Ex-Vivo Analysis of CD8+ T Cells Infiltrating Colorectal Tumors Identifies a Major Effector-Memory Subset with Low Perforin Content

2006 ◽  
Vol 26 (5) ◽  
pp. 447-456 ◽  
Author(s):  
SHENG-WEI YE ◽  
YU WANG ◽  
DANILA VALMORI ◽  
MAHA AYYOUB ◽  
YAN HAN ◽  
...  
2019 ◽  
Vol 5 (3) ◽  
pp. 63
Author(s):  
Alice Bayiyana ◽  
Samuel Okurut ◽  
Rose Nabatanzi ◽  
Godfrey Zziwa ◽  
David R. Boulware ◽  
...  

Despite improvement in the prognosis of HIV/AIDS (human immunodeficiency virus/acquired immune deficiency syndrome) patients on antiretroviral therapy (ART), cryptococcal meningitis (CM) still causes 10–15% mortality among HIV-infected patients. The immunological impact of ART on the CD4+ and CD8+ T cell repertoire during cryptococcal co-infection is unclear. We determined longitudinal phenotypic changes in T cell subsets among patients with CM after they initiated ART. We hypothesized that ART alters the clonotypic phenotype and structural composition of CD4+ and CD8+ T cells during CM co-infection. For this substudy, peripheral blood mononuclear cells (PBMC) were isolated at four time points from CM patients following ART initiation during the parent study (ClinicalTrials.gov number, NCT01075152). Phenotypic characterization of CD4+ and CD8+ T cells was done using T cell surface marker monoclonal antibodies by flow cytometry. There was variation in the expression of immunophenotypic markers defining central memory (CD27+CD45R0+), effector memory (CD45R0+CD27–), immune activation (CD38+ and Human Leucocyte Antigen DR (HLA-DR+), and exhaustion (Programmed cell death protein one (PD-1) in the CD4+ T cell subset. In comparison to the CD4+ T cell population, the CD8+ central memory subset declined gradually with minimal increase in the effector memory subset. Both CD4+ and CD8+ T cell immune exhaustion and activation markers remained elevated over 12 weeks. The relative surge and decline in the expression of T cell surface markers outlines a variation in the differentiation of CD4+ T cells during ART treatment during CM co-infection.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1071-1071
Author(s):  
Melody M. Smith ◽  
Cynthia R. Giver ◽  
Edmund K. Waller ◽  
Christopher R. Flowers

Abstract Ex vivo modification of donor lymphocytes with purine analogs (mDL) may help to minimize graft versus host disease (GvHD) while providing beneficial graft versus leukemia (GvL) effects. In a murine model system, we have shown that allogeneic donor splenocytes, treated with fludarabine ex vivo have significantly reduced GvHD activity when transferred to irradiated recipient mice, and retain anti-viral and GvL activities (Giver, 2003). This effect appears to be mediated by relative depletion of donor CD4 CD44low, “naive” T-cells. As a first step toward developing mDL for use in patients, we sought to evaluate the effects of ex vivo fludarabine exposure on human T-cell subsets, and to determine the minimum dose of fludarabine required to achieve this effect. Methods: Peripheral blood mononuclear cell samples from 6 healthy volunteers were evaluated at 0, 24, 48, and 72 hour time points after ex vivo incubation in varying dosages of fludarabine: 2, 5, and 10(n=3) mcg/ml. Fludarabine incubated samples were compared to samples that received no fludarabine (untreated). The total viable cell number was determined and the fractions and absolute numbers of viable CD4 and CD8 naïve and memory T-cells were determined using flow cytometry after incubation with 7-AAD (dead cell stain), CD4, CD8, CD45RA, CD62L, and CCR7 antibodies, and measuring the total viable cells/ml. Results: The numbers of viable CD4 and CD8 T-cells remained relatively stable in control cultures. Without fludarabine, the average viability at 72 hr of naive and memory T-cells were 92% and 77% for CD4 and 86% and 63% for CD 8 (Fig. 1A). Naive CD4 T-cells were more sensitive to fludarabine-induced death than memory CD4 cells. At 72 hr, the average viability of fludarabine-treated naive CD4 T-cells was 33% at 2 mcg/ml (8.2X the reduction observed in untreated cells) and 30% at 5 mcg/ml, while memory CD4 T-cells averaged 47% viability at 2 mcg/ml (2.3X the reduction observed in untreated cells) (Fig. 1B) and 38% at 5 mcg/ml. The average viability of naive CD8 T-cells at 72 hr was 27% at 2 mcg/ml and 20% at 5 mcg/ml, while memory CD8 T-cell viability was 22% at 2 mcg/ml and 17% at 5 mcg/ml. Analyses on central memory, effector memory, and Temra T-cells, and B-cell and dendritic cell subsets are ongoing. The 5 and 10 mcg/ml doses also yielded similar results in 3 initial subjects, suggesting that 2 mcg/ml or a lower dose of fludarabine is sufficient to achieve relative depletion of the naive T-cell subset. Conclusions: Future work will determine the minimal dose of fludarabine to achieve this effect, test the feasibility of using ex vivo nucleoside analog incubation to reduce alloreactivity in samples from patient/donor pairs, and determine the maximum tolerated dose of mDL in a phase 1 clinical trial with patients at high risk for relapse and infectious complications following allogeneic transplantation. Figure Figure


