Positive Impact of T-Cell Clonal Expansion On Overall Survival in Patients with High-Risk Myelodysplastic Syndromes.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1572-1572 ◽  
Author(s):  
Amber Schmidt ◽  
Rami S. Komrokji ◽  
Jeffrey S. Painter ◽  
Dana E Rollison ◽  
Ling Zhang ◽  
...  

Abstract Abstract 1572 Poster Board I-598 Background Suppression of clonal T-cells has been linked to immunosuppressive therapy response in patients with lower-risk MDS and with other forms of autoimmune bone marrow failure suggesting that T-cell clonal expansion is pathogenic to bone marrow hematopoiesis. It is possible, however, that clonal T-cells expanding in response to leukemia-associated antigens (LAA) may ultimately suppress tumor progression through immunosurveillance. Our group previously identified clonal T-cell expansion in 50% of MDS patients (n=52) compared to 5% in age matched control (n=20) (results published by Epling-Burnette et al, Leukemia 21:659, 2007). There was no statistically significant association between clonal T cell expansion and WHO subtype, IPSS, karyotype, transfusion dependency, age or gender in a cross-sectional analysis. Identification of the prognostic importance of clonal T-cell expansion is an important decision point concerning immunosuppressive therapies. We report here on overall survival for both lower and higher-risk MDS patients based on clonal T-cell expansion status. Materials and Methods The original study enrolled 52 patients diagnosed with MDS and 20 healthy volunteers. Peripheral mononuclear cells were isolated and clonal T cells were identified through analysis of the complementarity determining region (CDR)-3 of the T-cell receptor (TCR) using multiplex TCR-Vβ(CDR3) PCR on genomic DNA and by flow-cytometric analysis of Vβ expanded families. In this study, we retrospectively reviewed records of the 52 patients identified previously after obtaining IRB approval. We compared outcome of patients with evidence of clonal T-cell expansion in the peripheral blood to patients without clonal T-cell expansion. Data collected included demographic characteristics, WHO subtype, cytopenias at diagnosis, karyotype, and IPSS, subsequent therapies, disease progression, and survival. All analyses were conducted using SPSS version 15.0. (SPSS Inc, Chicago, IL). Kaplan–Meier curves were used for estimates of median overall survival. Results Long-term follow up data were available on 48 of the original 52 MDS patients and 50% had evidence of clonal T-cell expansion (n=24). RCMD was the commonest WHO subtype among both groups (n=24, 50%) and more patients were lower-risk based on IPSS (n=32, 67%). No difference in median overall survival (OS) was observed among the two groups based on the status of clonal T-cell expansion (55.8 mo with clonal T-cell expansion and 58.3 mo without clonal T-cell expansion, P-value 0.76. Patients were then stratified by IPSS into low/int-1 (lower risk) and int-2/high (higher risk) groups for subset analysis. In patients with a low/int-1 IPSS risk classification, the trend in median OS (median OS 79 mo in the positive group n=14; and 76 mo in the negative cohort n=18) was similar in both groups and consistent with previously published data from the International MDS risk analysis workshop (IMRAW/IPSS) for patients with low/int-1 IPSS risk classification. Interestingly, patients with clonal T-cell expansion in the int-2/high (higher risk) IPSS MDS group (n=10) had a median OS of 30 mo compared to 12 month in the same IPSS risk group with no clonal T-cell expansion (n=6). Therefore, the presence of clonal T-cells in higher risk MDS patients was associated with an improved outcome compared to reported median OS in higher risk IPPS MDS patients in the IMRAW database. Out of the 48 patients 14 received azacitidine; only two of the 6 patients with clonal T cell expansion (33%) had a response to azacitidine compared to 5 out of 8 patients (63%) without clonal T cell expansion. Conclusions The exact etiology of the clonal T-cell expansion is not known but may be autoimmune, homeostatic or antigen-driven. Improved survival in higher-risk patients with clonal T-cell expansion suggests a distinct pathophysiological mechanism. T-cell response may be generated to LAA providing immuneosurveillance that may be an important mechanism to slow disease progression. Furthermore, certain immunological signatures may be used as predictive tools for response to treatment. This study suggests that clonal T-cell expansion may be an important molecular determinant associate of improved survival outcome and a prospective larger cohort is warranted to confirm these observations. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3210-3210
Author(s):  
Takayuki Inouye ◽  
Motoko Koyama ◽  
Ensbey Kathleen ◽  
Nicholas Greene ◽  
Luke Samson ◽  
...  

