Expansion of Polyclonal CD4+CD25high Foxp3+ Regulatory T-Cells in High Risk Myelodysplastic Syndromes (MDS).

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2641-2641
Author(s):  
Shahram Y. Kordasti ◽  
Wendy Ingram ◽  
Janet Hayden ◽  
David Darling ◽  
Linda Barber ◽  
...  

Abstract Foxp3+ regulatory T-cells(Tregs) play a central role in maintaining tolerance. A reduction in Tregs activity is a key feature of autoimmune diseases, whereas their expansion in malignant diseases leads to suppression of the host anti-tumour responses. Expansion of oligoclonal cytotoxic T cells, inhibition of MDS CD34+ progenitors proliferation and response to immunosuppressive therapy suggest that autoimmunity contributes to the pathophysiology of low risk MDS. However, this is not generally a feature of high risk MDS. We hypothesise that in high risk MDS patients an increased number of Tregs may suppress immune responses against the dysplastic clone. In early MDS, these cells may be reduced and or be associated with coexistent autoimmune diseases, an uncommon but recognised association of low risk MDS. We therefore studied CD4+ CD25high Foxp3+ and CD8+ CD25+Foxp3+ Tregs in peripheral blood of MDS patients and examined correlation with bone marrow blasts, cytogenetic Status, IPSS score and progression to AML. Clonality of CD4+CD25+ Tregs was assessed by TCR spectratype analysis of CDR3 size distribution and by CDR3 sequence analysis. 52 patients with MDS (30 male, 22 female) with a mean age of 62 years (range 40 to 82 years) were studied. According to WHO classification, 5/52 (10%) had a diagnosis of 5q− syndrome, 9/52(17%) refractory anaemia (RA), 18/52 (35%) refractory cytopenia with multilineage dysplasia (RCMD), 16/52 (31%) refractory anaemia with excess blasts (RAEB) and 4/52 (7%) patients MDS/MPD (one with JAK2 V617F Mutation). Cytogenetic study shows normal pattern in 57%, 5q- in 22%, stable cytogenetic in 10% and complex findings in 11%. All samples were taken at diagnosis prior to any treatment. In addition 5 samples were analyzed pre and post 5-Azacytidine therapy. The absolute number of CD4+CD25highFoxp3+ regulatory T cells in 5q- syndrome was 0.5±0.28×107/l, RA0.6±0.56×107/l, RCMD1.42±0.97×107/l, RAEB2.8±2.2×107/l and MDS/MPD 2.9±2×107/l. In cases with <5% bone marrow blasts (RA, 5q− & RCMD)absolute number and percentage of Tregs was significantly lower than those with ≥5% BM blasts (p=0.001). The mean number was also significantly lower at 0.73±0.57×107/l in low risk cases(IPSS 0) compared with 2±1.5×107/l in intermediate and high risk groups (p=0.008). CD8+ Tregs were not significantly different between the subtypes of MDS and between low and high risk IPSS subgroups. Tregs number did not differ significantly between various cytogenetic subgroups. The spectratype of CD4+CD25+ TCR amplicons, showed a polyclonal pattern and the overall complexity of Vβ subfamilies was not different between low risk and high risk MDS, suggesting that the expanded Tregs in high risk MDS are not clonal and likely to arise by peripheral expansion rather than an antigen-driven response. CD4+ CD25high Foxp3+ Tregs in five patients were studied pre and post 5-Azacytidine. The numbers were significantly decreased after treatment in the one patient who responded to treatment (p=0.001), whereas Treg numbers were unchanged or increased in non responsive cases. In 5 RCMD cases with concomitant autoimmune diseases the percentage of Tregs was lower than other patients within the same subgroup of MDS, however, this did not achieve statistical significance. The findings indicate Tregs are altered in MDS and may be important in the pathophysiology of MDS. Monitoring of Tregs numbers can be a useful indicator for disease progression and response to immunosuppressive therapy.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 637-637
Author(s):  
Shahram Y Kordasti ◽  
Behdad Afzali ◽  
Ziyi Lim ◽  
Wendy Ingram ◽  
Janet Hayden ◽  
...  