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3268-3268 ◽  
Author(s):  
Anne Richter ◽  
Liane Preussner ◽  
Verena Traska ◽  
Michaela Peters ◽  
Ayse Oysal ◽  
...  

Abstract Introduction Adoptive transfer of virus-specific T cells is an encouraging strategy to manage severe and fatal infections in immunocompromised patients. To generalize this approach, a cGMP- compliant enrichment process of both CD4+ and CD8+ viral-specific T cells is necessary. Here, we used a newly established automated manufacturing process for rapid and efficient ex vivo selection of multi-virus-specific CD4+ and CD8+ T cells. We show how the isolated virus-specific T cells retain their original effector/memory status and their effector functions. Method Leukapheresis from healthy donors were used as starting material. Multi-virus or cytomegalovirus pp65 peptide-specific T cell products were generated in a novel closed cell-processing device with a fully automated manufacturing procedure. During this process white blood cells were stimulated with either a combination of peptide pools covering cytomegalovirus pp65, Epstein-Barr-Virus EBNA-1, BZLF1, and LMP2, and adenovirus hexon protein (n=6) or with a single pp65 peptide for four hours (n=4). Subsequently, virus-specific CD4+ and CD8+ T cells were magnetically enriched using the IFN-g secretion assay technology. In parallel, the reversible MHC/peptide multimer technology, which is restricted to CD8+ peptide-specific T cell enrichment, was used for comparison in a manual magnetic selection procedure for pp65 peptide-specific CD8+ T cells (n=4). All virus-specific T cell products were rested in vitro in the presence of T-cell-depleted PBMCs without addition of cytokines or antigens for up to 4 days. Expression of CD45RA, CCR7, CD28, CD69, CD137 as well as IFN-g production with and without cognate antigen(s)-restimulation were analyzed by flow cytometry before and up to 4 days after the selection process. Results Manufacturing of multi-virus and pp65 peptide-specific T cells using the IFN-g secretion assay technology requires a short period of antigen stimulation and IFN-g expression, therefore, up to 96% of T cells produced IFN-g in the enriched fraction. However, after a few days resting phase in culture, IFN-g production decreased drastically. In addition, we detected an upregulation of CD69 and CD137 in the IFN-g enriched T cell products directly and 24 hours after the selection process, respectively. The transient nature of activation could again be confirmed, as both, CD69 and CD137 were downregulated during the resting phase. Results were compared to pp65-peptide specific CD8+ T cell products generated by the MHC/peptide multimer technology, which does not require an antigen incubation step. Activation was also seen for these enriched T cells, even when the MHC/peptide complexes were released, while unprocessed and cultured PBMC did not show IFN-g secretion or activation marker expression; indicating that cell processing and not the culture conditions triggered the activation. To test the functionality of the generated T cell products, we re-incubated three days resting cells with the corresponding antigens. In all samples, independent of the technology used for selection, induction of IFN-g expression in up to 100% of T cells was observed. Thus, T cells in all the products were able to maintain their in vivo imprinted physiological role, i.e. IFN-g production after antigen contact. Furthermore we examined if cell processing influences the effector/memory status of virus-specific T cells. Because the MHC/peptide multimer technology is restricted to the selection of single peptide-specific CD8+ T cells only, we monitored CD45RA, CD28 and CCR7 expression on pp65-peptide specific CD8+ T cells either identified by IFN-g secretion or by MHC/peptide multimer staining before and directly after the enrichment. The frequency of CD45RA+ and CD28+ cell populations varied between the donors and CCR7 was not detected at all, but importantly the enrichment process did not induce phenotypic changes. This result demonstrates the phenotype is stable during the manufacturing process. Conclusion A newly developed automated manufacturing process for direct ex vivo enrichment of multi-virus-specific CD4+ and CD8+ T cell populations via the IFN-g secretion assay technology provides a product for immunotherapy, where the original phenotypic and functional characteristics of the cells are conserved. Hence this cellular product is expected to fight efficiently against viral infections upon adoptive transfer. Disclosures: Richter: Miltenyi Biotec GmbH: Employment. Preussner:Miltenyi Biotec: Employment. Traska:Miltenyi Biotec: Employment. Peters:Miltenyi Biotec: Employment. Oysal:Miltenyi Biotec: Employment. Ruhnke:Miltenyi Biotec: Employment. Brauns:Miltenyi Biotec: Employment. Kramer:Miltenyi Biotec: Employment. Schmitz:Miltenyi Biotec: Employment. Assenmacher:Miltenyi Biotec: Employment.