Leukemia relapse represents a failure of graft-versus-leukemia (GVL) and remains the major limitation of allogeneic stem cell/bone marrow transplantation (BMT). Graft-versus-host disease (GVHD) within the gastrointestinal (GI) tract is the principal determinant of transplant-related mortality and is initiated by a network of alloantigen presentation by professional and non-professional APC that prime donor T cells in the GI tract and related lymphoid structures. Since GVL and lethal GVHD are mediated by donor T cells at spatially distinct sites; bone marrow (BM) and the GI tract respectively, we sought tractable approaches to spatially separate alloreactive responses at these two locations. The administration of high dose steroids in the peri-transplant period is permissive of T cell replete HLA-haploidentical BMT and significant GVL effects (Ogawa H, et al. BBMT. 2006). We utilized murine haploidentical BMT models (B6D2F1 → B6C3F1, B6 → B6D2F1) with recipient background MLL/AF9 primary acute myeloid leukemia (AML), with or without dexamethasone (Dex) administration (5 mg/kg/day i.p., days -1 to +5). Dex-treatment improved transplant survival (from 25% to 68% at day 100, P=0.0012) with significant reductions in GVHD histopathology specifically in the colon (histopathology scores 8.7±1.0 vs 4.6±0.8, P< 0.05), despite excellent leukemia control. To understand this paradox, we analyzed the kinetics of donor T cell expansion after BMT. In the mesenteric lymph node (mLN), Dex treatment significantly suppressed the expansion of both CD4 and CD8 T cells (3.3±0.3 x 105 vs 1.4±0.3 x 105, P< 0.001 and 4.2±0.4 x 105 vs 2.1±0.4 x 105, P< 0.01 respectively) and the activation of CD4 T cells (CD25 MFI: 2021±146 vs 1056±102, P< 0.01). In contrast, donor effector/memory CD44+ CD8 T cells were expanded in the BM of Dex treated recipients (1.9±0.3 x 105 vs 3.1±0.4 x 105, P< 0.05) that demonstrated high per cell cytolytic activity against leukemia (specific lysis: 65±2.4 % vs 62±2.6 % in untreated vs Dex-treated, P> 0.05). Surprisingly, there was no difference in proliferation (cell tracking dye dilution: 63±5.5 % vs 57±5.5 % in untreated vs Dex-treated, P> 0.05) or apoptosis (caspase-3: 6.6±0.4 % vs 6.1±0.6 %, caspase-8: 20±1.6 % vs 17±3.3 % in untreated vs Dex-treated, respectively, P> 0.05) of CD4 T cells in the mLN between the two groups. We undertook experiments with luciferase expressing T cells and noted that Dex-treatment preferentially inhibited T cell accumulation in the GI tract, but not marrow after BMT. Thus, it appeared that Dex treatment preferentially re-distributed donor T cells from the GI tract to the bone marrow. We next determined if Dex exerted effects via direct signaling to the donor T cell. We thus transplanted glucocorticoid receptor (GR)-deficient or intact T cells (GRfl/fl lck-Cre mice). Dex-treatment reduced donor CD4 T cell expansion in the mLN independent of their expression of the GR (untreated vs Dex-treated: 2.8±0.6 x 105 vs 1.2±0.3 x 105, lckCREGRfl/fl and 2.4±0.3 x 105 vs 1.4±0.4 x 105, GRfl/fl littermates, P< 0.05 both groups). Thus steroid effects were mediated indirectly, putatively via effects on recipient alloantigen presentation. There was a marked reduction in recipient dendritic cells (DC) and macrophages expressing the Ea peptide within MHC class II in the GI tract of Dex-treated recipients (terminal Ileum YAe+ DC number 896±93 vs 356±40, P< 0.01, YAe+ macrophage number 1035±136 vs 355±97, P< 0.01). In conjunction with this, expression of the gut homing integrin a4b7 expression was reduced in CD4 T cells from Dex treated recipient mLN (25±1.6 % vs 17±1.7 %, P< 0.01), while the marrow homing integrin VLA-4 (a4b1) was increased (a4: 62±2.2 % vs 75±1.6 %, P< 0.001, b1: 52±2.5 % vs 61±1.6 %, P< 0.05) in donor CD8 T cells from Dex treated recipient BM. Finally, Dex treatment enhanced GVL against a second primary AML (BCR/ABL-NUP98/HOXA9) relative to untreated recipients and those receiving post-transplant cyclophosphamide (PT-Cy) (relapse rate: 0% vs 40% vs 100% at day 35 in Dex vs untreated vs PT-Cy, PT-Cy vs Dex-treated, P< 0.0001; untreated vs Dex-treated, P=0.029). These data suggest a potential therapeutic strategy to modulate antigen presentation in the GI tract and consequent integrin imprinting that minimizes GVHD lethality whilst enhancing GVL within BM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 121-121
Author(s):  
Jichun Chen ◽  
Neal S. Young

