Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neoplasia ; 31: 100812, 2022 09.
Article in English | MEDLINE | ID: mdl-35667149

ABSTRACT

Radiation-induced lymphopenia is a common occurrence in radiation oncology and an established negative prognostic factor, however the mechanisms underlying the relationship between lymphopenia and inferior survival remain elusive. The relevance of lymphocyte co-irradiation as critical normal tissue component at risk is an emerging topic of high clinical relevance, even more so in the context of potentially synergistic radiotherapy-immunotherapy combinations. The impact of the radiotherapy treatment volume on the lymphocytes of healthy and tumor-bearing mice was investigated in a novel mouse model of radiation-induced lymphopenia. Using an image-guided small-animal radiotherapy treatment platform, translationally relevant tumor-oriented volumes of irradiation with an anatomically defined increasing amount of normal tissue were irradiated, with a focus on the circulating blood and lymph nodes. In healthy mice, the influence of irradiation with increasing radiotherapy treatment volumes was quantified on the level of circulating blood cells and in the spleen. A significant decrease in the lymphocytes was observed in response to irradiation, including the minimally irradiated putative tumor area. The extent of lymphopenia correlated with the increasing volumes of irradiation. In tumor-bearing mice, differential radiotherapy treatment volumes did not influence the overall therapeutic response to radiotherapy alone. Intriguingly, an improved treatment efficacy in mice treated with draining-lymph node co-irradiation was observed in combination with an immune checkpoint inhibitor. Taken together, our study reveals compelling data on the importance of radiotherapy treatment volume in the context of lymphocytes as critical components of normal tissue co-irradiation and highlights emerging challenges at the interface of radiotherapy and immunotherapy.


Subject(s)
Lymphopenia , Neoplasms , Animals , Disease Models, Animal , Lymphocytes/pathology , Lymphopenia/etiology , Lymphopenia/pathology , Mice , Neoplasms/radiotherapy , Treatment Outcome
2.
Cell Rep ; 37(5): 109911, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731632

ABSTRACT

Suppressive regulatory T cell (Treg) differentiation is controlled by diverse immunometabolic signaling pathways and intracellular metabolites. Here we show that cell-permeable α-ketoglutarate (αKG) alters the DNA methylation profile of naive CD4 T cells activated under Treg polarizing conditions, markedly attenuating FoxP3+ Treg differentiation and increasing inflammatory cytokines. Adoptive transfer of these T cells into tumor-bearing mice results in enhanced tumor infiltration, decreased FoxP3 expression, and delayed tumor growth. Mechanistically, αKG leads to an energetic state that is reprogrammed toward a mitochondrial metabolism, with increased oxidative phosphorylation and expression of mitochondrial complex enzymes. Furthermore, carbons from ectopic αKG are directly utilized in the generation of fatty acids, associated with lipidome remodeling and increased triacylglyceride stores. Notably, inhibition of either mitochondrial complex II or DGAT2-mediated triacylglyceride synthesis restores Treg differentiation and decreases the αKG-induced inflammatory phenotype. Thus, we identify a crosstalk between αKG, mitochondrial metabolism and triacylglyceride synthesis that controls Treg fate.


Subject(s)
Cell Differentiation/drug effects , Energy Metabolism/drug effects , Ketoglutaric Acids/pharmacology , Lipid Metabolism/drug effects , Mitochondria/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Fibrosarcoma/metabolism , Fibrosarcoma/therapy , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeostasis , Humans , Immunotherapy, Adoptive , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism
3.
FEBS Lett ; 591(19): 3104-3118, 2017 10.
Article in English | MEDLINE | ID: mdl-28901530

