scholarly journals Peripheral Blood Plasmacytoid Dendritic Cells at Day 100 Can Predict Outcome after Allogeneic Stem Cell Transplantation

2015 ◽  
Vol 21 (8) ◽  
pp. 1431-1436 ◽  
Author(s):  
Zinaida Peric ◽  
Xavier Cahu ◽  
Florent Malard ◽  
Eolia Brissot ◽  
Patrice Chevallier ◽  
...  
2015 ◽  
Vol 21 (7) ◽  
pp. 1223-1229 ◽  
Author(s):  
Matheus Vescovi Gonçalves ◽  
Mihoko Yamamoto ◽  
Eliza Yurico Sugano Kimura ◽  
Vergílio Antônio Rensi Colturato ◽  
Mair Pedro de Souza ◽  
...  

Blood ◽  
2008 ◽  
Vol 111 (2) ◽  
pp. 534-536 ◽  
Author(s):  
Markus Mezger ◽  
Michael Steffens ◽  
Melanie Beyer ◽  
Carolin Manger ◽  
Johannes Eberle ◽  
...  

Patients after allogeneic stem-cell transplantation (alloSCT) have an increased risk for invasive aspergillosis (IA). Here, recipients of an allograft with IA (n = 81) or without IA (n = 58) were screened for 84 single nucleotide polymorphisms in 18 immune relevant genes. We found 3 markers in chemokine (C-X-C motif) ligand 10 (CXCL10, 4q21, 11 101 C > T, P = .007; 1642 C < G, P = .003; −1101 A < G, P = .001) significantly associated with an increased risk of developing IA. Furthermore, immature dendritic cells (iDCs) exposed to Aspergillus fumigatus germlings showed markedly higher CXCL10 expression, if carrying the wild type genotype, compared with the “CGAG” high risk haplotype. In addition, serum from patients with proven/probable IA showed increased serum levels of CXCL10, compared with immunocompromised patients without IA. Thus, polymorphisms in CXCL10 determine chemokine secretion by iDCs upon exposure to A fumigatus and most likely thereby genetically determine the risk of IA after alloSCT.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5206-5206
Author(s):  
Djordje Atanackovic ◽  
Yanran Cao ◽  
Christiane Faltz ◽  
Katrin Bartels ◽  
Christine Wolschke ◽  
...  

Abstract BACKGROUND: Immunosuppressive CD4+Foxp3+ T regulatory cells (Treg) play a vital role in immune regulation. Thus, Treg contribute to the prevention of autoimmune disease and graft-versus-host reactions following allogeneic stem cell transplantation (alloSCT) but also to the inhibition of effective anti-tumor T cell responses. It has previously been suggested that the frequency of Treg is increased in the peripheral blood of patients with multiple myeloma (MM). However, little is known about the presence of Treg in the bone marrow and it is unclear whether allogeneic stem cell transplantation might deplete Treg from this immune compartment. METHODS: In the present study, we analyzed percentages of CD4+Foxp3+ Treg as well as Treg expression of CD45RA and CCR7 in the bone marrow (BM) and in the peripheral blood of MM patients who had received alloSCT (N=42), in newly diagnosed MM patients (N=18), and in healthy controls (N=15) using flow cytometry. In addition, we performed inhibition assays in order to test the functional relevance of peripheral and BM-residing Treg. RESULTS: While newly diagnosed MM patients and healthy controls showed no significant difference in the proportions of CD4+Foxp3+ Treg in the bone marrow, percentages of BM-residing CD4+Foxp3+ T regulatory cells were markedly higher (p<0.001 and p<0.01) in patients post alloSCT (3.3±0.3%) than in normal BM (1.0±0.3%) or in BM of untreated MM patients (1.8±0.4%). In both groups of patients (p<0.05) as well as in the healthy controls (p<0.001) percentages of Treg were higher in the peripheral blood than in the bone marrow. While there were no differences regarding the percentages of peripheral Treg between the remaining groups, patients post alloSCT had higher percentages of peripheral Treg than newly diagnosed patients (5.6±0.8 vs. 3.2±0.7%, p<0.05). More than 90% of these donor-derived peripheral and BM-residing Treg expressed a memory T cell phenotype, being negative for CD45RA and CCR7. Importantly, peripheral as well as BM-residing Treg of patients post alloSCT were capable of inhibiting the proliferation of autologous non-Treg CD4+ T cells. CONCLUSION: Our study demonstrates for the first time an increased frequency of immunosuppressive Treg in the bone marrow of MM patients. Remarkably, in our patients these memory-type Treg were all donor-derived and led to an efficient replenishment of Treg in the periphery. These Treg might be necessary for the prevention of graft-versus-host disease in the transplanted MM patients, however, they might also contribute to the failure of an effective graft-versus-myeloma effect in the majority of the patients.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 340-340
Author(s):  
Martin Bornhaeuser ◽  
Uta Oelschlaegel ◽  
Gesine Bug ◽  
Uwe Platzbecker ◽  
Karin Lutterbeck ◽  
...  