Abstract We have produced a murine model of immune-mediated bone marrow (BM) failure based on minor histocompatability antigen mismatch. Infusion of C57BL/6 (B6) lymph node (LN) cells into congenic C.B10 mice results 2–5 weeks later in severe marrow hypoplasia and fatal pancytopenia. Expansion of pathogenic T cells, especially cytotoxic T cells specific for the dominant minor antigen H60, is critical in disease progression: infusion of LN cells from H60-matched congenic B6 donors failed to induce BM failure in C.B10 recipients. We observed that the proportion of CD4+CD25+ T cells in the BM increased significantly in LN-cell-infused animals, coordinate with BM invasion by CD4 and CD8 T cells. However, a very large fraction (60–80%) of CD4+CD25+ T cells from BM failure mice did not express intracellular FoxP3, in contrast to CD4+CD25+ cells from normal BM which were 80–90% FoxP3+. In the mouse model, there were significant declines in the ratios between regulatory T cells and total T cells in the BM: CD4+CD25+FoxP3+ (Treg) to CD4+CD25+FoxP3− (activated CD4 cells) ratio decreased from 1 : 0.29 to 1 : 2.10; CD4+CD25+FoxP3+ to CD4+ ratio decreased from 1 : 7.01 to 1 : 19.87 whereas CD4+CD25+FoxP3+ to CD8+ ratio decreased from 1 : 7.94 to 1 : 46.71 when unaffected animals and BM failure mice were compared. Infusion of as few as 5000 CD4+CD25+ T cells obtained from normal B6 mouse spleen (&gt;80% FoxP3+) with allogeneic B6 LN cells prevented H60-specific T cell expansion in C.B10 recipients and suppressed marrow destruction. A reduced regulatory T cell to activated T cell ratio accompanies pathogenic T cell expansion in this model of immune-mediated marrow destruction. Added regulatory T cells can suppress T cell expansion and prevent murine aplastic anemia. Our model concurs with recent observations in human aplastic anemia, in which numbers and activity of circulating regulatory T cells are much reduced in comparison to healthy controls (Solomou EE et al. ASH abstract submitted). Regulatory T cells might be useful in cellular therapy of autoimmune diseases. Figure Figure


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 7-7
Author(s):  
Carlotta Welters ◽  
Meng-Tung Hsu ◽  
Christian Alexander Stein ◽  
Livius Penter ◽  
María Fernanda Lammoglia Cobo ◽  
...  