ABSTRACT

T cells are stimulated by the engagement of antigen, cytokine, pathogen, and hormone receptors. While research performed over many years has focused on deciphering the molecular components of these pathways, recent data underscore the importance of the metabolic environment in conditioning responses to receptor engagement. The ability of T cells to undergo a massive proliferation and cytokine secretion in response to receptor signals requires alterations to their bioenergetic homeostasis, allowing them to meet new energetic and biosynthetic demands. The metabolic reprogramming of activated T cells is regulated not only by changes in intracellular nutrient uptake and utilization but also by nutrient and oxygen concentrations in the extracellular environment. Notably, the extracellular environment can be profoundly altered by pathological conditions such as infections and tumors, thereby perturbing the metabolism and function of antigen-specific T lymphocytes. This review highlights the interplay between diverse metabolic networks and the transcriptional/epigenetic states that condition T-cell differentiation, comparing the metabolic features of T lymphocytes with other immune cells. We further address recent discoveries in the metabolic pathways that govern T-cell function in physiological and pathological conditions.


Subject(s)
Cell Differentiation , Cell Lineage , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Epigenesis, Genetic , Humans , Metabolome/genetics , Signal Transduction
4.
Immunol Cell Biol ; 95(4): 356-363, 2017 04.
Article in English | MEDLINE | ID: mdl-28003642

ABSTRACT

The potential for immunotherapy as a treatment option for cancer is clear from remarkable responses of some leukemia patients to adoptive cell transfer using autologous T cells genetically modified to express chimeric antigen receptors (CARs). However, the vast majority of cancers, in particular the more common solid cancers, such as those of the breast, colon and lung, fail to respond significantly to infusions of CAR T cells. Solid cancers present some formidable barriers to adoptive cell transfer, including suppression of T-cell function and inhibition of T-cell localization. In this review, we discuss the current state of CAR T-cell therapy in solid cancers, the variety of concepts being investigated to overcome these barriers as well as approaches aimed at increasing the specificity and safety of adoptive cell transfer.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/immunology , Colonic Neoplasms/therapy , Immunotherapy, Adoptive/methods , Lung Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Cell Movement , Colonic Neoplasms/immunology , Genetic Engineering , Humans , Immune Tolerance , Lung Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/transplantation , Tumor Escape
5.
Oncotarget ; 7(23): 34582-98, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27153556

ABSTRACT

While adoptive immunotherapy using chimeric antigen receptor (CAR)-modified T cells can induce remission of some tumors, the role of other CAR-modified leukocytes is not well characterized. In this study, we characterize the function of leukocytes including natural killer (NK) cells, macrophages and CAR T cells from transgenic mice expressing a CAR under the control of the pan-hematopoietic promoter, vav, and determine the ability of these mice to respond to ERB expressing tumors. We demonstrate the anti-tumor functions of leukocytes, including antigen specific cytotoxicity and cytokine secretion. The adoptive transfer of CAR T cells provided a greater survival advantage in the E0771ERB tumor model than their wildtype (WT) counterparts. In addition, CAR NK cells and CAR T cells also mediated increased survival in the RMAERB tumor model. When challenged with Her2 expressing tumors, F38 mice were shown to mount an effective immune response, resulting in tumor rejection and long-term survival. This was shown to be predominantly dependent on both CD8+ T cells and NK cells. However, macrophages and CD4+ T cells were also shown to contribute to this response. Overall, this study highlights the use of the vav-CAR mouse model as a unique tool to determine the anti-tumor function of various immune subsets, either alone or when acting alongside CAR T cells in adoptive immunotherapy.


Subject(s)
Adoptive Transfer/methods , Cytotoxicity, Immunologic/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Macrophages/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Cytokines/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/mortality , Proto-Oncogene Proteins c-vav/genetics , Receptor, ErbB-2/metabolism
6.
PLoS One ; 10(10): e0140543, 2015.
Article in English | MEDLINE | ID: mdl-26505904

