrate that adoptive immunotherapy against gp100 decreases the growth of RM1-gp100 but not RM1. RESULTS: Expressing gp100 did not change the growth of RM1 cell in vitro or in vivo. The DCs pulsed with RM1-gp100 could be used to stimulate Pmel-1 lymphocyte proliferation and activation. Pmel-1 lymphocytes could be adoptively transferred into C57Bl/6 mice that were treated with DCs pulsed with RM1-gp100. The resulting Pmel-1 lymphocytes were monitored to assess the primary cellular immune response and memory response. CONCLUSION: We describe a murine model for prostate adenocarcinoma with a well-characterized antigen that can be used in an immunologically intact mice to monitor the temporal CD8+ lymphocyte-mediated antitumor immunity.


Subject(s)
Cancer Vaccines/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , gp100 Melanoma Antigen/immunology , Animals , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Disease Models, Animal , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/genetics , T-Lymphocytes/immunology , Transfection , gp100 Melanoma Antigen/genetics
3.
Am J Respir Cell Mol Biol ; 62(6): 793-804, 2020 06.
Article in English | MEDLINE | ID: mdl-32078336

ABSTRACT

Patients with lymphangioleiomyomatosis (LAM) develop pulmonary cysts associated with neoplastic, smooth muscle-like cells that feature neuroendocrine cell markers. The disease preferentially affects premenopausal women. Existing therapeutics do not cure LAM. As gp100 is a diagnostic marker expressed by LAM lesions, we proposed to target this immunogenic glycoprotein using TCR transgenic T cells. To reproduce the genetic mutations underlying LAM, we cultured Tsc2-/- kidney tumor cells from aged Tsc2 heterozygous mice and generated a stable gp100-expressing cell line by lentiviral transduction. T cells were isolated from major histocompatibility complex-matched TCR transgenic pmel-1 mice to measure cytotoxicity in vitro, and 80% cytotoxicity was observed within 48 hours. Antigen-specific cytotoxicity was likewise observed using pmel-1 TCR-transduced mouse T cells, suggesting that transgenic T cells may likewise be useful to treat LAM in vivo. On intravenous injection, slow-growing gp100+ LAM-like cells formed lung nodules that were readily detectable in severe combined immunodeficient/beige mice. Adoptive transfer of gp100-reactive but not wild-type T cells into mice significantly shrunk established lung tumors, even in the absence of anti-PD-1 therapy. These results demonstrate the treatment potential of adoptively transferred T cells to eliminate pulmonary lesions in LAM.


Subject(s)
Immunotherapy, Adoptive , Lymphangioleiomyomatosis/therapy , T-Lymphocyte Subsets/transplantation , Animals , Cell Line , Cell Line, Tumor , Coculture Techniques , Gene Knockout Techniques , Immunocompetence , Kidney Neoplasms , Lymphangioleiomyomatosis/immunology , Male , Melanoma/immunology , Melanoma/therapy , Mice , Mice, Mutant Strains , Mice, SCID , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/genetics , Vesicular Transport Proteins/deficiency , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/immunology
4.
Cancer Immunol Res ; 8(1): 7-18, 2020 01.
Article in English | MEDLINE | ID: mdl-31719059

ABSTRACT

Loss of target antigens in tumor cells has become one of the major hurdles limiting the efficacy of adoptive cell therapy (ACT)-based immunotherapies. The optimal approach to overcome this challenge includes broadening the immune response from the initially targeted tumor-associated antigen (TAA) to other TAAs expressed in the tumor. To induce a more broadly targeted antitumor response, we utilized our previously developed Re-energized ACT (ReACT), which capitalizes on the synergistic effect of pathogen-based immunotherapy and ACT. In this study, we showed that ReACT induced a sufficient endogenous CD8+ T-cell response beyond the initial target to prevent the outgrowth of antigen loss variants in a B16-F10 melanoma model. Sequentially, selective depletion experiments revealed that Batf3-driven cDC1s were essential for the activation of endogenous tumor-specific CD8+ T cells. In ReACT-treated mice that eradicated tumors, we observed that endogenous CD8+ T cells differentiated into memory cells and facilitated the rejection of local and distal tumor rechallenge. By targeting one TAA with ReACT, we provided broader TAA coverage to counter antigen escape and generate a durable memory response against local relapse and metastasis.See related Spotlight on p. 2.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Listeria monocytogenes/pathogenicity , Listeriosis/complications , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , gp100 Melanoma Antigen/immunology , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Female , Immunologic Memory , Listeriosis/immunology , Listeriosis/microbiology , Melanoma, Experimental/metabolism , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Skin Neoplasms/metabolism , Skin Neoplasms/microbiology , Skin Neoplasms/therapy
5.
Nanomedicine ; 22: 102114, 2019 11.
Article in English | MEDLINE | ID: mdl-31655203

