scholarly journals Novel Simultaneous Single Cell mRNA and Protein Expression Profiling Identifies Distinct Treg and T Effector Signatures in the Bone Marrow of MDS Patients

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 ◽  
1991 ◽  
Vol 78 (8) ◽  
pp. 2139-2149 ◽  
Author(s):  
JH Antin ◽  
BE Bierer ◽  
BR Smith ◽  
J Ferrara ◽  
EC Guinan ◽  
...  

Seventy-one patients with hematologic malignancies received bone marrow from a histocompatible sibling (n = 48) or a partially matched relative (n = 23) that had been depleted of CD5+ T cells with either an anti-CD5 mooclonal antibody (MoAb) plus complement (anti-Leu1 + C) or an anti- CD5 MoAb conjugated to ricin A chain (ST1 immunotoxin [ST1-IT]). These patients received intensive chemoradiotherapy consisting of cytosine arabinoside, cyclophosphamide, and fractionated total body irradiation. Both anti-Leu1 + C and ST1-IT ex vivo treatments effectively depleted bone marrow of T cells (97% and 95%, respectively). Overall, primary and late graft failure each occurred in 4% of evaluable patients. The diagnosis of myelodysplasia was a significant risk factor for graft failure (P less than .001), and if myelodysplastic patients were excluded, there were no graft failures in major histocompatibility complex (MHC)-matched patients and 2 of 23 (8.7%) in MHC-mismatched patients. The actuarial risk of grade 2 to 4 acute graft-versus-host disease (GVHD) was 23% in MHC-matched patients and 50% in MHC- mismatched patients. In MHC-matched patients, acute GVHD tended to be mild and treatable with corticosteroids. Chronic GVHD was observed in 6 of 36 (17%) MHC-matched patients and none of 11 MHC-mismatched patients. There were no deaths attributable to GVHD in the MHC-matched group. Epstein-Barr virus-associated lymphoproliferative disorders were observed in 3 of 23 MHC-mismatched patients. The actuarial event-free survival was 38% in the MHC-matched patients versus 21% in the MHC- mismatched patients. However, if outcome is analyzed by risk of relapse, low-risk patients had a 62% actuarial survival compared with 11% in high-risk patients. These data indicate that the use of anti-CD5 MoAbs can effectively control GVHD in histocompatible patients, and that additional strategies are required in MHC-mismatched and high-risk patients.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3224-3224 ◽  
Author(s):  
Laura Vann ◽  
Milcah Larks ◽  
Christopher Flowers ◽  
Sagar Lonial ◽  
Jonathan Kaufman ◽  
...  

Abstract Background: Successful reconstitution of cellular immunity following allogeneic HPCT reduces the risk of relapse and confers protection against opportunistic infections. We performed an IRB-approved retrospective analysis of patients who underwent allogeneic HPCT and in whom the content of immune cells were measured in the graft and in post-transplant blood samples. Methods: The study population consisted of 122 patients with hematologic disorders (71 acute leukemia; 14 chronic leukemia; 18 lymphoma, 12 MDS; 3 aplastic anemia; and 4 other) who underwent HPCT with a non-T cell depleted graft from an HLA matched related (73) or unrelated (49) donor. 47 patients had low risk disease (AA, ALL CR1, AML CR1, CML CP1), while 75 had high risk disease (all others). The conditioning regimen was non-myeloablative in 38 (31%), included ATG in 18 (15%), and included TBI in 54 (44%). Peripheral blood was drawn at a median of 101 days post-HPCT and analyzed for T-cell subsets, B-cells, NK cells, and dendritic cells. Subjects were divided into three strata based upon the maximal value for the content of each cell subset in the blood. Univariate and multi-variable stepwise logistic regression analyses were performed to test the association of pre-transplant clinical factors, the cells in the graft, and the numbers of immune cells in the blood post-transplant with overall survival. Results: The estimated three-year survival for all subjects was 53%, with death in 49/122 patients (40%) due to progressive disease (37%), infection (29%), GVHD (20%), and other causes (14%). Univariate factors associated with death included high risk, age, the use of reduced intensity conditioning regimen, the use of TBI, the use of ATG during conditioning and the measurement of lower numbers of total T-cells, CD4+ T-cells, CD8+ T-cells, γδ T-cells, DC1 and DC2 in the peripheral blood during the first 200 days post-transplant. A multi-variable Cox model identified non-myeloablative conditioning (HR 2.2, 95% CI 1.2–3.9), TBI (HR 1.9, 95% CI 1.1–3.3), transplant risk strata (HR 1.9, 95% CI 1.0–3.6), and a blood CD3+ T-cell count of less than 600 cells/mcL (HR 1.8, 95% CI 1.2–2.5) as independent risk factors for post-transplant death. The presence of acute GVHD (all grades) or graft constituents was not significantly associated with survival. Limiting the study population to those subjects who survived at least 100 days showed that blood CD3+ T-cells, non-myeloablative conditioning, the use of TBI remained significantly associated with survival. Conclusions: Higher CD3+ counts in the early post-transplant period predict better survival. Patients who fail to achieve a blood CD3+ T-cell count of >600/mcL in the first 200 days post-transplant may be appropriate subjects for adoptive cellular immunotherapy. Low Risk Patients Low Risk Patients High Risk Patients High Risk Patients


1997 ◽  
Vol 15 (1) ◽  
pp. 44-51 ◽  
Author(s):  
K Seiter ◽  
E J Feldman ◽  
H D Halicka ◽  
F Traganos ◽  
Z Darzynkiewicz ◽  
...  

PURPOSE To determine the maximal-tolerated dose (MTD) of topotecan with cytarabine in acute leukemia patients, and to evaluate leukemia cell apoptosis in these patients. PATIENTS AND METHODS Fifty-three patients with acute leukemia not responsive to standard therapy were treated at eight dose levels of topotecan (2.5 mg/m2/d to 7.75 mg/m2/d). Topotecan was given as a 30-minute infusion daily with cytarabine 1 g/m2/d, both for 5 days. Using a flow-cytometric technique, the percent apoptotic cells in blood and bone marrow samples was determined, and the cell cycle distribution of the leukemic cells studied. RESULTS Oropharyngeal mucositis was dose-limiting. The MTD of topotecan was 4.75 mg/m2/d for 5 days in high-risk patients and 7.0 mg/m2/d for 5 days in low-risk patients. The mean percent apoptotic cells in the peripheral blood reached a peak of 18.8%, a median of 48 hours following the first dose of topotecan. Patients with higher S-phase fractions, either before treatment or following cytarabine, were more likely to achieve bone marrow aplasia than those with lower S-phase fractions (P = .01 and P < .05, respectively). Clinical responses were seen in four of 39 patients with acute myelogenous leukemia (AML; of whom 32 had received prior high-dose cytarabine), three of six with acute lymphoblastic leukemia (ALL), and one of eight with chronic myelogenous leukemia in blast phase (CML-BP). CONCLUSION The recommended phase II dose of topotecan with intermediate-dose cytarabine is 4.75 mg/m2/d for high-risk patients and 7.0 mg/m2/d for low-risk patients. The percentage of cells in S phase was important in determining response to treatment.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1903-1903
Author(s):  
Vivian H Y Lee ◽  
Erica Peterson ◽  
Leslie Zypchen ◽  
Lynda M Foltz

Abstract Abstract 1903 Poster Board I-926 Introduction: The discovery of the JAK2 V617F gene mutation has significantly altered the clinical diagnostic approach to the myeloproliferative neoplasms, reflected in the revised 2008 WHO diagnostic criteria. Both the 2001 and 2008 diagnostic criteria for essential thrombocythemia (ET) require a bone marrow biopsy showing megakaryocytic proliferation to make a diagnosis of ET. Published expert opinion based on clinical studies suggests that ET patients > 60 years old or with history of thrombosis should be characterized as high risk and treated with hydroxyurea. It is unclear whether physicians in clinical practice utilize the WHO diagnostic criteria or follow expert treatment recommendations for ET. Methods: We conducted a retrospective chart review of all patients with a clinical diagnosis of ET made by a hematologist who were seen in clinic from 2006 to 2008 at two university teaching hospitals in Vancouver, Canada. Data collected included demographic information, thrombosis history, diagnostic tests performed and treatment administered. Testing for JAK2 V617F became locally available in 2006, so for assessment of diagnostic tests performed, patients were divided into cohorts of diagnosis pre-2006 and 2006–2008. Patients were characterized as high risk if > 60 y or history of thrombosis at the time of diagnosis. All other patients were considered low risk. Results: Diagnostic information was available for 116 patients diagnosed prior to the availability of testing for JAK2 V617F. 65% (75/116) of patients in this cohort had a bone marrow biopsy performed (table 1). 