GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Society of Hematology  (10)
  • 1
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4594-4594
    Abstract: Background: Since the first publication at the John Hopkins Hospital, (Luznik et al. BBMT 2008), Haploidentical T-cell replete Stem Cell Transplantation (HaploSCT) with post transplantation cyclophosphamide (PT-Cy) has become a reproducible and feasibility therapeutic option for many patients (pts) with hematologic malignancies , notably because of the low incidence of GVHD and infections, without increased graft failure. Initially, bone marrow (BM) was considered as the favorite source of hematopoietic stem cells in order to minimize the risk of GVHD. In a retrospective study, we previously showed no difference in terms of increased events (GVHD, NRM), regardless of the hematopoietic stem cell source (PBSC or BM) (Castagna et al. BMT 2014). Some recent publication seems to confirm this observation (Sugita J. BBMT 2015 , Solomon R. Adv in Hem. 2016), although no randomized study so far has been conducted. Here, we retrospectively analyze the incidence and the characteristics of GVHD in the setting of HaploSCT using PT-Cy, after infusion of PBSC. Methods: Inclusion criteria were: adult pts with hematologic malignancies receiving a PBSC HaploSCT from 2012 to 2015 in 4 centers (3 in France and 1 in Italy) with PT-Cy as part of the GVHD prophylaxis. PBSC infusion at day 0 was followed by PT-Cy 50 mg/kg on days +3 and +4 in association with calcineurin inhibitors (cyclosporine A or Tacrolimus) and mycophenolate mofetil (MMF), started at day +5. All patients received G-CSF support from day+5 until neutrophil recovery. Study end points were the cumulative incidences of acute (a) and chronic (c) GVHD, with a specific organ grading evaluation, non-relapse mortality (NRM), relapse (CIR) as well as progression free (PFS) and overall survival (OS). Additionally, we analyzed the composite endpoint "GVHD and relapse free survival" (GFRS) for which the occurrence of relapse, death or severe chronic GVHD was considered as relevant events. Correlation between CD 34+ and CD 3+ and the incidence of aGVHD and cGVHD was studied by a linear continuous variable. Results: Between March 2012 and December 2015, 192 pts with a median age of 57 years (range: 16-73) received T-cell replete PBSC HaploSCT for hematologic malignancies (myeloid: n= 55%; lymphoid: n=45%) in 4 centers. Patient's characteristics are shown in table 1. Pts received non myeloablative (according to Baltimore regimen) or busulfan-based reduced intensity conditioning, in 56% and 44% of cases, respectively. All, but 3 pts, engrafted, with a median time of 19 days (range, 14-47) to neutrophil recovery (ANC 〉 500 x106/L) and 22 days (range, 14-252) to platelet recovery (PLT 〉 20 G x 109/L). The median CD34+ x 106/Kg and CD3+ x 106/Kg cells infused were 5.5 (range, 1.5-14.8) and 404 (range, 38-704), respectively. No relevant correlation was observed between the CD34+ and CD3+ infused cells and the incidence of GVHD, studied by linear continues variable. We noted only a trend to develop severe cGVHD with an increasing number of CD3+ cells infused. This result has to be considered with caution, because of the small events (6 pts affected by severe cGVHD). Complete donor T cell chimerisme was evaluable in 162 pts (83%) and achieved by day +30. The incidence of aGVHD was 38% at 100 days (all grades), whereas grade II-IV and III-IV were 24% and 10%, respectively. Concerning patients with aGVHD grade 2-4, the most affected organ was skin (19%), followed by gut (9%) and liver (2%). The incidence of 3-year cGVHD according to NIH classification was 15% (all grades). Three percent of pts developed severe cGVHD (lung n=1; liver, n=1). The most frequent involved organs were skin and mucosae (70%). No patient showed gut cGVHD. Finally, in univariate analysis, busulfan-based conditioning seems to negatively impact on severe cGVHD (p = 0.03; HR=3.37 [1.09 -10.46]) After a median follow up of 20 (range, 