Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 133(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-36951942

ABSTRACT

Patients with small cell lung cancer (SCLC) generally have a poor prognosis and a median overall survival of only about 13 months, indicating the urgent need for novel therapies. Delta-like protein 3 (DLL3) has been identified as a tumor-specific cell surface marker on neuroendocrine cancers, including SCLC. In this study, we developed a chimeric antigen receptor (CAR) against DLL3 that displays antitumor efficacy in xenograft and murine SCLC models. CAR T cell expression of the proinflammatory cytokine IL-18 greatly enhanced the potency of DLL3-targeting CAR T cell therapy. In a murine metastatic SCLC model, IL-18 production increased the activation of both CAR T cells and endogenous tumor-infiltrating lymphocytes. We also observed an increased infiltration, repolarization, and activation of antigen-presenting cells. Additionally, human IL-18-secreting anti-DLL3 CAR T cells showed an increased memory phenotype, less exhaustion, and induced durable responses in multiple SCLC models, an effect that could be further enhanced with anti-PD-1 blockade. All together, these results define DLL3-targeting CAR T cells that produce IL-18 as a potentially promising novel strategy against DLL3-expressing solid tumors.


Subject(s)
Carcinoma, Neuroendocrine , Interleukin-18 , Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Humans , Mice , Cell Line, Tumor , Immunotherapy, Adoptive , Interleukin-18/pharmacology , Interleukin-18/therapeutic use , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , T-Lymphocytes/pathology , Xenograft Model Antitumor Assays
2.
Leukemia ; 35(2): 506-521, 2021 02.
Article in English | MEDLINE | ID: mdl-32447345

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy has shown remarkable responses in B-cell malignancies. However, many patients suffer from limited response and tumor relapse due to lack of persisting CAR T cells and immune escape. These clinical challenges have compromised the long-term efficacy of CAR T-cell therapy and call for the development of novel CAR designs. We demonstrated that CAR T cells secreting a cytokine interleukin-36γ (IL-36γ) showed significantly improved CAR T-cell expansion and persistence, and resulted in superior tumor eradication compared with conventional CAR T cells. The enhanced cellular function by IL-36γ was mediated through an autocrine manner. In addition, activation of endogenous antigen-presenting cells (APCs) and T cells by IL-36γ aided the formation of a secondary antitumor response, which delayed the progression of antigen-negative tumor challenge. Together, our data provide preclinical evidence to support the translation of this design for an improved CAR T-cell-mediated antitumor response.


Subject(s)
Immunity, Cellular/immunology , Immunotherapy, Adoptive/methods , Interleukin-1/metabolism , Lymphocyte Activation/immunology , Lymphoma, T-Cell/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Animals , Antigen-Presenting Cells , Apoptosis , Cell Proliferation , Female , Humans , Interleukin-1/genetics , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myeloid Differentiation Factor 88/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Nat Commun ; 11(1): 6171, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33268774

ABSTRACT

While effective in specific settings, adoptive chimeric antigen receptor (CAR) T cell therapy for cancer requires further improvement and optimization. Our previous results show that CD40L-overexpressing CAR T cells mobilize endogenous immune effectors, resulting in improved antitumor immunity. However, the cell populations required for this protective effect remain to be identified. Here we show, by analyzing Batf3-/- mice lacking the CD103+ conventional dendritic cell type 1 (cDC1) subpopulation important for antigen cross-presentation, that CD40L-overexpressing CAR T cells elicit an impaired antitumor response in the absence of cDC1s. We further find that CD40L-overexpressing CAR T cells stimulate tumor-resident CD11b-CD103- double-negative (DN) cDCs to proliferate and differentiate into cDC1s in wild-type mice. Finally, re-challenge experiments show that endogenous CD8+ T cells are required for protective antitumor memory in this setting. Our findings thus demonstrate the stimulatory effect of CD40L-overexpressing CAR T cells on innate and adaptive immune cells, and provide a rationale for using CD40L-overexpressing CAR T cells to improve immunotherapy responses.


