Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Res Sq ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38645165

ABSTRACT

Interleukin-15 (IL15) promotes the survival of T lymphocytes and enhances the antitumor properties of CAR T cells in preclinical models of solid neoplasms in which CAR T cells have limited efficacy1-4. Glypican-3 (GPC3) is expressed in a group of solid cancers5-10, and here we report the first evaluation in humans of the effects of IL15 co-expression on GPC3-CAR T cells. Cohort 1 patients (NCT02905188/NCT02932956) received GPC3-CAR T cells, which were safe but produced no objective antitumor responses and reached peak expansion at two weeks. Cohort 2 patients (NCT05103631/NCT04377932) received GPC3-CAR T cells that co-expressed IL15 (15.CAR), which mediated significantly increased cell expansion and induced a disease control rate of 66% and antitumor response rate of 33%. Infusion of 15.CAR T cells was associated with increased incidence of cytokine release syndrome, which was rapidly ameliorated by activation of the inducible caspase 9 safety switch. Compared to non-responders, tumor-infiltrating 15.CAR T cells from responders showed repression of SWI/SNF epigenetic regulators and upregulation of FOS and JUN family members as well as genes related to type I interferon signaling. Collectively, these results demonstrate that IL15 increases the expansion, intratumoral survival, and antitumor activity of GPC3-CAR T cells in patients.

3.
Nat Med ; 29(6): 1379-1388, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37188782

ABSTRACT

Vα24-invariant natural killer T cells (NKTs) have anti-tumor properties that can be enhanced by chimeric antigen receptors (CARs). Here we report updated interim results from the first-in-human phase 1 evaluation of autologous NKTs co-expressing a GD2-specific CAR with interleukin 15 (IL15) (GD2-CAR.15) in 12 children with neuroblastoma (NB). The primary objectives were safety and determination of maximum tolerated dose (MTD). The anti-tumor activity of GD2-CAR.15 NKTs was assessed as a secondary objective. Immune response evaluation was an additional objective. No dose-limiting toxicities occurred; one patient experienced grade 2 cytokine release syndrome that was resolved by tocilizumab. The MTD was not reached. The objective response rate was 25% (3/12), including two partial responses and one complete response. The frequency of CD62L+NKTs in products correlated with CAR-NKT expansion in patients and was higher in responders (n = 5; objective response or stable disease with reduction in tumor burden) than non-responders (n = 7). BTG1 (BTG anti-proliferation factor 1) expression was upregulated in peripheral GD2-CAR.15 NKTs and is a key driver of hyporesponsiveness in exhausted NKT and T cells. GD2-CAR.15 NKTs with BTG1 knockdown eliminated metastatic NB in a mouse model. We conclude that GD2-CAR.15 NKTs are safe and can mediate objective responses in patients with NB. Additionally, their anti-tumor activity may be enhanced by targeting BTG1. ClinicalTrials.gov registration: NCT03294954 .


Subject(s)
Natural Killer T-Cells , Neuroblastoma , Receptors, Chimeric Antigen , Child , Animals , Mice , Humans , Cytotoxicity, Immunologic , Receptors, Chimeric Antigen/genetics , Neuroblastoma/therapy , Immunotherapy, Adoptive/methods
4.
J Immunother Cancer ; 11(3)2023 03.
Article in English | MEDLINE | ID: mdl-36882225

