Abstract B050: Functional characterization of virus specific stem cell like memory T cells, their maturation in vitro, and potential contribution to secondary T cell responses

Author(s):  
Tzu-yun Kuo ◽  
Aisha N. Hasan ◽  
Richard J. O'Reilly
Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3502-3502
Author(s):  
Liane Bergmann ◽  
Matthias Staudinger ◽  
Christiane Pott ◽  
Ingrid Bolz ◽  
Martin Gramatzki ◽  
...  

Abstract Patients with mantle cell lymphoma (MCL) and MRD after intensive radiochemotherapy and autologous stem cell transplantation have a high risk of relapse. Allogeneic stem cell transplantation offers the possibility of cure but is associated with a high risk of severe “graft versus host disease”(GvHD). A way to decrease the risk of GvHD while augmenting the “graft versus lymphoma” effect may be the in vitro activation and subsequent transplantation of allogeneic idiotyp-specific T-cells. This study was set out to determine whether cytotoxic T-cell responses specific for peptides derived from the mantle cell idiotype immunoglobulin can be activated in healthy individuals. In four patients with MCL treated in the European Mantle Cell Lymphoma Study Group the immunoglobulin heavy chain (IgH) gene family was amplified in lymphoma samples by PCR and sequenced. Using bioinformatics, the corresponding aminoacid sequence was analyzed for nonapeptides potentially binding to the individual HLA-haplotype. Peptides with a Rammensee-score >20 were synthesized. To determine whether these peptides could indeed elicit CD8+ T-cell responses they were used for dendritic cell (DC) pulsation and subsequent T-cell activation. The specificity of the CD8+ T-cells was tested against idiotype-pulsed DC and measured by flow cytometric intracellular interferon (IFN)-gamma staining. The lymphoma specific IgH rearrangements were successfully amplified and sequenced in all patients. In a HLA-A3 positive patient who was in remission after intensive radiochemotherapy and autologous hematopoietic stem cell transplantation three different idiotype HLA-matching peptides with a HLA-A3 binding score >20 were predicted from the VH-region, one additional nonapeptide was overlapping to the N-region of the immunoglobulin, rendering this peptide lymphoma-specific. This pool of peptides was synthesized and used for pulsation of monocyte derived dendritic cells (moDC) in two healthy HLA-A3 positive individuals. The maturation of the DC was done according to a standard protocol using proinflammatory cytokines (IL-6, IL-1 beta, TNF-alpha, PGE2). After 2–3 weekly stimulations of lymphocytes that had been depleted of regulatory T-cells 2.1% idiotype-specific CD8+ T-cells were activated in both healthy donors. Interestingly, T-cell stimulation using moDC matured with CD40− and TLR7/8-ligands was more efficient in comparison to the standard protocol and resulted in 12.3% IFN-gamma positive CD8+ cells. In summary, these data suggest, that idiotype-specific T-cells can be activated from healthy individuals by standard lymphocyte stimulating protocols in vitro. Moreover, the ability of moDC to activate idiotype-specific T-cells is exceeded by DC maturation using CD40− in combination with TLR7/8-ligands. These findings may help to improve immunotherapy in the settings of allogeneic transplantation strategies in relapsed MCL patients.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3047-3047
Author(s):  
Judith Feucht ◽  
Kathrin Opherk ◽  
Cornelia Neinhaus ◽  
Simone Kayser ◽  
Wolfgang A. Bethge ◽  
...  

