Anti-Tumor Cytotoxicity of Blood γδ T Cells Expanded from Leukemia Patients Against Autologous Leukemia Cells—Enhancement of the Anti-Tumor Cytotoxicity by Type I IFN.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2385-2385
Author(s):  
Anri Saito ◽  
Miwako Narita ◽  
Norihiro Watanabe ◽  
Ayumi Yokoyama ◽  
Asuka Sekiguchi ◽  
...  

Abstract In order to establish an efficient anti-tumor cellular immunotherapy using blood γδ T cells, we investigated the cytotoxic activity of γδ T cells expanded from patients with leukemia against autologous leukemia cells and explored the potent methods for enhancing the anti-tumor cytotoxic activity of γδ T cells. We clarified that γδ T cells generated from leukemia patients possess the cytotoxic activity against autologous leukemia cells. Besides, anti-tumor cytotoxic activity of expanded γδ T cells was enhanced by the short-term culture of γδ T cells with type I IFN (IFN-α and IFN-β). The sensitivity of target leukemia cells to γδ T cells was enhanced by the exposure of the target cells to bisphosphonate such as zoledronate, which is one of the antigens recognized by γδ T cells and elevates the content of potent antigen for γδ T cells, isoprenyl pyrophosphate (IPP), in tumor cells. Blood γδ T cells were expanded from anti-CD3 microbead-separated T cells or anti-γδ TCR microbead-separated γδ T cells in the patients with acute myelogenous leukemia by the culture with zoledronate and a low concentration of IL-2 for 1–2 weeks. For the activation of expanded γδ T cells, cultured γδ T cells were exposed with type I IFN for 1–3 days. The supernatant prepared from the culture of type I IFN-activated γδ T cells was assayed for cytokine (IFN-γ, TNF-α, IL-4, IL-5, IL-10) concentration by cytometric bead array. Anti-tumor cytotoxicity of γδ T cells was evaluated by 51Cr-release assay by using purified γδ T cells as effector cells and autologous leukemia cells as target cells. In most patients with acute leukemia, γδ T cells could be markedly expanded by the culture with zoledronate and IL-2 and almost all the expanded γδ T cells possessed Vδ2 TCR. Expanded and purified γδ T cells derived from the patients with leukemia were demonstrated to be cytotoxic against autologous leukemia cells. By the culture of expanded γδ T cells with type I IFN, the expression of the activation marker CD69 and the apoptosis molecule Trail was enhanced at the concentration dependent of type I IFN especially IFN-β. The expanded γδ T cells were shown to produce a remarkable amount of IFN-γ and a considerable amount of TNF-α and the cytokine production was increased by the addition of type I IFN. In addition, the cytotoxic activity of γδ T cells was enhanced by incubating target leukemia cells with zoledronate for 1–2 days. The present study demonstrated that γδ T cells expanded from patient’s blood are cytotoxic to patient’s leukemia cells. It is also demonstrated that there are two methods practically available for enhancing the cytotoxic activity of expanded γδ T cells against leukemia cells, one of which is activating γδ T cells by using type I IFN, and the other is elevating the sensitivity of target cells by using bisphosphonate. These findings implied the possibility that type I IFN-activated γδ T cells could be efficiently applied for cellular immunotherapy in the patients with hematological malignancies who is being administered with bisphosphonate. Moreover, in vivo administration of bisphosphpnate, a low dose of IL-2 and type I IFN could be effective for tumors as γδ T cell-based cellular immunotherapy.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3895-3895
Author(s):  
Anri Saito ◽  
Miwako Narita ◽  
Norihiro Watanabe ◽  
Nozomi Tochiki ◽  
Yumi Hiroi ◽  
...  

