scholarly journals Determination of Optimal Target Antigen and Immune Modulatory Approaches to Improve and Standardize Epstein-Barr Virus-Specific Adoptive T Cell Therapy

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5817-5817
Author(s):  
Monica L Mitchell ◽  
John T. Patton ◽  
Lynn O'Donnell ◽  
Robert A. Baiocchi

Abstract Epstein-Barr Virus (EBV) is a ubiquitous gamma herpesvirus that infects lymphocytes and epithelial cells, establishing lifelong latency. Immunosuppression is associated with a transition from a quiescent, latent EBV infection to either a lytic or active latent life cycle, which involves transformation of memory B cells to rapidly growing lymphoblastoid cell lines (LCLs). Due to the immunogenicity of EBV viral proteins, cells expressing most latent or lytic peptides are eradicated through immune surveillance mechanisms. EBV-associated lymphoproliferative disorders (LPDs) arise in immunocompromised individuals and are facilitated by the expression of the same viral proteins that are expressed in the immune tolerant environment. Furthermore, EBV-LPDs acquire various mechanisms to suppress immune function, such as expression of the programmed death ligand (PD-L1), CTLA-4, amino acid depletion, and propagation of suppressive immune cell subsets, including tumor-associated macrophages (TAMs) that attenuate T cell receptor signaling and cytokine production. These immunosuppressive mechanisms increase the risk of acquiring EBV viremia and can facilitate the development of LPDs. In other work reported by our group (Patton et al, ASH abstract 2014), a TAM-like population has recently been discovered to spontaneously expand in peripheral blood mononuclear cell (PBMC)/LCL co-cultures. These TAM-like macrophages are capable of potent cytotoxicity against EBV-specific T cells, thus novel strategies to counteract this negative regulatory network are needed. One effective strategy developed to reestablish EBV-specific immunity is adoptive T cell therapy. However, the optimal combination of target antigens required to protect against EBV-LPD is not well characterized. It remains plausible that expansion of cytotoxic and/or helper T cell populations specific for multiple EBV antigens will improve the efficiency of the T cell response. The suppression of negative immune regulators may further enhance the effectiveness of the adoptive therapies to provide optimal control of viremia and effectively treat EBV-driven malignancies. Here, we generated EBV-specific T cell preparations by culturing PBMCs supplemented with interleukins 4 and 7 (IL4, IL7) in the presence of latent (EBNA1, EBNA3, LMP1, LMP2) or lytic (BZLF1, BRLF, BMLF1, BMRF1) PepMixes (complete protein-spanning pools of overlapping peptides). Cells were separated into two groups, a test and expansion group, to determine optimal ex vivo expansion. Activity of T cells was measured by direct flow-based cytotoxicity against LCL targets and interferon gamma (IFNγ) production via Enzyme-Linked Immuno Spot (ELISpot). Preliminary ELISpot assays from two donors in the test group showed that BZLF1-specific T cells produced as much as 34-fold more IFNγ in comparison to other EBV antigens (standardized to a negative control). T cells produced in culture with a peptivator PepMix, a collection of 13 latent and lytic peptide pools, on average exhibited the second highest levels of IFNγ release, expressing as much as 1.4-fold more IFNγ than the next highest antigen-specific response, depending on the donor. Additionally, LMP1 produced as much as 24-fold higher levels of IFNγ. Therefore, given the heterogeneous EBV viral gene expression profiles and variation of immune dominance across human leukocyte antigen (HLA) types, use of polyclonal immunogen-specific T cells may be more effective in improving survival and long-term protection from EBV-driven complications via improved memory T cell surveillance. Additional ex vivo work is currently underway examining combinations of lytic and latent antigens based on immunodominance analysis across HLA types. Finally, use of immune modulatory mechanisms to address newly discovered TAM-like populations contributing to reduced immune effector function against EBV-LPD are currently being evaluated. We are testing strategies to promote PD-L1 and CTLA-4 blockade, inhibition of amino acid depletion, and down regulation of regulatory T cells, to allow for enhanced expansion of antigen-specific T cell populations. Confirmation of these hypotheses will be directed toward developing streamlined methods for efficient, rapid, and personalized T cell preparation that can be standardized for translation into clinical trials. Disclosures No relevant conflicts of interest to declare.