Multiple myeloma is a malignancy of monoclonal plasma cells accumulating in the bone marrow. The critical influence of tumor-infiltrating T cells on disease control and therapeutic responses has been shown in a variety of malignancies, however, the role of multiple myeloma bone marrow-infiltrating T cells is incompletely understood. Although it has been shown that multiple myeloma neo-antigen-specific T cells can be expanded in vitro, little is known about functions and specificities of clonally expanded multiple myeloma-infiltrating bone marrow T cells. Here we asked at the single cell level whether clonally expanded T cells i) were detectable in multiple myeloma bone marrow and peripheral blood, ii) showed characteristic immune phenotypes, and iii) recognized antigens selectively presented on multiple myeloma cells. A total of 6,744 single bone marrow T cells from 13 treatment-naïve patients were index-sorted and sequenced using our methodologies for determination of paired T cell receptor (TCR) αβ sequences along with immune phenotype, transcription factor and cytokine expression. Clonal T cell expansion occurred predominantly within the CD8+ compartment. Phenotypes of clonally expanded T cells were distinctive of cytolytic effector differentiation and significantly different from non-expanded CD8+ T cells. Less than 25% of expanded CD8+ T cell clones expressed the immune checkpoint molecules programmed death-1 (PD-1), cytotoxic T lymphocyte antigen-4 (CTLA-4), or T cell immunoglobulin and mucin-domain containing-3 (TIM-3), while B and T lymphocyte attenuator (BTLA) was expressed on more than half of the expanded clones. Clonal T cell expansion did not correlate with neo-antigen load as determined by whole exome and RNA sequencing of purified multiple myeloma cells. Furthermore, peripheral blood TCRβ repertoire sequencing from five selected patients with substantial bone marrow T cell expansion identified 90% of expanded bone marrow T cell clones overlapping with peripheral blood. To determine whether clonally expanded bone marrow T cells recognized antigens selectively presented on multiple myeloma cells, 71 dominant TCRs from five selected patients with substantial clonal T cell expansion were re-expressed in 58α-β- T-hybridoma reporter T cells and co-incubated with CD38-enriched multiple myeloma cells from the same patients. Only one of these TCRs recognized antigens selectively presented on multiple myeloma cells and this TCR was not neo-antigen-specific. Hypothesizing that the target antigen was a non-mutated self-antigen, we could show that this TCR also recognized the plasma cell leukemia cell line U-266 in an HLA-A*02:01-restricted manner. In summary, clonally expanded T cells in multiple myeloma bone marrow of newly diagnosed patients show cytolytic effector differentiation. In the majority of patients, clonally expanded bone marrow T cells do not recognize antigens presented on multiple myeloma cells and are not neo-antigen-specific. Our findings are relevant for the design of future therapeutics and clinical trials. The identified TCR, which recognizes a multiple myeloma antigen shared with U-266 in an HLA-A*02:01-restricted manner, could be a promising candidate for T cell therapy. Disclosures Bullinger: Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Hexal: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Menarini: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Seattle Genetics: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3171-3171
Author(s):  
Madhusudhanan Sukumar ◽  
Andrea Wilke ◽  
Josef Mautner ◽  
Hans-Jochem Kolb ◽  
Georg Bornkamm ◽  
...  

Abstract Allogeneic stem cell transplantation for high grade lymphoma typically suffers from high transplant related mortality and minimal success in achieving long term complete remission. The immunological aspects behind rejection of high grade lymphoma are poorly understood, partly due to the lack of animal models. Using a transgenic mouse lymphoma model, where the proto-oncogene c-myc is driven by parts of the immunoglobulin lambda locus representing a t(8;22) translocation as found in Burkitt’s lymphoma, we developed a haploidentical transplantation model. c-myc-lambda transgenic C57/BL6 mice were crossbred with DBA mice, giving rise to a B6D2F1 generation which develops high grade lymphoma spontaneously within the first 6 months post birth. Primary F1-lymphoma cell lines displayed low MHC class I and class II expression compared to wild type B-cells. When transferred into immune-competent, healthy C57/BL6 parental mice, F1-lymphoma cells were rejected even at high doses of 50 Mio. cells, whereas the syngeneic transfer of 10,000 cells into B6D2F1 mice resulted in 100% mortality due to lymphoma growth. Immunosuppression of C57/BL6 mice by lethal total body irradiation and transplantation of T-cell depleted bone marrow allowed F1-lymphoma to grow when animals were challenged at day +1 after bone marrow transplantation with 1 Mio. cells. Addback of 2.5 or 10 Mio. parental T-cells together with F1-lymphoma on day +1 completely prevented lymphoma growth, and resulted in long term survival for more than 60 days. When T-cell addback was delayed until day +3 or +6 after lymphoma challenge, animals died due to lymphoma progression 7–8 days later. By using “green” T-cells from GFP transgenic C57/BL6 mice we could show that spleens 7 days after T-cell addback contained only 0.5% (15% of total CD3+ cells) GFP positive T-cells, whereas up to 15% (55%–75% of total CD3+ cells) of total splenocytes were GFP positive in long term survivors that rejected lymphoma. Similar results were obtained when T-cells were labelled with CFDA. This indicates that T-cell expansion is associated with lymphoma rejection; animals that receive T-cells after lymphoma has been established do not show any T-cell expansion, even when lymphoma cells display a haplo-mismatch and mature T-cells are co-localized within the spleen. T-Cell expansion is suppressed when lymphoma cells are present in splenic tissue, indicating efficient immune escape. Similar results were observed when bone marrow and T-cells from C57BL/6 donors were transplanted into irradiated B6D2F1 mice and 1 Mio. F1-lymphoma cells were injected on day +1 post transplantation. Despite the presence of GvHD when low number of T-cells (0.5–2.0 Mio.) were used lymphoma growth was measurable and animals eventually died due to disease progression. These results point to the crucial role of pretransplant lymphoma burden for the success of allogeneic stem cell transplantation in high grade lymphoma.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A812-A812
Author(s):  
Pia Aehnlich ◽  
Per Thor Straten ◽  
Ana Micaela Carnaz Simoes ◽  
Signe Skadborg ◽  
Gitte Olofsson