44 patients received a new diagnosis of ET from 2006–2008. Only 48% (21/44) patients in this cohort had a bone marrow biopsy performed, significantly less than in the historical cohort (p = 0.019). 41/44 had JAK2 V617F testing performed: 41% (17/41) were JAK2 V617F negative, 56% (23/41) positive and 1 equivocal. Bone marrow biopsy was performed in 59% (10/17) of JAK2 V617F negative patients and 39% (9/23) of JAK2 V617F positive patients (p = 0.055) (table 1). Bone marrow biopsy was also performed in 1 patient with equivocal JAK2 V617F testing and 1 patient not tested for JAK2 V617F. 170 patients diagnosed with ET were seen in follow up 2006–2008. 64% (109/170) were high risk due to age > 60 y or history of thrombosis. The remaining 36% (61/170) were considered low risk. Hydroxyurea was used preferentially over anagrelide for treatment of ET (table 2). Only 76% of high risk patients were receiving cytoreductive treatment. 23% of low risk patients received cytoreductive treatment. ASA was prescribed to 89% of high risk and 79% of low risk patients. Conclusion: Despite the requirement for a bone marrow biopsy to meet the WHO criteria for ET, hematologists performed a bone marrow biopsy in less than half of patients they diagnosed with ET since 2006. Hematologists performed bone marrow biopsy less frequently after JAK2 V617F testing became available, particularly in JAK2 V617F positive patients. A substantial portion of high risk patients (24%) were receiving no cytoreductive therapy, contrary to expert recommendation. Further study is required to understand the barriers to implementing treatment recommendations in clinical practice. This study highlights the challenges in translating published diagnostic criteria and treatment guidelines into changes in patient care. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1991 ◽  
Vol 78 (8) ◽  
pp. 2139-2149 ◽  
Author(s):  
JH Antin ◽  
BE Bierer ◽  
BR Smith ◽  
J Ferrara ◽  
EC Guinan ◽  
...  

Abstract Seventy-one patients with hematologic malignancies received bone marrow from a histocompatible sibling (n = 48) or a partially matched relative (n = 23) that had been depleted of CD5+ T cells with either an anti-CD5 mooclonal antibody (MoAb) plus complement (anti-Leu1 + C) or an anti- CD5 MoAb conjugated to ricin A chain (ST1 immunotoxin [ST1-IT]). These patients received intensive chemoradiotherapy consisting of cytosine arabinoside, cyclophosphamide, and fractionated total body irradiation. Both anti-Leu1 + C and ST1-IT ex vivo treatments effectively depleted bone marrow of T cells (97% and 95%, respectively). Overall, primary and late graft failure each occurred in 4% of evaluable patients. The diagnosis of myelodysplasia was a significant risk factor for graft failure (P less than .001), and if myelodysplastic patients were excluded, there were no graft failures in major histocompatibility complex (MHC)-matched patients and 2 of 23 (8.7%) in MHC-mismatched patients. The actuarial risk of grade 2 to 4 acute graft-versus-host disease (GVHD) was 23% in MHC-matched patients and 50% in MHC- mismatched patients. In MHC-matched patients, acute GVHD tended to be mild and treatable with corticosteroids. Chronic GVHD was observed in 6 of 36 (17%) MHC-matched patients and none of 11 MHC-mismatched patients. There were no deaths attributable to GVHD in the MHC-matched group. Epstein-Barr virus-associated lymphoproliferative disorders were observed in 3 of 23 MHC-mismatched patients. The actuarial event-free survival was 38% in the MHC-matched patients versus 21% in the MHC- mismatched patients. However, if outcome is analyzed by risk of relapse, low-risk patients had a 62% actuarial survival compared with 11% in high-risk patients. These data indicate that the use of anti-CD5 MoAbs can effectively control GVHD in histocompatible patients, and that additional strategies are required in MHC-mismatched and high-risk patients.