ABSTRACT

BACKGROUND: Tumor progression and resistance to therapy in children with neuroblastoma (NB), a common childhood cancer, are often associated with infiltration of monocytes and macrophages that produce inflammatory cytokines. However, the mechanism by which tumor-supportive inflammation is initiated and propagated remains unknown. Here, we describe a novel protumorigenic circuit between NB cells and monocytes that is triggered and sustained by tumor necrosis factor alpha (TNF-α). METHODS: We used NB knockouts (KOs) of TNF-α and TNFRSF1A mRNA (TNFR1)/TNFRSF1B mRNA (TNFR2) and TNF-α protease inbitor (TAPI), a drug that modulates TNF-α isoform expression, to assess the role of each component in monocyte-associated protumorigenic inflammation. Additionally, we employed NB-monocyte cocultures and treated these with clinical-grade etanercept, an Fc-TNFR2 fusion protein, to neutralize signaling by both membrane-bound (m) and soluble (s)TNF-α isoforms. Further, we treated NOD/SCID/IL2Rγ(null) mice carrying subcutaneous NB/human monocyte xenografts with etanercept and evaluated the impact on tumor growth and angiogenesis. Gene set enrichment analysis (GSEA) was used to determine whether TNF-α signaling correlates with clinical outcomes in patients with NB. RESULTS: We found that NB expression of TNFR2 and monocyte membrane-bound tumor necrosis factor alpha is required for monocyte activation and interleukin (IL)-6 production, while NB TNFR1 and monocyte soluble TNF-α are required for NB nuclear factor kappa B subunit 1 (NF-κB) activation. Treatment of NB-monocyte cocultures with clinical-grade etanercept completely abrogated release of IL-6, granulocyte colony-stimulating factor (G-CSF), IL-1α, and IL-1ß and eliminated monocyte-induced enhancement of NB cell proliferation in vitro. Furthermore, etanercept treatment inhibited tumor growth, ablated tumor angiogenesis, and suppressed oncogenic signaling in mice with subcutaneous NB/human monocyte xenografts. Finally, GSEA revealed significant enrichment for TNF-α signaling in patients with NB that relapsed. CONCLUSIONS: We have described a novel mechanism of tumor-promoting inflammation in NB that is strongly associated with patient outcome and could be targeted with therapy.


Subject(s)
Neuroblastoma , Receptors, Tumor Necrosis Factor, Type II , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Carcinogenesis , Etanercept , Mice, Inbred NOD , Mice, SCID , Monocytes , Neuroblastoma/drug therapy , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics
5.
Cancer Immunol Res ; 11(2): 171-183, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36484736

ABSTRACT

Vα24-invariant natural killer T cells (NKT) possess innate antitumor properties that can be exploited for cancer immunotherapy. We have shown previously that the CD62L+ central memory-like subset of these cells drives the in vivo antitumor activity of NKTs, but molecular mediators of NKT central memory differentiation remain unknown. Here, we demonstrate that relative to CD62L- cells, CD62L+ NKTs express a higher level of the gene encoding the Wnt/ß-catenin transcription factor lymphoid enhancer binding factor 1 (LEF1) and maintain active Wnt/ß-catenin signaling. CRISPR/Cas9-mediated LEF1 knockout reduced CD62L+ frequency after antigenic stimulation, whereas Wnt/ß-catenin activator Wnt3a ligand increased CD62L+ frequency. LEF1 overexpression promoted NKT expansion and limited exhaustion following serial tumor challenge and was sufficient to induce a central memory-like transcriptional program in NKTs. In mice, NKTs expressing a GD2-specific chimeric-antigen receptor (CAR) with LEF1 demonstrated superior control of neuroblastoma xenograft tumors compared with control CAR-NKTs. These results identify LEF1 as a transcriptional activator of the NKT central memory program and advance development of NKT cell-based immunotherapy. See related Spotlight by Van Kaer, p. 144.


Subject(s)
Natural Killer T-Cells , Receptors, Chimeric Antigen , Humans , Animals , Mice , Natural Killer T-Cells/immunology , beta Catenin , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphocyte Activation/immunology
6.
Blood ; 141(8): 869-876, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36347021

ABSTRACT

T cells expressing chimeric antigen receptors (CARs) have achieved major clinical success in patients with hematologic malignancies. However, these treatments remain largely ineffective for solid cancers and require significant time and resources to be manufactured in an autologous setting. Developing alternative immune effector cells as cancer immunotherapy agents that can be employed in allogeneic settings is crucial for the advancement of cell therapy. Unlike T cells, Vα24-invariant natural killer T cells (NKTs) are not alloreactive and can therefore be generated from allogeneic donors for rapid infusion into numerous patients without the risk of graft-versus-host disease. Additionally, NKT cells demonstrate inherent advantages over T-cell products, including the ability to traffic to tumor tissues, target tumor-associated macrophages, transactivate NK cells, and cross-prime tumor-specific CD8 T cells. Both unmodified NKTs, which specifically recognize CD1d-bound glycolipid antigens expressed by certain types of tumors, and CAR-redirected NKTs are being developed as the next generation of allogeneic cell therapy products. In this review, we describe studies on the biology of NKTs and other types of innate-like T cells and summarize the clinical experiences of unmodified and CAR-redirected NKTs, including recent interim reports on allogeneic NKTs.


