Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(2): 101421, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38340727

ABSTRACT

Chimeric antigen receptor T cell (CAR T) therapy is a potent treatment for relapsed/refractory (r/r) B cell lymphomas but provides lasting remissions in only ∼40% of patients and is associated with serious adverse events. We identify an upregulation of CD80 and/or CD86 in tumor tissue of (r/r) diffuse large B cell lymphoma (DLBCL) patients treated with tisagenlecleucel. This finding leads to the development of the CAR/CCR (chimeric checkpoint receptor) design, which consists of a CD19-specific first-generation CAR co-expressed with a recombinant CTLA-4-linked receptor with a 4-1BB co-stimulatory domain. CAR/CCR T cells demonstrate superior efficacy in xenograft mouse models compared with CAR T cells, superior long-term activity, and superior selectivity in in vitro assays with non-malignant CD19+ cells. In addition, immunocompetent mice show an intact CD80-CD19+ B cell population after CAR/CCR T cell treatment. The results reveal the CAR/CCR design as a promising strategy for further translational study.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , T-Lymphocytes , Humans , Animals , Mice , CTLA-4 Antigen , Lymphoma, Large B-Cell, Diffuse/therapy , Lymphoma, Large B-Cell, Diffuse/etiology , Immunotherapy, Adoptive/methods , B-Lymphocytes , Antigens, CD19/genetics
2.
Blood Cancer Discov ; 4(1): 78-97, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36346827

ABSTRACT

Genomic profiling revealed the identity of at least 5 subtypes of diffuse large B-cell lymphoma (DLBCL), including the MCD/C5 cluster characterized by aberrations in MYD88, BCL2, PRDM1, and/or SPIB. We generated mouse models harboring B cell-specific Prdm1 or Spib aberrations on the background of oncogenic Myd88 and Bcl2 lesions. We deployed whole-exome sequencing, transcriptome, flow-cytometry, and mass cytometry analyses to demonstrate that Prdm1- or Spib-altered lymphomas display molecular features consistent with prememory B cells and light-zone B cells, whereas lymphomas lacking these alterations were enriched for late light-zone and plasmablast-associated gene sets. Consistent with the phenotypic evidence for increased B cell receptor signaling activity in Prdm1-altered lymphomas, we demonstrate that combined BTK/BCL2 inhibition displays therapeutic activity in mice and in five of six relapsed/refractory DLBCL patients. Moreover, Prdm1-altered lymphomas were immunogenic upon transplantation into immuno-competent hosts, displayed an actionable PD-L1 surface expression, and were sensitive to antimurine-CD19-CAR-T cell therapy, in vivo. SIGNIFICANCE: Relapsed/refractory DLBCL remains a major medical challenge, and most of these patients succumb to their disease. Here, we generated mouse models, faithfully recapitulating the biology of MYD88-driven human DLBCL. These models revealed robust preclinical activity of combined BTK/BCL2 inhibition. We confirmed activity of this regimen in pretreated non-GCB-DLBCL patients. See related commentary by Leveille et al., p. 8. This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Myeloid Differentiation Factor 88 , Humans , Mice , Animals , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , B-Lymphocytes , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/therapy , Plasma Cells/metabolism , Plasma Cells/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/therapeutic use
3.
Front Immunol ; 13: 934343, 2022.
Article in English | MEDLINE | ID: mdl-36032080

ABSTRACT

CAR (Chimeric Antigen Receptor) T-cell therapy has revolutionized the field of oncology in recent years. This innovative shift in cancer treatment also provides the opportunity to improve therapies for many patients suffering from various autoimmune diseases. Recent studies have confirmed the therapeutic suppressive potential of regulatory T cells (Tregs) to modulate immune response in autoimmune diseases. However, the polyclonal character of regulatory T cells and their unknown TCR specificity impaired their therapeutic potency in clinical implementation. Genetical engineering of these immune modulating cells to express antigen-specific receptors and using them therapeutically is a logical step on the way to overcome present limitations of the Treg strategy for the treatment of autoimmune diseases. Encouraging preclinical studies successfully demonstrated immune modulating properties of CAR Tregs in various mouse models. Still, there are many concerns about targeted Treg therapies relating to CAR target selectivity, suppressive functions, phenotype stability and safety aspects. Here, we summarize recent developments in CAR design, Treg biology and future strategies and perspectives in CAR Treg immunotherapy aiming at clinical translation.