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 ◽  
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 ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1562-1562
Author(s):  
Florian Nolte ◽  
Claudia Kunz ◽  
Stephanie Fey ◽  
Julia Obländer ◽  
Susanne Brendel ◽  
...  

Abstract Introduction Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal myeloid malignancies characterized by ineffective hematopoiesis and an increased risk of transformation into acute myeloid leukemia. Particularly early stage MDS are at least in part characterized by an increased apoptosis of myeloid and erythroid progenitors that causes peripheral cytopenia. APG101 is a glycosylated fusion protein consisting of the extracellular domain of human CD95 (Fas receptor) and the Fc domain of human IgG1. APG101 effectively binds to the CD95 ligand (CD95L) expressed on effector cells as well as to functionally active ligand in solution, by that blocking the interaction between CD95 and its ligand. The aim of our study was to evaluate whether APG101 treatment of primary CD34+ reduces the apoptotic rate and improves the differentiation capacity of these cells. Methods Bone marrow cells were obtained during routine bone marrow aspiration after all patients gave their written informed consent. Isolated primary CD34+ cells from 11 MDS patients were cultured in complete supplemented IMDM medium for 6 days with increasing concentrations of APG101 (1 µg/mL, 3 µg/mL, 10 µg/mL, 30 µg/mL, 100 µg/mL, 200 µg/mL, 300 µg/mL). After incubation time, cells were multicolor- stained with the following dye combination: Annexin-FITC + CD235a-PE + CD34-PECy7 + CD71-APC + 7-AAD and analyzed immediately on a flow cytometer (FACSCanto, BD Bioscience, Heidelberg, Germany). Analysis of raw FACS data was done with the FACSDiva software. To analyze the differentiation capacity of CD34+ progenitors, methylcellulose assays were performed in parallel to the aforementioned experiments. However, due to limited cell numbers, colony assays were performed on 9 MDS patients only. Cells were cultured in triplicates with increasing concentrations of APG101 for 14 days. Colonies were counted and the mean number of colonies was determined. Results Treatment of differentiating CD34+ cells with APG101 led to a decreased apoptosis in both CD34+ cells and CD71+ cells, respectively, indicated by decreased Annexin-FITC fluorescence. Interestingly, this effect was particularly seen at low APG101 concentrations (maximum of 10 µg/ml), while the effect was abrogated at higher APG101 concentrations. The anti-apoptotic effect was more pronounced in low risk MDS patients compared to high risk MDS patients. No effect was seen when the CD235a+ fraction of cells was analyzed. With regard to colony formation, an improvement of erythroid differentiation, indicated by an increase in CFU-E, was found in 3 out of 4 low risk patients (less than 5% blasts in the bone marrow). No effect was seen on erythroid differentiation in high risk patients (more than 5% blasts in the bone marrow). Conclusion APG101 shows promising in vitro activity in viable CD34+ cells with regard to inhibition of apoptosis and promotion of differentiation. The observation that the anti-apoptotic effect was more pronounced in low risk MDS patients as compared to high risk MDS patients supports the concept of increased apoptosis particularly in early stage MDS progenitors. Although the numbers in the differentiation experiments are small, we found a promising effect of APG101 on CFU-E formation at lower doses in patients with less than 5% bone marrow blasts. Moreover, the same dose-dependent effect was observed in the apoptosis assays. Since the activation of the CD95 pathway seems not only to be involved in apoptosis induction, but is also required for terminal erythroid differentiation in normal hematopoiesis, this dose-dependent effect might particularly reflect these ambivalent roles of CD95 and its ligand in both MDS and healthy hematopoiesis, repsectively. Disclosures: Kunz: APOGENIX GmbH: Employment. Fricke:APOGENIX GmbH: Employment.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2895-2895