Abstract Relapse of hematological malignancy remains a major complication after allogeneic stem cell transplantation. This is especially true for patients receiving minimal or reduced-intensity conditioning therapy. Analysis of donor chimerism (DC) is an important diagnostic tool to assess the risk of relapse after allogeneic stem cell transplantation, especially in patients lacking a specific marker suitable for monitoring of minimal residual disease. The sensitivity of a standard PCR assay amplifying short-tandem-repeat sequences can be improved significantly by investigating sorted CD34+ peripheral blood cells. We prospectively compared the serial analysis of DC in selected CD34+ cells and unmanipulated whole blood (WB) within a randomised study in 131 patients with CD34+ hematological malignancies (AML, ALL and MDS) surviving more than 100 days after transplantation. The primary end-point was the association between decreasing CD34+ DC and haematological relapse. Whenever a decreasing CD34+ or WB DC was confirmed and no signs of active GvHD were present, a rapid taper of CsA or tracolimus (50% every 5–7 days) was suggested. If no GvHD occurred within 14 days after the stop of CsA or tacolimus, patients were scheduled to receive donor lymphocyte infusions (DLI) in incremental doses. The cumulative incidence of relapse was significantly increased in patients with decreasing or incomplete CD34+ DC (62% vs. 38%, p=0.01). This was associated with a lower probability of overall survival (20% vs. 39%, p=0.03). The interval between the decrease in CD34+ DC and hematological relapse was 35 days (range 0–567) in the study group compared to only 8 days (range 0–63) in the control group monitored by WB DC analysis (p=0.05). The median time between a decrease in CD34+ DC and WB DC was 14 days (range, 0 to 445). Patients receiving preemptive therapy triggered by decreasing CD34+ DC had a significantly higher probability of disease-free survival compared to cases monitored and treated according to WB DC (19% vs. 0%, p=0.009). Multivariate analysis revealed age, disease-risk and decreasing CD34+ DC as independent risk-factors for overall survival in the study group. In summary, we could demonstrate that the quantification of DC in CD34+ selected cells is a sensitive method to predict relapse and survival after allogeneic SCT. Although this technology opens a window for preemptive therapy, new treatment approaches have to be employed to improve the overall outcome of patients with recurrence of residual disease after allogeneic stem cell transplantation.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 867-867
Author(s):  
Pablo A Ramirez ◽  
Claudio Brunstein ◽  
Brian Miller ◽  
Todd E. DeFor ◽  
Daniel J. Weisdorf

Abstract Abstract 867 Introduction: Several reports have shown that despite prompt neutrophil engraftment after allogeneic stem cell transplantation, there can be delayed platelet recovery, especially in umbilical cord blood (UCB) stem cell transplant recipients. This dichotomy in platelets and neutrophils engraftment is poorly explained. In addition, the risk factors and complications associated with delayed platelet recovery are not clearly identified. Methods: We conducted a retrospective analysis to characterize the frequency of delayed platelet recovery and its associated risk factors and related complications. All allografts at the University of Minnesota between 2000 and 2005 were included. Results: 875 patients with hematologic malignancies or benign disorders were included. Myeloablative conditioning was used in 576 (66%) patients and non-myeloablative conditioning in 299 (34%) patients. 343 (39%) patients received related donor and 532 (61%) unrelated grafts. The source of the graft was bone marrow in 226 (26%) patients, peripheral blood in 255 (29%) patients and UCB in 394 (45%) patients. 150 (17%) patients were excluded due to early death (n=87, 10%), graft failure (n=62, 7%) and second transplant without engraftment (n=1). The 60 day cumulative incidence of platelet recovery by donor type was UCB single cord 39% (n=180, CI 31-47%), UCB double cord 40% (n=206, CI 32-48%), URD mismatched 57% (n=28, CI 39-75%), URD matched 56% (n=117, 48-64%) and sibling 74% (n=319, 65-85%). Overall, 232 (32%) had delayed platelet recovery (platelets<50K by day 60) and 475 (67%) had successful recovery (platelets>50K by day 60). Cox regression analysis showed that variables significantly associated with delayed platelet recovery were donor type (UCB RR 0.3 [CI 0.2-0.35], p<0.01 vs sibling RR 1.0), ABO match (major mismatch RR 0.8 [CI 0.6-1.0], p<0.01 vs matched RR 1.0), CMV status (positive RR 0.8 [CI 0.6-1.0], p=0.04 vs negative RR 1.0) and the vs negative RR 1.0). Transplant related mortality (TRM) at 12 months was also higher in patients with delayed platelet recovery compared to patients with successful platelet recovery (30% vs 11%) (p<0.01). Risk factors for TRM by proportional hazard regression were delayed platelet engraftment (RR 3.9 [2.6-6.0], p=<0.01 vs early RR 1.0), HLA mismatched unrelated donor (RR 3.7 [CI 2.5-7.1], p=0.01 vs sibling RR 1.0), severe aGVHD (RR 1.8 [CI 1.2-2.8], p=0.02 vs no severe RR 1.0) and age > 35 (RR 1.6 [CI 1.0-2.5], p=0.03 vs age < 35 RR 1.0). Successful platelet recovery was associated with a 77% 1 year survival versus only 59% in patients with delayed platelet recovery (p<0.01). Conclusions: These results suggest that delayed platelet recovery and poor graft function are common complications of allogeneic stem cell transplantation, especially after UCB grafts. Further study is needed to determine if modification of these associated risk factors will reduce the risks of severe complications and improve survival. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2096-2096
Author(s):  
Alan M Hanash ◽  
Sean M. Devlin ◽  
Molly Maloy ◽  
Kristina M. Knapp ◽  
Vincent A. Miller ◽  
...  