Subject(s)
Autoimmune Diseases , T-Lymphocytes, Regulatory , Animals , Immunotherapy , Immunotherapy, Adoptive , Mice , Receptors, Antigen
4.
Front Immunol ; 13: 865424, 2022.
Article in English | MEDLINE | ID: mdl-35784280

ABSTRACT

T cells modified for expression of Chimeric Antigen Receptors (CARs) were the first gene-modified cell products approved for use in cancer immunotherapy. CAR-T cells engineered with gammaretroviral or lentiviral vectors (RVs/LVs) targeting B-cell lymphomas and leukemias have shown excellent clinical efficacy and no malignant transformation due to insertional mutagenesis to date. Large-scale production of RVs/LVs under good-manufacturing practices for CAR-T cell manufacturing has soared in recent years. However, manufacturing of RVs/LVs remains complex and costly, representing a logistical bottleneck for CAR-T cell production. Emerging gene-editing technologies are fostering a new paradigm in synthetic biology for the engineering and production of CAR-T cells. Firstly, the generation of the modular reagents utilized for gene editing with the CRISPR-Cas systems can be scaled-up with high precision under good manufacturing practices, are interchangeable and can be more sustainable in the long-run through the lower material costs. Secondly, gene editing exploits the precise insertion of CARs into defined genomic loci and allows combinatorial gene knock-ins and knock-outs with exciting and dynamic perspectives for T cell engineering to improve their therapeutic efficacy. Thirdly, allogeneic edited CAR-effector cells could eventually become available as "off-the-shelf" products. This review addresses important points to consider regarding the status quo, pending needs and perspectives for the forthright evolution from the viral towards gene editing developments for CAR-T cells.


Subject(s)
Gene Editing , Receptors, Chimeric Antigen , CRISPR-Cas Systems , Immunotherapy , T-Lymphocytes
5.
Mol Ther ; 30(2): 593-605, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34678512

ABSTRACT

Chimeric antigen receptor (CAR)-redirected T cell therapy often fails to control tumors in the long term due to selecting cancer cells that downregulated or lost CAR targeted antigen. To reprogram the functional capacities specifically of engineered CAR T cells, we inserted IL12 into the extracellular moiety of a CD28-ζ CAR; both the CAR endodomain and IL12 were functionally active, as indicated by antigen-redirected effector functions and STAT4 phosphorylation, respectively. The IL12-CAR reprogrammed CD8+ T cells toward a so far not recognized natural killer (NK) cell-like signature and a CD94+CD56+CD62Lhigh phenotype closely similar, but not identical, to NK and cytokine induced killer (CIK) cells. In contrast to conventional CAR T cells, IL12-CAR T cells acquired antigen-independent, human leukocyte antigen E (HLA-E) restricted cytotoxic capacities eliminating antigen-negative cancer cells in addition to eliminating cancer cells with CAR cognate antigen. Simultaneous signaling through both the CAR endodomain and IL12 were required for inducing maximal NK-like cytotoxicity; adding IL12 to conventional CAR T cells was not sufficient. Antigen-negative tumors were attacked by IL12-CAR T cells, but not by conventional CAR T cells. Overall, we present a prototype of a new family of CARs that augments tumor recognition and elimination through expanded functional capacities by an appropriate cytokine integrated into the CAR exodomain.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy, Adoptive , Interleukin-12 , Neoplasms , CD8-Positive T-Lymphocytes/immunology , Humans , Interleukin-12/immunology , Killer Cells, Natural/immunology , Neoplasms/therapy
6.
Nat Commun ; 12(1): 240, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431832