Abstract In order to establish an efficient anti-tumor cellular immunotherapy using blood In order to establish an efficient anti-tumor cellular immunotherapy using blood γδ T cells, we investigated the in vitro expansion of γδ T cells in the patients with myeloma and lymphoma by the culture of PB-MNC with bisphosphonate and a low dose of IL-2 and we demonstrated the cytotoxic activity of the expanded γδ T cells against myeloma/lymphoma cells. Simultaneously we explored the potent methods for enhancing the anti-tumor cytotoxic activity of γδ T cells by both directions of activating the expanded γδ T cells and making target tumor cells sensitive to γδ T cells. For the activation of γδ T cells, expanded γδ T cells were exposed with type I IFN, monocyte-derived dendritic cells (mo-DC), or plasmacytoid dendritic cell like cell line PMDC05 (leukemia cell line established from CD4+ CD56+ acute leukemia in our laboratory) for 2 days. For the enhancement of sensitivity of target tumor cell to γδ T cells, we aimed to increase the content of IPP (the potent pyrophosphate antigen for γδ T cells) in tumor cells by decreasing the metabolic downstream of IPP. For decreasing the downstream of IPP, we tried to suppress FPP synthetase, which is involved in downstream metabolism of IPP, by using nitrogen-containing bisphosphonate. In addition, the expression of stress-induced molecules such as MICA/B on target tumor cells was evaluated in association with the level of cytotoxicity of γδ T cells against the tumor cells. Compared with normal control, the patients with myeloma (n=8) demonstrated decreased percentage and counts of PB γδ T cells. Patients with lymphoma (n=7) showed a wide range of values in PB γδ T cells, covering a normal range. Amplification rate of PB γδ T cells by culture with zoledronate and IL-2 varied markedly from patient to patient up to 120 times in myeloma and 90 times in lymphoma. Expanded γδ T cells generated in patients with myeloma/lymphoma were demonstrated to possess the cytotoxic activity against myeloma/lymphoma cells by 51Cr-release assay and CFSE-labeled target cell. The cytotoxic activity of expanded γδ T cells was enhanced by the exposure of γδ T cells with type I IFN (IFN-α and IFN-β). The activation of γδ T cells, which was evaluated by the elevation of CD69 expression, was observed by the exposure of γδ T cells with type I IFN, mo-DC, or PMDC05 for 2 days. The sensitivity of target myeloma/lymphoma cells to γδ T cells was enhanced by the exposure of the target cells to bisphosphonate such as zoledronate. The expression level of MICA/B on target tumor cells was demonstrated to be associated with the potency of cytotoxicity of γδ T cells against the tumor cells. The present study demonstrated that γδ T cells expanded from myeloma/lymphoma patient’s blood are cytotoxic to myeloma/lymphoma cells. There are two methods practically available for enhancing the cytotoxic activity of expanded γδ T cells against myeloma/lymphoma cells, one of which is activating γδ T cells and the other is elevating the sensitivity of target cells by using bisphosphonate.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4762-4762
Author(s):  
Anri Saito ◽  
Miwako Narita ◽  
Norihiro Watanabe ◽  
Nozomi Tochiki ◽  
Noriyuki Satoh ◽  
...  

Abstract In order to establish an efficient gd T cell-mediated immunotherapy for hematological malignancies, we tried to clarify whether γδ T cells could be expanded from blood cells of patients with myeloma, lymphoma and acute leukemia by culture with zoledronate and a low dose of IL-2 and whether the expanded patients’ γδ T cells could kill tumor cells including self tumor cells with sparing normal clone cells. In addition, we explored the methods to enhance the anti-tumor cytotoxicity of the expanded γδ T cells by activating them with type I IFN, monocyte-derived dendritic cells (mo-DCs), or ab T cells. Although γδ T cells could be expanded in patients with myeloma, lymphoma and leukemia as well as normal persons, the amplification rates of gd T cells before and after the culture were varied from patient to patient in the patients with hematological malignancies. γδ T cells generated in patients with myeloma and lymphoma showed a potent cytotoxic ability against myeloma/lymphoma cell lines (RPMI8226, Daudi) as shown in γδ T cells generated in normal persons. In addition, γδ T cells generated in a patient with myeloma and acute leukemia showed a cytotoxic ability against self myeloma or leukemia cells freshly prepared from bone marrow. However, the same γδ T cells were not cytotoxic to normal lymphocytes of the patients. Then the expanded γδ T cells were stimulated with type I IFN, mo-DCs, or αβ T cells and the activation (CD69 expression) and cytotoxicity against tumor cells were examined. By the stimulation with type I IFN, the expression of CD69 and Trail of γδ T cells was increased and the cytotoxic ability of γδ T cells was enhanced at dose-dependent manner of type I IFN. CD69 expression on γδ T cells was enhanced by co-culture with both immature and mature mo-DCs in a cell-number-dependent fashion. CD69 expression was enhanced after the addition of mo-DCs of either autologous or allogeneic origin. Activation of γδ T cells with mo-DCs enhanced anti-tumor cytotoxicity of γδ T cells against RPMI8226 and CML blastic crisis cell line (C2F8) in an effector-to-target ratio-dependent manner. Although CD69 expression of γδ T cells was enhanced by the co-culture with allogeneic ab T cells, autologous ab T cells couldn’t activate γδ T cells. However, autologous ab T cells stimulated with IL-2 or PHA could induce the activation of γδ T cells. The activation of γδ T cells with stimulated αβ T cells required cell-to-cell interaction. These findings suggested that αβ T cells stimulated by allogeneic γδ T cells could activate the same allogeneic γδ T cells. The present data demonstrated that γδ T cells, which could be expanded in vitro from blood cells of the patients with myeloma, lymphoma and leukemia by culture with zoledronate and IL-2, possess an enough cytotoxic ability against tumor cells including self tumor cells with sparing normal cells. These findings suggested that in vitro generated patients’ γδ T cells could be applied to γδ T cell-mediated immunotherapy for hematological malignancies. Besides, potent γδ T cells activated by type I IFN, mo-DCs or activated αβ T cells were considered to be applicable for γδ T cell-mediated immunotherapy.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2383-2383
Author(s):  
Norihiro Watanabe ◽  
Miwako Narita ◽  
Ayumi Yokoyama ◽  
Asuka Sekiguchi ◽  
Anri Saito ◽  
...  

