scholarly journals Ex Vivo Expanded Human Vγ9Vδ2 T-Cells Can Suppress Epithelial Ovarian Cancer Cell Growth

2019 ◽  
Vol 20 (5) ◽  
pp. 1139 ◽  
Author(s):  
Tsui Mao ◽  
Carol Miao ◽  
Yi Liao ◽  
Ying Chen ◽  
Chia Yeh ◽  
...  

γδ-T-cells have attracted attention because of their potent cytotoxicity towards tumors. Most γδ-T-cells become activated via a major histocompatibility complex (MHC)-independent pathway by the interaction of their receptor, Natural Killer Group 2 Member D (NKG2D) with the tumor-specific NKG2D ligands, including MHC class I-related chain A/B (MICA/B) and UL16-binding proteins (ULBPs), to kill tumor cells. However, despite their potent antitumor effects, the treatment protocols specifically targeting ovarian tumors require further improvements. Ovarian cancer is one of the most lethal and challenging female malignancies worldwide because of delayed diagnoses and resistance to traditional chemotherapy. In this study, we successfully enriched and expanded γδ-T-cells up to ~78% from peripheral blood mononuclear cells (PBMCs) with mostly the Vγ9Vδ2-T-cell subtype in the circulation. We showed that expanded γδ-T-cells alone exerted significant cytotoxic activities towards specific epithelial-type OVCAR3 and HTB75 cells, whereas the combination of γδ-T cells and pamidronate (PAM), a kind of aminobisphosphonates (NBPs), showed significantly enhanced cytotoxic activities towards all types of ovarian cancer cells in vitro. Furthermore, in tumor xenografts of immunodeficient NSG mice, γδ-T-cells not only suppressed tumor growth but also completely eradicated preexisting tumors with an initial size of ~5 mm. Thus, we concluded that γδ-T-cells alone possess dramatic cytotoxic activities towards epithelial ovarian cancers both in vitro and in vivo. These results strongly support the potential of clinical immunotherapeutic application of γδ-T-cells to treat this serious female malignancy.

2008 ◽  
Vol 76 (10) ◽  
pp. 4538-4545 ◽  
Author(s):  
William W. Kwok ◽  
Junbao Yang ◽  
Eddie James ◽  
John Bui ◽  
Laurie Huston ◽  
...  

ABSTRACT Cellular immune responses against protective antigen (PA) of Bacillus anthracis in subjects that received the anthrax vaccine adsorbed (AVA) vaccine were examined. Multiple CD4+ T-cell epitopes within PA were identified by using tetramer-guided epitope mapping. PA-reactive CD4+ T cells with a CD45RA− phenotype were also detected by direct ex vivo staining of peripheral blood mononuclear cells (PBMC) with PA-specific tetramers. Surprisingly, PA-specific T cells were also detected in PBMC of nonvaccinees after a single cycle of in vitro PA stimulation. However, PA-reactive CD4+ T cells in nonvaccinees occurred at lower frequencies than those in vaccinees. The majority of PA-reactive T cells from nonvaccinees were CD45RA+ and exhibited a Th0/Th1 cytokine profile. In contrast, phenotyping and cytokine profile analyses of PA-reactive CD4+ T cells from vaccinees indicated that vaccination leads to commitment of PA-reactive T cells to a Th2 lineage, including generation of PA-specific, pre-Th2 central memory T cells. These results demonstrate that the current AVA vaccine is effective in skewing the development of PA CD4+ T cells to the Th2 lineage. The data also demonstrated the feasibility of using class II tetramers to analyze CD4+ cell responses and lineage development after vaccination.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 5119-5119
Author(s):  
Annie Levesque ◽  
Ann-Louise Savard ◽  
Denis-Claude Roy ◽  
Francine Foss ◽  
Christian Scotto