ABSTRACT

CAR-T cell therapy targeting CD19 demonstrated strong activity against advanced B cell leukemia, however shows less efficacy against lymphoma with nodal dissemination. To target both B cell Non-Hodgkin's lymphoma (B-NHLs) and follicular T helper (Tfh) cells in the tumor microenvironment (TME), we apply here a chimeric antigen receptor (CAR) that recognizes human CXCR5 with high avidity. CXCR5, physiologically expressed on mature B and Tfh cells, is also highly expressed on nodal B-NHLs. Anti-CXCR5 CAR-T cells eradicate B-NHL cells and lymphoma-supportive Tfh cells more potently than CD19 CAR-T cells in vitro, and they efficiently inhibit lymphoma growth in a murine xenograft model. Administration of anti-murine CXCR5 CAR-T cells in syngeneic mice specifically depletes endogenous and malignant B and Tfh cells without unexpected on-target/off-tumor effects. Collectively, anti-CXCR5 CAR-T cells provide a promising treatment strategy for nodal B-NHLs through the simultaneous elimination of lymphoma B cells and Tfh cells of the tumor-supporting TME.


Subject(s)
B-Lymphocytes/immunology , Lymphoma, Non-Hodgkin/immunology , Neoplasms/immunology , Receptors, CXCR5/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cell Proliferation , Cell Survival , HEK293 Cells , Hep G2 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Xenograft Model Antitumor Assays
7.
Sci Rep ; 9(1): 8410, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182802

ABSTRACT

The demand for recombinant proteins for analytic and therapeutic purposes is increasing; however, most currently used bacterial production systems accumulate the recombinant proteins in the intracellular space, which requires denaturating procedures for harvesting and functional testing. We here present a novel FimH-based expression system that enables display of fully functional eukaryotic proteins while preventing technical difficulties in translocating, folding, stabilizing and isolating the displayed proteins. As examples, Gaussia Luciferase (GLuc), epidermal growth factor (EGF), transforming growth factor-α (TGF-α) and epiregulin (EPRG) were expressed as FimH fusion proteins on the surface of E. coli bacteria. The fusion proteins were functionally active and could be released from the bacterial surface by specific proteolytic cleavage into the culture supernatant allowing harvesting of the produced proteins. EGFR ligands, produced as FimH fusion proteins and released by proteolytic cleavage, bound to the EGF receptor (EGFR) on cancer cells inducing EGFR phosphorylation. In another application of the technology, GLuc-FimH expressed on the surface of bacteria was used to track tumor-infiltrating bacteria by bioluminescence imaging upon application to mice, thereby visualizing the colonization of transplanted tumors. The examples indicate that the FimH-fusion protein technology can be used in various applications that require functionally active proteins to be displayed on bacterial surfaces or released into the culture supernatant.


Subject(s)
Adhesins, Escherichia coli/metabolism , Cell Surface Display Techniques , Escherichia coli/metabolism , Fimbriae Proteins/metabolism , Recombinant Proteins/metabolism , Animals , Biocatalysis , ErbB Receptors/metabolism , Escherichia coli/ultrastructure , Ligands , Luciferases/metabolism , Mice, Inbred C57BL , Phosphorylation , Transformation, Genetic
8.
Stem Cells ; 36(11): 1752-1763, 2018 11.
Article in English | MEDLINE | ID: mdl-30063808

ABSTRACT

The trabecular extracellular matrix (ECM) forms a three-dimensional scaffold to stabilize the bone marrow, provide substrates for cell-matrix interactions and retain, present or release signals to modulate hematopoietic stem and progenitor cell development. However, the impact of trabecular ECM components on hematopoiesis has been poorly studied. Using collagen IX alpha1 - deficient (Col9a1(-/-) ) mice, we revealed that a lack of collagen IX alpha1 results in a disorganized trabecular network enriched in fibronectin, and in a reduction in myeloid cells, which was accompanied by a decrease in colony-stimulating factor 1 receptor expression on monocytes from the bone marrow. In contrast, B-cell numbers in the bone marrow and T-cell numbers in the thymus remained unchanged. Alterations in the bone marrow microenvironment may not only reduce myeloid cell numbers, but also have long-term implications for myeloid cell function. Mice were infected with Listeria moncytogenes to analyze the function of myeloid cells. In this case, an inadequate macrophage-dependent clearance of bacterial infections was observed in Col9a1(-/-) mice in vivo. This was mainly caused by an impaired interferon-gamma/tumor necrosis factor-alpha-mediated activation of macrophages. The loss of collagen IX alpha1 therefore destabilizes the trabecular bone network, impairs myeloid cell differentiation, and affects the innate immune response against Listeria. Stem Cells 2018;36:1752-1763.