Subject(s)
Hematopoietic Stem Cell Transplantation , Natural Killer T-Cells , Neoplasms , Receptors, Chimeric Antigen , Humans , Allogeneic Cells , Neoplasms/therapy , Killer Cells, Natural , Immunotherapy, Adoptive
7.
Contrast Media Mol Imaging ; 2021: 6641384, 2021.
Article in English | MEDLINE | ID: mdl-34220380

ABSTRACT

Objective: Tumor-associated macrophages (TAMs) within the tumor immune microenvironment (TiME) of solid tumors play an important role in treatment resistance and disease recurrence. The purpose of this study was to investigate if nanoradiomics (radiomic analysis of nanoparticle contrast-enhanced images) can differentiate tumors based on TAM burden. Materials and Methods: In vivo studies were performed in transgenic mouse models of neuroblastoma with low (N = 11) and high (N = 10) tumor-associated macrophage (TAM) burden. Animals underwent delayed nanoparticle contrast-enhanced CT (n-CECT) imaging at 4 days after intravenous administration of liposomal-iodine agent (1.1 g/kg). CT imaging-derived conventional tumor metrics (tumor volume and CT attenuation) were computed for segmented tumor CT datasets. Nanoradiomic analysis was performed using a PyRadiomics workflow implemented in the quantitative image feature pipeline (QIFP) server containing 900 radiomic features (RFs). RF selection was performed under supervised machine learning using a nonparametric neighborhood component method. A 5-fold validation was performed using a set of linear and nonlinear classifiers for group separation. Statistical analysis was performed using the Kruskal-Wallis test. Results: N-CECT imaging demonstrated heterogeneous patterns of signal enhancement in low and high TAM tumors. CT imaging-derived conventional tumor metrics showed no significant differences (p > 0.05) in tumor volume between low and high TAM tumors. Tumor CT attenuation was not significantly different (p > 0.05) between low and high TAM tumors. Machine learning-augmented nanoradiomic analysis revealed two RFs that differentiated (p < 0.002) low TAM and high TAM tumors. The RFs were used to build a linear classifier that demonstrated very high accuracy and further confirmed by 5-fold cross-validation. Conclusions: Imaging-derived conventional tumor metrics were unable to differentiate tumors with varying TAM burden; however, nanoradiomic analysis revealed texture differences and enabled differentiation of low and high TAM tumors.


Subject(s)
Contrast Media/pharmacology , Nanoparticles/chemistry , Neuroblastoma/diagnostic imaging , Tomography, X-Ray Computed , Animals , Contrast Media/chemistry , Humans , Iodine Radioisotopes/chemistry , Iodine Radioisotopes/pharmacology , Machine Learning , Mice , Mice, Transgenic , Neuroblastoma/pathology , Radiometry , Tumor Burden/radiation effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Tumor-Associated Macrophages
8.
Nat Med ; 26(11): 1686-1690, 2020 11.
Article in English | MEDLINE | ID: mdl-33046868

ABSTRACT

Vα24-invariant natural killer T (NKT) cells have shown potent anti-tumor properties in murine tumor models and have been linked to favorable outcomes in patients with cancer. However, low numbers of these cells in humans have hindered their clinical applications. Here we report interim results from all three patients enrolled on dose level 1 in a phase 1 dose-escalation trial of autologous NKT cells engineered to co-express a GD2-specific chimeric antigen receptor (CAR) with interleukin-15 in children with relapsed or resistant neuroblastoma (NCT03294954). Primary and secondary objectives were to assess safety and anti-tumor responses, respectively, with immune response evaluation as an additional objective. We ex vivo expanded highly pure NKT cells (mean ± s.d., 94.7 ± 3.8%) and treated patients with 3 × 106 CAR-NKT cells per square meter of body surface area after lymphodepleting conditioning with cyclophosphamide/fludarabine (Cy/Flu). Cy/Flu conditioning was the probable cause for grade 3-4 hematologic adverse events, as they occurred before CAR-NKT cell infusion, and no dose-limiting toxicities were observed. CAR-NKT cells expanded in vivo, localized to tumors and, in one patient, induced an objective response with regression of bone metastatic lesions. These initial results suggest that CAR-NKT cells can be expanded to clinical scale and safely applied to treat patients with cancer.