Author(s):  
Alena Moudra ◽  
Sona Hubackova ◽  
Jiri Bartek ◽  
Zdenek Hodny ◽  
Anna Jonasova

Abstract Introduction Treatment with 5-azacytidine (5-AC) is indicated for high-risk MDS patients. Besides the inhibitory effects of 5-AC on DNA and RNA methylation, 5-AC has been recently shown to induce DNA damage and apoptosis in cultured cells. However, in vivo effects of 5-AC remain to be elucidated. Several recent publications implicate aberrant bone marrow (BM) microenvironment and inflammation-related changes in the occurrence and/or progression of the MDS. To provide more insights into this emerging concept, we assessed: i) the extent to which inflammation related cytokines may contribute to MDS progression, and ii) potential changes of cytokine abundance in response to 5-AC therapy. Patients and methods We have collected BM samples from 30 high-risk MDS patients (IPSS int II or IPSS high, 16 females, 14 males) treated by 5-AC at the Hematology Clinic, General University Hospital in Prague. Patients' mean age was 72y (range 55-85) and the WHO 2008 diagnoses were: 15 RAEB II, 5 RAEB I, 2 CMML II, 2 RCMD, 1 U-MDS/MPN and 5 AML/MDS with < 30% myeloblasts. We analyzed BM aspirates collected before 5-AC therapy and at day 7 after the completion of respectively the 4th and 8th cycle at which time initial response was also assessed. BM plasma was immediately separated from cells and kept in liquid nitrogen until the time of analysis. As controls we used BM samples from 4 healthy subjects (males, mean age 42y, range 32-59), along with BM samples from 6 low-risk 5q- MDS patients (females, mean age 68y, range 46-80). For the presence of inflammation-related cytokines, BM plasma was analyzed using Human Inflammation 11-Plex (IFNγ, IL1α, IL1β, IL6, IL8, IL10, IL12p70, IL27, IP10, MCP1, and TNFα; YSLBio) via flow cytometry. For the purposes of data analysis, 5-AC treated patients were divided into 2 groups depending on their response to therapy: responders (hematological improvement, partial remission, complete remission, complete remission with incomplete BM recovery) and non-responders (stable disease, progressive disease). Obtained cytokine values were transformed using Box-Cox procedure, and repeated measurements, analyzed using linear models with mixed effects. Comparisons of 5-AC-treated patients, 5q- MDS low-risk patients and controls were subjected to a Kruskal-Wallis test. P-values less than 0.05 were considered as statistically significant. Analyses were conducted using the R statistical package, version 3.1.2, R Core Team (2014). Results Among the 11 cytokines analyzed, 3 (IL27, IP10 and MCP1) displayed significantly altered levels when comparing high-risk 5-AC treated patients, low-risk MDS patients and healthy controls. First, IL27 was elevated in low-risk MDS in comparison to 5-AC or healthy controls (p = 0.041); Figure 1. For IP10, 5-AC MDS patients before therapy showed higher levels (p = 0.005) compared to the low-risk group and healthy controls, respectively. The difference for IP10 was also significant after 4 cycles of 5-AC therapy (p = 0.005), but insignificant after 8 cycles (p = 0.288). The difference in IP10 levels between the two treated groups (4 vs. 8 cycles) were not significant, likely reflecting insufficient sample size, thereby masking the presumably lower levels of IP10 in responders (Figure 2). Further, the 5-AC treated patients showed higher levels of MCP1 than MDS low-risk patients and healthy controls, a difference apparent before therapy (p = 0.011), after 4 cycles (p = 0.003), but not after 8 cycles of therapy (p = 0.058). Also, MCP1 levels changed (p = 0.030) during the treatment, yet irrespective of clinical responses to therapy (Figure 3). Conclusions The IL27 level was higher in low-risk MDS patients compared to high-risk MDS 5-AC patients. Levels of IP10 and MCP1 were higher in high-risk MDS 5-AC patients. Levels of MCP1 changed significantly during the 8 cycles of 5-AC therapy. The observed correlation of IP10 with responses to 5-AC therapy should be further validated. Acknowledgment and Institutional support: This study was supported by grant from Internal Grant Agency of Ministry of Health of the Czech republic (Project NT14174-3) and by Institutional grant (Project RVO 68378050). Figure 1. Figure 1. Figure 2. Figure 2. Figure 3. Figure 3. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5390-5390