Subject(s)
Collagen/metabolism , Myeloid Cells/metabolism , Animals , Disease Models, Animal , Female , Flow Cytometry , Humans , Mice
9.
Mol Ther ; 26(8): 1906-1920, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30078440

ABSTRACT

Autologous T cells genetically modified with a chimeric antigen receptor (CAR) redirected at CD19 have potent activity in the treatment of B cell leukemia and B cell non-Hodgkin's lymphoma (B-NHL). Immunotherapies to treat multiple myeloma (MM) targeted the B cell maturation antigen (BCMA), which is expressed in most cases of MM. We developed a humanized CAR with specificity for BCMA based on our previously generated anti-BCMA monoclonal antibody. The targeting single-chain variable fragment (scFv) domain exhibited a binding affinity in the low nanomolar range, conferring T cells with high functional avidity. Redirecting T cells by this CAR allowed us to explore BCMA as an alternative target for mature B-NHLs. We validated BCMA expression in diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma, and chronic lymphocytic leukemia. BCMA CAR T cells triggered target cell lysis with an activation threshold in the range of 100 BCMA molecules, which allowed for an efficient eradication of B-NHL cells in vitro and in vivo. Our data corroborate BCMA is a suitable target in B cell tumors beyond MM, providing a novel therapeutic option for patients where BCMA is expressed at low abundance or where anti-CD19 immunotherapies have failed due to antigen loss.


Subject(s)
B-Cell Maturation Antigen/immunology , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Jurkat Cells , Lymphoma, B-Cell/immunology , Mice , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
10.
Cell Rep ; 21(11): 3205-3219, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241547

ABSTRACT

Adoptive therapy with chimeric antigen receptor (CAR)-redirected T cells has achieved remarkable efficacy in the treatment of hematopoietic malignancies. However, eradicating large solid tumors in advanced stages of the disease remains challenging. We explored augmentation of the anti-tumor immune reaction by establishing an acute inflammatory reaction. Systematic screening indicates that IL-18 polarizes CAR T cells toward T-bethigh FoxO1low effectors with an acute inflammatory response. CAR T cells engineered with inducible IL-18 release exhibited superior activity against large pancreatic and lung tumors that were refractory to CAR T cells without cytokines. IL-18 CAR T cell treatment was accompanied by an overall change in the immune cell landscape associated with the tumor. More specifically, CD206- M1 macrophages and NKG2D+ NK cells increased in number, whereas Tregs, suppressive CD103+ DCs, and M2 macrophages decreased, suggesting that "iIL18 TRUCKs" can be used to sensitize large solid tumor lesions for successful immune destruction.


Subject(s)
Forkhead Box Protein O1/immunology , Immunotherapy, Adoptive/methods , Interleukin-18/immunology , Lung Neoplasms/therapy , Pancreatic Neoplasms/therapy , T-Box Domain Proteins/immunology , T-Lymphocytes/immunology , Animals , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Cell Engineering , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Forkhead Box Protein O1/genetics , Gene Expression , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Primary Cell Culture , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Survival Analysis , T-Box Domain Proteins/genetics , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Transgenes , Tumor Cells, Cultured
11.
Front Immunol ; 8: 1125, 2017.
Article in English | MEDLINE | ID: mdl-28955341