ABSTRACT

Genetically modified CD8+ T lymphocytes have shown significant anti-tumor effects in the adoptive immunotherapy of cancer, with recent studies highlighting a potential role for a combination of other immune subsets to enhance these results. However, limitations in present genetic modification techniques impose difficulties in our ability to fully explore the potential of various T cell subsets and assess the potential of other leukocytes armed with chimeric antigen receptors (CARs). To address this issue, we generated a transgenic mouse model using a pan-hematopoietic promoter (vav) to drive the expression of a CAR specific for a tumor antigen. Here we present a characterization of the immune cell compartment in two unique vav-CAR transgenic mice models, Founder 9 (F9) and Founder 38 (F38). We demonstrate the vav promoter is indeed capable of driving the expression of a CAR in cells from both myeloid and lymphoid lineage, however the highest level of expression was observed in T lymphocytes from F38 mice. Lymphoid organs in vav-CAR mice were smaller and had reduced cell numbers compared to the wild type (WT) controls. Furthermore, the immune composition of F9 mice differed greatly with a significant reduction in lymphocytes found in the thymus, lymph node and spleen of these mice. To gain insight into the altered immune phenotype of F9 mice, we determined the chromosomal integration site of the transgene in both mouse strains using whole genome sequencing (WGS). We demonstrated that compared to the 7 copies found in F38 mice, F9 mice harbored almost 270 copies. These novel vav-CAR models provide a ready source of CAR expressing myeloid and lymphoid cells and will aid in facilitating future experiments to delineate the role for other leukocytes for adoptive immunotherapy against cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , Recoverin/genetics , Animals , Cell Lineage/immunology , Humans , Mice , Mice, Transgenic , Neoplasms/immunology , Recoverin/biosynthesis , Recoverin/immunology , Signal Transduction , Thymocytes/immunology , Thymus Gland/immunology
7.
PLoS One ; 10(9): e0136817, 2015.
Article in English | MEDLINE | ID: mdl-26334628

ABSTRACT

The development of antigen-targeted therapeutics is dependent on the preferential expression of tumor-associated antigens (TAA) at targetable levels on the tumor. Tumor-associated antigens can be generated de novo or can arise from altered expression of normal basal proteins, such as the up-regulation of human epidermal growth factor receptor 2 (Her2/ErbB2). To properly assess the development of Her2 therapeutics in an immune tolerant model, we previously generated a transgenic mouse model in which expression of the human Her2 protein was present in both the brain and mammary tissue. This mouse model has facilitated the development of Her2 targeted therapies in a clinically relevant and suitable model. While heterozygous Her2+/- mice appear to develop in a similar manner to wild type mice (Her2-/-), it has proven difficult to generate homozygous Her2+/+ mice, potentially due to embryonic lethality. In this study, we performed whole genome sequencing to determine if the integration site of the Her2 transgene was responsible for this lethality. Indeed, we report that the Her2 transgene had integrated into the Pds5b (precocious dissociation of sisters) gene on chromosome 5, as a 162 copy concatemer. Furthermore, our findings demonstrate that Her2+/+ mice, similar to Pds5b-/- mice, are embryonic lethal and confirm the necessity for Pds5b in embryonic development. This study confirms the value of whole genome sequencing in determining the integration site of transgenes to gain insight into associated phenotypes.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Lethal , Genes, erbB-2 , Homozygote , Transcription Factors/genetics , Animals , Cell Line, Tumor , Chromosome Mapping , Exons , Fetal Death , Humans , Mice , Mice, Transgenic , Phenotype
8.
Cancer Immunol Res ; 3(5): 483-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25711536