Blood ◽  
2004 ◽  
Vol 104 (2) ◽  
pp. 478-486 ◽  
Author(s):  
Michelina Nascimbeni ◽  
Eui-Cheol Shin ◽  
Luis Chiriboga ◽  
David E. Kleiner ◽  
Barbara Rehermann

Abstract Although an increased frequency of CD4+CD8+ T cells has been observed in the peripheral blood during viral infections, their role, function, and biologic significance are still poorly understood. Here we demonstrate that the circulating CD4+CD8+ T-cell population contains mature effector memory lymphocytes specific for antigens of multiple past, latent, and high-level persistent viral infections. Upon in vitro antigenic challenge, a higher frequency of CD4+CD8+ than single-positive cells displayed a T helper 1/T cytotoxic 1 (Th1/Tc1) cytokine profile and proliferated. Ex vivo, more double-positive than single-positive cells exhibited a differentiated phenotype. Accordingly, their lower T-cell receptor excision circles (TREC) content and shorter telomeres proved they had divided more frequently than single-positive cells. Consistent with expression of the tissue-homing marker CXCR3, CD4+CD8+ T cells were demonstrated in situ at the site of persistent viral infection (ie, in the liver during chronic hepatitis C). Finally, a prospective analysis of hepatitis C virus (HCV) infection in a chimpanzee, the only animal model for HCV infection, showed a close correlation between the frequency of activated CD4+CD8+ T cells and viral kinetics. Collectively, these findings demonstrate that peripheral CD4+CD8+ T cells take part in the adaptive immune response against infectious pathogens and broaden the perception of the T-cell populations involved in antiviral immune responses. (Blood. 2004;104:478-486)


Blood ◽  
2003 ◽  
Vol 101 (3) ◽  
pp. 1024-1029 ◽  
Author(s):  
Yvonne M. Mueller ◽  
Paul M. Bojczuk ◽  
E. Scott Halstead ◽  
Alfred H. J. Kim ◽  
James Witek ◽  
...  