ABSTRACT

Cellular therapy with chimeric antigen receptor (CAR)-redirected cytotoxic T cells has shown impressive efficacy in the treatment of hematologic malignancies. We explored a regulatory T cell (Treg)-based therapy in the treatment of allergic airway inflammation, a model for asthma, which is characterized by an airway hyper-reactivity (AHR) and a chronic, T helper-2 (Th2) cell-dominated immune response to allergen. To restore the immune balance in the lung, we redirected Tregs by a CAR toward lung epithelia in mice upon experimentally induced allergic asthma, closely mimicking the clinical situation. Adoptively transferred CAR Tregs accumulated in the lung and in tracheobronchial lymph nodes, reduced AHR and diminished eosinophilic airway inflammation, indicated by lower cell numbers in the bronchoalveolar lavage fluid and decreased cell infiltrates in the lung. CAR Treg cells furthermore prevented excessive pulmonary mucus production as well as increase in allergen-specific IgE and Th2 cytokine levels in exposed animals. CAR Tregs were more efficient in controlling asthma than non-modified Tregs, indicating the pivotal role of specific Treg cell activation in the affected organ. Data demonstrate that lung targeting CAR Treg cells ameliorate key features of experimental airway inflammation, paving the way for cell therapy of severe allergic asthma.

12.
Mol Ther ; 24(8): 1423-34, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27112062

ABSTRACT

Recent clinical trials with chimeric antigen receptor (CAR) redirected T cells targeting CD19 revealed particular efficacy in the treatment of leukemia/lymphoma, however, were accompanied by a lasting depletion of healthy B cells. We here explored CD30 as an alternative target, which is validated in lymphoma therapy and expressed by a broad variety of Hodgkin's and non-Hodgkin's lymphomas. As a safty concern, however, CD30 is also expressed by lymphocytes and hematopoietic stem and progenitor cells (HSPCs) during activation. We revealed that HRS3scFv-derived CAR T cells are superior since they were not blocked by soluble CD30 and did not attack CD30(+) HSPCs while eliminating CD30(+) lymphoma cells. Consequently, normal hemato- and lymphopoiesis was not affected in the long-term in the humanized mouse; the number of blood B and T cells remained unchanged. We provide evidence that the CD30(+) HSPCs are protected against a CAR T-cell attack by substantially lower CD30 levels than lymphoma cells and higher levels of the granzyme B inactivating SP6/PI9 serine protease, which furthermore increased upon activation. Taken together, adoptive cell therapy with anti-CD30 CAR T cells displays a superior therapeutic index in the treatment of CD30(+) malignancies leaving healthy activated lymphocytes and HSPCs unaffected.

13.
J Immunol ; 196(2): 759-66, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26673145

ABSTRACT

Treatment of cancer patients by adoptive T cell therapy has yielded promising results. In solid tumors, however, T cells encounter a hostile environment, in particular with increased inflammatory activity as a hallmark of the tumor milieu that goes along with abundant reactive oxygen species (ROS) that substantially impair antitumor activity. We present a strategy to render antitumor T cells more resilient toward ROS by coexpressing catalase along with a tumor specific chimeric Ag receptor (CAR) to increase their antioxidative capacity by metabolizing H2O2. In fact, T cells engineered with a bicistronic vector that concurrently expresses catalase, along with the CAR coexpressing catalase (CAR-CAT), performed superior over CAR T cells as they showed increased levels of intracellular catalase and had a reduced oxidative state with less ROS accumulation in both the basal state and upon activation while maintaining their antitumor activity despite high H2O2 levels. Moreover, CAR-CAT T cells exerted a substantial bystander protection of nontransfected immune effector cells as measured by CD3ζ chain expression in bystander T cells even in the presence of high H2O2 concentrations. Bystander NK cells, otherwise ROS sensitive, efficiently eliminate their K562 target cells under H2O2-induced oxidative stress when admixed with CAR-CAT T cells. This approach represents a novel means for protecting tumor-infiltrating cells from tumor-associated oxidative stress-mediated repression.