BackgroundAdoptive cell therapy (ACT) is an approved treatment option for certain hematological cancers and has also shown success for some solid cancers. Still, benefit and eligibility do not extend to all patients. ACT with Vγ9Vδ2 T cells is a promising approach to overcome this hurdle.MethodsIn this study, we explored the effect of different cytokine conditions on the expansion of Vγ9Vδ2 T cells in vitro.ResultsWe could show that Vγ9Vδ2 T cell expansion is feasible with two different cytokine conditions: (a) 1000U/ml interleukin (IL)-2 and (b) 100U/ml IL-2+100U/ml IL-15. We did not observe differences in expansion rate or Vγ9Vδ2 T cell purity between the conditions; however, IL-2/IL-15-expanded Vγ9Vδ2 T cells displayed enhanced cytotoxicity against tumor cells, also in hypoxia. While this increase in killing capacity was not reflected in phenotype, we demonstrated that IL-2/IL-15-expanded Vγ9Vδ2 T cells harbor increased amounts of perforin, granzyme B and granulysin in a resting state and release more upon activation. IL-2/IL-15-expanded Vγ9Vδ2 T cells also showed higher levels of transcription factor T-bet, which could indicate that T-bet and cytotoxic molecule levels confer the increased cytotoxicity.ConclusionsThese results advocate the inclusion of IL-15 into ex vivo Vγ9Vδ2 T cell expansion protocols in future clinical studies.


2021 ◽  
pp. annrheumdis-2021-220435
Author(s):  
Theresa Graalmann ◽  
Katharina Borst ◽  
Himanshu Manchanda ◽  
Lea Vaas ◽  
Matthias Bruhn ◽  
...  

ObjectivesThe monoclonal anti-CD20 antibody rituximab is frequently applied in the treatment of lymphoma as well as autoimmune diseases and confers efficient depletion of recirculating B cells. Correspondingly, B cell-depleted patients barely mount de novo antibody responses during infections or vaccinations. Therefore, efficient immune responses of B cell-depleted patients largely depend on protective T cell responses.MethodsCD8+ T cell expansion was studied in rituximab-treated rheumatoid arthritis (RA) patients and B cell-deficient mice on vaccination/infection with different vaccines/pathogens.ResultsRituximab-treated RA patients vaccinated with Influvac showed reduced expansion of influenza-specific CD8+ T cells when compared with healthy controls. Moreover, B cell-deficient JHT mice infected with mouse-adapted Influenza or modified vaccinia virus Ankara showed less vigorous expansion of virus-specific CD8+ T cells than wild type mice. Of note, JHT mice do not have an intrinsic impairment of CD8+ T cell expansion, since infection with vaccinia virus induced similar T cell expansion in JHT and wild type mice. Direct type I interferon receptor signalling of B cells was necessary to induce several chemokines in B cells and to support T cell help by enhancing the expression of MHC-I.ConclusionsDepending on the stimulus, B cells can modulate CD8+ T cell responses. Thus, B cell depletion causes a deficiency of de novo antibody responses and affects the efficacy of cellular response including cytotoxic T cells. The choice of the appropriate vaccine to vaccinate B cell-depleted patients has to be re-evaluated in order to efficiently induce protective CD8+ T cell responses.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Sasan Ghaffari ◽  
Monireh Torabi-Rahvar ◽  
Sajjad Aghayan ◽  
Zahra Jabbarpour ◽  
Kobra Moradzadeh ◽  
...  