Subject(s)
Bone Neoplasms/drug therapy , Natural Killer T-Cells/drug effects , Neuroblastoma/drug therapy , Receptors, Chimeric Antigen/genetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Child , Cyclophosphamide/administration & dosage , Drug Resistance, Neoplasm/immunology , Humans , Immunity/drug effects , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Male , Natural Killer T-Cells/immunology , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/pathology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
9.
Cancer Immunol Res ; 8(3): 309-320, 2020 03.
Article in English | MEDLINE | ID: mdl-31953246

ABSTRACT

Hepatocellular carcinoma (HCC) is the fourth most common cause of cancer-related death in the world, and curative systemic therapies are lacking. Chimeric antigen receptor (CAR)-expressing T cells induce robust antitumor responses in patients with hematologic malignancies but have limited efficacy in patients with solid tumors, including HCC. IL15 and IL21 promote T-cell expansion, survival, and function and can improve the antitumor properties of T cells. We explored whether transgenic expression of IL15 and/or IL21 enhanced glypican-3-CAR (GPC3-CAR) T cells' antitumor properties against HCC. We previously optimized the costimulation in GPC3-CARs and selected a second-generation GPC3-CAR incorporating a 4-1BB costimulatory endodomain (GBBz) for development. Here, we generated constructs encoding IL15, IL21, or both with GBBz (15.GBBz, 21.GBBz, and 21.15.GBBz, respectively) and examined the ability of transduced T cells to kill, produce effector cytokines, and expand in an antigen-dependent manner. We performed gene-expression and phenotypic analyses of GPC3-CAR T cells and CRISPR-Cas9 knockout of the TCF7 gene. Finally, we measured GPC3-CAR T-cell antitumor activity in murine xenograft models of GPC3+ tumors. The increased proliferation of 21.15.GBBz T cells was at least in part dependent on the upregulation and maintenance of TCF-1 (encoded by TCF7) and associated with a higher percentage of stem cell memory and central memory populations after manufacturing. T cells expressing 21.15.GBBz had superior in vitro and in vivo expansion and persistence, and the most robust antitumor activity in vivo These results provided preclinical evidence to support the clinical evaluation of 21.15.GPC3-CAR T cells in patients with HCC.


Subject(s)
Carcinoma, Hepatocellular/therapy , Glypicans/immunology , Immunotherapy, Adoptive/methods , Interleukin-15/immunology , Interleukins/immunology , Liver Neoplasms/therapy , Animals , Apoptosis/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation/physiology , Female , Glypicans/genetics , Humans , Interleukin-15/biosynthesis , Interleukin-15/genetics , Interleukins/biosynthesis , Interleukins/genetics , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 25(23): 7126-7138, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31484667

ABSTRACT

PURPOSE: Vα24-invariant natural killer T cells (NKT) are attractive carriers for chimeric antigen receptors (CAR) due to their inherent antitumor properties and preferential localization to tumor sites. However, limited persistence of CAR-NKTs in tumor-bearing mice is associated with tumor recurrence. Here, we evaluated whether coexpression of the NKT homeostatic cytokine IL15 with a CAR enhances the in vivo persistence and therapeutic efficacy of CAR-NKTs. EXPERIMENTAL DESIGN: Human primary NKTs were ex vivo expanded and transduced with CAR constructs containing an optimized GD2-specific single-chain variable fragment and either the CD28 or 4-1BB costimulatory endodomain, each with or without IL15 (GD2.CAR or GD2.CAR.15). Constructs that mediated robust CAR-NKT cell expansion were selected for further functional evaluation in vitro and in xenogeneic mouse models of neuroblastoma. RESULTS: Coexpression of IL15 with either costimulatory domain increased CAR-NKT absolute numbers. However, constructs containing 4-1BB induced excessive activation-induced cell death and reduced numeric expansion of NKTs compared with respective CD28-based constructs. Further evaluation of CD28-based GD2.CAR and GD2.CAR.15 showed that coexpression of IL15 led to reduced expression levels of exhaustion markers in NKTs and increased multiround in vitro tumor cell killing. Following transfer into mice bearing neuroblastoma xenografts, GD2.CAR.15 NKTs demonstrated enhanced in vivo persistence, increased localization to tumor sites, and improved tumor control compared with GD2.CAR NKTs. Importantly, GD2.CAR.15 NKTs did not produce significant toxicity as determined by histopathologic analysis. CONCLUSIONS: Our results informed selection of the CD28-based GD2.CAR.15 construct for clinical testing and led to initiation of a first-in-human CAR-NKT cell clinical trial (NCT03294954).