Abstract Although the risk of graft versus host disease (GvHD) can be reduced by improved donor-recipient matching and by the depletion of T cells before transplantation, GvHD still develops in 30–70% of allogeneic hematopoietic stem cell transplantation (HSCT) patients. The chronic phase of the disease (cGvHD), for which the pathogenesis is similar to autoimmune diseases, involves profound immune dysregulation leading to both immunodeficiency and autoimmunity. Standard therapies for cGvHD such as corticosteroids and immunosuppressants are associated with high toxicity and have demonstrated limited efficacy in patients with extensive disease. Extracorporeal photopheresis (ECP) has been shown by others in the clinic as a non-aggressive and beneficial alternative treatment for cGvHD, inducing Th1/Th2 immunomodulation that restores immunological tolerance. Celmed has developed an alternative approach to eliminate immunoreactive T cells using the Theralux™ photodynamic cell therapy (PDT) system based on the use of the rhodamine-123 derivative TH9402 illuminated ex vivo with a visible light source (λ =514nm). It has been suggested that the apoptotic cells, when returned to the patient, may be able to modulate the immune system as seen with other ECP methods. We aimed to evaluate in vivo and in vitro the possibility of also using the Theralux™ system in the ECP setting. A preliminary mouse model suggested that splenic T cells pre-treated with the Theralux™ system were able to induce an improvement of overall survival (p<0.05) in mice with acute GvHD. Additionally, we developed a simplified PDT process and conducted a series of experiments with peripheral blood mononuclear cells (PBMCs) isolated from healthy volunteers. These studies have shown that the intra- and inter-donor variability in TH9402 incorporation are very low (~5% and 10%, respectively). A dose-effect study has shown a relationship of the PDT conditions with the levels of cell death, allowing significant control of the level of apoptosis induced. Phenotypic analyses have shown that this process results in an increase of AnnexinV positive cells as well as a decrease in the absolute number of CD3+ cells, CD19+/CD20+ cells and CD14+ cells and an increase in CD11c+ cells. This would suggest that apoptosis could be induced in both autoreactive T and B cells which could potentially stimulate an immune response against them. Moreover, the increase in CD11c+ cells combined with the decrease in CD14+ cells could reflect the maturation of macrophages into dendritic cells that are very potent antigen presenting cells. The mechanism by which these specific PDT conditions induce cell death is still under investigation but preliminary studies have shown that the cell death in unselected resting PBMCs may be caspase-independent. Finally, the evaluation of the effect of PDT on samples from cGvHD patients also demonstrated the capacity of this treatment strategy to induce apoptosis in these cells. Based on these data, we intend to begin a pilot clinical study evaluating two controlled PDT conditions inducing different levels of apoptosis in order to assess the safety and biological effect of the Theralux™ ECP system to treat patients with cGvHD.


2019 ◽  
Vol 17 (1) ◽  
Author(s):  
Leonardo Mirandola ◽  
Maurizio Chiriva-Internati ◽  
Robert Bresalier ◽  
Lucia Piccotti ◽  
Fabio Grizzi ◽  
...  

Abstract Background The inefficacy of standard therapeutic strategies for ovarian cancer is reflected by the enduring poor prognosis of this malignancy. Due to the potential for exquisite specificity, sensitivity and long-term memory, immunotherapy offers an alternative modality for durable control of the disease, provided appropriate antigens can be identified and presented in the right context. Methods We tested a novel dendritic cell vaccine formulation to reprogram autologous antigen-specific T-cells in vitro, in vivo in a murine model of ovarian cancer, and ex vivo using human cells from patients. Results We show that dendritic cells (DCs) treated with a p38 MAPK inhibitor and transduced with a recombinant adenovirus associated vector (AAV) expressing Sperm protein (Sp) 17 are highly effective in generating antigen-specific T-cell cytotoxic response against ovarian cancer cells. Additionally, these DCs enhanced the differentiation of effector T-cells while reducing the frequency of Foxp3+ T-reg cells in vitro. Conclusions This work provides a rationale for translation of pharmacologically reprogrammed DCs into clinical trials for prevention of tumor recurrence and progression in high-risk ovarian cancer patients.