Abstract Immunological responses are increasingly recognized as important in the initiation and progression of MDS. Indeed, autoimmune diseases commonly occur in association with low risk MDS. Different studies, including our own, have shown that Foxp3+ Tregs play a role in the inhibition of tumour immune surveillance in high risk MDS; however, less clear are the potential factors which are inducing inflammatory response in low risk MDS that may also predispose to autoimmunity. CD4+IL 17 producing T cells (Th17) are considered as important pro-inflammatory T cells and contribute to the pathogenesis of a number of autoimmune diseases as well as some malignancies. The aim of this study was to investigate the potential role of Th17 cells in low risk MDS. We show for the first time that Th17 cells are markedly increased in low risk MDS in comparison to high risk disease and healthy age matched controls. We also describe an inverse relationship between numbers of Th17 cells and naturally occurring Tregs in MDS. 88 patients with newly diagnosed MDS, and 15 age matched healthy donors, were recruited. Analysis of Th17 cells was performed in a subset of 43 patients. WHO classifications were as follows: 19% had RA, 33% had RCMD, 32% had RAEB I & II and 16% had 5q- syndrome. A clear difference was observed in the median percentage and absolute number of Th17 cells between patients with low risk (based on IPSS) compared to high risk MDS (p&lt;0.01). In low risk, but not high risk MDS, CD4+ cells had greater median number of committed Th17 cells than healthy donors (p&lt;0.01). The absolute number of polyclonal Foxp3+ Tregs inversely correlated with Th17 cells, with higher Tregs in the high risk MDS when compared to low risk MDS or healthy donors (p&lt;0.001), consistent with our previous report. Analysis according to IPSS, BM blast percentage or WHO classification revealed the same patterns of Th17 and Tregs expression, with the higher risk phenotypes being associated with significantly higher Treg number and lower risk phenotypes with greater Th17 cells. This finding was specific for Th17 as there was no significant difference in the number of circulating Th1 and Th2 cells between MDS subtypes. Co-culture of Tregs from patients with MDS with their own T effectors lead to suppression of supernatant IFNγ levels. Importantly, Tregs did not have an inhibitory effect on IL17 production and indicates that reduced Th17 number in the presence of high Tregs does not indicate active suppression of the Th17 phenotype. To investigate differences in serum cytokines between low and high risk MDS and healthy donors, serum samples from patients were analyzed by Luminex for multiple cytokines. The cytokines IL12(p&lt;0.01) IL7(p&lt;0.005) IFNγ(p&lt;0.01) and RANTES (p&lt;0.005) were significantly higher in patients with low risk compared to high risk MDS and healthy controls. The inhibitory cytokine IL10 and soluble IL2 receptor were present at higher levels in the serum of patients with high risk MDS (p&lt;0.01 and p&lt;0.005 respectively). To confirm that the peripheral blood cytokines are a true reflection of cytokine concentration in the bone marrow, the concentration of 30 different cytokines was measured by Luminex in 10 matched bone marrow and peripheral blood samples from these patients and there was no significant difference between these two sets of samples. In addition, the level of apoptosis, assessed by TUNEL assay, was also higher in the bone marrow of low risk patients. In conclusion, the “unfavourable” Th17 to Tregs ratio in low risk MDS may explain the higher risk of autoimmunity and bone marrow apoptosis. These findings may help identify subtype of low risk MDS patients likely to respond to immunosuppressive therapy.


Blood ◽  
2007 ◽  
Vol 110 (3) ◽  
pp. 847-850 ◽  
Author(s):  
Shahram Y. Kordasti ◽  
Wendy Ingram ◽  
Janet Hayden ◽  
David Darling ◽  
Linda Barber ◽  
...  