Subject(s)
Cytotoxicity, Immunologic/immunology , Gangliosides/immunology , Immunotherapy, Adoptive/methods , Interleukin-15/immunology , Natural Killer T-Cells/transplantation , Neuroblastoma/therapy , Receptors, Chimeric Antigen/immunology , Animals , Apoptosis , Cell Proliferation , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Natural Killer T-Cells/immunology , Neuroblastoma/immunology , Neuroblastoma/metabolism , Receptors, Antigen, T-Cell/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
J Immunol ; 201(7): 2141-2153, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30111631

ABSTRACT

T cells expressing CD19-specific chimeric Ag receptors (CARs) produce high remission rates in B cell lymphoma, but frequent disease recurrence and challenges in generating sufficient numbers of autologous CAR T cells necessitate the development of alternative therapeutic effectors. Vα24-invariant NKTs have intrinsic antitumor properties and are not alloreactive, allowing for off-the-shelf use of CAR-NKTs from healthy donors. We recently reported that CD62L+ NKTs persist longer and have more potent antilymphoma activity than CD62L- cells. However, the conditions governing preservation of CD62L+ cells during NKT cell expansion remain largely unknown. In this study, we demonstrate that IL-21 preserves this crucial central memory-like NKT subset and enhances its antitumor effector functionality. We found that following antigenic stimulation with α-galactosylceramide, CD62L+ NKTs both expressed IL-21R and secreted IL-21, each at significantly higher levels than CD62L- cells. Although IL-21 alone failed to expand stimulated NKTs, combined IL-2/IL-21 treatment produced more NKTs and increased the frequency of CD62L+ cells versus IL-2 alone. Gene expression analysis comparing CD62L+ and CD62L- cells treated with IL-2 alone or IL-2/IL-21 revealed that the latter condition downregulated the proapoptotic protein BIM selectively in CD62L+ NKTs, protecting them from activation-induced cell death. Moreover, IL-2/IL-21-expanded NKTs upregulated granzyme B expression and produced more TH1 cytokines, leading to enhanced in vitro cytotoxicity of nontransduced and anti-CD19-CAR-transduced NKTs against CD1d+ and CD19+ lymphoma cells, respectively. Further, IL-2/IL-21-expanded CAR-NKTs dramatically increased the survival of lymphoma-bearing NSG mice compared with IL-2-expanded CAR-NKTs. These findings have immediate translational implications for the development of NKT cell-based immunotherapies targeting lymphoma and other malignancies.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukins/metabolism , Lymphoma, B-Cell/therapy , Natural Killer T-Cells/immunology , Th1 Cells/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic , Galactosylceramides/immunology , Granzymes/metabolism , Humans , Interleukin-2/metabolism , L-Selectin/metabolism , Lymphocyte Activation , Lymphoma, B-Cell/immunology , Mice , Natural Killer T-Cells/transplantation , Neoplasm Transplantation , Receptors, Antigen, T-Cell, alpha-beta/metabolism
12.
J Clin Invest ; 126(6): 2341-55, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183388

ABSTRACT

Vα24-invariant natural killer T cells (NKTs) localize to tumors and have inherent antitumor properties, making them attractive chimeric antigen receptor (CAR) carriers for redirected cancer immunotherapy. However, clinical application of CAR-NKTs has been impeded, as mechanisms responsible for NKT expansion and the in vivo persistence of these cells are unknown. Here, we demonstrated that antigen-induced expansion of primary NKTs in vitro associates with the accumulation of a CD62L+ subset and exhaustion of CD62L- cells. Only CD62L+ NKTs survived and proliferated in response to secondary stimulation. When transferred to immune-deficient NSG mice, CD62L+ NKTs persisted 5 times longer than CD62L- NKTs. Moreover, CD62L+ cells transduced with a CD19-specific CAR achieved sustained tumor regression in a B cell lymphoma model. Proliferating CD62L+ cells downregulated or maintained CD62L expression when activated via T cell receptor alone or in combination with costimulatory receptors. We generated HLAnull K562 cell clones that were engineered to express CD1d and costimulatory ligands. Clone B-8-2 (HLAnullCD1dmedCD86high4-1BBLmedOX40Lhigh) induced the highest rates of NKT expansion and CD62L expression. B-8-2-expanded CAR-NKTs exhibited prolonged in vivo persistence and superior therapeutic activities in models of lymphoma and neuroblastoma. Therefore, we have identified CD62L as a marker of a distinct NKT subset endowed with high proliferative potential and have developed artificial antigen-presenting cells that generate CD62L-enriched NKTs for effective cancer immunotherapy.