2001 ◽  
Vol 69 (1) ◽  
pp. 89-96 ◽  
Author(s):  
Allister J. Smyth ◽  
Michael D. Welsh ◽  
R. Martyn Girvin ◽  
John M. Pollock

ABSTRACT It is generally accepted that protective immunity against tuberculosis is generated through the cell-mediated immune (CMI) system, and a greater understanding of such responses is required if better vaccines and diagnostic tests are to be developed. γδ T cells form a major proportion of the peripheral blood mononuclear cells (PBMC) in the ruminant system and, considering data from other species, may have a significant role in CMI responses in bovine tuberculosis. This study compared the in vitro responses of αβ and γδ T cells from Mycobacterium bovis-infected and uninfected cattle. The results showed that, following 24 h of culture of PBMC withM. bovis-derived antigens, the majority of γδ T cells from infected animals became highly activated (upregulation of interleukin-2R), while a lower proportion of the αβ T-cell population showed activation. Similar responses were evident to a lesser degree in uninfected animals. Study of the kinetics of this response showed that γδ T cells remained significantly activated for at least 7 days in culture, while activation of αβ T cells declined during that period. Subsequent analysis revealed that the majority of activated γδ T cells expressed WC1, a 215-kDa surface molecule which is not expressed on human or murine γδ T cells. Furthermore, in comparison with what was found for CD4+ T cells, M. bovis antigen was found to induce strong cellular proliferation but relatively little gamma interferon release by purified WC1+ γδ T cells. Overall, while the role of these cells in protective immunity remains unclear, their highly activated status in response to M. bovis suggests an important role in antimycobacterial immunity, and the ability of γδ T cells to influence other immune cell functions remains to be elucidated, particularly in relation to CMI-based diagnostic tests.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1901-1901 ◽  
Author(s):  
Yongxian Hu ◽  
Yanjun Gu ◽  
Lixia Sheng ◽  
Huarui Fu ◽  
Kangni Wu ◽  
...  