Abstract Foxp3+ regulatory T cells (Tregs) play a central role in maintaining immune tolerance. A reduction in the function of Tregs is a key feature of autoimmune diseases, whereas their expansion in malignant diseases leads to the suppression of host antitumor responses. We analyzed the absolute number of CD4+ and CD8+ Tregs in the peripheral blood of 52 patients with myelodysplastic syndrome (MDS) and show a significant correlation between increased number of CD4+ Tregs and MDS subgroups with 5% or more bone marrow blasts (P &lt; .001), high International Prognostic Scoring System (IPSS) score (P &lt; .001), and disease progression (P &lt; .001), whereas no correlation between CD8+ Tregs and prognostic variables was observed. The CD4+ Tregs showed a polyclonal spectratype, and the percentage of the naive subset was significantly higher in the high-risk patients compared with low-risk or healthy age-matched donors (P = .032). Our data suggest that CD4+ Treg expansion is a feature of high-risk MDS and progression to aggressive subtypes of the disease.


2018 ◽  
Vol 185 (2) ◽  
pp. 357-360 ◽  
Author(s):  
Angela Giovazzino ◽  
Stefania Leone ◽  
Valentina Rubino ◽  
Anna Teresa Palatucci ◽  
Giuseppe Cerciello ◽  
...  

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2445-2445
Author(s):  
Shahram Y. Kordasti ◽  
Wendy Ingram ◽  
Janet Hayden ◽  
David Darling ◽  
Linda Barber ◽  
...  

Abstract Regulatory T-cells (Tregs) are important in the immune surveillance of malignancies. In MDS Tregs may inhibit effective immune responses against the dysplastic clone thereby facilitating disease progression. We studied the number (CD4+ and CD8+), function and clonality of CD4+Tregs in the peripheral blood of 75 MDS patients with different subtypes of MDS and 9 healthy volunteers as controls. The number of Tregs was also compared between different cytogenetic abnormalities. The phenotype of the expanded Tregs was analysed by assessment of the naïve vs memory subpopulations (CD25highFoxp3+CD27+CD45RO− and CD25highFoxp3+CD27+CD45RO+ respectively) in low and high risk MDS. The absolute number of CD4+CD25highFoxp3+ and CD4+/CD8+CD25+Foxp3+ was calculated. The median number of CD4+CD25highFoxp3+ Tregs in 5q-syndrome was 0.7×107/l (range, 0.2–2.2x107), Refractory Anemia (RA) 0.7×107/l (range, 0.5–1.6x107), Refractory Cytopenia with Multilineage Dysplasia (RCMD) 1.3×107/l (range, 0.2–2.6x107), Refractory Anemia with Excess Blast (RAEB) 2.2×107/l (range, 0.6–7.0x107) and Myelodysplastic/Myeloproliferative Disease (MDS/MPD) 3.1×107/l (range, 0.8–5.0x107). CD4+Tregs were higher in patients with ≥5% BM blasts vs &lt;5% BM blasts (p&lt;0.001), in high vs low/intermediate IPSS (p&lt;0.001), disease progression vs stable disease (p&lt;0.001). CD4+ Tregs were lower in 5q- syndrome, RCMD and RA. However, these did not differ significantly from normal controls (p=0.6), whereas RAEB and MDS/MPD had higher CD4+ Tregs than normal donors (p&lt;0.001, p=0.02). The number of Tregs has also been correlated with cytogenetic abnormalities (based on IPSS definition). In patients with isolated 5q- Tregs were significantly lower than those with complex (p=0.004) or intermediate risk karyotypes (p&lt;0.001). There was no difference in the number of CD8+ Tregs between MDS subtypes (p=0.28), IPSS (p=0.19), or disease progression (p=0.19). The percentage of naïve Tregs was significantly higher in high risk patients compared with low risk and healthy volunteers (p=0.032). The ratio of naïve to memory Tregs was also significantly higher in the high risk than low risk (p=0.016) or control groups (p=0.032). The spectratype of CD4+CD25+ TCR amplicons showed a polyclonal pattern and the overall complexity of Vβ spectratypes was not different between low and high risk group (p=0.54). By contrast the spectratype of CD8+Tcells was skewed on average in 6/24 Vβ subfamilies indicating the clonal expansion of these cells. Functionality of the expanded Tregs was demonstrated by inhibition of IFN-γ secretion by effector T-cells, confirmed by both intracellular staining and ELISA. We demonstrate that expansion of Tregs occurs frequently in high risk MDS and disease progression. By contrast, in low risk MDS the Treg population tends to be lower, thereby permitting the emergence of autoimmune responses. Although the increased number of Tregs in high IPSS MDS is an important indication of immune suppression, Karyotype and bone marrow blast percentage can influence the number of Tregs independently.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3792-3792 ◽  
Author(s):  
Christos K Kontos ◽  
Vassiliki Pappa ◽  
Diamantina Vasilatou ◽  
Maria-Angeliki S Pavlou ◽  
Frida Kontsioti ◽  
...  