Author(s):  
Catherine M Claussen ◽  
Tammy Chuang ◽  
Jatin Shah ◽  
Hans Lee ◽  
Nina Shah ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is characterized by malignant plasma cell (PC) proliferation. Gene expression profiling (GEP) of CD138+ bone marrow PC has emerged as a new way to identify patients with worse clinical outcomes so that an individualized treatment approach can be undertaken in the clinic. The majority of data with GEP come from clinical trials. We aimed to evaluate the use of GEP in a standard clinical setting. Methods: We retrospectively searched our database of newly diagnosed MM patients with GEP completed prior to initial treatment. 35MM patients from April 2014 until June 2015 were identified and included in our analysis. GEP was performed through MyPRS® (Signal Genetics, Little Rock, AR). Fisher's exact test was used to evaluate the associations between complete response status and other categorical variables. The Wilcoxon rank sum test was used to evaluate the difference in continuous variables between patients that achieved a complete remission after 6 months of initial treatment and those who did not. Responses were assessed using IMWG criteria. Results: Median age was 60 (38-76). Patients presented with lytic lesions (60%), anemia (80%), kidney dysfunction (11%) and hypercalcemia (20%). All patients with known initial therapy were treated with bortezomib (n=29) or carfilzomib (n=1) based therapy. 10 patients had upfront autologous stem cell transplant. 18 patients had available response at 6 months. 37% (n=13) of patients were characterized as high risk by GEP, of which 46% (n=6) had the proliferation (PR) subtype. Most low risk patients had hyperdiploidy (HY) subtype (n=12, 55%). Patients with high risk GEP presented more often with complex karyotypes whereas low risk GEP patients most often had normal or hyperdiploid karyotypes. FISH abnormalities that are usually present in high risk myeloma were also present in patients classified as GEP low risk (Table 1). At 6 months after diagnosis, lower baseline total protein, serum M-spike, serum free light chain ratio and serum kappa light chain levels were significantly associated with achieving a stringent complete response (sCR) (p<0.05). High risk patients were more likely to achieve a CR (n= 6/10, 60%) than low risk patients (n=5/16, 31%) at 6 months. Despite this, high risk patients seemed more likely to lose CR and relapse or die. Within one year of diagnosis, one patient relapsed after achieving a VGPR (high risk, PR subtype) and one patient died due to MM progressive disease after achieving an sCR (high risk, CD-1 subtype) (Table 1). Conclusion: In a standard clinical setting, GEP seems to identify MM patients that are at higher risk of adverse clinical outcomes early after diagnosis. GEP may be an adjunct to cytogenetics/FISH in MM risk stratification, as high risk FISH abnormalities were also found in low risk GEP patients. Larger studies with longer follow up may help address the particular role of GEP in the individualized treatment of MM. Figure 1. Figure 1. Disclosures Orlowski: Array BioPharma: Consultancy, Research Funding; Acetylon: Membership on an entity's Board of Directors or advisory committees; Onyx Pharmaceuticals: Consultancy, Research Funding; Spectrum Pharmaceuticals: Research Funding; Celgene: Consultancy, Research Funding; Genentech: Consultancy; Millennium Pharmaceuticals: Consultancy, Research Funding; Forma Therapeutics: Consultancy; BioTheryX, Inc.: Membership on an entity's Board of Directors or advisory committees; Janssen Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Consultancy, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document