2016 ◽  
Vol 39 (3) ◽  
pp. 140-148 ◽  
Author(s):  
Hyeon-Seok Eom ◽  
Beom K. Choi ◽  
Youngjoo Lee ◽  
Hyewon Lee ◽  
Tak Yun ◽  
...  

Blood ◽  
2002 ◽  
Vol 99 (9) ◽  
pp. 3302-3309 ◽  
Author(s):  
Qi Sun ◽  
Robert L. Burton ◽  
Kenneth G. Lucas

Abstract Ex vivo expanded Epstein-Barr virus (EBV)–specific T cells have been successfully applied clinically for adoptive immunotherapy. However, the role of CD4+ T cells in the therapeutic T-cell culture has not been established for the reconstitution of EBV-specific immunity. We isolated and characterized CD4+ T-cell lines from the ex vivo T-cell cultures. Monoclonal line PD-F4 and oligoclonal lines ND-R4 and TD-B4 were CD3+CD4+CD8−. Cytolytic tests with targets of mismatched major histocompatibility complex (MHC) and anti-MHC antibodies confirmed that the cytotoxicity of these CD4+ cells was restricted by MHC class II. Single cells of ND-R4 expressed interferon-γ (IFN-γ, or interleukin 4 (IL-4), but rarely coexpressed these 2 cytokines. In contrast, PD-F4 coexpressed IFN-γ, IL-2, and IL-4. Kinetic studies with PD-F4 showed that expression of the 3 cytokines plateaued 5 hours upon stimulation and was then drastically reduced, with a pattern consistent with independent modulation and differential off-cycle signal requirements. The cytotoxicity of these CD4+ cells was largely resistant to brefeldin A, an inhibitor for cytolytic pathways by Fas-ligand family molecules. Although sensitive to concanamycin A and ethyleneglycotetraacetic acid, which inhibit cytotoxicity by granule exocytosis, the CD4+ cytotoxic T lymphocytes (CTLs) did not express perforin, suggesting a cytotoxic mechanism independent of perforin although involving exocytosis. Flow cytometric analysis showed that the CD4+ CTLs expressed granulysin, a recently identified cytolytic molecule associated with exocytotic cytolytic granules. These data suggested that CD4+ T cells in the therapeutic B-lymphoblastoid cell lines–primed T-cell culture are diverse in producing TH1 and TH2 cytokines, and may exert specific cytotoxicity via exocytosis of granulysin.


Blood ◽  
2008 ◽  
Vol 111 (3) ◽  
pp. 1334-1343 ◽  
Author(s):  
Sumita Bhaduri-McIntosh ◽  
Marisa J. Rotenberg ◽  
Benjamin Gardner ◽  
Marie Robert ◽  
George Miller

AbstractAnswers to questions about frequency and repertoire of immune cells, relative contributions made by different types of immune cells toward the total Epstein-Barr virus (EBV)–directed response and the variation of such responses in healthy persons have been elusive because of disparities in assays, antigen presenting cells, and antigenic sources used in previous experiments. In this study, we addressed these questions using an assay that allowed direct comparison of responses generated by different types of cells of the immune system. This short-term (20-hour) ex vivo assay measured interferon-γ production by blood cells in response to autologous EBV-transformed lymphoblastoid cell lines (LCLs). Our experiments defined the variation in responses among persons and clearly distinguished 10 healthy EBV-immune from 10 healthy EBV-naive persons. In EBV-immune persons, 33% of responding cells were CD4+, 43.3% were CD8+, and 12.9% were γ-δ T cells. LCL-reactive CD8+ T cells were only 1.7-fold more frequent than similarly reactive CD4+T cells. Responses by γ-δ T cells were 6-fold higher in seropositive than in seronegative persons. Our findings emphasize the importance of CD4+ and γ-δ T-cell responses and have implications for immunotherapy and for identifying defects in T-cell populations that might predispose to development of EBV-associated lymphomas.