ABSTRACT

Plasmid DNA gp100 is able to act as an available vaccine against metastatic melanoma, but its administration is usually limited to parenteral route. Since oral delivery of plasmid DNA is intervened by various physical obstacles, here we constructed a nanogel (Alg-Tat-gp100) with multi-faceted functions through blending-by-blending method. Due to the cooperation of alginate and Tat peptide, Alg-Tat-gp100 demonstrated the significant improvement of stability in the stomach, mucus penetration ability in intestine, and transport across mucus layer and MDCK cells. Moreover, the bone marrow-derived cells were activated with an enhanced co-stimulatory molecule expression. Following immunization using Alg-Tat-gp100 nanogels in C57BL/6 mice, the secretion of IFN-γ and the activation of cytotoxic T cells were significantly improved. Benefiting from those cases, the B16F10 tumor inhibition rate achieved 42.5% by this oral DNA vaccine, suggesting that this multi-faceted nanogel prepared by simple blending-by-blending method may provide a new strategy for oral DNA vaccine delivery.


Subject(s)
Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Nanogels/chemistry , Plasmids/administration & dosage , Vaccines, DNA/administration & dosage , gp100 Melanoma Antigen/immunology , Administration, Oral , Alginates/chemistry , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dogs , Female , Immunization , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mucus/metabolism , Neoplasm Metastasis , Permeability , Phenotype , Protein Transport , Spleen/immunology , T-Lymphocytes/immunology , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/ultrastructure
6.
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
7.
J Immunol Methods ; 472: 55-64, 2019 09.
Article in English | MEDLINE | ID: mdl-31207210

ABSTRACT

Introduction of a tumor antigen-specific T cell receptor (TCR) into patient-derived lymphocytes has already exhibited promising results for the treatment of melanoma and other malignancies in clinical trials. However, insufficient or unsuccessful ex vivo manufacturing of engineered T cells due to low expansion and/or transduction rate can still be observed in some patients. Thus, we isolated human CD8+ T cells from healthy donors and equipped them with a gp100-specific TCR using a lentiviral construct in combination with a novel chemical lentiviral transduction enhancer (Lentiboost) to increase the rate of transduced cells. Following experiments to determine the ideal multiplicity of infection (MOI) and to analyze the efficacy of the transduction enhancer using a GFP-encoding lentivirus, we analyzed in the next step the transduction rate, cell count, and functionality of gp100 TCR-transduced T cells, i.e. antigen-specific cytokine secretion and lytic capacity. In order to increase the number of transduced cells, antigen-specific stimulation was performed, either once for 1 week (1st activation) or twice for another week (2nd activation). In general, each cycle of antigen-specific stimulation resulted in expansion of TCR-positive cells, while no further significant increase of transduced cells was observed after 2nd activation. Cytokine production pattern of transduced cells after antigen encounter, however, revealed significant antigen-specific secretion of TNF and IFNγ after the 1st as well as the 2nd activation. Furthermore, TCR T cells, either activated once or twice, showed significant cytotoxicity towards antigen-positive tumor cells. Taken together, these results show that it is feasible to transduce human T cells using a lentiviral construct in combination with this novel lentiviral transduction enhancer, which shows potential in the growing field of cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lentivirus/genetics , Melanoma/immunology , Transduction, Genetic , gp100 Melanoma Antigen/immunology , Cytokines/biosynthesis , Humans , Melanoma/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
8.
Cancer Treat Rev ; 77: 35-43, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31207478