Subject(s)
Catalase/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Oxidative Stress/physiology , T-Lymphocytes/immunology , Blotting, Western , Bystander Effect/immunology , Cell Line , Cell Separation , Humans , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Transfection
14.
Expert Opin Biol Ther ; 15(8): 1145-54, 2015.
Article in English | MEDLINE | ID: mdl-25985798

ABSTRACT

INTRODUCTION: Adoptive cell therapy of malignant diseases takes advantage of the cellular immune system to recognize and destroy cancer cells. This is impressively demonstrated by redirecting T cells with a chimeric antigen receptor (CAR) towards CD19, inducing complete and lasting remission of leukemia in more than two-thirds of patients in early phase trials. AREAS COVERED: We outline how the CAR strategy is highly specific in redirecting T cells towards pre-defined target cells, however, reaches its limits when targeting solid tumors with a tremendous phenotypic heterogeneity. After initial tumor reduction by CAR T cells, antigen-negative cancer cells not recognized by CAR may give rise to tumor relapse. The situation may be overcome by CAR-mediated activation of T cells in the tumor, releasing inducible IL-12 which augments T-cell activation and attracts and activates innate immune cells to eliminate antigen-negative cancer cells in the targeted lesion. EXPERT OPINION: CAR T cells with a transgenic 'payload', so-called TRUCK T cells or the 'fourth-generation' CAR T cells, are worthwhile to explore to shape the tumor environment by the inducible release of transgenic immune modifiers. Such TRUCK T cells are moreover envisioned to be applied in fields beyond cancer therapy including the therapy of virus infections, auto-immune diseases or metabolic disorders.


Subject(s)
Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Humans , Immunotherapy, Adoptive/methods , Interleukin-12/immunology , Leukemia/immunology , Lymphocyte Activation/immunology
15.
J Natl Cancer Inst ; 107(1): 364, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25424197

ABSTRACT

BACKGROUND: One bottleneck for adoptive T cell therapy (ACT) is recruitment of T cells into tumors. We hypothesized that combining tumor-specific T cells, modified with a marker antigen and a bispecific antibody (BiAb) that selectively recognizes transduced T cells and tumor cells would improve T cell recruitment to tumors and enhance therapeutic efficacy. METHODS: SV40 T antigen-specific T cells from T cell receptor (TCR)-I-transgenic mice were transduced with a truncated human epidermal growth factor receptor (EGFR) as a marker protein. Targeting and killing by combined ACT and anti-EGFR-anti-EpCAM BiAb therapy was analyzed in C57Bl/6 mice (n = six to 12 per group) carrying subcutaneous tumors of the murine gastric cancer cell line GC8 (SV40(+) and EpCAM(+)). Anti-EGFR x anti-c-Met BiAb was used for targeting of human tumor-specific T cells to c-Met(+) human tumor cell lines. Differences between experimental conditions were analyzed using the Student's t test, and differences in tumor growth with two-way analysis of variance. Overall survival was analyzed by log-rank test. All statistical tests were two-sided. RESULTS: The BiAb linked EGFR-transduced T cells to tumor cells and enhanced tumor cell lysis. In vivo, the combination of ACT and Biab produced increased T cell infiltration of tumors, retarded tumor growth, and prolonged survival compared with ACT with a control antibody (median survival 95 vs 75 days, P < .001). In human cells, this strategy enhanced recruitment of human EGFR-transduced T cells to immobilized c-Met and recognition of tyrosinase(+) melanoma cells by TCR-, as well as of CEA(+) colon cancer cells by chimeric antigen receptor (CAR)-modified T cells. CONCLUSIONS: BiAb recruitment of tumor-specific T cells transduced with a marker antigen to tumor cells may enhance efficacy of ACT.