AbstractHIV-specific CD8+ T cells are prone to undergo apoptosis, and this may affect their ability to control HIV infection. Because CD8-mediated immune responses play a key role in controlling HIV infection, enhancing the survival and effector function of HIV-specific CD8+ T cells may augment their ability to control HIV virus. We show here that interleukin 15 (IL-15) potently inhibits spontaneous and CD95/Fas-induced apoptosis of HIV-specific CD8+ T cells. IL-15 inhibits apoptosis in both CD45RA−CD62L− and CD45RA+CD62L− effector memory subpopulations of these cells. Furthermore, IL-15 greatly enhances the survival of HIV-specific CD8+ T cells in long-term cultures. Finally, IL-15 directly enhances activation, interferon γ (IFNγ) production, and direct ex vivo cytotoxicity of HIV-specific CD8+ T cells. Thus, IL-15 potently enhances the survival and effector function of HIV-specific CD8+ T cells and, therefore, may prove useful in augmenting the antiviral function of these cells.


Author(s):  
Jennifer R Habel ◽  
Thi H O Nguyen ◽  
Carolien E van de Sandt ◽  
Jennifer A Juno ◽  
Priyanka Chaurasia ◽  
...  

An improved understanding of human T-cell-mediated immunity in COVID-19 is important if we are to optimize therapeutic and vaccine strategies. Experience with influenza shows that infection primes CD8+ T-cell memory to shared peptides presented by common HLA types like HLA-A2. Following re-infection, cross-reactive CD8+ T-cells enhance recovery and diminish clinical severity. Stimulating peripheral blood mononuclear cells from COVID-19 convalescent patients with overlapping peptides from SARS-CoV-2 Spike, Nucleocapsid and Membrane proteins led to the clonal expansion of SARS-CoV-2-specific CD8+ and CD4+ T-cells in vitro, with CD4+ sets being typically robust. For CD8+ T-cells taken directly ex vivo, we identified two HLA-A*02:01-restricted SARS-CoV-2 epitopes, A2/S269-277 and A2/Orf1ab3183-3191. Using peptide-HLA tetramer enrichment, direct ex vivo assessment of the A2/S269+CD8+ and A2/Orf1ab3183+CD8+ populations indicated that the more prominent A2/S269+CD8+ set was detected at comparable frequency (1.3x10-5) in acute and convalescent HLA-A*02:01+ patients. But, while the numbers were higher than those found in uninfected HLA-A*02:01+ donors (2.5x10-6), they were low when compared with frequencies for influenza-specific (A2/M158) and EBV-specific (A2/BMLF1280) (1.38x10-4) populations. Phenotypic analysis ex vivo of A2/S269+CD8+ T-cells from COVID-19 convalescents showed that A2/S269+CD8+ T-cells were predominantly negative for the CD38, HLA-DR, PD-1 and CD71 activation markers, although the majority of total CD8+ T-cells were granzyme and/or perforin-positive. Furthermore, the bias towards naive, stem cell memory and central memory A2/S269+CD8+ T-cells rather than effector memory populations suggests that SARS-CoV2 infection may be compromising CD8+ T-cell activation. Priming with an appropriate vaccine may thus have great value for optimizing protective CD8+ T-cell immunity in COVID-19.


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.


Blood ◽  
2007 ◽  
Vol 110 (13) ◽  
pp. 4360-4366 ◽  
Author(s):  
Edward C. P. Waller ◽  
Nicola McKinney ◽  
Ray Hicks ◽  
Andrew J. Carmichael ◽  
J. G. Patrick Sissons ◽  
...  