Abstract Abstract 3792 Introduction: MicroRNAs are single, small non-coding RNA molecules of approximately 21–26 nucleotides, which regulate the expression of numerous genes. miRNAs may act either at the post-transcriptional or the post-translational level to repress gene expression; still, upregulation of gene expression has been noticed in some cases as a direct effect of miRNA function. The importance of miRNAs in carcinogenesis is emphasized by the association of cancers with alterations in miRNA expression. Many miRNAs, including let-7a and those of the miR-17-92 cluster (miR-17, miR-20a, etc.), have been shown or are predicted to affect the activities of targeted mRNAs encoding proteins that have oncogenic or anti-oncogenic functions. let-7a downregulates KRAS, while miR-17 and miR-20a downregulate E2F1. Both these proteins are overexpressed in myelodysplastic syndromes (MDS) and have been shown to be involved in the pathobiology of the disease. Purpose: In the current study, we examined the prognostic value of let-7a, miR-17 and miR-20a levels in MDS and their potential as novel molecular biomarkers. Furthermore, we investigated the protein expression levels of validated targets of these three miRNAs in bone marrow CD34+ cells of MDS patients. Material and Methods: We evaluated 43 patients with MDS (34 men, 9 women) with a median age of 73 years (range 45–87). According to WHO classification, 12 patients (27.9%) were diagnosed with RA, 6 (13.9%) RCMD, 8 (18.6%) with RAEB-I, 7 (16.3%) with RAEB-II, 8 (18.6%) with AML, and 2 (4.7%) with CMML. According to IPSS, 13 patients (32.5%) had low risk, 14 (35.0%) intermediate I risk, 6 (15.0%) intermediate II, and 7 (17.5%) high risk disease. WPSS classification was: 8 (23.5%) very low risk, 5 (14.7%) low risk, 8 (23.5%) intermediate, 9 (26.5%) high risk, and 4 (11.8%) very high risk. We isolated CD34+ cells from bone marrow mononuclear cells from MDS patients, as well as from peripheral blood of donors of CD34+ cells for stem cell transplantation, using magnetic beads. Extraction of small RNA-containing total RNA from CD34+ cells was performed and cDNA of let-7a, miR-17 and miR-20a was synthesized using specific primers. miRNA expression levels were determined using quantitative real-time PCR, the TaqMan® chemistry and the relative quantification (2−ΔΔCT) method. The snoRNA RNU48 was used as reference gene. Furthermore, total protein was extracted from CD34+ cells using a lysis buffer and subsequently quantified using the Bradford assay. Western blot analysis was carried out for MYC, E2F1, Cyclin D1 (CCND1), BCL2 and KRAS, while Actin was used as reference protein. Results: In MDS patients, let-7a expression levels were 0.053–506.1 copies/RNU48 copies, while miR-17 and miR-20a expression levels were 0.005–2694.5 and 0.003–3116.7 copies/103RNU48 copies, respectively. No significant differences were found between patients and controls regarding let-7a, miR-17 and miR-20a expression. let-7a underexpression was associated with high (>10%) bone marrow blasts percentage (P =0.036), presence of WHO classification subtypes with poor prognosis (RAEB-I, RAEB-II and AML) (P =0.020), and high IPSS (P =0.037). Furthermore, miR-17 underexpression was related to high (>10%) bone marrow blasts percentage (P =0.008), intermediate and/or high risk karyotype (P =0.018) and high IPSS (P =0.016). Moreover, miR-20a underexpression was associated with high IPSS (P =0.037) and WPSS (P =0.013). Interestingly, protein expression levels of all targets analyzed in the current study were shown to be lower in samples overexpressing let-7a, miR-17 and/or miR-20a, in comparison with the corresponding protein levels noticed in specimens showing lower expression of these three miRNAs. Conclusion: To the best of our knowledge, this is the first study showing that expression levels of let-7a, miR-17 and miR-20a are associated with established prognostic factors in MDS, including IPSS and WPSS. Furthermore, these three miRNAs seem to be implicated in the pathogenesis of the disease, most probably by finely tuning the expression of target proteins that are involved in highly important molecular pathways, therefore affecting key cellular functions, such as cell cycle control, apoptosis, cell proliferation, and regulation of gene expression. Undoubtedly, further studies are needed to confirm the present findings and clarify their association with the pathogenesis of different MDS subgroups. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2980-2980
Author(s):  
Jessica Ann Timms ◽  
Susann Winter ◽  
Steven Hargreaves ◽  
Donal P. McLornan ◽  
Uwe Platzbecker ◽  
...  