ABSTRACT

Immunotherapeutic strategies have revolutionised cancer therapy in recent years, bringing meaningful improvements in outcomes for patients with previously intractable conditions. These successes have, however, been largely limited to certain types of liquid tumours and a small subset of solid tumours that are known to be particularly immunogenic. Broadening these advances across the majority of tumour indications, which are characterised by an immune-excluded, immune-deserted or immune-suppressed ('cold') phenotype, will require alternative approaches that are able to specifically address this unique biological environment. Several newer therapeutic modalities, including adoptive cell therapy and T cell redirecting bispecific molecules, are considered to hold particular promise and are being investigated in early phase clinical trials across various solid tumour indications. ImmTAC molecules are a novel class of T cell redirecting bispecific biologics that exploit TCR-based targeting of tumour cells; providing potent and highly specific access to the vast landscape of intracellular targets. The first of these reagents to reach the clinic, tebentafusp (IMCgp100), has generated demonstrable clinical efficacy in an immunologically cold solid tumour with a high unmet need. Here, we highlight the key elements of the ImmTAC platform that make it ideally positioned to overcome the cold tumour microenvironment in an off-the-shelf format.


Subject(s)
Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Biological Products/administration & dosage , Humans , Immunotherapy, Adoptive/methods , Proteins/immunology , Single-Chain Antibodies/immunology , gp100 Melanoma Antigen/immunology
9.
Cancer Immunol Res ; 7(7): 1188-1201, 2019 07.
Article in English | MEDLINE | ID: mdl-31113806

ABSTRACT

Antigen-presenting cells expand antigen-specific T cells ex vivo and in vivo for tumor immunotherapy, but are time-consuming to generate and, as live cells, raise biosafety concerns. An alternative is found in cell-free artificial antigen-presenting cells (aAPC), but these only present two or three kinds of immune molecules. Here, we describe a multipotent artificial antigen-presenting cell (MaAPC) that delivered 11 kinds of immune moleclues. This MaAPC simulated natural APCs through the concurent coupling of target antigens (H-2Kb/TRP2180-188-Ig dimers and H-2Db/gp10025-33-Ig dimers), costimulatory molecules (anti-CD28, anti-4-1BB, and anti-CD2), and "self-marker" CD47-Fc onto surface-modified polylactic-co-glycolic acid microparticles (PLGA-MP). These PLGA-MPs also encapsulated cytokines (IL2 and IL15), a chemokine (CCL21), and checkpoint inhibitors (anti-CTLA-4 and anti-PD-1). Culture of MaAPCs with naïve T cells for 1 week elevated the frequencies of TRP2180-188-specific and gp10025-33-specific CTLs to 51.0% and 43.3%, respectively, with enhanced cytotoxicity. Three infusions of MaAPCs inhibited subcutaneous melanoma growth in a mouse model and expanded TRP2180-188 and gp10025-33-specific CTLs 59-86-fold in peripheral blood, 76-77-fold in spleen, and 205-212-fold in tumor tissue, in an antigen-specific manner. Compared with conventional aAPCs carrying two or three immune molecules, the 11-signal MaAPCs exerted greater impact on T cells, including activation, proliferation, cytotoxicity, differentiation to memory CTLs or regulatory T cells and cytokines profiles, without detected side effects. Such MaAPCs could be used to individualize tumor immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , gp100 Melanoma Antigen/immunology , Animals , Antibodies, Monoclonal/immunology , Apoptosis , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Proliferation , Chemokines/immunology , Cytokines/immunology , Female , Immunotherapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Tumor Cells, Cultured
10.
Cancer Immunol Immunother ; 68(5): 709-720, 2019 May.
Article in English | MEDLINE | ID: mdl-30783693