Subject(s)
Adoptive Transfer , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , ErbB Receptors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Antigen, T-Cell/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , T-Lymphocytes/immunology , Analysis of Variance , Animals , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , ErbB Receptors/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-met/immunology , Transduction, Genetic
16.
Cancer Res ; 74(23): 6796-805, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25297631

ABSTRACT

Adoptive T-cell therapy using chimeric antigen receptor-modified T cells (CAR-T therapy) has shown dramatic efficacy in patients with circulating lymphoma. However, eradication of solid tumors with CAR-T therapy has not been reported yet to be efficacious. In solid tumors, stroma destruction, due to MHC-restricted cross-presentation of tumor antigens to T cells, may be essential. However, CAR-Ts recognize antigens in an MHC-independent manner on cancer cells but not stroma cells. In this report, we show how CAR-Ts can be engineered to eradicate large established tumors with provision of a suitable CD28 costimulatory signal. In an HER2-dependent tumor model, tumor rejection by HER2-specific CAR-Ts was associated with sustained influx and proliferation of the adoptively transferred T cells. Interestingly, tumor rejection did not involve natural killer cells but was associated instead with a marked increase in the level of M1 macrophages and a requirement for IFNγ receptor expression on tumor stroma cells. Our results argue that CAR-T therapy is capable of eradicating solid tumors through a combination of antigen-independent stroma destruction and antigen-specific tumor cell targeting.


Subject(s)
Interferon-gamma/immunology , Interferon-gamma/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cell Differentiation/immunology , Cell Line, Tumor , Cell Proliferation , Cell- and Tissue-Based Therapy/methods , Humans , Immunotherapy, Adoptive/methods , Macrophages/immunology , Macrophages/metabolism , Mice , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocytes/metabolism
17.
Immunol Rev ; 257(1): 83-90, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24329791

ABSTRACT

Adoptive T-cell therapy recently achieved impressive efficacy in early phase trials, in particular in hematologic malignancies, strongly supporting the notion that the immune system can control cancer. A current strategy of favor is based on ex vivo-engineered patient T cells, which are redirected by a chimeric antigen receptor (CAR) and recognize a predefined target by an antibody-derived binding domain. Such CAR T cells can substantially reduce the tumor burden as long as the targeted antigen is present on the cancer cells. However, given the tremendous phenotypic diversity in solid tumor lesions, a reasonable number of cancer cells are not recognized by a given CAR, considerably reducing the therapeutic success. This article reviews a recently described strategy for overcoming this shortcoming of the CAR T-cell therapy by modulating the tumor stroma by a CAR T-cell-secreted transgenic cytokine like interleukin-12 (IL-12). The basic process is that CAR T cells, when activated by their CAR, deposit IL-12 in the targeted tumor lesion, which in turn attracts an innate immune cell response toward those cancer cells that are invisible to CAR T cells. Such TRUCKs, T cells redirected for universal cytokine-mediated killing, exhibited remarkable efficacy against solid tumors with diverse cancer cell phenotypes, suggesting their evaluation in clinical trials.


Subject(s)
Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cytokines/metabolism , Cytotoxicity, Immunologic , Humans , Immunity, Innate , Immunologic Factors/genetics , Immunologic Factors/metabolism , Immunomodulation , Immunotherapy, Adoptive , Interleukin-12/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Stromal Cells/metabolism
18.
Front Immunol ; 4: 371, 2013.
Article in English | MEDLINE | ID: mdl-24273543

ABSTRACT

Adoptive T-cell therapy has recently shown promise in initiating a lasting anti-tumor response with spectacular therapeutic success in some cases. Specific T-cell therapy, however, is limited since a number of cancer cells are not recognized by T cells due to various mechanisms including the limited availability of tumor-specific T cells and deficiencies in antigen processing or major histocompatibility complex (MHC) expression of cancer cells. To make adoptive cell therapy applicable for the broad variety of cancer entities, patient's T cells are engineered ex vivo with pre-defined specificity by a recombinant chimeric antigen receptor (CAR) which consists in the extracellular part of an antibody-derived domain for binding with a "tumor-associated antigen" and in the intracellular part of a T-cell receptor (TCR)-derived signaling moiety for T-cell activation. The specificity of CAR-mediated T-cell recognition is defined by the antibody domain, is independent of MHC presentation and can be extended to any target for which an antibody is available. We discuss the advantages and limitations of MHC-independent T-cell targeting by an engineered CAR in comparison to TCR modified T cells and the impact of the CAR activation threshold on redirected T-cell activation. Finally we review most significant progress recently made in early stage clinical trials to treat cancer.