Background Regulatory T cells (Tregs) are a non-redundant, suppressive population of CD4+ T cells that are essential for the prevention of autoimmune diseases in humans. They may also contribute to overall immune suppression in malignancies and prevent effective immune surveillance against malignant cells. The correlation between the increased number of Tregs and higher-risk MDS has been shown before (Kordasti, Blood 2007). Expansion of effector T cells (Teff) also signifies the lower-risk disease (Kordasti, BJH, 2009). Using deep phenotyping and unbiased automated clustering we have previously identified novel populations of Tregs with distinct functional properties (Kordasti, Blood 2016), including a CD161-expressing subpopulation (Povoleri, Nature Imm, 2018). The aim of this study was to interrogate Treg and Teff subpopulations in more depth and identify transcriptomic profiles of Tregs and Teff in bone marrow (BM) and peripheral blood (PB) of MDS patients, and potential differences between PB and BM residing cells. Single-cell RNA sequencing (scRNAseq) in combination with oligonucleotide-tagged antibodies and an in-house pipeline utilizing Seurat for QC, data integration and analysis were used for this purpose. Methods PB and BM were collected from a low (del(5q)) and a high-risk (monosomy 7) MDS patient. These samples were magnetically enriched for CD4+ T cells, followed by flow sorting of Tregs (CD4+, CD25high, and CD127low) and Teff (CD4+, CD25low, and CD127high). In total, 8 cell fractions were used for downstream scRNAseq and concomitant protein expression profiling using the BD Rhapsody Single-Cell Analysis System (Becton Dickinson, CA, USA). Cell fractions were first labelled by oligo-tagged antibodies (CD95, CD45RO, CD38, CTLA-4, PD-1, HLA-DR) and a sample tag (for sample multiplexing). After labelling, all samples were combined for cartridge-based single-cell capture and molecular indexing of mRNA transcripts on magnetic barcoded beads. Following bead retrieval and on-bead cDNA synthesis, parallel targeted mRNA (259 T cell-related genes) and Abseq sequencing libraries were generated. Deep sequencing was performed using the HiSeq400 platform. Nonlinear dimensionality reduction was carried out using uniform manifold approximation and projection (UMAP), which produced UMAP layouts and heatmaps to identify populations. Tregs were further stratified according to high/low expression of the CD95 protein, CD161, and CD150 mRNA expression. The Wilcoxon Rank Sum test was used for statistics followed by Bonferroni corrections. The Gene Set Variation Analysis (GSVA) package was used for pathway enrichment analysis. Results and discussion Unbiased analysis by UMAP was able to clearly separate the high- and low-risk patients based on the Treg and Teff signature in BM, which was not the case in PB (Figure 1a and 1b). At protein level, BM Tregs from the high-risk patient had a significantly higher expression of CD95 (p = 7.23 x 10-17), CD45RO (p = 1.06 x 10-51), CTLA4 (p = 4.47 x 10-83) PD-1(p = 5.42 x 10-81), and CD38 (p = 4.16 x 10-63) (Figure 1c), suggesting a more functional Treg profile. GSVA analysis of transcriptomes showed significant changes between high- and low-risk patients, for both Tregs (102 pathways p≤0.05) and Teffs (89 pathways p≤0.05) in BM. Specifically, Tregs from high-risk MDS were more enriched with non-TCR-mediated activation pathways, including leukocyte differentiation, Jak-STAT signaling, and positive regulation of leukocyte cell-cell adhesion (Figure 1d). A deeper investigation of Tregs showed that subsets of interest, in particular the IL-17 secreting CD161+ Tregs and the hematopoietic stem cell quiescence-maintaining CD150+ Tregs (Hirata, 2018) were found in low abundance in the BM of both high- and low-risk patients; 16% and 10%; and, 1% and 7%, respectively. In summary, we have developed an analytical pipeline and provide evidence for the ability of simultaneous single-cell transcriptomics and protein expression to identify different BM-specific T cell signatures in high- versus low-risk MDS. Interestingly, Tregs from high risk MDS BM show higher expression of Treg-related proteins and are more enriched in non-TCR-specific activation pathways, which may reflect a myeloid derived inflammatory environment. Overall, these findings suggest a bone marrow-specific Treg signature in MDS, which could distinguish low- and high-risk disease. Disclosures McLornan: Jazz Pharmaceuticals: Honoraria, Speakers Bureau; Novartis: Honoraria. Platzbecker:Celgene: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria. Kordasti:Celgene: Research Funding; Novartis: Research Funding; Boston Biomed: Consultancy; API: Consultancy.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1659-1659 ◽  
Author(s):  
Elias Jabbour ◽  
Guillermo Garcia-Manero ◽  
Jianqin Shan ◽  
Susan O’Brien ◽  
Jorge Cortes ◽  
...  