Abstract Immune competent cells which possess both abilities to present tumor antigens to effector αβ T cells and to act by themselves as effector cells of anti-tumor immune reaction, if they would exist, are thought to be useful cells for establishing an efficient anti-tumor cellular immunotherapy. We identified human Vγ2Vδ2 T cells to be the multi-functional cells with the abilities of antigen presentation for antigen-specific immune reaction and anti-tumor cytotoxicity using non-peptides as antigens. Human γδ T cells were expanded from human peripheral blood mononuclear cells by the culture with zoledronate or isopentenyl pyrophosphate (IPP) and a low concentration of IL-2 for 1–2 weeks. Immature dendritic cells were generated from blood non-adherent cells or anti-CD14 microbead-separated monocytes by the culture with GM-CSF and IL-4 for 6–8 days and mature dendritic cells were induced from immature dendritic cells by the additional 2 days-culture with TNF-α, IL-1α, IL-6 and PGE2. Cultured γδ T cell-containing cells were analyzed for the expression of surface phenotypes and cytoplasmic molecules by flow cytometry. The supernatant of the culture of anti-γδ TCR microbead-separated γδ T cells were assayed for the concentration of IFN-γ, TNF-α, IL-4, IL-5, and IL-2 by cytometric bead array. The uptake ability of γδ T cells were investigated by flow cytometry using FITC-dextran and Lucifer yellow. Antigen presenting ability of γδ T cells was evaluated by mixed lymphocyte culture by using purified γδ T cells as stimulator cells and blood non-adherent cells or naïve αβ T cells as responder cells in both autologous and allogeneic settings. In autologous settings, KLH or PPD were added as exogenous antigens to γδ T cells or dendritic cells for the whole duration of the culture. Anti-tumor cytotoxicity of γδ T cells was evaluated by 51Cr-release assay by using purified γδ T cells as effector cells and leukemia/myeloma cell lines or fresh acute leukemia leukemia cells as target cells. Seventy to eighty percent of the cultured cells generated from blood mononuclear cells were positive for γδ TCR and almost all of the γδ TCR+ cells possessed Vδ2 TCR. Ten to twenty percient of γδ T cells were positive for CD40/CD80 and most of γδ T cells were positive for CD86/CD54/CD58/HLA-DR. The expanded γδT cells were shown to produce a remarkable amount of IFN-γ and a considerable amount of TNF-α and the cytokine production was much increased by the addition of a low dose of IL-2. Cultured γδ T cells were shown to be able to uptake FITC-Dextran and lucifer yellow at a comparable level of immature dendritic cells. Purified and cultured γδ T cells were demonstrated to possess an allogeneic and antigen-specific autologous antigen presenting ability at a similar level of mature dendritic cells. Zoledronate/IL-2-expanded γδ T cells were demonstrated to be cytotoxic against leukemia or myeloma cell lines and fresh acute leukemia leukemia cells at effector to target ratio dependent. The present study revealed that human γδ T cells, which could be markedly expanded by the culture with zoledronate or IPP and IL-2, possess both functions of antigen presentation and anti-tumor cytotoxicity and might be efficiently applied to anti-tumor cellular immunotherapy.


Cytotherapy ◽  
2006 ◽  
Vol 8 (2) ◽  
pp. 118-129 ◽  
Author(s):  
N. Watanabe ◽  
M. Narita ◽  
A. Yokoyama ◽  
A. Sekiguchi ◽  
A. Saito ◽  
...  