ABSTRACT

Adoptive therapy with chimeric antigen receptor (CAR) T cells shows great promise clinically. However, there are important aspects of CAR-T-cell biology that have not been explored, particularly with respect to the kinetics of activation, immune synapse formation, and tumor cell killing. Moreover, the effects of signaling via the endogenous T-cell receptor (TCR) or CAR on killing kinetics are unclear. To address these issues, we developed a novel transgenic mouse (designated CAR.OT-I), in which CD8(+) T cells coexpressed the clonogenic OT-I TCR, recognizing the H-2K(b)-presented ovalbumin peptide SIINFEKL, and an scFv specific for human HER2. Primed CAR.OT-I T cells were mixed with SIINFEKL-pulsed or HER2-expressing tumor cells and visualized in real-time using time-lapse microscopy. We found that engagement via CAR or TCR did not affect cell death kinetics, except that the time from degranulation to CAR-T-cell detachment was faster when CAR was engaged. We showed, for the first time, that individual CAR.OT-I cells can kill multiple tumor cells ("serial killing"), irrespective of the mode of recognition. At low effector:target ratios, the tumor cell killing rate was similar via TCR or CAR ligation over the first 20 hours of coincubation. However, from 20 to 50 hours, tumor cell death mediated through CAR became attenuated due to CAR downregulation throughout the time course. Our study provides important insights into CAR-T-tumor cell interactions, with implications for single- or dual receptor-focused T-cell therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Receptors, Antigen/immunology , Animals , Cell Line, Tumor , Lysosomal-Associated Membrane Protein 1/immunology , Mice, Transgenic , Perforin/immunology , Receptor, ErbB-2/genetics , Receptors, Antigen, T-Cell/immunology
9.
Mol Immunol ; 67(2 Pt A): 46-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25595028

ABSTRACT

The immune system plays a critical role in the elimination and suppression of pathogens. Although the endogenous immune system is capable of immune surveillance resulting in the elimination of cancer cells, tumor cells have developed a variety of mechanisms to escape immune recognition often resulting in tumor outgrowth. The presence of immune infiltrate in tumors has been correlated with a good prognosis following treatment (Sato et al., 2005; Loi et al., 2013; Clemente et al., 1996; Galon et al., 2006). As such, immune cells such as T cells, have been harnessed in order to target cancer. Tumor reactive lymphocytes, called tumor-infiltrating lymphocytes (TILs) have been isolated and expanded from the tumor and reinfused back into patients for the treatment of melanoma. The promise of adoptive immunotherapy utilizing TILs as a robust treatment for cancer has been highlighted in patients with advanced melanoma with greater than 50% of patients responding to treatment (Dudley et al., 2005). Although TIL therapy has shown promising results in melanoma patients, it has proved difficult to translate this approach to other cancers, given that the numbers of TILs that can be isolated are generally low. To broaden this therapy for other cancers, T cells have been genetically modified to endow them with tumor reactivity using either a T cell receptor (TCR) (Parkhurst et al., 2009, 2011; Chinnasamy et al., 2011) or a chimeric antigen receptor (CAR) (Grupp et al., 2013; Park et al., 2007). This review will outline the origins and development of adoptive immunotherapy utilizing TILs leading to genetic modification strategies to redirect T cells to cancer. Potential hurdles and novel strategies will be discussed for realizing the full potential of adoptive immunotherapy becoming a standard of care treatment for cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/transplantation , Neoplasms/therapy , Transgenes/immunology , Humans , Immunotherapy, Adoptive/trends , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Models, Immunological , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Transgenes/genetics , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends , Treatment Outcome
10.
PLoS One ; 9(9): e108670, 2014.
Article in English | MEDLINE | ID: mdl-25264896

ABSTRACT

The transcription factor Foxp3 represents the most specific functional marker of CD4+ regulatory T cells (TRegs). However, previous reports have described Foxp3 expression in other cell types including some subsets of macrophages, although there are conflicting reports and Foxp3 expression in cells other than Treg is not well characterized. We performed detailed investigations into Foxp3 expression in macrophages in the normal tissue and tumor settings. We detected Foxp3 protein in macrophages infiltrating mouse renal cancer tumors injected subcutaneously or in the kidney. Expression was demonstrated using flow cytometry and Western blot with two individual monoclonal antibodies. Further analyses confirmed Foxp3 expression in macrophages by RT PCR, and studies using ribonucleic acid-sequencing (RNAseq) demonstrated a previously unknown Foxp3 messenger (m)RNA transcript in tumor-associated macrophages. In addition, depletion of Foxp3+ cells using diphtheria toxin in Foxp3DTR mice reduced the frequency of type-2 macrophages (M2) in kidney tumors. Collectively, these results indicate that tumor-associated macrophages could express Foxp3.