2003 ◽  
Vol 110 (2-3) ◽  
pp. 149-153 ◽  
Author(s):  
M. Helen Huls ◽  
Cliona M. Rooney ◽  
Helen E. Heslop

2009 ◽  
Vol 83 (12) ◽  
pp. 6192-6198 ◽  
Author(s):  
Corey Smith ◽  
Leone Beagley ◽  
Rajiv Khanna

ABSTRACT Latent membrane antigen 1 and -2 (LMP-1/2)-specific CD8+ T cells from newly diagnosed and relapsed Hodgkin's lymphoma (HL) patients display a selective functional impairment. In contrast, CD8+ T cells specific for Epstein-Barr virus (EBV) nuclear proteins and lytic antigens retain normal T-cell function. Reversion to a dysfunctional phenotype of LMP-1/2-specific T cells is coincident with the regression of HL. To delineate the potential basis for this differential susceptibility for the loss of function, we have carried out a comprehensive functional analysis of EBV-specific T cells using ex vivo multiparametric flow cytometry in combination with assessment of antigen-driven proliferative potential. This analysis revealed that LMP-1/2-specific T cells from healthy virus carriers display a deficient polyfunctional profile compared to that of T cells specific for epitopes derived from EBV nuclear proteins and lytic antigens. Furthermore, LMP-specific T-cells are highly susceptible to galectin-1-mediated immunosuppression and are less likely to degranulate following exposure to cognate peptide epitopes and poorly recognized endogenously processed epitopes from virus-infected B cells. More importantly, ex vivo stimulation of these T cells with an adenoviral vector encoding multiple minimal CD8+ T-cell epitopes as a polyepitope, in combination with a γC cytokine, interleukin-2, restored polyfunctionality and shielded these cells from the inhibitory effects of galectin-1.


Blood ◽  
1997 ◽  
Vol 89 (6) ◽  
pp. 1978-1986 ◽  
Author(s):  
Amy P. Sing ◽  
Richard F. Ambinder ◽  
Doley J. Hong ◽  
Michael Jensen ◽  
Wendy Batten ◽  
...  

Abstract A subset of Hodgkin's disease (HD) patients have detectable Epstein-Barr virus (EBV) genomes in the malignant Reed-Sternberg (R-S) cells. R-S cells express only a limited set of latent EBV proteins, but only LMP1 and LMP2 can potentially elicit a CD8+ cytotoxic T-lymphocyte (CTL) response. We have evaluated if either of these proteins could be used as targets for specific adoptive T-cell therapy for EBV-positive (EBV+) HD. The success of this strategy requires that R-S cells are susceptible to lysis by CD8+ CTL, and that CTL specific for LMP1 and LMP2 can be detected and potentially amplified in HD patients. Antigen presentation and CTL sensitivity was evaluated with an in vitro maintained, phenotypically representative R-S cell line, HDLM-2. The R-S cells were able to process and present viral proteins, and to be efficiently lysed by specific CTL in a Class I–restricted manner. Since CTL responses to LMP1 and LMP2 do not represent the dominant responses to EBV, we examined if CTL clones specific for these proteins could be isolated despite the presence of weak or nondetectable responses in polyclonal T-cell lines. LMP-specific clones were generated from individuals either by cloning from the polyclonal EBV-reactive T-cell lines or by direct stimulation of peripheral blood mononuclear cells (PBMC) with cells expressing LMP1 or LMP2 as the only EBV protein. Our ability to isolate CTL specific for LMP proteins from individuals with HD and the sensitivity of R-S cells for CTL-mediated lysis suggest that the pursuit of specific adoptive immunotherapy represents a viable strategy for the subset of HD patients with EBV+ tumors.


2021 ◽  
Vol 39 (5) ◽  
pp. 514-524
Author(s):  
Helen E. Heslop ◽  
Sandhya Sharma ◽  
Cliona M. Rooney

2020 ◽  
Vol 154 (Supplement_1) ◽  
pp. S81-S81
Author(s):  
J Lanceta ◽  
W Xue ◽  
M Hurford ◽  
H Wu