Abstract Disease relapse remains the greatest cause of mortality following allogeneic stem cell transplantation (SCT). Improved predictive markers are needed to identify patients most likely to benefit from SCT. Several mutations reported recently in MDS and AML have potential prognostic importance, however the relevance of these mutations to clinical outcome after SCT is poorly understood. In order to evaluate mutations present in transplant patients and provide an initial assessment of their clinical significance, we performed next-generation sequencing of myeloid malignancies from 55 patients (23 MDS, 32 AML) treated with SCT at MSKCC from 2010-2013. Median recipient age was 56 (20-69); 22/55 patients were transplanted in remission. Stem cell sources were CD34-selected (36) or unmanipulated (14) peripheral blood, unmanipulated marrow (2), or cord blood (3). 40/55 allografts were HLA matched (20 related, 20 unrelated). Sequencing was performed on peripheral blood or marrow aspirates in patients with >20% blasts (AML) or >20% dysplastic cells (MDS). Adaptor ligated sequencing libraries were captured with two custom baitsets targeting 374 cancer-related genes and 24 genes often rearranged for DNA-seq, and 272 genes often rearranged for RNA-seq. Captured libraries were sequenced to high depth (Illumina HiSeq), averaging >590X for DNA and >20,000,000 total pairs for RNA. Statistics included cumulative incidence functions for relapse, Kaplan-Meier estimates for relapse-free survival (RFS), and outcome comparison with a permutation-based logrank test. Only mutations observed in at least 5 patients were analyzed. No adjustments were made for multiple comparisons. Median follow-up of survivors was 16.2 months (5.5-32.8). We detected genetic variants in each patient, suggesting the utility of this approach for identifying somatic mutations to track minimal residual disease (MRD) post-SCT. Six patients had known FLT3 mutations detected by a CLIA-certified test; all 6 of these mutations were identified by the sequencing platform. In addition, 3 FLT3 mutations and 1 FLT3 amplification were identified in patients who previously tested FLT3 negative. We identified 13 patients with IDH mutations (5 IDH1, 8 IDH2), eight with NPM1 mutations (all in AML), and 10 with DNMT3A mutations. We identified MAP kinase pathway mutations in 12 patients, including NRAS (7), KRAS (5), and NF1 (4), and we identified mutations in novel genes previously implicated in MDS/AML, including STAT4, CASP8, APC, and ALK. We next evaluated if specific mutations were associated with SCT outcome. Patients with IDH mutations (all of whom had normal karyotype) demonstrated significantly less relapse than patients with wild-type (WT) IDH (1 yr incidence: 0% vs 29%, p=.027, Fig 1). This translated into improved RFS (p=.037) in patients with IDH mutant AML (Fig 2). Treatment-related mortality (TRM) was similar with and without IDH mutations, suggesting the improved outcome was due to reduced relapse. For FLT3, 5/10 patients with FLT3 abnormalities relapsed. All 5 that relapsed were IDH WT. In contrast, IDH mutations were present in 4/5 FLT3+ AMLs that did not relapse, suggesting that IDH mutations may predict for improved SCT outcomes in patients with intermediate cytogenetic risk AML and in patients with FLT3 mutations. Mutant KRAS correlated with reduced overall survival in AML (p=.008), but the significance of this was unclear due to the absence of relapses and high TRM, including infection and GVHD, in this group. We also evaluated disease progression in 2 AML patients who relapsed post-transplant with archived samples collected pre-SCT and at relapse. In both patients we observed a distinct mutational profile pre and post-transplant consistent with clonal evolution. Of note, 1 patient gained a NF1 mutation post-SCT, while the other patient lost a NF-1 mutation, although when detected, both mutations were present at a frequency less than 10%. In summary, we performed mutational profiling in SCT patients using a novel high throughput platform, which allowed us to identify clinically relevant mutations, including some not detected by clinical laboratory testing. Notably, we found that IDH mutations may predict for favorable outcome after SCT, even in FLT3 mutant AML. These data suggest that mutational profiling can identify clinically relevant biomarkers pre-SCT and identify mutations for tracking MRD. Disclosures: Miller: Foundation Medicine, Inc: Employment. Lipson:Foundation Medicine, Inc: Employment. Stephens:Foundation Medicine, Inc: Employment. Otto:Foundation Medicine, Inc: Employment. Yelensky:Foundation Medicine, Inc: Employment. Nahas:Foundation Medicine, Inc: Employment. Wang:Foundation Medicine, Inc: Employment. Levine:Foundation Medicine, Inc: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document