ABSTRACT

We used four-color ImmunoSpot® assays, in conjunction with peptide pools that cover the sequence of tyrosinase (Tyr), melanoma-associated antigen A3 (MAGE-A3), melanocyte antigen/melanoma antigen recognized by T cells 1 (Melan-A/MART-1), glycoprotein 100 (gp100), and New York esophageal squamous cell carcinoma-1 (NY-ESO-1) to characterize the melanoma antigen (MA)-specific CD8 + cell repertoire in PBMC of 40 healthy human donors (HD). Tyr triggered interferon gamma (IFN-γ)-secreting CD8 + T cells in 25% of HD within 24 h of antigen stimulation ex vivo. MAGE-A3, Melan-A/MART-1, and gp100 also induced recall responses in 10%, 7.5%, and 2.5% of HD, respectively. At this time point, these CD8 + T cells did not yet produce GzB (granzyme B). However, they engaged in GzB production after 72 h of antigen stimulation. By this 72-h time point, 57.5% of the HD responded to at least one, and typically several, of the MA. A closer characterization of the Tyr-specific CD8 + T cell repertoire indicated that it was low-affinity, and to primarily entail a stem cell-like subpopulation. Collectively, our data reveal pre-existing endogenous T cell immunity against melanoma antigens in healthy donors, and analogous to natural autoantibodies, we have termed this "natural T cell autoreactivity".


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunospot Assay/methods , Monophenol Monooxygenase/immunology , Antigens, Neoplasm/immunology , Autoimmunity , Cell Proliferation , Clone Cells , Healthy Volunteers , Humans , Immunologic Memory , Interferon-gamma/metabolism , Lymphocyte Activation , MART-1 Antigen/immunology , Neoplasm Proteins/immunology , gp100 Melanoma Antigen/immunology
11.
J Immunol ; 201(7): 1875-1888, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30135181

ABSTRACT

Cancer immunotherapy has been flourishing in recent years with remarkable clinical success. But as more patients are treated, a shadow is emerging that has haunted other cancer therapies: tumors develop resistance. Resistance is often caused by defects in the MHC class I Ag presentation pathway critical for CD8 T cell-mediated tumor clearance. TAP and tapasin, both key players in the pathway, are frequently downregulated in human cancers, correlating with poor patient survival. Reduced dependence on these factors may promote vaccine efficiency by limiting immune evasion. In this study, we demonstrate that PMEL209-217, a promising phase 3 trial-tested antimelanoma vaccine candidate, is robustly presented by various TAP- and/or tapasin-deficient cell lines. This striking characteristic may underlie its potency as a vaccine. Surprisingly, cytosolic proteasomes generate the peptide even for TAP-independent presentation, whereas tripeptidyl peptidase 2 (TPP2) efficiently degrades the epitope. Consequently, inhibiting TPP2 substantially boosts PMEL209-217 presentation, suggesting a possible strategy to improve the therapeutic efficacy of the vaccine.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Melanoma/immunology , Vaccines/immunology , Aminopeptidases/metabolism , Antigen Presentation , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Cytosol/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-A2 Antigen/metabolism , Humans , Immune Evasion , Membrane Transport Proteins/genetics , Oligopeptides/genetics , Serine Endopeptidases/metabolism , gp100 Melanoma Antigen/immunology , gp100 Melanoma Antigen/metabolism
12.
Int Immunol ; 30(4): 141-154, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617862

ABSTRACT

Immunotherapies have led to the successful development of novel therapies for cancer. However, there is increasing concern regarding the adverse effects caused by non-tumor-specific immune responses. Here, we report an effective strategy to generate high-avidity tumor-antigen-specific CTLs, using Cas9/single-guide RNA (sgRNA) ribonucleoprotein (RNP) delivery. As a proof-of-principle demonstration, we selected the gp100 melanoma-associated tumor antigen, and cloned the gp100-specific high-avidity TCR from gp100-immunized mice. To enable rapid structural dissection of the TCR, we developed a 3D protein structure modeling system for the TCR/antigen-major histocompatibility complex (pMHC) interaction. Combining these technologies, we efficiently generated gp100-specific PD-1(-) CD8+ T cells, and demonstrated that the genetically engineered CD8+ T cells have high avidity against melanoma cells both in vitro and in vivo. Our methodology offers computational prediction of the TCR response, and enables efficient generation of tumor antigen-specific CD8+ T cells that can neutralize tumor-induced immune suppression leading to a potentially powerful cancer therapeutic.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CRISPR-Cas Systems , Gene Editing , Neoplasms/genetics , Neoplasms/immunology , T-Cell Antigen Receptor Specificity/immunology , Animals , Antigens, Neoplasm/chemistry , Cell Line, Tumor , Female , Gene Knockout Techniques , Genes, Reporter , Melanoma, Experimental , Mice , Models, Molecular , Multiprotein Complexes , Neoplasms/metabolism , Peptides/chemistry , Peptides/immunology , Peptides/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , gp100 Melanoma Antigen/chemistry , gp100 Melanoma Antigen/immunology , gp100 Melanoma Antigen/metabolism
13.
J Clin Invest ; 128(4): 1338-1354, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29480817