Abstract Background The successful ex vivo expansion of T-cells in great numbers is the cornerstone of adoptive cell therapy. We aimed to achieve the most optimal T-cell expansion condition by comparing the expansion of T-cells at various seeding densities, IL-2 concentrations, and bead-to-cell ratios. we first expanded the peripheral blood mononuclear cells (PBMCs) of a healthy donor at a range of 20 to 500 IU/mL IL-2 concentrations, 125 × 103 to 1.5 × 106 cell/mL, and 1:10 to 10:1 B:C (Bead-to-cell) ratios and compared the results. We then expanded the PBMC of three healthy donors using the optimized conditions and examined the growth kinetics. On day 28, CD3, CD4, and CD8 expression of the cell populations were analyzed by flow cytometry. Results T-cells of the first donor showed greater expansion results in IL-2 concentrations higher than 50 IU/mL compared to 20 IU/mL (P = 0.02). A seeding density of 250 × 103 cell/mL was superior to higher or lower densities in expanding T-cells (P = 0.025). Also, we witnessed a direct correlation between the B:C ratio and T-cell expansion, in which, in 5:1 and 10:1 B:C ratios T-cell significantly expanded more than lower B:C ratios. The results of PBMC expansions of three healthy donors were similar in growth kinetics. In the optimized condition, 96–98% of the lymphocyte population expressed CD3. While the majority of these cells expressed CD8, the mean expression of CD4 in the donors was 19.3, 16.5, and 20.4%. Conclusions Our methodology demonstrates an optimized culture condition for the production of large quantities of polyclonal T-cells, which could be useful for future clinical and research studies.


2021 ◽  
Vol 6 (59) ◽  
pp. eabh1516
Author(s):  
Marion Moreews ◽  
Kenz Le Gouge ◽  
Samira Khaldi-Plassart ◽  
Rémi Pescarmona ◽  
Anne-Laure Mathieu ◽  
...  

Multiple Inflammatory Syndrome in Children (MIS-C) is a delayed and severe complication of SARS-CoV-2 infection that strikes previously healthy children. As MIS-C combines clinical features of Kawasaki disease and Toxic Shock Syndrome (TSS), we aimed to compare the immunological profile of pediatric patients with these different conditions. We analyzed blood cytokine expression, and the T cell repertoire and phenotype in 36 MIS-C cases, which were compared to 16 KD, 58 TSS, and 42 COVID-19 cases. We observed an increase of serum inflammatory cytokines (IL-6, IL-10, IL-18, TNF-α, IFNγ, CD25s, MCP1, IL-1RA) in MIS-C, TSS and KD, contrasting with low expression of HLA-DR in monocytes. We detected a specific expansion of activated T cells expressing the Vβ21.3 T cell receptor β chain variable region in both CD4 and CD8 subsets in 75% of MIS-C patients and not in any patient with TSS, KD, or acute COVID-19; this correlated with the cytokine storm detected. The T cell repertoire returned to baseline within weeks after MIS-C resolution. Vβ21.3+ T cells from MIS-C patients expressed high levels of HLA-DR, CD38 and CX3CR1 but had weak responses to SARS-CoV-2 peptides in vitro. Consistently, the T cell expansion was not associated with specific classical HLA alleles. Thus, our data suggested that MIS-C is characterized by a polyclonal Vβ21.3 T cell expansion not directed against SARS-CoV-2 antigenic peptides, which is not seen in KD, TSS and acute COVID-19.


Blood ◽  
2008 ◽  
Vol 111 (5) ◽  
pp. 2497-2498
Author(s):  
Susumu Nakae ◽  
Keisuke Oboki ◽  
Hirohisa Saito

IgE/antigen-FcϵRI crosslinking promotes antigen internalization and apoptosis in mouse mast cells. Dendritic cells uptake the apoptotic mast cells carrying internalized antigens, and thus can efficiently present the antigens to memory T cells.


2004 ◽  
Vol 231 (1-2) ◽  
pp. 75-84 ◽  
Author(s):  
Kenneth Flanagan ◽  
Dorota Moroziewicz ◽  
Heesun Kwak ◽  
Heidi Hörig ◽  
Howard L. Kaufman

Sign in / Sign up

Export Citation Format

Share Document