2010 ◽  
Vol 184 (7) ◽  
pp. 3755-3767 ◽  
Author(s):  
Thomas Henry ◽  
Girish S. Kirimanjeswara ◽  
Thomas Ruby ◽  
Jonathan W. Jones ◽  
Kaitian Peng ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1347-1347
Author(s):  
Kiyoshi Sato ◽  
Shinya Kimura ◽  
Takeshi Yuasa ◽  
Hiromi Wada ◽  
Taira Maekawa

Abstract Bisphosphonates (BPs), widely used to treat bone diseases, have recently been attracted much interest for their antitumor activity and have been reported to exert direct antitumor effects on several cancer cell lines via the inactivation of Ras proteins. BPs inhibit farnesyl pyrophosphate (FPP) synthetase in the mevalonate pathway and deplete cellular pools of PPs such as farnesyl PP and geranylgeranyl PP which are indispensable for the activation of Ras proteins. In addition to their direct antitumor activity, BPs expand γδ T-cells which are potent effector cells and also induce the accumulation of isopentenyl PP as a tumor antigen in target cancer cells. The purpose of this study was to clarify the cytotoxic activity of γδ T-cells expanded ex vivo by the potent third generation BP zoledronate (ZOL). Peripheral blood mononuclear cells of five healthy donors were incubated with different concentrations of ZOL and interleukin-2. After 14 days incubation, 1 μM ZOL increased the absolute number of γδ T-cells 500–800 fold. Expanded γδ T-cells were of the Vγ9Vδ2 subset and cytokine levels of IL-2, -4, -5, -10, TNF-α and IFN-γ were not elevated at resting i.e. before contact to target cancer cells. In vitro cytotoxic activities of γδ T-cells against the luciferase-labeled small cell lung cancer (SCLC) cell line SBC-5 were examined by a newly developed cytotoxic assay using an in vivo imaging system (Xenogen, Alameda, CA) and video microscopy, Leica AS MDW (Leica Microsystems Inc., Bannockburn, IL). γδ T-cells killed SBC-5 cells pre-treated with 5 μM ZOL for 12 h after 1.5–3.0 h contact with the target cells whereas untreated SBC-5 were rarely killed. SBC-5 cells pretreated with 5 μM ZOL showed a marked increase in their sensitivity to lysis by γδ T-cells, percentages of specific lysis were 42% and 55% at effector/target (E/T) ratios of 5:1 and 10:1, respectively, while those of untreated SBC-5 cells were 8% and 13% at E/T ratios of 5:1 and 10:1, respectively. In vivo efficacy of γδ T-cells was investigated in mice xenografted subcutaneously with SBC-5 cells. Pretreatment with 80 μg/kg ZOL enhanced significantly antitumor activity of γδ T-cells also in vivo. These findings showed that ZOL stimulated the proliferation of γδ T-cells significantly and that the cytotoxic activity of γδ T-cells required pre-treatment of target cells with ZOL, indicating the potential use of autologous ex vivo expanded γδ T-cells for cancer immunotherapy.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Norzawani Buang ◽  
Lunnathaya Tapeng ◽  
Victor Gray ◽  
Alessandro Sardini ◽  
Chad Whilding ◽  
...  

AbstractThe majority of patients with systemic lupus erythematosus (SLE) have high expression of type I IFN-stimulated genes. Mitochondrial abnormalities have also been reported, but the contribution of type I IFN exposure to these changes is unknown. Here, we show downregulation of mitochondria-derived genes and mitochondria-associated metabolic pathways in IFN-High patients from transcriptomic analysis of CD4+ and CD8+ T cells. CD8+ T cells from these patients have enlarged mitochondria and lower spare respiratory capacity associated with increased cell death upon rechallenge with TCR stimulation. These mitochondrial abnormalities can be phenocopied by exposing CD8+ T cells from healthy volunteers to type I IFN and TCR stimulation. Mechanistically these ‘SLE-like’ conditions increase CD8+ T cell NAD+ consumption resulting in impaired mitochondrial respiration and reduced cell viability, both of which can be rectified by NAD+ supplementation. Our data suggest that type I IFN exposure contributes to SLE pathogenesis by promoting CD8+ T cell death via metabolic rewiring.


Blood ◽  
2009 ◽  
Vol 114 (20) ◽  
pp. 4422-4431 ◽  
Author(s):  
Georg Gruenbacher ◽  
Hubert Gander ◽  
Andrea Rahm ◽  
Walter Nussbaumer ◽  
Nikolaus Romani ◽  
...  