Abstract Abstract 1901 Regulatory γδ T cells (γδ Tregs) is a novel subset of cells with immunosuppressive function while methods for γδ Treg induction is rarely introduced and its role in graft-versus-host disease (GVHD) prevention remains unkown. Decitabine, a kind of hypomethylating agents, can act synergistically with TGF-β1 to convert a variety of αβ T cells to regulatory αβ T cells with suppressive function but its role in induction and function of γδ Tregs has not been reported. We show here the role of decitabine for the induction of γδ Tregs. Moreover, we provide functional analysis and underlying mechanisms of decitabine-induced γδ Tregs relative to γδ Tregs without decitabine induction as well as in vivo evidences of their preventions on GVHD. Human peripheral blood mononuclear cells (PBMCs) were cultured with IL-2, IL-15, TGF-β1 and zoledronic acid (ZOL). On day 2, 0.5μmol/ml decitabine was added to aliquots of PBMCs. On days 4, 7, and 10, half of the supernatant volume was replaced with media containing cytokines. On day 11, frequencies of γδ Tregs were detected by flow cytometry (FACS). We found the frequency of γδ Tregs was 36.2% in TGF-β1/IL-15/ZOL stimulated group (referred to as common γδ Tregs below) and 59.9% in IL-2/TGF-β1/IL-15/ZOL/decitabine stimulated group (referred to as decitabine-induced γδ Tregs) (p<0.05). In order to compare immunosuppressive function of the two populations, γδ T cells containing γδ Tregs were isolated by magnetic cell sorting system (MACS) and tested for their ability to suppress proliferation of alloreactive PBMCs using CFSE-based assay. After 5 days of in vitro culture, CFSE-labeled PBMCs proliferation was significantly reduced in the presence of enriched γδ Tregs even at 8:1(PBMCs: γδ Tregs) ratio. The inhibition rate was significantly different (decitabine-induced γδ Tregs VS common γδ Tregs at ratio 1:1 is 81.3% VS 68.2%, p<0.05). To clarify the underlying mechanisms we performed ELISA to measure levels of inhibitory cytokines IL-10, IL-4 and TGF-β1 in supernatant of CFSE-based assay. We noted an elevated IL-10 secretion in the decitabine-induced γδ Tregs group compared with common γδ Tregs group (92.7±11pg/ml VS 10.3±2pg/ml at ratio 1:1, p<0.01). We confirmed the result by intracellular IL-10 detection using FACS. Previous reports showed high levels of inducible T-cell costimulator (ICOS) were correlated with IL-10 synthesis. So γδ Tregs were monitored for ICOS expression by FACS. The result revealed that ICOS expression was up-regulated in decitabine-induced γδ Tregs in contrast to common γδ Tregs (MFI: 268 VS 54). Stability of Foxp3 is a critical factor in the immunosuppressive ability of Tregs. Thus we evaluated the frequency of γδ Tregs after 5 days in CFSE-based assay. We observed loss of Foxp3 expression in decitabine-induced γδ Tregs was negligible (<3%) while 15.5% common γδ Tregs lost foxp3 expression. To confirm the results in vitro we tested the functional ability to prevent GVHD in vivo. GVHD was induced in NOD/SCID mice following busulfan and anti-CD122 condition and 1×107 PBMCs transfusion. Animals were co-injected with either decitabine-induced γδ Tregs or common γδ Tregs at a ratio of 1:1. Survival time and GVHD manifestations of the transplanted mice were evaluated. As a result, transplantation of human PBMCs alone induced lethal GVHD with average survival time 25± 8 days while the survival time was 43± 5 days and 58±7 days in mice co-injected with common γδ Tregs and decitabine-induced γδ Tregs, respectively (p<0.05). Clinical manifestations such as hunched back, diarrhea, and body weight loss were statistically different among 3 groups. To investigate the infiltration of human lymphocytes into nonlymphoid tissues in GVHD mice, we performed immunohistochemical analysis of the liver and intestines using anti-human CD45. Remarkably abundant invasion of human CD45+ cells was observed around the veins in the liver and intestines transplanted with PBMCs alone while less invasion in mice co-injected with common γδ Tregs and the lest invasion in mice co-injected with decitabine-induced γδ Tregs. Altogether, our findings reveal that decitabine and the cytokines can efficiently syngenerize to induce γδ Tregs with enhanced immunosuppression on GVHD which are via higher levels of IL-10 production due to ICOS up-regulation as well as stability of Foxp3 expression. Thus γδ Tregs may be potentially exploited therapeutically in a variety of transplantation settings. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2000 ◽  
Vol 96 (1) ◽  
pp. 275-281 ◽  
Author(s):  
Christophe Nicot ◽  
Renaud Mahieux ◽  
Shigeki Takemoto ◽  
Genoveffa Franchini

Human T lymphotropic virus type I (HTLV-I) is the etiological agent of adult T-cell lymphocytic leukemia (ATLL), whereas HTLV-II has not been associated with hematopoietic malignancies. The control of apoptotic pathways has emerged as a critical step in the development of many cancer types. As a result, the underlying mechanism of long-term survival of HTLV-I and HTLV-II was studied in infected T cells in vitro and in ex vivo ATLL samples. Results indicate that HTLV-I– and HTLV-II–infected T cells in vitro express high levels of the antiapoptotic protein Bcl compared with other human leukemic T cell lines or uninfected peripheral blood mononuclear cells. The levels of proapoptotic proteins Bax, BAD, and Bak were not significantly altered. HTLV-I and HTLV-II viral transactivators, Tax1 and Tax2, are known to increase expression of cellular genes. These proteins were tested for increased transcription from the human Bcl2 and Bcl-XL promoters. Whereas no effect was observed on the Bcl2 promoter, both Tax1 and Tax2 increased transcription of the Bcl-XL promoter in T cells, although Tax1 appeared to be more efficient than Tax2. The biological significance of these observations was validated by the finding of an increased expression of Bcl-XL in ex vivo ATLL cells, especially from patients unresponsive to various chemotherapy regimens. Altogether, these data suggest that overexpression of Bcl-XL in vivomay be in part responsible for the resistance of ATLL cells to chemotherapy. In addition, inefficient activation of the Bcl-XL promoter by Tax2 may result in a shorter survival time of HTLV-II–infected cells in vivo and a diminished risk of leukemia development.