Abstract Background and aims: Prognosis in pts with MDS post decitabine failure is reported to be poor, but there are no published data pertaining to survival, particularly in relation to pts who might benefit from specific strategies such as allogeneic stem cell transplantation (SCT) or investigational agents. In this regard, we report the results from our institution. Materials and Methods: A total of 87 pts with MDS (n=67) and CMML (n=20) after failure of decitabine regimens from 2003 until June 2007 were analyzed. Responses to decitabine and subsequent therapies were coded according to the modified International Working Group criteria (IWG). Acute myeloid leukemia (AML) was defined by 30% or more of bone marrow blasts. Ps were classified according to the IPSS score and the MD Anderson score (MDAS) at the time of start of decitabine therapy and at the time of failure. Results: Twenty-five (29%) pts had secondary MDS. Among 49 evaluable pts for IPSS risk category at the start of decitabine, 13 (26%) were int-1, 23 (48%) int-2, and 13 (26%) high-risk disease. According to the MDAS, 32 (37%) were high-risk, 31 (36%) int-2, 17 (19%) int-1, and 7 (8%) low-risk disease. Cytogenetics were diploid in 35 (40%) pts; 39 (45%) pts had ≥ 10% bone marrow blasts; 35 (40%) pts had primary resistance to decitabine and 52 (60%) had disease recurrence. Best response to decitabine was complete response (CR) in 21 (24%) pts, partial response (PR) in 2 (2%) pts, a bone marrow CR in 14 (16%) pts, and hematologic improvement (HI) in 14 (16%) pts. The median number of cycles of decitabine administered was 7 (range, 1–20). After a median of 21 months (range, 6 to 39) from decitabine failure, 13 (15%) pts remained alive. The median survival post decitabine failure was 4.3 months; the estimated 12-month survival was 28%. Upon failure, 22 (25%) pts transformed into AML, of whom only 3 were alive. At failure 15 (42%) were high-risk, 15 (42%) were int-2, and 6 (8%) were int-1 among 36 patients assessed for the IPSS score. Among 58 pts assessed by the MDAS, 32 (55%) were high-risk, 17 (29%) int-2, 7 (12%) int-1, and 2 (4%) were low-risk disease. The estimated 12-month survival rates were 27%, 33%, and 33% for respectively, patients with high-risk, int-2, and int-1 risk disease (p=0.99). The 12-month survival rates were 100%, 54%, 41%, and 18% for respectively, pts with low, int-1, int-2, and high-risk disease according to the MDAS (p=0.01). Post decitabine failure, 20 pts received idarubicin and cytarabine (IA); the objective response rate (ORR) was 20%; 39 pts received investigational agents with an ORR of 26%; 5 pts received an allogeneic SCT: 3 achieved and remained in CR. At the last follow-up, 13 pts were alive, 5 in CR post allogeneic SCT in 2, post clofarabine in 1, and post IA in 1, and 1 in marrow CR post IA. Conclusion: Outcome of pts post decitabine failure is poor with a median survival of 4.3 months. The MDAS can predict survival at the start of decitabine therapy and upon decitabine failure, with patients with low-risk disease having favorable outcome.