ABSTRACT

Anticancer vaccination is a promising approach to increase the efficacy of cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed death ligand 1 (PD-L1) checkpoint blockade therapies. However, the landmark FDA registration trial for anti-CTLA-4 therapy (ipilimumab) revealed a complete lack of benefit of adding vaccination with gp100 peptide formulated in incomplete Freund's adjuvant (IFA). Here, using a mouse model of melanoma, we found that gp100 vaccination induced gp100-specific effector T cells (Teffs), which dominantly forced trafficking of anti-CTLA-4-induced, non-gp100-specific Teffs away from the tumor, reducing tumor control. The inflamed vaccination site subsequently also sequestered and destroyed anti-CTLA-4-induced Teffs with specificities for tumor antigens other than gp100, reducing the antitumor efficacy of anti-CTLA-4 therapy. Mechanistically, Teffs at the vaccination site recruited inflammatory monocytes, which in turn attracted additional Teffs in a vicious cycle mediated by IFN-γ, CXCR3, ICAM-1, and CCL2, dependent on IFA formulation. In contrast, nonpersistent vaccine formulations based on dendritic cells, viral vectors, or water-soluble peptides potently synergized with checkpoint blockade of both CTLA-4 and PD-L1 and induced complete tumor regression, including in settings of primary resistance to dual checkpoint blockade. We conclude that cancer vaccine formulation can dominantly determine synergy, or lack thereof, with CTLA-4 and PD-L1 checkpoint blockade therapy for cancer.


Subject(s)
B7-H1 Antigen/immunology , CTLA-4 Antigen/immunology , Cancer Vaccines/pharmacology , Cell Cycle Checkpoints/immunology , Melanoma/therapy , Neoplasms, Experimental/therapy , Peptides/immunology , gp100 Melanoma Antigen/pharmacology , Animals , B7-H1 Antigen/genetics , CTLA-4 Antigen/genetics , Cancer Vaccines/immunology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Peptides/pharmacology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , gp100 Melanoma Antigen/immunology
14.
J Immunother ; 41(4): 181-189, 2018 May.
Article in English | MEDLINE | ID: mdl-29334492

ABSTRACT

The chemokine MIP3α (CCL20) binds to CCR6 on immature dendritic cells. Vaccines fusing MIP3α to gp100 have been shown to be effective in therapeutically reducing melanoma tumor burden and prolonging survival in a mouse model. Other studies have provided evidence that interleukin-10 (IL-10) neutralizing antibodies (αIL-10) enhance immunologic melanoma therapies by modulating the tolerogenic tumor microenvironment. In the current study, we have utilized the B16F10 syngeneic mouse melanoma model to demonstrate for the first time that a therapy neutralizing IL-10 enhances the antitumor efficacy of a MIP3α-gp100 DNA vaccine, leading to significantly smaller tumors, slower growing tumors, and overall increases in mouse survival. The additive effects of αIL-10 were not shown to be correlated to vaccine-specific tumor-infiltrating lymphocytes (TILs), total TILs, or regulatory T cells. However, we discovered an upregulation of IFNα-4 transcripts in tumors and a correlation of increased plasmacytoid dendritic cell numbers with reduced tumor burden in αIL-10-treated mice. Interferon α receptor knockout (IFNαR1) mice received no benefit from αIL-10 treatment, demonstrating that the additional therapeutic value of αIL-10 is primarily mediated by type I IFNs. Efficient targeting of antigen to immature dendritic cells with a chemokine-fusion vaccine provides an effective anticancer therapeutic. Combining this approach with an IL-10 neutralizing antibody therapy enhances the antitumor efficacy of the therapy in a manner dependent upon the activity of type I IFNs. This combination of a vaccine and immunomodulatory agent provides direction for future optimization of a novel cancer vaccine therapy.