Viruses ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 1300
Author(s):  
Jose Mateus ◽  
Alba Grifoni ◽  
Hannah Voic ◽  
Michael A. Angelo ◽  
Elizabeth Phillips ◽  
...  

Yellow fever virus (YFV) is a mosquito-borne member of the genus flavivirus, including other important human-pathogenic viruses, such as dengue, Japanese encephalitis, and Zika. Herein, we report identifying 129 YFV Class II epitopes in donors vaccinated with the live attenuated YFV vaccine (YFV-17D). A total of 1156 peptides predicted to bind 17 different common HLA-DRB1 allelic variants were tested using IFNγ ELISPOT assays in vitro re-stimulated peripheral blood mononuclear cells from twenty-six vaccinees. Overall, we detected responses against 215 YFV epitopes. We found that the capsid and envelope proteins, as well as the non-structural (NS) proteins NS3 and NS5, were the most targeted proteins by CD4+ T cells from YF-VAX vaccinated donors. In addition, we designed and validated by flow cytometry a CD4+ mega pool (MP) composed of structural and non-structural epitopes in an independent cohort of vaccinated donors. Overall, this study provides a comprehensive prediction and validation of YFV epitopes in a cohort of YF-17D vaccinated individuals. With the design of a CD4 epitope MP, we further provide a useful tool to detect ex vivo responses of YFV-specific CD4 T cells in small sample volumes.


2020 ◽  
Vol 8 (2) ◽  
pp. e001237
Author(s):  
Adekunle Odunsi ◽  
A J Robert McGray ◽  
Anthony Miliotto ◽  
Yali Zhang ◽  
Jianming Wang ◽  
...  

BackgroundImmune checkpoint blockers (ICBs) have been approved by the Food and Drug Administration to be used alone in front-line therapies or in combination with other regimens for certain advanced cancers. Since ICB only works in a subset of patients and has limited efficacy in treating ovarian cancer (OVC), developing preclinical models that help to understand which patients may derive benefit from ICB would be of tremendous benefit in OVC.MethodsHere, we generated preclinical human OVC models from freshly resected tumors, which include six patient-derived xenografts (PDXs) from six different patient tumors, three transplantable OVC PD spheroid lines (PD-sphs), and 3 cell lines (PD-CLs). We tested the therapeutic combination of anti-PD1/CTLA4 antibodies with (1) autologous tumor-associated leukocytes (TALs) on the growth of PD-sphs in a coculture system in vitro, (2) with adoptively transferred autologous peripheral blood mononuclear cells or TALs in patient-derived OVC models using partially humanized mice, NSG-HHDxSGM3 (N-HSGM3).ResultsWe show that PD-1 and CTLA-4 dual blockade when combined with autologous TALs effectively reduced PD-sph number in a co-culture system and led to regression of established PD-CLs and PDXs in the N-HSGM3 mice. Combinatorial PD-1 and CTLA-4 blockade increased the frequency and function of tumor-specific CD8 T cells. These CD8 T cells persisted in the tumor microenvironment, exhibited memory phenotype and protected animals from tumor growth on tumor rechallenge. Gene expression analysis of tumors resistant to dual PD1/CTLA4 blockade treatment identified upregulation of antigen processing and presentation pathways and downregulation of extracellular matrix organization genes.ConclusionsThese findings describe a novel platform for developing patient-derived preclinical tumor models suitable for rationally testing combinatorial ICB in the context of autologous tumor-reactive T cells. This platform can be further developed for testing additional targeted therapies relevant to OVC.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A185-A185
Author(s):  
Maho Nagasawa ◽  
Remco Bos ◽  
Haoxiao Zuo ◽  
Kiave Yune Ho Wang Yin ◽  
Marie-José van Lierop ◽  
...  