Abstract Casestudy Epstein-Barr virus (EBV)-associated peripheral T-cell lymphomas are a group of aggressive neoplasms with a geographic predilection for South America and Asia, but are very rare in Western populations. Results We report a case of a 74-year-old Caucasian female who presented with pancytopenia and B symptoms with EBV-IgG detected on admission. Past medical history included: ITP, chronic urticaria, and recently diagnosed myelodysplastic syndrome (MDS) on bone marrow biopsy one month prior to admission. Excisional biopsies of an enlarged right neck lymph node (repeated within 6 months) and right axillary lymph node five years ago were negative for a lymphoproliferative disorder at the time. Repeated bone marrow biopsy, performed during the current admission, confirmed the diagnosis of MDS, with scattered T-cells without aberrant immunophenotype. Despite aggressive treatment from multiple specialties, the patient deteriorated and expired four weeks later from complications of MDS. At autopsy, there was diffuse lymphadenopathy involving the mediastinum, axilla, pelvis and peripancreatic fat. Lymph node sections demonstrated nodal architecture effacement by diffuse, vaguely nodular lymphoid infiltrates. Histologically, the infiltrates were composed of medium to large lymphocytes with round to slight irregular nuclei, rare Reed-Sternberg-like multinucleated cells, clumped chromatin, and indistinct nucleoli. Individual cell necrosis was abundant with mitotic figures readily identifiable. Immunohistochemistry revealed CD2+ CD3+ neoplastic T-cells that co-express MUM1 and a subset of CD30, while negative for CD4, CD5, CD8, CD56, ALK1, and TDT. EBV-encoded RNA in-situ hybridization was focally positive. The final postmortem diagnosis was peripheral T-cell lymphoma, not otherwise specified (NOS), with focal EBV positivity. Conclusion Co-existence of a de-novo MDS and non-Hodgkin lymphoma without any prior chemotherapeutic exposure is a highly unusual finding, although MDS-like presentations can occur with EBV-associated lymphomas. Peripheral T-cell lymphoma, NOS is an aggressive lymphoma and EBV positivity has been found correlated with a poor prognosis. This case demonstrates how postmortem examination remains an important tool in clinical- pathological correlation and highlights the potential pathogenetic role EBV plays in MDS and T-cell lymphoma.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii111-ii111
Author(s):  
Lan Hoang-Minh ◽  
Angelie Rivera-Rodriguez ◽  
Fernanda Pohl-Guimarães ◽  
Seth Currlin ◽  
Christina Von Roemeling ◽  
...  

Abstract SIGNIFICANCE Adoptive T cell therapy (ACT) has emerged as the most effective treatment against advanced malignant melanoma, eliciting remarkable objective clinical responses in up to 75% of patients with refractory metastatic disease, including within the central nervous system. Immunologic surrogate endpoints correlating with treatment outcome have been identified in these patients, with clinical responses being dependent on the migration of transferred T cells to sites of tumor growth. OBJECTIVE We investigated the biodistribution of intravenously or intraventricularly administered T cells in a murine model of glioblastoma at whole body, organ, and cellular levels. METHODS gp100-specific T cells were isolated from the spleens of pmel DsRed transgenic C57BL/6 mice and injected intravenously or intraventricularly, after in vitro expansion and activation, in murine KR158B-Luc-gp100 glioma-bearing mice. To determine transferred T cell spatial distribution, the brain, lymph nodes, heart, lungs, spleen, liver, and kidneys of mice were processed for 3D imaging using light-sheet and multiphoton imaging. ACT T cell quantification in various organs was performed ex vivo using flow cytometry, 2D optical imaging (IVIS), and magnetic particle imaging (MPI) after ferucarbotran nanoparticle transfection of T cells. T cell biodistribution was also assessed in vivo using MPI. RESULTS Following T cell intravenous injection, the spleen, liver, and lungs accounted for more than 90% of transferred T cells; the proportion of DsRed T cells in the brains was found to be very low, hovering below 1%. In contrast, most ACT T cells persisted in the tumor-bearing brains following intraventricular injections. ACT T cells mostly concentrated at the periphery of tumor masses and in proximity to blood vessels. CONCLUSIONS The success of ACT immunotherapy for brain tumors requires optimization of delivery route, dosing regimen, and enhancement of tumor-specific lymphocyte trafficking and effector functions to achieve maximal penetration and persistence at sites of invasive tumor growth.


Sign in / Sign up

Export Citation Format

Share Document