Abstract Abstract 3047 Allogeneic stem cell transplantation (SCT) can expose patients to a transient but marked immunosuppression, during which viral infections are an important cause of morbidity and mortality. The control of these infections will ultimately depend on the restoration of adequate T-cell immunity. Most viral infections after SCT are caused by endogenous reactivation of persistent pathogens such as cytomegalovirus (CMV), adenovirus (ADV) and Epstein-Barr-virus (EBV). Risk of viral complications is even higher under GvHD treatment or prophylaxis like calcineurin inhibitors and steroids. Post transplant often the immunosuppression needs to be reduced to improve viral complications with the risk of GvHD. The virus-specific T-cell responses in peripheral blood have been shown to be a good marker of immunological protection, but has not been used for clinical decision making and the guidance of drug plasma levels. Therefore, we performed a prospective clinical trial in 33 adult and pediatric patients after allogeneic stem cell transplantation receiving pharmacologic immunosuppression with steroids, Cyclosporin A, Tacrolimus, Everolimus or Mycophenolate. Median Age was 16 years. T-cell responses were analyzed ex vivo against Cytomegalovirus (pp65), Adenovirus (hexon antigen) and Epstein-Barr Virus (EBNA, LMP) using intracellular cytokine staining. In addition in vitro analysis of the proliferation responses using CFSE were performed. Responses were compared to healthy donors. The T-cell responses in vitro under low, high and supraphysiologic plasma concentrations of the respective drugs were investigated. Under the direct influence of steroids, activated, virus-specific T-cells underwent apoptosis. Among the Calcineurin inhibitors, Tacrolimus had the strongest inhibition on virus-specific T-cell immunity, followed by Cyclosporin A. But, under low therapeutic levels, Virus speciffic T-cell responses have been able to develop in PBMCs. Mycophenolate had only in high concentrations a strong effect on the T-cell response against viral pathogens. Relevant differences in the frequency of virus-specific T-cells secreting IFN-g could be detected within the CD4 compartment in correlation to the level of immunosuppression. In conclusion we could show that detection of virus-specific T-cells could be used to guide the level of immunosuppression in case of viral complications after allogeneic stem cell transplantation, since emergence of in vivo T-cell responses was closely associated with a clearance or reduction of the viral load. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (24) ◽  
pp. 5071-5080 ◽  
Author(s):  
Phillip Scheinberg ◽  
Jan J. Melenhorst ◽  
Jason M. Brenchley ◽  
Brenna J. Hill ◽  
Nancy F. Hensel ◽  
...  

Abstract The successful reconstitution of adaptive immunity to human cytomegalovirus (CMV) in hematopoietic stem cell transplantation (HSCT) recipients is central to the reduction of viral reactivation-related morbidity and mortality. Here, we characterized the magnitude, specificity, phenotype, function, and clonotypic composition of CMV-specific T-cell responses in 18 donor-recipient pairs both before and after HSCT. The principal findings were: (1) the specificity of CMV-specific T-cell responses in the recipient after HSCT mirrors that in the donor; (2) the maintenance of these targeting patterns reflects the transfer of epitope-specific T-cell clonotypes from donor to recipient; (3) less differentiated CD27+CD57− CMV-specific memory T cells are more likely to persist in the recipient after HSCT compared with more terminally differentiated CD27− CD57+ CMV-specific memory T cells; (4) the presence of greater numbers of less differentiated CD8+ CMV-specific T cells in the donor appears to confer protection against viral reactivation in the recipient after HSCT; and (5) CMV-specific T cells acquire a more differentiated phenotype and a restricted functional profile after HSCT. Overall, these findings define the immunologic factors that influence the successful adoptive transfer of antigen-specific T-cell immunity during HSCT, which enables the identification of recipients at particular risk of CMV reactivation after HSCT.


2021 ◽  
Vol 9 (9) ◽  
pp. e002754
Author(s):  
Eva Bräunlein ◽  
Gaia Lupoli ◽  
Franziska Füchsl ◽  
Esam T Abualrous ◽  
Niklas de Andrade Krätzig ◽  
...  

BackgroundNeoantigens derived from somatic mutations correlate with therapeutic responses mediated by treatment with immune checkpoint inhibitors. Neoantigens are therefore highly attractive targets for the development of therapeutic approaches in personalized medicine, although many aspects of their quality and associated immune responses are not yet well understood. In a case study of metastatic malignant melanoma, we aimed to perform an in-depth characterization of neoantigens and respective T-cell responses in the context of immune checkpoint modulation.MethodsThree neoantigens, which we identified either by immunopeptidomics or in silico prediction, were investigated using binding affinity analyses and structural simulations. We isolated seven T-cell receptors (TCRs) from the patient’s immune repertoire recognizing these antigens. TCRs were compared in vitro by multiparametric analyses including functional avidity, multicytokine secretion, and cross-reactivity screenings. A xenograft mouse model served to study in vivo functionality of selected TCRs. We investigated the patient’s TCR repertoire in blood and different tumor-related tissues over 3 years using TCR beta deep sequencing.ResultsSelected mutated peptide ligands with proven immunogenicity showed similar binding affinities to the human leukocyte antigen complex and comparable disparity to their wild-type counterparts in molecular dynamic simulations. Nevertheless, isolated TCRs recognizing these antigens demonstrated distinct patterns in functionality and frequency. TCRs with lower functional avidity showed at least equal antitumor immune responses in vivo. Moreover, they occurred at high frequencies and particularly demonstrated long-term persistence within tumor tissues, lymph nodes and various blood samples associated with a reduced activation pattern on primary in vitro stimulation.ConclusionsWe performed a so far unique fine characterization of neoantigen-specific T-cell responses revealing defined reactivity patterns of neoantigen-specific TCRs. Our data highlight qualitative differences of these TCRs associated with function and longevity of respective T cells. Such features need to be considered for further optimization of neoantigen targeting including adoptive T-cell therapies using TCR-transgenic T cells.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3269-3269
Author(s):  
Anniek B. van der Waart ◽  
Noortje van der Weem ◽  
Luca Gattinoni ◽  
Nicolaas PM Schaap ◽  
Robbert van der Voort ◽  
...  