Abstract CD56+ human dendritic cells (DCs) have recently been shown to differentiate from monocytes in response to GM-CSF and type 1 interferon in vitro. We show here that CD56+ cells freshly isolated from human peripheral blood contain a substantial subset of CD14+CD86+HLA-DR+ cells, which have the appearance of intermediate-sized lymphocytes but spontaneously differentiate into enlarged DC-like cells with substantially increased HLA-DR and CD86 expression or into fully mature CD83+ DCs in response to appropriate cytokines. Stimulation of CD56+ cells containing both DCs and abundant γδ T cells with zoledronate and interleukin-2 (IL-2) resulted in the rapid expansion of γδ T cells as well as in IFN-γ, TNF-α, and IL-1β but not in IL-4, IL-10, or IL-17 production. IFN-γ, TNF-α, and IL-1β production were almost completely abolished by depleting CD14+ cells from the CD56+ subset before stimulation. Likewise, depletion of CD14+ cells dramatically impaired γδ T-cell expansion. IFN-γ production could also be blocked by neutralizing the effects of endogenous IL-1β and TNF-α. Conversely, addition of recombinant IL-1β, TNF-α, or both further enhanced IFN-γ production and strongly up-regulated IL-6 production. Our data indicate that CD56+ DCs from human blood are capable of stimulating CD56+ γδ T cells, which may be harnessed for immunotherapy.


2021 ◽  
Vol 11 (9) ◽  
pp. 923
Author(s):  
Josephine G. M. Strijker ◽  
Ronja Pscheid ◽  
Esther Drent ◽  
Jessica J. F. van der Hoek ◽  
Bianca Koopmans ◽  
...  

Currently ~50% of patients with a diagnosis of high-risk neuroblastoma will not survive due to relapsing or refractory disease. Recent innovations in immunotherapy for solid tumors are highly promising, but the low MHC-I expression of neuroblastoma represents a major challenge for T cell-mediated immunotherapy. Here, we propose a novel T cell-based immunotherapy approach for neuroblastoma, based on the use of TEG002, αβ-T cells engineered to express a defined γδ-T cell receptor, which can recognize and kill target cells independent of MHC-I. In a co-culture killing assay, we showed that 3 out of 6 neuroblastoma organoids could activate TEG002 as measured by IFNγ production. Transcriptional profiling showed this effect correlates with an increased activity of processes involved in interferon signaling and extracellular matrix organization. Analysis of the dynamics of organoid killing by TEG002 over time confirmed that organoids which induced TEG002 activation were efficiently killed independent of their MHC-I expression. Of note, efficacy of TEG002 treatment was superior to donor-matched untransduced αβ-T cells or endogenous γδ-T cells. Our data suggest that TEG002 may be a promising novel treatment option for a subset of neuroblastoma patients.


2021 ◽  
Vol 22 (16) ◽  
pp. 8910
Author(s):  
Masatsugu Miyashita ◽  
Teruki Shimizu ◽  
Eishi Ashihara ◽  
Osamu Ukimura

Human γδ T cells show potent cytotoxicity against various types of cancer cells in a major histocompatibility complex unrestricted manner. Phosphoantigens and nitrogen-containing bisphosphonates (N-bis) stimulate γδ T cells via interaction between the γδ T cell receptor (TCR) and butyrophilin subfamily 3 member A1 (BTN3A1) expressed on target cells. γδ T cell immunotherapy is classified as either in vivo or ex vivo according to the method of activation. Immunotherapy with activated γδ T cells is well tolerated; however, the clinical benefits are unsatisfactory. Therefore, the antitumor effects need to be increased. Administration of γδ T cells into local cavities might improve antitumor effects by increasing the effector-to-target cell ratio. Some anticancer and molecularly targeted agents increase the cytotoxicity of γδ T cells via mechanisms involving natural killer group 2 member D (NKG2D)-mediated recognition of target cells. Both the tumor microenvironment and cancer stem cells exert immunosuppressive effects via mechanisms that include inhibitory immune checkpoint molecules. Therefore, co-immunotherapy with γδ T cells plus immune checkpoint inhibitors is a strategy that may improve cytotoxicity. The use of a bispecific antibody and chimeric antigen receptor might be effective to overcome current therapeutic limitations. Such strategies should be tested in a clinical research setting.


Sign in / Sign up

Export Citation Format

Share Document