Subject(s)
L-Selectin/metabolism , Natural Killer T-Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans , Immunotherapy, Adoptive , Lymphocyte Activation , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Natural Killer T-Cells/classification , Neuroblastoma/immunology , Neuroblastoma/therapy , Receptors, Antigen/immunology , Recombinant Fusion Proteins/immunology , Xenograft Model Antitumor Assays
13.
Cancer Res ; 74(21): 6260-70, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25213323

ABSTRACT

Vaccines explored for cancer therapy have been based generally on injectable vector systems used to control foreign infectious pathogens, to which the immune system evolved to respond naturally. However, these vectors may not be effective at presenting tumor-associated antigens (TAA) to the immune system in a manner that is sufficient to engender antitumor responses. We addressed this issue with a novel orally administered Salmonella-based vector that exploits a type III secretion system to deliver selected TAA in the cytosol of professional antigen-presenting cells in situ. A systematic comparison of candidate genes from the Salmonella Pathogenicity Island 2 (SPI2) locus was conducted in the vaccine design, using model antigens and a codon-optimized form of the human TAA survivin (coSVN), an oncoprotein that is overexpressed in most human cancers. In a screen of 20 SPI2 promoter:effector combinations, a PsifB::sseJ combination exhibited maximal potency for antigen translocation into the APC cytosol, presentation to CD8 T cells, and murine immunogenicity. In the CT26 mouse model of colon carcinoma, therapeutic vaccination with a lead PsifB::sseJ-coSVN construct (p8032) produced CXCR3-dependent infiltration of tumors by CD8 T cells, reversed the CD8:Treg ratio at the tumor site, and triggered potent antitumor activity. Vaccine immunogenicity and antitumor potency were enhanced by coadministration of the natural killer T-cell ligand 7DW8-5, which heightened the production of IL12 and IFNγ. Furthermore, combined treatment with p8032 and 7DW8-5 resulted in complete tumor regression in A20 lymphoma-bearing mice, where protective memory was demonstrated. Taken together, our results demonstrate how antigen delivery using an oral Salmonella vector can provide an effective platform for the development of cancer vaccines.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Neoplasms/drug therapy , Salmonella typhimurium/immunology , Vaccines, Attenuated/administration & dosage , Animals , Antigen-Presenting Cells/immunology , Bacterial Proteins/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Humans , Membrane Proteins/genetics , Mice , Natural Killer T-Cells/immunology , Neoplasms/immunology , Neoplasms/prevention & control , Salmonella typhimurium/genetics , Vaccines, Attenuated/immunology
14.
Blood ; 124(18): 2824-33, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25049283

ABSTRACT

Advances in the design of chimeric antigen receptors (CARs) have improved the antitumor efficacy of redirected T cells. However, functional heterogeneity of CAR T cells limits their therapeutic potential and is associated with toxicity. We proposed that CAR expression in Vα24-invariant natural killer T (NKT) cells can build on the natural antitumor properties of these cells while their restriction by monomorphic CD1d limits toxicity. Primary human NKT cells were engineered to express a CAR against the GD2 ganglioside (CAR.GD2), which is highly expressed by neuroblastoma (NB). We compared CAR.GD2 constructs that encoded the CD3ζ chain alone, with CD28, 4-1BB, or CD28 and 4-1BB costimulatory endodomains. CAR.GD2 expression rendered NKT cells highly cytotoxic against NB cells without affecting their CD1d-dependent reactivity. We observed a striking T helper 1-like polarization of NKT cells by 4-1BB-containing CARs. Importantly, expression of both CD28 and 4-1BB endodomains in the CAR.GD2 enhanced in vivo persistence of NKT cells. These CAR.GD2 NKT cells effectively localized to the tumor site had potent antitumor activity, and repeat injections significantly improved the long-term survival of mice with metastatic NB. Unlike T cells, CAR.GD2 NKT cells did not induce graft-versus-host disease. These results establish the potential of NKT cells to serve as a safe and effective platform for CAR-directed cancer immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy , Natural Killer T-Cells/immunology , Neuroblastoma/immunology , Neuroblastoma/therapy , Receptors, Antigen/immunology , 4-1BB Ligand/chemistry , 4-1BB Ligand/metabolism , Animals , Antigens, CD1d/metabolism , CD28 Antigens/metabolism , Cell Line , Cell Proliferation , Cytokines/metabolism , Cytotoxicity, Immunologic , Gangliosides , Graft vs Host Disease/immunology , Humans , Lymphocyte Activation/immunology , Macrophages/metabolism , Mice , Neoplasm Metastasis , Neuroblastoma/pathology , Protein Structure, Tertiary , Retroviridae/genetics , Transduction, Genetic , Treatment Outcome
15.
Cancer Res ; 73(13): 4134-46, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23687340