4 - 52) months, NRM at 100 days and 1 year was 10% and 20%, respectively. Three-year OS, PFS, CIR and GRFS were 63%, 55%, 25% and 49%, respectively. Conclusion: This retrospective study shows a very low incidence of severe cGVHD after HaploSCT even with PBSC as stem cell source, suggesting that the use of PT-Cy may overcome the anticipated increased incidence of cGVHD, contrary to as previously reported in the HLA identical setting (Mohty et al. Leukemia 2003). Similar to HLA identical sibling and unrelated donor transplantation, the most frequent organs involved are skin and mucosae Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood Advances, American Society of Hematology, Vol. 3, No. 24 ( 2019-12-23), p. 4238-4251
    Abstract: Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive leukemia for which we developed a nationwide network to collect data from new cases diagnosed in France. In a retrospective, observational study of 86 patients (2000-2013), we described clinical and biological data focusing on morphologies and immunophenotype. We found expression of markers associated with plasmacytoid dendritic cell origin (HLA-DRhigh, CD303+, CD304+, and cTCL1+) plus CD4 and CD56 and frequent expression of isolated markers from the myeloid, B-, and T-lymphoid lineages, whereas specific markers (myeloperoxidase, CD14, cCD3, CD19, and cCD22) were not expressed. Fifty-one percent of cytogenetic abnormalities impact chromosomes 13, 12, 9, and 15. Myelemia was associated with an adverse prognosis. We categorized chemotherapeutic regimens into 5 groups: acute myeloid leukemia (AML)–like, acute lymphoid leukemia (ALL)–like, lymphoma (cyclophosphamide, doxorubicin, vincristine, and prednisone [CHOP])–like, high-dose methotrexate with asparaginase (Aspa-MTX) chemotherapies, and not otherwise specified (NOS) treatments. Thirty patients received allogeneic hematopoietic cell transplantation (allo-HCT), and 4 patients received autologous hematopoietic cell transplantation. There was no difference in survival between patients receiving AML-like, ALL-like, or Aspa-MTX regimens; survival was longer in patients who received AML-like, ALL-like, or Aspa-MTX regimens than in those who received CHOP-like regimens or NOS. Eleven patients are in persistent complete remission after allo-HCT with a median survival of 49 months vs 8 for other patients. Our series confirms a high response rate with a lower toxicity profile with the Aspa-MTX regimen, offering the best chance of access to hematopoietic cell transplantation and a possible cure.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 160-160
    Abstract: Abstract 160 Background: Until the early 2000s, treatment prior to transplant in candidates to allo-ST varied according to cytogenetic score and percentage of BM blasts, from best supportive care (BSC) to AML-type chemotherapy (intensive CT). Recently AZA has become a reference treatment of IPSS higher risk MDS not candidates for allo-SCT, but its role prior to transplant in order to reduce the tumor burden remains uncertain. Methods: We report a retrospective study on 470 consecutive MDS patients who underwent allo-SCT between Jan 1999 and Dec 2009 in 26 French and Belgian centers. Inclusion criteria were: age 〉 18 and allo-CST from either sibling (n=285) or HLA-A, -B, -C, -DRB1 and -DQB1 allele matched unrelated donor (10/10) (n=172) for MDS or AML/RAEB-t (with 20–30% BM blasts). Data quality was ensured using computerized discrepancy errors and vigorous on-site data verification of every single file. HLA matching was double-checked by the French Bone Marrow Donor Registry. Results: The first 405 files analyzed until now are presented, including 250 males and 155 females. At diagnosis, WHO was: 102 RA/RARS/RCMD, 136 RAEB-1, 146 RAEB-2 and 18 RAEB-t/AML; Cytogenetic IPSS were favorable (n=205), intermediate (n=104), unfavorable (n=86) and missing (n=10); 216 patients had IPSS Int-2 or High. One hundred and therty-three patients had progressed to a more advanced disease before allo-SCT. Prior