Subject(s)
CD40 Ligand/genetics , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/genetics , Adaptive Immunity , Animals , Antigen Presentation , Antigens, CD/genetics , Antigens, CD/immunology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD11b Antigen/deficiency , CD11b Antigen/genetics , CD11b Antigen/immunology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Female , Gene Expression , Immunity, Innate , Immunophenotyping , Integrin alpha Chains/deficiency , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Transplantation , Receptors, Chimeric Antigen/immunology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Repressor Proteins/immunology
4.
Blood Cancer Discov ; 1(2): 146-154, 2020 09.
Article in English | MEDLINE | ID: mdl-33089218

ABSTRACT

CAR T-cell therapy for multiple myeloma (MM) targeting B-cell maturation antigen (TNFRSF17; BCMA) induces high overall response rates; however, relapse occurs commonly. Implicated in relapse is a reservoir of MM if cells lacking sufficient BCMA surface expression (antigen escape). We demonstrate that simultaneous targeting of an additional antigen-here, G protein-coupled receptor class-C group-5 member-D (GPRC5D)-can prevent BCMA escape-mediated relapse in a model of MM. To identify an optimal approach, we compare subtherapeutic doses of different forms of dual-targeted cellular therapy. These include (1) parallel-produced and pooled mono-targeted CAR T-cells, (2) bicistronic constructs expressing distinct CARs from a single vector, and (3) a dual-scFv "single-stalk" CAR design. When targeting BCMA-negative disease, bicistronic and pooled approaches had the highest efficacy, whereas for dual-antigen-expressing disease, the bicistronic approach was more efficacious than the pooled approach. Mechanistically, expressing two CARs on a single cell enhanced the strength of CAR T-cell/target cell interactions.


Subject(s)
Immunotherapy, Adoptive , Multiple Myeloma , B-Cell Maturation Antigen/genetics , Humans , Multiple Myeloma/genetics , Neoplasm Recurrence, Local , Receptors, G-Protein-Coupled/genetics
5.
Cancer Immunol Res ; 8(6): 732-742, 2020 06.
Article in English | MEDLINE | ID: mdl-32213625

ABSTRACT

Although clinical responses with CD19-targeting chimeric antigen receptor (CAR) T-cell treatment have been observed in patients with certain hematologic malignancies, high rates of disease relapse highlight the necessity to understand and improve mechanisms of CAR T-cell failure. Because T-cell dysfunction is thought to contribute to CAR T-cell treatment failure, understanding what mechanisms drive T cells into this dysfunctional state may aid optimal design of efficacious CAR T cells. Dysfunctional CAR T cells have been characterized as having upregulated inhibitory receptors and decreased cytolytic capabilities. Previous studies have identified a role for sustained CAR CD3ζ signaling in CAR T-cell dysfunction. Here, we demonstrate a mechanism that drives dysfunction in CAR T cells through excessive costimulation. Fully activated CD19-targeted CAR T cells were rendered dysfunctional upon stimulation with both endogenous CD28 stimulation and CAR-mediated CD28 costimulation. Costimulation-driven dysfunction of CAR T cells was demonstrated in a syngeneic immunocompetent mouse model, in which CAR T cells were activated with signals 1 (CD3ζ), 2 (CD28), and 3 (IL12). Thus, we show that CAR T-cell dysfunction can be driven through excessive CD28 and 4-1BB costimulation.See related article by Drakes et al., p. 743.


Subject(s)
CD28 Antigens/immunology , Immunotherapy, Adoptive/methods , Interleukin-12/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Animals , Antigens, CD19/immunology , Antigens, CD19/metabolism , Apoptosis , CD28 Antigens/metabolism , Cell Proliferation , Cytokines , Humans , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancer Immunol Res ; 8(6): 743-755, 2020 06.
Article in English | MEDLINE | ID: mdl-32209638