ABSTRACT

Neuroblastoma is a neural crest-derived embryonal malignancy, which accounts for 13% of all pediatric cancer mortality, primarily due to tumor recurrence. Therapy-resistant cancer stem cells are implicated in tumor relapse, but definitive phenotypic evidence of the existence of these cells has been lacking. In this study, we define a highly tumorigenic subpopulation in neuroblastoma with stem cell characteristics, based on the expression of CSF3R, which encodes the receptor for granulocyte colony-stimulating factor (G-CSF). G-CSF receptor positive (aka G-CSFr(+) or CD114(+)) cells isolated from a primary tumor and the NGP cell line by flow cytometry were highly tumorigenic and capable of both self-renewal and differentiation to progeny cells. CD114(+) cells closely resembled embryonic and induced pluripotent stem cells with respect to their profiles of cell cycle, miRNA, and gene expression. In addition, they reflect a primitive undifferentiated neuroectodermal/neural crest phenotype revealing a developmental hierarchy within neuroblastoma tumors. We detected this dedifferentiated neural crest subpopulation in all established neuroblastoma cell lines, xenograft tumors, and primary tumor specimens analyzed. Ligand activation of CD114 by the addition of exogenous G-CSF to CD114(+) cells confirmed intact STAT3 upregulation, characteristic of G-CSF receptor signaling. Together, our data describe a novel distinct subpopulation within neuroblastoma with enhanced tumorigenicity and a stem cell-like phenotype, further elucidating the complex heterogeneity of solid tumors such as neuroblastoma. We propose that this subpopulation may represent an additional target for novel therapeutic approaches to this aggressive pediatric malignancy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/metabolism , Neuroblastoma/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Female , Granulocyte Colony-Stimulating Factor/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Phenotype , Proto-Oncogene Proteins/genetics , STAT3 Transcription Factor/metabolism , Side-Population Cells/metabolism , Transcriptome , Tumor Suppressor Protein p53/metabolism
17.
J Clin Invest ; 122(6): 2221-33, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22565311

ABSTRACT

Vα24-invariant NKT cells inhibit tumor growth by targeting tumor-associated macrophages (TAMs). Tumor progression therefore requires that TAMs evade NKT cell activity through yet-unknown mechanisms. Here we report that a subset of cells in neuroblastoma (NB) cell lines and primary tumors expresses membrane-bound TNF-α (mbTNF-α). These proinflammatory tumor cells induced production of the chemokine CCL20 from TAMs via activation of the NF-κB signaling pathway, an effect that was amplified in hypoxia. Flow cytometry analyses of human primary NB tumors revealed selective accumulation of CCL20 in TAMs. Neutralization of the chemokine inhibited in vitro migration of NKT cells toward tumor-conditioned hypoxic monocytes and localization of NKT cells to NB grafts in mice. We also found that hypoxia impaired NKT cell viability and function. Thus, CCL20-producing TAMs served as a hypoxic trap for tumor-infiltrating NKT cells. IL-15 protected antigen-activated NKT cells from hypoxia, and transgenic expression of IL-15 in adoptively transferred NKT cells dramatically enhanced their antimetastatic activity in mice. Thus, tumor-induced chemokine production in hypoxic TAMs and consequent chemoattraction and inhibition of NKT cells represents a mechanism of immune escape that can be reversed by adoptive immunotherapy with IL-15-transduced NKT cells.