Subject(s)
Forkhead Transcription Factors/biosynthesis , Kidney Neoplasms/immunology , Macrophages/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Forkhead Transcription Factors/genetics , Mice , Mice, Inbred BALB C , Mice, SCID , RNA, Messenger/biosynthesis
11.
Curr Opin Immunol ; 27: 46-52, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24534448

ABSTRACT

The immune system can be induced to respond against cancer with some success reported in clinical trials using a range of approaches including vaccines and antibodies. In addition to these approaches, cell based therapies are demonstrating much promise as potential therapies for cancer. In cell therapies autologous patient leukocytes are isolated and manipulated in vitro before transfer back to the patient in adoptive transfer regimens. The majority of approaches utilize conventional T cells or dendritic cells, but a wide variety of other types of leukocytes exist which can possess anti-cancer activity. In this review, we present a brief overview of T cell adoptive cell transfer followed by a review of approaches using alternate lymphocyte subsets and other leukocytes including neutrophils, macrophages and eosinophils.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Neoplasms/immunology
12.
Mol Ther ; 22(1): 18-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24048441

ABSTRACT

The tumor microenvironment can promote tumor growth and reduce treatment efficacy. Tumors can occur in many sites in the body, but how surrounding normal tissues at different anatomical sites affect tumor microenvironments and their subsequent response to therapy is not known.We demonstrated that tumors from renal, colon, or prostate cell lines in orthotopic locations responded to immunotherapy consisting of three agonist antibodies, termed Tri-mAb, to a much lesser extent than the same tumor type located subcutaneously. A tissue-specific response to Tri-mAb was confirmed by ex vivo separation of subcutaneous (SC) or orthotopic tumor cells from stromal cells, followed by reinjection of tumor cells into the opposite site. Compared with SC tumors, orthotopic tumors had a microenvironment associated with a type 2 immune response, related to immunosuppression, and an involvement of alternatively activated macrophages in the kidney model. Orthotopic kidney tumors were more highly vascularized than SC tumors. Neutralizing the macrophage- and Th2-associated molecules chemokine (C-C motif) ligand 2 or interleukin-13 led to a significantly improved therapeutic effect. This study highlights the importance of the tissue of implantation in sculpting the tumor microenvironment. These are important fundamental issues in tumor biology and crucial factors to consider in the design of experimental models and treatment strategies.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/pathology , Tumor Microenvironment/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/immunology , Cell Line, Tumor , Chemokine CCL2/immunology , Colonic Neoplasms/immunology , Disease Models, Animal , Gene Expression , Interleukin-13/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Neoplasms/mortality , Neoplasms/therapy , Neovascularization, Pathologic/immunology , Organ Specificity/immunology , Prostate/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
13.
Clin Cancer Res ; 19(20): 5636-46, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23873688

ABSTRACT

PURPOSE: To determine the antitumor efficacy and toxicity of a novel combination approach involving adoptive T-cell immunotherapy using chimeric antigen receptor (CAR) T cells with an immunomodulatory reagent for blocking immunosuppression. EXPERIMENTAL DESIGN: We examined whether administration of a PD-1 blocking antibody could increase the therapeutic activity of CAR T cells against two different Her-2(+) tumors. The use of a self-antigen mouse model enabled investigation into the efficacy, mechanism, and toxicity of this combination approach. RESULTS: In this study, we first showed a significant increase in the level of PD-1 expressed on transduced anti-Her-2 CD8(+) T cells following antigen-specific stimulation with PD-L1(+) tumor cells and that markers of activation and proliferation were increased in anti-Her-2 T cells in the presence of anti-PD-1 antibody. In adoptive transfer studies in Her-2 transgenic recipient mice, we showed a significant improvement in growth inhibition of two different Her-2(+) tumors treated with anti-Her-2 T cells in combination with anti-PD-1 antibody. The therapeutic effects observed correlated with increased function of anti-Her-2 T cells following PD-1 blockade. Strikingly, a significant decrease in the percentage of Gr1(+) CD11b(+) myeloid-derived suppressor cells (MDSC) was observed in the tumor microenvironment of mice treated with the combination therapy. Importantly, increased antitumor effects were not associated with any autoimmune pathology in normal tissue expressing Her-2 antigen. CONCLUSION: This study shows that specifically blocking PD-1 immunosuppression can potently enhance CAR T-cell therapy that has significant implications for potentially improving therapeutic outcomes of this approach in patients with cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/genetics , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , Antigens/immunology , Antineoplastic Agents/administration & dosage , Autoimmunity , Cell Line, Tumor , Disease Models, Animal , Humans , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Myeloid Cells/immunology , Neoplasms/pathology , Neoplasms/therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology
14.
Immunotherapy ; 5(6): 577-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23725282