ABSTRACT

T-cell receptor (TCR)-modified T-cell gene therapy can target a variety of extracellular and intracellular tumor-associated antigens, yet has had little clinical success. A potential explanation for limited antitumor efficacy is a lack of T-cell activation in vivo We postulated that expression of proinflammatory cytokines in TCR-modified T cells would activate T cells and enhance antitumor efficacy. We demonstrate that expression of interleukin 18 (IL18) in tumor-directed TCR-modified T cells provides a superior proinflammatory signal than expression of interleukin 12 (IL12). Tumor-targeted T cells secreting IL18 promote persistent and functional effector T cells and a proinflammatory tumor microenvironment. Together, these effects augmented overall survival of mice in the pmel-1 syngeneic tumor model. When combined with sublethal irradiation, IL18-secreting pmel-1 T cells were able to eradicate tumors, whereas IL12-secreting pmel-1 T cells caused toxicity in mice through excessive cytokine secretion. In another xenograft tumor model, IL18 secretion enhanced the persistence and antitumor efficacy of NY-ESO-1-reactive TCR-modified human T cells as well as overall survival of tumor-bearing mice. These results demonstrate a rationale for optimizing the efficacy of TCR-modified T-cell cancer therapy through expression of IL18.See related commentary by Wijewarnasuriya et al., p. 732.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/standards , Interleukin-12/metabolism , Interleukin-18/metabolism , Lymphocyte Activation/immunology , Melanoma/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Humans , Immunotherapy, Adoptive/methods , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Knockout , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Cancer Cell ; 35(3): 473-488.e6, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30889381

ABSTRACT

Chimeric antigen receptor (CAR) T cells provide great efficacy in B cell malignancies. However, improved CAR T cell therapies are still needed. Here, we engineered tumor-targeted CAR T cells to constitutively express the immune-stimulatory molecule CD40 ligand (CD40L) and explored efficacy in different mouse leukemia/lymphoma models. We observed that CD40L+ CAR T cells circumvent tumor immune escape via antigen loss through CD40/CD40L-mediated cytotoxicity and induction of a sustained, endogenous immune response. After adoptive cell transfer, the CD40L+ CAR T cells displayed superior antitumor efficacy, licensed antigen-presenting cells, enhanced recruitment of immune effectors, and mobilized endogenous tumor-recognizing T cells. These effects were absent in Cd40-/- mice and provide a rationale for the use of CD40L+ CAR T cells in cancer treatment.


Subject(s)
CD40 Ligand/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Animals , Antigen-Presenting Cells/immunology , CD40 Ligand/genetics , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Receptors, Chimeric Antigen/genetics , Tumor Escape , Xenograft Model Antitumor Assays
8.
Appl Environ Microbiol ; 71(7): 3858-65, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16000798

ABSTRACT

Members of the family Geobacteraceae are commonly the predominant Fe(III)-reducing microorganisms in sedimentary environments, as well as on the surface of energy-harvesting electrodes, and are able to effectively couple the oxidation of acetate to the reduction of external electron acceptors. Citrate synthase activity of these organisms is of interest due to its key role in acetate metabolism. Prior sequencing of the genome of Geobacter sulfurreducens revealed a putative citrate synthase sequence related to the citrate synthases of eukaryotes. All citrate synthase activity in G. sulfurreducens could be resolved to a single 49-kDa protein via affinity chromatography. The enzyme was successfully expressed at high levels in Escherichia coli with similar properties as the native enzyme, and kinetic parameters were comparable to related citrate synthases (kcat= 8.3 s(-1); Km= 14.1 and 4.3 microM for acetyl coenzyme A and oxaloacetate, respectively). The enzyme was dimeric and was slightly inhibited by ATP (Ki= 1.9 mM for acetyl coenzyme A), which is a known inhibitor for many eukaryotic, dimeric citrate synthases. NADH, an allosteric inhibitor of prokaryotic hexameric citrate synthases, did not affect enzyme activity. Unlike most prokaryotic dimeric citrate synthases, the enzyme did not have any methylcitrate synthase activity. A unique feature of the enzyme, in contrast to citrate synthases from both eukaryotes and prokaryotes, was a lack of stimulation by K+ ions. Similar citrate synthase sequences were detected in a diversity of other Geobacteraceae members. This first characterization of a eukaryotic-like citrate synthase from a prokaryote provides new insight into acetate metabolism in Geobacteraceae members and suggests a molecular target for tracking the presence and activity of these organisms in the environment.


Subject(s)
Citrate (si)-Synthase , Deltaproteobacteria/enzymology , Geobacter/enzymology , Amino Acid Sequence , Citrate (si)-Synthase/chemistry , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/isolation & purification , Citrate (si)-Synthase/metabolism , Culture Media , DNA, Bacterial/analysis , Deltaproteobacteria/genetics , Deltaproteobacteria/growth & development , Eukaryotic Cells/enzymology , Geobacter/genetics , Geobacter/growth & development , Kinetics , Molecular Sequence Data , Phylogeny , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...