In healthy carriers of human cytomegalovirus (HCMV), the virus-specific memory CD8+ T-cell population is often dominated by CD28− CD45RAhi cells that exhibit direct ex vivo cytotoxicity but whose capacity for proliferation and generation of further memory cells has been questioned. We show that when highly purified CD28− CD45RAhi CD8+ T cells are stimulated with viral peptide presented by autologous monocytes, the virus-specific T cells show early up-regulation of CD137 (4–1BB) and CD278 (ICOS), re-express CD28, and proliferate with similarly high cloning efficiency in limiting dilution analysis as CD28+ CD45ROhi cells or CD28− CD45ROhi cells. Using peptide-pulsed autologous fibroblasts transfected with individual costimulatory ligands as antigen presenting cells, we showed CD137L to be a key costimulatory ligand for proliferation of CD28− CD45RAhi CD8+ T cells and not CD80, CD86, or CD275 (ICOSL). Therefore, CD28− CD45RAhi CD8+ T cells were not terminally differentiated but required a specific costimulatory signal for proliferation.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3048-3048
Author(s):  
Yi Zhang ◽  
Gerard Joe ◽  
Elizabeth Hexner ◽  
Stephen G. Emerson

Abstract Memory CD8+ T cells are an important component of long-term immunity against infectious pathogens because of their higher frequency of antigen-specific CD8+ T cells as well as their ability to proliferate, produce inflammatory cytokines, and kill target cells more rapidly upon secondary antigen encounter than naïve CD8+ T cells. How the pool of memory CD8+ T cells is generated and maintained is key issue to understanding and perhaps manipulating long-term memory response, such as graft-versus-host disease (GVHD) where host antigens persist. Using a major histocompatibility complex (MHC)-identical but minor histocmpatibility (miHA)-mismatched mouse model of human allogeneic BM transplantation (allo-BMT), we recently identified alloreactive memory CD8+ T cells responsible for persistent GVHD. We found that donor CD44hiCD62Llo effector memory and CD44hiCD62Lhi central memory CD8+ T cells recovered 42 days after allo-BMT (d42-CD8+ T cells) from B6 mice receiving normal C3H.SW CD44loCD8+ T cells and T cell-depleted (T−BM) caused lethal GVHD in secondary B6 recipient mice. Interestingly, in addition to these classical memory phenotypes, a third population of donor CD44loCD62LhiCD8+ T cells, which accounted for 2% to 6% of whole d42-donor CD8+ T cells, was identified in the spleens and livers of these B6 recipients with ongoing GVHD. When cultured in the presence of B6 dendritic cells (DCs)+IL-2+IL-15, these d42-CD44loCD62LhiCD8+ T cells rapidly and vigorously proliferated as compared to d42-CD44hiCD62Llo and d42-CD44hiCD62LhiCD8+ T cells. By day 15 following this ex vivo culture, d42-CD44loCD62LhiCD8+ T cells expanded as many as 92.0-fold, whereas CD44hiCD62Llo effector/effector memory and CD44hiCD62Lhi central memory CD8+ T cells only expanded 2.1-fold and 11.0-fold, respectively. Furthermore, ex vivo stimulation of d42-CD44hiCD62LloCD8+ T cells with B6 DCs+IL-2+IL-15 only induced the generation of CD44hiCD62Llo effector/effector memory CD8+ T cells, whereas d42-CD44hiCD62hiCD8+ T cells generated both CD44hiCD62Llo and CD44hiCD62Lhi cells. In contrast, d42-CD44loCD62LhiCD8+ T cells generated all three T cells subsets, e.g., CD44hiCD62Llo, CD44hiCD62Lhi and CD44loCD62Lhi CD8+ T cells. These data suggest that d42-CD44loCD62Lhi CD8+ T cells have more potent ability than any other CD8+ T memory cell subsets to proliferate and differentiate into effector/memory T cells upon re-exposure to specific host miHAs as well as the ability to self-renew, resembling to the property of stem cells. Of note, these d42-CD44loCD62Lhi CD8+ T cells expressed much higher levels of CD122 and CD127 than donor naive CD44loCD8+ T cells. When cultured in the presence of B6 DCs+IL-2+IL-15 for 5 days, there were significantly more donor CD8+ T cells recovered from d42-CD4loCD62LhiCD8+ T cell cultures than that from truly naive CD44loCD8+ T cells(7.9-fold vs. 2.1 fold). Thus, d42-CD44loCD62LhiCD8+ T cells found in B6 mice with GVHD are distinguishable from truly naïve CD44loCD8+ T cells of normal C3H.SW mice and are responsible for sustaining the generation of both alloreactive effector memory and central memory CD8+ T cells. In summary, these data identify a heretofore unrecognized population of CD8+ T memory stem cells that may be key cellular targets for the prevention and treatment of persistent acute and chronic GVHD.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A19.2-A20
Author(s):  
C Ripamonti ◽  
C Steinkuhler ◽  
G Fossati