Subject(s)
Cancer Vaccines/immunology , Chemokine CCL20/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon Type I/metabolism , Interleukin-10/antagonists & inhibitors , gp100 Melanoma Antigen/immunology , Animals , Cytokines/metabolism , Dendritic Cells/drug effects , Disease Models, Animal , Female , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental , Mice , Mice, Knockout , Treatment Outcome , Vaccination
15.
BMC Cancer ; 17(1): 551, 2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28818060

ABSTRACT

BACKGROUND: Adoptive T-cell therapy relying on conventional T cells transduced with T-cell receptors (TCRs) or chimeric antigen receptors (CARs) has caused substantial tumor regression in several clinical trials. However, genetically engineered T cells have been associated with serious side-effects due to off-target toxicities and massive cytokine release. To obviate these concerns, we established a protocol adaptable to GMP to expand and transiently transfect γ/δ T cells with mRNA. METHODS: PBMC from healthy donors were stimulated using zoledronic-acid or OKT3 to expand γ/δ T cells and bulk T cells, respectively. Additionally, CD8+ T cells and γ/δ T cells were MACS-isolated from PBMC and expanded with OKT3. Next, these four populations were electroporated with RNA encoding a gp100/HLA-A2-specific TCR or a CAR specific for MCSP. Thereafter, receptor expression, antigen-specific cytokine secretion, specific cytotoxicity, and killing of the endogenous γ/δ T cell-target Daudi were analyzed. RESULTS: Using zoledronic-acid in average 6 million of γ/δ T cells with a purity of 85% were generated from one million PBMC. MACS-isolation and OKT3-mediated expansion of γ/δ T cells yielded approximately ten times less cells. OKT3-expanded and CD8+ MACS-isolated conventional T cells behaved correspondingly similar. All employed T cells were efficiently transfected with the TCR or the CAR. Upon respective stimulation, γ/δ T cells produced IFNγ and TNF, but little IL-2 and the zoledronic-acid expanded T cells exceeded MACS-γ/δ T cells in antigen-specific cytokine secretion. While the cytokine production of γ/δ T cells was in general lower than that of conventional T cells, specific cytotoxicity against melanoma cell lines was similar. In contrast to OKT3-expanded and MACS-CD8+ T cells, mock-electroporated γ/δ T cells also lysed tumor cells reflecting the γ/δ T cell-intrinsic anti-tumor activity. After transfection, γ/δ T cells were still able to kill MHC-deficient Daudi cells. CONCLUSION: We present a protocol adaptable to GMP for the expansion of γ/δ T cells and their subsequent RNA-transfection with tumor-specific TCRs or CARs. Given the transient receptor expression, the reduced cytokine release, and the equivalent cytotoxicity, these γ/δ T cells may represent a safer complementation to genetically engineered conventional T cells in the immunotherapy of melanoma (Exper Dermatol 26: 157, 2017, J Investig Dermatol 136: A173, 2016).


Subject(s)
RNA , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adult , Cell Culture Techniques , Cytokines/metabolism , Cytotoxicity, Immunologic , Electroporation , Genetic Engineering , HLA-A2 Antigen/immunology , Healthy Volunteers , Humans , Immunomagnetic Separation , Immunophenotyping , Immunotherapy, Adoptive , Melanoma/genetics , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Cell Antigen Receptor Specificity , Transfection , Young Adult , gp100 Melanoma Antigen/immunology
16.
Immunology ; 152(3): 425-438, 2017 11.
Article in English | MEDLINE | ID: mdl-28640942