Life ◽  
2021 ◽  
Vol 11 (3) ◽  
pp. 245
Author(s):  
Daniil Shevyrev ◽  
Valeriy Tereshchenko ◽  
Elena Blinova ◽  
Nadezda Knauer ◽  
Ekaterina Pashkina ◽  
...  

Homeostatic proliferation (HP) is a physiological process that reconstitutes the T cell pool after lymphopenia involving Interleukin-7 and 15 (IL-7 and IL-15), which are the key cytokines regulating the process. However, there is no evidence that these cytokines influence the function of regulatory T cells (Tregs). Since lymphopenia often accompanies autoimmune diseases, we decided to study the functional activity of Tregs stimulated by HP cytokines from patients with rheumatoid arthritis as compared with that of those from healthy donors. Since T cell receptor (TCR) signal strength determines the intensity of HP, we imitated slow HP using IL-7 or IL-15 and fast HP using a combination of IL-7 or IL-15 with anti-CD3 antibodies, cultivating Treg cells with peripheral blood mononuclear cells (PBMCs) at a 1:1 ratio. We used peripheral blood from 14 patients with rheumatoid arthritis and 18 healthy volunteers. We also used anti-CD3 and anti-CD3 + IL-2 stimulation as controls. The suppressive activity of Treg cells was evaluated in each case by the inhibition of the proliferation of CD4+ and CD8+ cells. The phenotype and proliferation of purified CD3+CD4+CD25+CD127lo cells were assessed by flow cytometry. The suppressive activity of the total pool of Tregs did not differ between the rheumatoid arthritis and healthy donors; however, it significantly decreased in conditions close to fast HP when the influence of HP cytokines was accompanied by anti-CD3 stimulation. The Treg proliferation caused by HP cytokines was lower in the rheumatoid arthritis (RA) patients than in the healthy individuals. The revealed decrease in Treg suppressive activity could impact the TCR landscape during lymphopenia and lead to the proliferation of potentially self-reactive T cell clones that are able to receive relatively strong TCR signals. This may be another explanation as to why lymphopenia is associated with the development of autoimmune diseases. The revealed decrease in Treg proliferation under IL-7 and IL-15 exposure can lead to a delay in Treg pool reconstitution in patients with rheumatoid arthritis in the case of lymphopenia.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 237.1-238
Author(s):  
M. Rosenzwajg ◽  
R. Lorenzon ◽  
P. Cacoub ◽  
F. Pitoiset ◽  
S. Aractingi ◽  
...  