ABSTRACT

AIM: The aim of the current study was to fully elucidate the functions of T cells genetically modified with an erbB2-specific chimeric antigen receptor (CAR). MATERIAL & METHODS: In this study, key functional parameters of CAR T cells were examined following antigen-specific stimulation of the chimeric anti-erbB2 receptor. RESULTS: Gene-modified T cells produced the cytokines IFN-γ, IL-2, IL-4, IL-10, TNF-α and IL-17, and the chemokine RANTES upon CAR ligation. A multifunctional capacity of these CAR T cells was also demonstrated, where 13.7% of cells were found to simultaneously express IFN-γ and CD107a, indicative of cytolytic granule release. In addition, the CAR T cells were able to respond to a greater degree on the second ligation of CAR, which has not been previously shown. IFN-γ secretion levels were significantly higher on second ligation than those secreted following initial ligation. CAR-expressing T cells were also demonstrated to lyze erbB2-expressing tumor cells in the absence of activity against bystander cells not expressing the erbB2 antigen, thereby demonstrating exquisite specificity. CONCLUSION: This study demonstrates the specificity of CAR gene-engineered T cells and their capacity to deliver a wide range of functions against tumor cells with an enhanced response capability after initial receptor engagement.


Subject(s)
CD28 Antigens/metabolism , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptor, ErbB-2/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/transplantation , Animals , CD28 Antigens/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , HCT116 Cells , Humans , Immunologic Memory , Lymphocyte Activation/genetics , Mice , Protein Engineering , Receptor, ErbB-2/genetics , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology
15.
PLoS One ; 8(5): e63037, 2013.
Article in English | MEDLINE | ID: mdl-23667569

ABSTRACT

Genetic engineering of cellular function holds much promise for the treatment of a variety of diseases including gene deficiencies and cancer. However, engineering the full complement of cellular functions can be a daunting genetic exercise since many molecular triggers need to be activated to achieve complete function. In the case of T cells, genes encoding chimeric antigen receptors (CARs) covalently linking antibodies to cytoplasmic signaling domains can trigger some, but not all, cellular functions against cancer cells. To date, relatively few CAR formats have been investigated using a candidate molecule approach, in which rationally chosen molecules were trialed one by one. Therefore, to expedite this arduous process we developed an innovative screening method to screen many thousands of CAR formats to identify genes able to enhance the anticancer ability of T cells. We used a directional in-frame library of randomly assembled signaling domains in a CAR specific for the tumor associated antigen erbB2. Several new and original CARs were identified, one of which had an enhanced ability to lyse cancer cells and inhibit tumor growth in mice. This study highlights novel technology that could be used to screen a variety of molecules for their capacity to induce diverse functions in cells.


Subject(s)
Cell Engineering/methods , Gene Library , Receptors, Antigen/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/cytology , Animals , Cell Line, Tumor , Genes, erbB-2/genetics , Genetic Vectors/genetics , Humans , Mice , Receptors, Immunologic/genetics , Reproducibility of Results , Retroviridae/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...