Subject(s)
Interleukin-15/immunology , Macrophages/immunology , Natural Killer T-Cells/immunology , Neuroblastoma/immunology , Adolescent , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/immunology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Child , Child, Preschool , Female , Humans , Immunotherapy, Adoptive , Interleukin-15/genetics , Macrophages/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Natural Killer T-Cells/pathology , Neoplasm Transplantation , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/therapy , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Transduction, Genetic , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
18.
Eur J Immunol ; 41(11): 3312-22, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21818755

ABSTRACT

Efficient induction of antigen-specific immunity is achieved by delivering multiple doses of vaccine formulated with appropriate adjuvants that can harness the benefits of innate immune mediators. The synthetic glycolipid α-galactosylceramide (α-GalCer) is a potent activator of NKT cells, a major innate immune mediator cell type effective in inducing maturation of DCs for efficient presentation of co-administered antigens. However, systemic administration of α-GalCer results in NKT cell anergy in which the cells are unresponsive to subsequent doses of α-GalCer. We show here that α-GalCer delivered as an adjuvant by the intranasal route, as opposed to the intravenous route, enables repeated activation of NKT cells and DCs, resulting in efficient induction of cellular immune responses to co-administered antigens. We show evidence that after intranasal delivery,α-GalCer is selectively presented by DCs for the activation of NKT cells, not B cells. Furthermore, higher levels of PD-1 expression, a potential marker for functional exhaustion of the NKT cells when α-GalCer is delivered by the intravenous route, are not observed after intranasal delivery. These results support a mucosal route of delivery for the utility of α-GalCer as an adjuvant for vaccines, which often requires repeated dosing to achieve durable protective immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Galactosylceramides/administration & dosage , Lymphocyte Activation/drug effects , Natural Killer T-Cells/drug effects , Respiratory Mucosa/immunology , Administration, Intranasal , Animals , Cell Separation , Female , Flow Cytometry , Galactosylceramides/immunology , Immunity, Mucosal/drug effects , Immunity, Mucosal/immunology , Injections, Intravenous , Lung/cytology , Lung/drug effects , Lung/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/immunology , Respiratory Mucosa/drug effects , Vaccination/methods
19.
J Vis Exp ; (38)2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20414189

ABSTRACT

The rhesus macaque model is currently the best available model for HIV-AIDS with respect to understanding the pathogenesis as well as for the development of vaccines and therapeutics(1,2,3). Here, we describe a method for the detailed phenotypic and functional analyses of cellular immune responses, specifically intracellular cytokine production by CD4+ and CD8+ T cells as well as the individual memory subsets. We obtained precise quantitative and qualitative measures for the production of interferon gamma (INF-) and interleukin (IL) -2 in both CD4+ and CD8+ T cells from the rhesus macaque PBMC stimulated with PMA plus ionomycin (PMA+I). The cytokine profiles were different in the different subsets of memory cells. Furthermore, this protocol provided us the sensitivity to demonstrate even minor fractions of antigen specific CD4+ and CD8+ T cell subsets within the PBMC samples from rhesus macaques immunized with an HIV envelope peptide cocktail vaccine developed in our laboratory. The multicolor flow cytometry technique is a powerful tool to precisely identify different populations of T cells (4,5) with cytokine-producing capability(6) following non-specific or antigen-specific stimulation (5,7).


Subject(s)
Flow Cytometry/methods , Macaca mulatta/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Immunity, Cellular , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Leukocytes, Mononuclear/immunology , Macaca mulatta/blood , T-Lymphocyte Subsets/immunology
20.
Proc Natl Acad Sci U S A ; 106(39): 16776-81, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19805372

ABSTRACT

Mucosal dendritic cells have been implicated in the capture, storage, and transmission of HIV to CD4(+) T cells as well as in the promotion of HIV replication in activated CD4(+) T cells during the cognate T-cell and DC interaction. We report that HIV induces human genital mucosal epithelial cells to produce thymic stromal lymphopoietin (TSLP) via activation of the NFkappaB signaling pathway. The TSLP secreted by HIV exposed epithelial cells activated DC, which promoted proliferation and HIV-1 replication of co-cultured autologous CD4(+) T cells. In rhesus macaques, we observed dramatic increases in TSLP expression concurrent with an increase in viral replication in the vaginal tissues within the first 2 weeks after vaginal SIV exposure. These data suggest that HIV-mediated TSLP production by mucosal epithelial cells is a critical trigger for DC-mediated amplification of HIV-infection in activated CD4(+) T cells. The cross talk between mucosal epithelial cells and DC, mediated by HIV-induced TSLP, may be an important mechanism for the high rate of HIV infection in women through the vaginal mucosa.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , HIV-1/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , HIV-1/pathogenicity , Humans , Macaca mulatta , NF-kappa B/metabolism , Simian Immunodeficiency Virus/pathogenicity , Thymic Stromal Lymphopoietin
SELECTION OF CITATIONS
SEARCH DETAIL
...