19.
Gastroenterology ; 145(2): 456-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639914

ABSTRACT

BACKGROUND & AIMS: Antiviral agents suppress hepatitis B virus (HBV) replication but do not clear the infection. A strong effector T-cell response is required to eradicate HBV, but this does not occur in patients with chronic infection. T cells might be directed toward virus-infected cells by expressing HBV-specific receptors and thereby clear HBV and help to prevent development of liver cancer. In mice, we studied whether redirected T cells can engraft after adoptive transfer, without prior T-cell depletion, and whether the large amounts of circulating viral antigens inactivate the transferred T cells or lead to uncontrolled immune-mediated damage. METHODS: CD8(+) T cells were isolated from mice and stimulated using an optimized protocol. Chimeric antigen receptors (CARs) that bind HBV envelope proteins (S-CAR) and activate T cells were expressed on the surface of cells using retroviral vectors. S-CAR-expressing CD8(+) T cells, which carried the marker CD45.1, were injected into CD45.2(+) HBV transgenic mice. We compared these mice with mice that received CD8(+) T cells induced by vaccination, cells that express a CAR without a proper signaling domain, or cells that express a CAR that does not bind HBV proteins (controls). RESULTS: CD8(+) T cells that expressed HBV-specific CARs recognized different HBV subtypes and were able to engraft and expand in immune-competent HBV transgenic mice. After adoptive transfer, the S-CAR-expressing T cells localized to and functioned in the liver and rapidly and efficiently controlled HBV replication compared with controls, causing only transient liver damage. The large amount of circulating viral antigen did not impair or overactivate the S-CAR-grafted T cells. CONCLUSIONS: T cells with a CAR specific for HBV envelope proteins localize to the liver in mice to reduce HBV replication, causing only transient liver damage. This immune cell therapy might be developed for patients with chronic hepatitis B, regardless of their HLA type.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Hepatitis B virus/immunology , Liver/immunology , Receptors, Antigen, T-Cell/immunology , Viral Envelope Proteins/immunology , Virus Replication/immunology , Animals , Hepatitis B virus/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Recombinant Fusion Proteins/immunology
20.
Hum Gene Ther ; 24(3): 259-69, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23350854

ABSTRACT

Adoptive therapy of cancer with genetically redirected T cells showed spectacular efficacy in recent trials. A body of preclinical and clinical data indicate that young effector and central memory T cells perform superior in a primary antitumor response; repetitive antigen engagement, however, drives T-cell maturation to terminally differentiated cells associated with the loss of CCR7, which enables T cells to persist in peripheral tissues. In this work, we explored the antitumor efficacy of CCR7(-) T cells when redirected in an antigen-dependent fashion by a chimeric antigen receptor (CAR) toward tumors in the periphery. CAR-engineered CCR7(-) T cells more efficiently accumulated at the tumor site, secreted more IFN-γ, expressed higher amounts of cytotoxic molecules, and showed superior tumor cell lysis compared to the younger CCR7(+) cells. CCR7(-) T cells, however, were more prone to spontaneous and activation-induced cell death, which could be counteracted by simultaneous CD28 and OX40 (CD134) costimulation. Consequently, the combined CD28-ζ-OX40 signaling CAR rescued CCR7(-) T cells from apoptosis, which then produced more efficient antitumor efficacy than CCR7(+) T cells redirected by the same CAR. Data suggest that T-cell therapy will benefit from combined CD28-ζ-OX40 stimulation in the long-term by rescuing continuously generated CCR7(-) T cells for an antitumor attack.


Subject(s)
CD28 Antigens/immunology , Immunotherapy, Adoptive , Receptors, CCR7/deficiency , Receptors, OX40/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , CD28 Antigens/metabolism , Cell Death/immunology , Humans , Immunologic Memory , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Knockout , Neoplasms/immunology , Neoplasms/therapy , Phenotype , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, OX40/genetics , Receptors, OX40/metabolism , Signal Transduction , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...