BackgroundOvarian cancer (OC) is the gynecological malignancy with the highest mortality due to the late diagnosis of disease and a high rate of relapse following initial therapy. Immunotherapy in combination with standard treatment modalities has emerged as an encouraging treatment approach to surmount this unmet medical need. DCP-001 is a cancer relapse vaccine derived from the DCOne human leukemic cell line and is currently progressing through clinical trials in hematological malignancies. During manufacturing, DCOne cells are shifted towards a mature dendritic cell phenotype, rendering the cells highly immunogenic and providing the basis for DCP-001, which is administered as an intradermal vaccine. DCOne cells express multiple common tumor associated antigens (TAA) such as WT-1, RHAMM, PRAME and MUC-1, which have been documented as potential target antigens in ovarian cancer. This observation suggests that DCP-001 vaccination may also have an anti-tumor effect in OC. To support this hypothesis, we assessed the capacity of DCP-001 to induce immune responses against OC in human peripheral blood mononuclear cells (PBMCs) and a humanized mouse model for OC.MethodsThe effect of DCP-001 on T cells from OC patients or healthy donors was evaluated after a 3 week culture of peripheral blood mononuclear cells (PBMCs) with or without DCP-001. Cytotoxic activity was analysed by specific IFNg production and CD107a expression when these cells were subsequently cultured with the OC cell line SKOV3. The effect of DCP-001 vaccination in vivo was evaluated in humanised NCG mouse subcutaneously engrafted with SKOV3 OC cells. Mice received intra-peritoneal (i.p.) vaccination with DCP-001 either after or prior to SKOV3 engraftment and tumor size was measured to evaluate the efficacy of DCP-001.ResultsIn vitro, DCP-001 was shown to activate both CD4+ as well as CD8+ T cells and to induce formation of memory T cells. Importantly, DCP-001-stimulated CD8+ T cells from OC patients were shown to exert a HLA class I dependent, cytotoxic immune response to OC cells. In vivo, in an ovarian tumor mouse model, significant reduction of tumor growth rate and partial or even complete tumor regressions were observed in mice vaccinated with DCP-001, particularly when administered as relapse vaccine (prior to tumor engraftment), as compared to PBS treated mice.ConclusionsThese pre-clinical in vitro and in vivo results support the potential use of DCP-001 as a cancer relapse vaccine in ovarian cancer, with the aim to reduce disease recurrence following initial standard of care therapy.


2021 ◽  
Vol 17 (3) ◽  
pp. 460-470
Author(s):  
Mohd Wajid Ali Khan ◽  

Human Vγ9/Vδ2 T cells (γδ T cells) are immune surveillance cells both in innate and adaptive immunity and are a possible target for anticancer therapies, which can induce immune responses in a variety of cancers. Small non-peptide antigens such as zoledronate can do activation and expansion of T cells in vitro. It is evident that for adoptive cancer therapies, large numbers of functional cells are needed into cancer patients. Hence, optimization of methods needs to be carried out for the efficient expansion of these T cells. Standardization of peripheral blood mononuclear cells (PBMCs) isolation was devised. Cytokines (interleukin 2 (IL-2) and interleukin 15 (IL-15)) and zoledronate were also standardized for different concentrations. It was found that an increased number of PBMCs were recovered when washing was done at 1100 revolution per minute (rpm). Significantly high expansion fold was (2524 ± 787 expansion fold) achieved when stimulation of PBMCs was done with 1 μM of zoledronate and both cytokines IL-2 and IL-15 supported the expansion and survival of cells ISSN 0973-2063 (online) 0973-8894 (print) Bioinformation 17(3): 460-469 (2021) ©Biomedical Informatics (2021) 461 at the concentrations of 100 IU/ml and 10 ng/ml respectively. 14-day cultures showed highly pure (91.6 ± 5.1%) and live (96.5 ± 2.5%) expanded γδ T cells. This study aimed to standardize an easy to manipulate technique for the expansion of γδ T cells, giving a higher yield.


Sign in / Sign up

Export Citation Format

Share Document