Background:Regulatory T cells (Tregs) prevent autoimmunity and control inflammation. As low-dose interleukin-2 (ld-IL2) expands and activates Tregs, it has a broad therapeutic potential for any autoimmune or inflammatory disease (AIID). We performed a disease-finding “basket trial” (TRANSREGNCT01988506) in patients affected by one of 11 different AIID and reported the outcome of the first 46 patients (Rosenzwajg et al, ARD 2019).Objectives:Here we analyzed and discussed results from deep immunophenotyping, of 78 patients, to comprehensively study the effect of ld-IL2 on the immune system of patients affected by various AIIDMethods:We performed a prospective, open label, phase I-IIa study in 78 patients with a mild to moderate form of one of 13 selected AIID. All patients received ld-IL2 (1 million IU/day) for 5 days, followed by fortnightly injections for 6 months. Deep immunophenotyping was performed before and after 5 days of ld-IL2.Results:ld-IL2 significantly expands both memory Tregs as well as naïve Tregs, including recent thymic emigrant Tregs. It also activates Tregs as demonstrated by the significantly increased expression of HLA-DR, CD39, CD73, GITR, CTLA-4. Similar results were observed across the different AIID.Conclusion:ld-IL2 “universally” improves Treg fitness across 13 autoimmune and inflammatory disease.References:[1]Rosenzwajg M#, Lorenzon R#, Cacoub P, Pham HP, Pitoiset F, El Soufi K, RIbet C, Bernard C, Aractingi S, Banneville B, Beaugerie L, Berenbaum F, Champey J, Chazouilleres O, Corpechot C, Fautrel B, Mekinian A, Regnier E, Saadoun D, Salem JE, Sellam J, Seksik P, Daguenel-Nguyen A, Doppler V, Mariau J, Vicaut E, Klatzmann D. Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a single, open clinical trial. Ann Rheum Dis. 2019 Feb;78(2):209-217. doi: 10.1136/annrheumdis-2018-214229. Epub 2018 Nov 24.Disclosure of Interests:Michelle Rosenzwajg: None declared, Roberta Lorenzon: None declared, Patrice cacoub: None declared, Fabien Pitoiset: None declared, Selim Aractingi: None declared, Beatrice Banneville Speakers bureau: Lilly, Novartis, Laurent Beaugerie: None declared, Francis Berenbaum Grant/research support from: TRB Chemedica (through institution), MSD (through institution), Pfizer (through institution), Consultant of: Novartis, MSD, Pfizer, Lilly, UCB, Abbvie, Roche, Servier, Sanofi-Aventis, Flexion Therapeutics, Expanscience, GSK, Biogen, Nordic, Sandoz, Regeneron, Gilead, Bone Therapeutics, Regulaxis, Peptinov, 4P Pharma, Paid instructor for: Sandoz, Speakers bureau: Novartis, MSD, Pfizer, Lilly, UCB, Abbvie, Roche, Servier, Sanofi-Aventis, Flexion Therapeutics, Expanscience, GSK, Biogen, Nordic, Sandoz, Regeneron, Gilead, Sandoz, Julien Champey: None declared, Olivier Chazouilleres: None declared, Christophe Corpechot: None declared, Bruno Fautrel Grant/research support from: AbbVie, Lilly, MSD, Pfizer, Consultant of: AbbVie, Biogen, BMS, Boehringer Ingelheim, Celgene, Lilly, Janssen, Medac MSD France, Nordic Pharma, Novartis, Pfizer, Roche, Sanofi Aventis, SOBI and UCB, Arsene Mekinian: None declared, Elodie Regnier: None declared, david Saadoun: None declared, Joe-Elie Salem: None declared, Jérémie SELLAM: None declared, Philippe Seksik: None declared, David Klatzmann Consultant of: ILTOO Pharma


Sign in / Sign up

Export Citation Format

Share Document