Abstract Allogeneic hematopoietic stem cell transplantation (allo-SCT) followed by donor lymphocyte infusion (DLI) is a potential curative treatment for patients suffering from a hematological malignancy. Efficacy is attributed to the graft-versus-tumor (GVT) response, during which engrafted donor T cells become activated by recipient minor histocompatibility antigens (MiHA) presented on dendritic cells (DC). Subsequently, these activated T cells expand, acquire effector functions and kill MiHA-positive tumor cells. However, persistence and recurrence of malignant disease is often observed, indicating that insufficient GVT immunity is induced. This imperfect alloreactive response is probably due to insufficient numbers of MiHA-specific effector T cells and/or defective antigen-presentation and costimulation. Therefore, adoptive transfer of potent ex vivo-generated MiHA-specific T cells, restricted to the hematopoietic system, would boost the GVT-effect without increasing the risk for GVHD. Although successful in vitro induction of MiHA-specific CD8+ T cells from naive precursors has been reported, the resulting antigen-experienced T cell population consist of fully differentiated effector-memory T cells (TEM). Over the past years it has been described that this T cell subset is not the most potent memory subset in anti-tumor responses in vivo following T cell transfer. In this regard, the less-differentiated memory subset called stem cell memory T cells (TSCM) with superior in vivo expansion, self-renewal capacity and plasticity to differentiate in potent effectors would generate a stronger GVT response. In this study, we aimed to investigate the in vivo availability and ex vivo generation of TSCM-like MiHA-specific T cells as additive treatment option for allo-SCT patients. First, we investigated whether in allo-SCT patients MiHA-specific T cells could be detected with a TSCM phenotype defined by the expression of CD45RO, CCR7, CD27 and CD95. Though TSCM cells could be clearly detected within CMV-specific CD8+ T cells in allo-SCT patients, similar to healthy controls, no MiHA-specific TSCM cells could be detected. This emphasises the need for more potent adoptive MiHA-specific T cell therapy following allo-SCT. Therefore, we next explored the possibility of generating TSCM-like CD8+ T cells by interfering with the Akt signalling pathway. Emerging findings indicate that the differentiation program of CD8+ T cells is dictated by the strength and duration of AKT activity. Therefore, we explored whether the pharmacological inhibition of this signaling pathway could results in the generation of TSCM-like CD8+ T cells. We stimulated CCR7+CD45RA+ naive CD8+ T cells with CD3/CD28 beads plus IL-2, IL-7 and/or IL-15 in the presence an Akt inhibitor. Interestingly, CD8+ T cells in these Akt-cultures were inhibited in their differentiation stage, expressing higher levels of CD45RA and CCR7 compared to controls. In addition, expression of CD95, IL2Rβ, and IL7Rα was also elevated confirming the TSCM-like phenotype. Although proliferation of the Akt-inhibited CD8+ T cells was decreased as shown by less PBSE dilution, expansion could be significantly preserved. Next, we investigated whether the established culture conditions could be used to generate MiHA-specific TSCM-like cells. Therefore, CD8+ T cells from MiHA-negative donors were primed using autologous MiHA peptide-loaded moDCs in the presence of the Akt-inhibitor. Interestingly, MiHA-specific T cell priming could be induced, consisting of mainly TCM and TSCM-like cells compared to almost entirely TEM cells in the control setting. Akt-inhibited MiHA-specific T cells showed higher expression of CCR7, CD45RA, CD62L, CD28, CD95, and IL7Rα. Importantly, for the Akt-inhibited MiHA-specific T cells, proliferation was reserved, resulting in robust proliferation capacity during restimulation after removal of the Akt-inhibitor. The resulting TEFF cells were highly functional, showing capacity to degranulate and produce IFNγ upon peptide restimulation. In conclusion, by inhibiting the Akt-pathway, in vitro CD8+ T cell differentiation can be reduced. Therefore, Akt signalling inhibition can be exploited for generating TSCM-like MiHA-specific T cells in adoptive immunotherapy after allo-SCT. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Jae Hyung Jung ◽  
Min-Seok Rha ◽  
Moa Sa ◽  
Hee Kyoung Choi ◽  
Ji Hoon Jeon ◽  
...  