ABSTRACT

The success of immune system-based cancer therapies depends on a broad immune response engaging a range of effector cells and mechanisms. Immune mobilizing monoclonal T cell receptors (TCRs) against cancer (ImmTAC™ molecules: fusion proteins consisting of a soluble, affinity enhanced TCR and an anti-CD3 scFv antibody) were previously shown to redirect CD8+ and CD4+ T cells against tumours. Here we present evidence that IMCgp100 (ImmTAC recognizing a peptide derived from the melanoma-specific protein, gp100, presented by HLA-A*0201) efficiently redirects and activates effector and memory cells from both CD8+ and CD4+ repertoires. Using isolated subpopulations of T cells, we find that both terminally differentiated and effector memory CD8+ T cells redirected by IMCgp100 are potent killers of melanoma cells. Furthermore, CD4+ effector memory T cells elicit potent cytotoxic activity leading to melanoma cell killing upon redirection by IMCgp100. The majority of T cell subsets belonging to both the CD8+ and CD4+ repertoires secrete key pro-inflammatory cytokines (tumour necrosis factor-α, interferon-γ, interleukin-6) and chemokines (macrophage inflammatory protein-1α-ß, interferon-γ-inducible protein-10, monocyte chemoattractant protein-1). At an individual cell level, IMCgp100-redirected T cells display a polyfunctional phenotype, which is a hallmark of a potent anti-cancer response. This study demonstrates that IMCgp100 induces broad immune responses that extend beyond the induction of CD8+ T cell-mediated cytotoxicity. These findings are of particular importance because IMCgp100 is currently undergoing clinical trials as a single agent or in combination with check point inhibitors for patients with malignant melanoma.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Melanoma/therapy , Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Skin Neoplasms/therapy , gp100 Melanoma Antigen/immunology , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Humans , Immunologic Memory/drug effects , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Phenotype , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , gp100 Melanoma Antigen/metabolism
17.
Cancer Immunol Res ; 5(8): 618-629, 2017 08.
Article in English | MEDLINE | ID: mdl-28630054

ABSTRACT

Cytotoxic T lymphocyte (CTL)-based immunotherapies have had remarkable success at generating objective clinical responses in patients with advanced metastatic melanoma. Although the melanocyte differentiation antigens (MDA) MART-1, PMEL, and tyrosinase were among the first melanoma tumor-associated antigens identified and targeted with immunotherapy, expression within normal melanocytes of the eye and inner ear can elicit serious autoimmune side effects, thus limiting their clinical potential as CTL targets. Using a tandem mass spectrometry (MS) approach to analyze the immunopeptidomes of 55 melanoma patient-derived cell lines, we identified a number of shared HLA class I-bound peptides derived from the melanocyte-specific transporter protein SLC45A2. Antigen-specific CTLs generated against HLA-A*0201- and HLA-A*2402-restricted SLC45A2 peptides effectively killed a majority of HLA-matched cutaneous, uveal, and mucosal melanoma cell lines tested (18/25). CTLs specific for SLC45A2 showed significantly reduced recognition of HLA-matched primary melanocytes that were, conversely, robustly killed by MART1- and PMEL-specific T cells. Transcriptome analysis revealed that SLC45A2 mRNA expression in normal melanocytes was less than 2% that of other MDAs, therefore providing a more favorable melanoma-to-melanocyte expression ratio. Expression of SLC45A2 and CTL sensitivity could be further upregulated in BRAF(V600E)-mutant melanoma cells upon treatment with BRAF or MEK inhibitors, similarly to other MDAs. Taken together, our study demonstrates the feasibility of using tandem MS as a means of discovering shared immunogenic tumor-associated epitopes and identifies SLC45A2 as a promising immunotherapeutic target for melanoma with high tumor selectivity and reduced potential for autoimmune toxicity. Cancer Immunol Res; 5(8); 618-29. ©2017 AACR.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy , Melanoma/therapy , Membrane Transport Proteins/immunology , Proto-Oncogene Proteins B-raf/genetics , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation/genetics , Antigen Presentation/immunology , Antigens, Neoplasm/genetics , Cytotoxicity, Immunologic , Epitopes/immunology , HLA-A2 Antigen/immunology , HLA-A24 Antigen/immunology , Humans , MART-1 Antigen/immunology , Melanocytes/immunology , Melanoma/immunology , Melanoma/pathology , Membrane Transport Proteins/genetics , Peptides/genetics , Peptides/immunology , Proto-Oncogene Proteins B-raf/immunology , Tandem Mass Spectrometry , Transcriptome/genetics , gp100 Melanoma Antigen/immunology
18.
J Immunother ; 40(5): 155-163, 2017 06.
Article in English | MEDLINE | ID: mdl-28368960