BackgroundCentral memory T cells show superior persistence and antitumor immunity compared to effector memory and effector T cells. T effector cells respond quickly to tumors, but they are terminally differentiated and undergo apoptosis upon killing activity. T memory differentiate rapidly into T effector cells and maintain a pool of cells that can continuously differentiate thus sustaining a more lasting response. In adoptive cell therapy (ACT), T cells infused into patients may have a limited time of activity if they are terminally differentiated, and may rapidly undergo exhaustion and apoptosis. The development of new strategies based on novel agents able to generate memory T cells ex-vivo is important for a successful clinical application of ACT.We have studied the effect of a potent and selective HDAC6 inhibitor, ITF3756, on CD8 T cells differentiation during an in vitro induced exhaustion process.Materials and MethodsTo induce exhaustion purified human CD8+ cells were stimulated twice with anti-CD3/CD28 beads (1:2) during 5 days, with or without ITF3756 1μM or 2μM added at all times of stimulation. At day 3 and 5 the expression of exhaustion, memory and effector T cells markers were analyzed by flow cytometry. Cells were also collected at day 5 for genes expression analysis. Expression of exhaustion, T phenotype, metabolic pathway and inflammatory cytokines were investigated by qPCR. Paired two-tailed t-tests was used to determine statistical significance between control versus treatment group at day 3 and 5 in 10 different donors. P-values ≤ 0.05 were considered significant.ResultsITF3756 1μM increased significantly the T central memory phenotype (CD45RO+CD62L+CCR7+) and decreased significantly the T effector phenotype (CD45RO+CD62L-CCR7-). The expression of CD62L in T central memory cells was significantly increased in agreement with the high expression of this marker in naïve and memory T cells. ITF3756 treatment decreased significantly the expression of exhaustion markers PD-1 and LAG-3. No effect was observed on TIM-3 expression. In agreement with the data obtained with protein analysis, treatment with ITF3756 reduced the mRNA level of Pd-1 and Lag-3. Gene expression of Tim-3 was also downmodulated, but this effect did not result in reduction of protein expression at the time of detection. ITF3756 reduced the expression of t-bet (Tbx21) driving T effector differentiation and increased genes related to T memory phenotype (Eomes, Lef-1 and albeit slightly, Tcf-7). T cell activation requires a metabolic reprogramming that supports highly proliferative phenotype and T effector differentiation. ITF3756 treatment decreased both Hif-1α and Glut-1 gene expression that are associated with TCR activation during the exhaustion process. T central memory cells produce less cytokines compared to T effector and effector memory cells. ITF3756 treatment decreased the genes expression of Il-2, Ifn-γ and Tnf-α. All these effects resulted dose dependent.ConclusionsThe selective inhibitor of HDAC6 ITF3756 delays the terminal differentiation of CD8 T cells and increases the percentage of memory T cells with a reduced expression of exhaustion markers in vitro. These results are the basis to further explore the possible use of ITF3756 as a safe ex vivo treatment of CD8 T cells for adoptive cell transfer.Disclosure InformationC. Ripamonti: A. Employment (full or part-time); Significant; Italfarmaco SpA. C. Steinkuhler: A. Employment (full or part-time); Significant; Italfarmaco SpA. G. Fossati: A. Employment (full or part-time); Significant; Italfarmaco SpA.


Sign in / Sign up

Export Citation Format

Share Document