AbstractMemory T cells contribute to rapid viral clearance during re-infection, but the longevity and differentiation of SARS-CoV-2-specific memory T cells remain unclear. We conducted direct ex vivo assays to evaluate SARS-CoV-2-specific CD4+ and CD8+ T cell responses in COVID-19 convalescents up to 254 days post-symptom onset (DPSO). Here, we report that memory T cell responses were maintained during the study period. In particular, we observed sustained polyfunctionality and proliferation capacity of SARS-CoV-2-specific T cells. Among SARS-CoV-2-specific CD4+ and CD8+ T cells detected by activation-induced markers, the proportion of stem cell-like memory T (TSCM) cells increased, peaking at approximately 120 DPSO. Development of TSCM cells was confirmed by SARS-CoV-2-specific MHC-I multimer staining. Considering the self-renewal capacity and multipotency of TSCM cells, our data suggest that SARS-CoV-2-specific T cells are long-lasting after recovery from COVID-19. The current study provides insight for establishing an effective vaccination program and epidemiological measurement.


2021 ◽  
Vol 22 (1) ◽  
pp. 459
Author(s):  
Asmaa A. Zidan ◽  
Mohammed Al-Hawwas ◽  
Griffith B. Perkins ◽  
Ghada M. Mourad ◽  
Catherine J. M. Stapledon ◽  
...  

Elucidation of the biological functions of extracellular vesicles (EVs) and their potential roles in physiological and pathological processes is an expanding field of research. In this study, we characterized USC–derived EVs and studied their capacity to modulate the human immune response in vitro. We found that the USC–derived EVs are a heterogeneous population, ranging in size from that of micro–vesicles (150 nm–1 μm) down to that of exosomes (60–150 nm). Regarding their immunomodulatory functions, we found that upon isolation, the EVs (60–150 nm) induced B cell proliferation and IgM antibody secretion. Analysis of the EV contents unexpectedly revealed the presence of BAFF, APRIL, IL–6, and CD40L, all known to play a central role in B cell stimulation, differentiation, and humoral immunity. In regard to their effect on T cell functions, they resembled the function of mesenchymal stem cell (MSC)–derived EVs previously described, suppressing T cell response to activation. The finding that USC–derived EVs transport a potent bioactive cargo opens the door to a novel therapeutic avenue for boosting B cell responses in immunodeficiency or cancer.


Vaccine ◽  
2010 ◽  
Vol 28 (13) ◽  
pp. 2556-2564 ◽  
Author(s):  
Erik Jongert ◽  
Arnaud Lemiere ◽  
Jo Van Ginderachter ◽  
Stéphane De Craeye ◽  
Kris Huygen ◽  
...  

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1097-1097
Author(s):  
Clemence Roux ◽  
Gaelle Saviane ◽  
Gihen Dhib ◽  
Jonathan Pini ◽  
Pierre-Simon Rohrlich ◽  
...  