ABSTRACT

We have previously shown that inhibition of the proteasome causes defective ribosomal products to be shunted into autophagosomes and subsequently released from tumor cells as defective ribosomal products in Blebs (DRibbles). These DRibbles serve as an excellent source of antigens for cross-priming of tumor-specific T cells. Here, we examine the role of ubiquitinated proteins (Ub-proteins) in this pathway. Using purified Ub-proteins from tumor cells that express endogenous tumor-associated antigen or exogenous viral antigen, we tested the ability of these proteins to stimulate antigen-specific T-cell responses, by activation of monocyte-derived dendritic cells generated from human peripheral blood mononuclear cells. Compared with total cell lysates, we found that purified Ub-proteins from both a gp100-specific melanoma cell line and from a lung cancer cell line expressing cytomegalovirus pp65 antigen produced a significantly higher level of IFN-γ in gp100- or pp65-specific T cells, respectively. In addition, Ub-proteins from an allogeneic tumor cell line could be used to stimulate tumor-infiltrating lymphocytes isolated and expanded from non-small cell lung cancer patients. These results establish that Ub-proteins provide a relevant source of antigens for cross-priming of antitumor immune responses in a variety of settings, including endogenous melanoma and exogenous viral antigen presentation, as well as antigen-specific tumor-infiltrating lymphocytes. Thus, ubiquitin can be used as an affinity tag to enrich for unknown tumor-specific antigens from tumor cell lysates to stimulate tumor-specific T cells ex vivo or to be used as vaccines to target short-lived proteins.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lung Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic , Aluminum Oxide/immunology , Antigens, Neoplasm/immunology , Autophagy , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cross-Priming , Humans , Interferon-gamma/metabolism , Lung Neoplasms/therapy , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma/therapy , Phosphoproteins/immunology , Ribosomes/immunology , T-Lymphocytes/transplantation , Ubiquitinated Proteins/immunology , Viral Matrix Proteins/immunology , gp100 Melanoma Antigen/immunology
19.
Clin Cancer Res ; 23(10): 2478-2490, 2017 May 15.
Article in English | MEDLINE | ID: mdl-27965307

ABSTRACT

Purpose: While adoptive transfer of T cells bearing a chimeric antigen receptor (CAR) can eliminate substantial burdens of some leukemias, the ultimate challenge remains the eradication of large solid tumors for most cancers. We aimed to develop an immunotherapy approach effective against large tumors in an immunocompetent, self-antigen preclinical mouse model.Experimental Design: In this study, we generated dual-specific T cells expressing both a CAR specific for Her2 and a TCR specific for the melanocyte protein (gp100). We used a regimen of adoptive cell transfer incorporating vaccination (ACTIV), with recombinant vaccinia virus expressing gp100, to treat a range of tumors including orthotopic breast tumors and large liver tumors.Results: ACTIV therapy induced durable complete remission of a variety of Her2+ tumors, some in excess of 150 mm2, in immunocompetent mice expressing Her2 in normal tissues, including the breast and brain. Vaccinia virus induced extensive proliferation of T cells, leading to massive infiltration of T cells into tumors. Durable tumor responses required the chemokine receptor CXCR3 and exogenous IL2, but were independent of IFNγ. Mice were resistant to tumor rechallenge, indicating immune memory involving epitope spreading. Evidence of limited neurologic toxicity was observed, associated with infiltration of cerebellum by T cells, but was only transient.Conclusions: This study supports a view that it is possible to design a highly effective combination immunotherapy for solid cancers, with acceptable transient toxicity, even when the target antigen is also expressed in vital tissues. Clin Cancer Res; 23(10); 2478-90. ©2016 AACR.


Subject(s)
Brain Neoplasms/therapy , Breast Neoplasms/therapy , Immunotherapy, Adoptive , Receptor, ErbB-2/immunology , gp100 Melanoma Antigen/immunology , Animals , Autoantigens/administration & dosage , Autoantigens/immunology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Mice , Mice, Transgenic , Receptor, ErbB-2/genetics , Receptors, Antigen, T-Cell/administration & dosage , Receptors, Antigen, T-Cell/immunology , Remission Induction , Vaccinia virus/genetics , Vaccinia virus/immunology , gp100 Melanoma Antigen/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...