Abstract Background One major problem of allogeneic hematopoietic stem cell transplantation (HSCT) is acute Graft versus Host Disease (aGVHD). aGVHD has been managed until now with HLA matching and a constant evolving repertoire of immunosuppressive drugs. One alternative would be to generate in the host a permanent tolerance state toward the graft. Tolerant inducing cell therapy has been proposed with adult mesenchymal stromal (MSCs) cells. Ex vivo isolated somatic MSCs have been implicated in immunoregulatory functions on cells from both the innate and adaptive immune system. They were proposed for cell therapies for the treatment of aGVHD, Nevertheless, their use is restricted because of the few number that can be recovered from adult tissues, their limited in vitro expansion, and the absence of a full phenotypic characterization. Therefore other sources of well-defined and unlimited number of MSCs are needed, and MSCs derived in vitro from human Induced pluripotent stem cell (huIPS) would be a valuable tool for therapeutic approaches. Aims Because of our expertise in pluripotent stem cell differentiation, we generated huiPS-MSCs that present a strong immunosuppressive activity on allogeneic T cell responses. Our objectives are: 1/ To evaluate and characterize in vitro this immunosuppression. 2/ To validate in vivo these results using a xenoGVHD model. Methods To characterize the huIPS-MSCs in vitro, FACS phenotyping and multipotency were tested. Their immunogenicity in vitro was monitored in co-cultures with allogenic peripheral blood mononuclear cells (PBMC). The in vivo immunosuppressive activity of huiPS-MSCs was evaluated using a xenoGVHD model in immunodeficient NSG (NOD/SCID/IL2rγKO) mice in which human PBMC were injected intra-peritonally. We established 3 groups : 1) huIPS-MSCs (control) 2) PBMC 3) PBMC + IPS-MSCs. We repeated huIPS-MSCs injection weekly with median number of injection n=3 (range 2-3). The activation state of human allogeneic T lymphocytes recovered from mice between 5 to 8 weeks after initial injection was evaluated and indicated the level of the xenoGVHD process and the efficiency of huiPS-MSCs to prevent it. Results a) In vitro characterization of huIPS-MSCs As expected, the huiPS-MSCs were positive for CD73, CD90, CD105, HLA-I Ags and negative for CD45, CD34, HLA-II Ags and they were capable of differentiation into the classical mesenchymal-derived cells (osteoblast, chondrocytes and adipocytes). To test their immunosuppressive properties, we analyzed their action on the proliferation of human T lymphocytes stimulated in an allogeneic manner (Fig 1). The stimulation of PBMC in mixed lymphocyte reaction resulted in CD4 and CD8 T cell proliferation (28 ± 7% and 47 ± 8%, respectively), which was significantly reduced in co-culture with huiPS-MSCs (4 ± 2% and 10 ± 2, respectively, n=3 p<0,05). We were able to demonstrate using blocking antibodies that part of the inhibition exerted by the iPS-MSCs is due to a) B7H1, a membrane receptor for the B7 family, known for its inhibitory action on the activation of T lymphocytes b) and B7H3 (from the same family) whose role remains controversial. b) In vivo characterization of huIPS-MSCs After sacrifice of mice, human circulating cells, those present in the peritoneal cavity and in the spleen were analysed by FACS. Mostly T lymphocytes were detected, and their number was significantly reduced in mice treated with huIPS-MSCs p<0,05 (Fig 2). Intracytoplasmic labelling of recovered T cells showed that untreated mice displayed high percentages of human differentiated T cells producing IFN  and TNF  (typical of a inflammatory Th1 cytokine polarization profile), while little or none produced low inflammatory (IL-4) or anti-inflammatory (IL-10) cytokines. In contrast, in mice treated with the huiPS-MSCs, the proportion of T cells of the Th1 type was substantially reduced, while that of T cells producing IL-4 and / or IL-10 was slightly increased (Fig 3). In parallel, T cells expressing FoxP3 appeared . Conclusion We were able to generate immune-modulatory huiPS-MSCs that can be used to reduce activation of T cells in a xeno-aGVHD model through a switch from a Th1 inflammatory differentiation pathway to a T cell regulatory pathway. Our results may favor the development of new tools and strategies based on the use of pluripotent stem cells and their derivatives to prevent aGVHD but also for the induction of specific tolerance. Figure 1 Figure 1. Figure 2 Figure 2. Figure 3 Figure 3. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Jae Hyung Jung ◽  
Min-Seok Rha ◽  
Moa Sa ◽  
Hee Kyoung Choi ◽  
Ji Hoon Jeon ◽  
...  

AbstractMemory T cells contribute to rapid viral clearance during re-infection, but the longevity and differentiation of SARS-CoV-2-specific memory T cells remain unclear. Here we conduct ex vivo assays to evaluate SARS-CoV-2-specific CD4+ and CD8+ T cell responses in COVID-19 convalescent patients up to 317 days post-symptom onset (DPSO), and find that memory T cell responses are maintained during the study period regardless of the severity of COVID-19. In particular, we observe sustained polyfunctionality and proliferation capacity of SARS-CoV-2-specific T cells. Among SARS-CoV-2-specific CD4+ and CD8+ T cells detected by activation-induced markers, the proportion of stem cell-like memory T (TSCM) cells is increased, peaking at approximately 120 DPSO. Development of TSCM cells is confirmed by SARS-CoV-2-specific MHC-I multimer staining. Considering the self-renewal capacity and multipotency of TSCM cells, our data suggest that SARS-CoV-2-specific T cells are long-lasting after recovery from COVID-19, thus support the feasibility of effective vaccination programs as a measure for COVID-19 control.


Sign in / Sign up

Export Citation Format

Share Document