to transplant, 77 patients had received AZA, either alone (AZA-alone-group; n=45) or AZA preceded or followed by intensive CT (AZA-chemo-group; n=32), generally due to failure of the first treatment given. The 328 remaining patients received BSC (n=162) or intensive CT (n=166) (no-AZA-group). In AZA groups, the drug was started 38 to 941 days after diagnosis (median 150) and discontinued 6 to 438 days before transplant (median 60) with a median number of 4 cycles (range 1–26). Overall, 178 patients (45%) were considered responders at transplant (in CR or PR), including 32/45 (71%) treated with AZA-alone, 19/32 (60%) with AZA-chemo, 6/162 (4%) with BSC and 121/166 (73%) with intensive CT, while 222 were transplanted with progressive disease (untreated, stable without hematological improvement, relapsed or refractory disease). Median age at allo SCT was 54 (range18–70). Patients received myeloablative (n=145) or nonmyeloablative (NMA) (n=260) conditioning with bone marrow (n=118) or PBSC (n=287) as source of stem cells. Compared with other treatment groups, patients belonging to AZA-alone-group were older (p =.025) and had more often Int-2 and high IPSS (p=.013) and poor cytogenetic IPSS (p 〈 .001) and received more often NMA conditioning (p =.005) from an unrelated donor (p=.007). As of April 1st, 2011, median FU was 4.6 years (range, 0.2–12.2). The estimated 3-year OS was 60%, 28%, 52% and 49% in the AZA-alone, AZA-chemo, BSC and intensive CT groups, respectively (p=.033); The estimated 3-year TRM was 13%, 29%, 34% and 20% in the AZA-alone, AZA-chemo, BSC and intensive CT groups, respectively (p=.055) and 3-year relapse was 31%, 41%, 29% and 38% in the AZA-alone, AZA-chemo, BSC and intensive CT groups respectively (p=.169). Multivariate analyses confirmed the influence of prior treatment with an unfavorable outcome in patients of the AZA-chemo group (who received CT before or after AZA) on OS and EFS (p=.003 and p=.0013, respectively), in spite of the fact that, 60% of the patients in that group had achieved at least PR at transplant. Conclusion: With the goal of down-staging underlying disease before allo-SCT, AZA treatment appears to be a valid therapeutic approach, but mainly in patients receiving AZA alone since allo-SCT in patients who required both AZA and chemotherapy had less satisfactory outcomes, possibly reflecting additional toxicity and/or more resistant disease. Disclosures: Michallet: Celgen: Honoraria. Mohty:celgene: Honoraria. Fenaux:Celgene: Honoraria, Research Funding. Yakoub-Agha:celgene: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 122, No. 14 ( 2013-10-03), p. 2402-2411
    Abstract: Caloric restriction reduces Mcl-1 expression and sensitizes lymphoma cells to ABT-737 in vivo. Caloric restriction mimetics can sensitize lymphomas to ABT-737–induced death independently of p53 and of the main BH3-only proteins.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1638-1638
    Abstract: Background: Nivolumab demonstrated remarkable activity in patients with relapse or refractory (R/R) Hodgkin lymphoma (HL). However, long term efficacy and the need for a consolidation with allogenic stem cell transplantation remain unclear. Patient and method: We retrospectively analyzed 78 patients with R/R HL treated with nivolumab in the French Early Access Program and compared their outcome according to subsequent alloHSCT. Results: After a median follow-up of 31.5 months, the best overall response rate was 64%, including 37.3% complete response (CR). The median progression-free survival (PFS) was 12.1 months and median overall survival (OS) was not reached. At 3 years, PFS and OS rates were 35% and 65%, respectively. Patients reaching a CR upon nivolumab had a significantly longer PFS than those reaching a PR (median = not reached vs 10.1 months). In our cohort, 17 patients underwent consolidation with allogenic stem cells transplantation (alloHSCT) after nivolumab therapy (Figure 1). At the time of transplantation, 8 patients were in CR, 5 in partial response (PR) and 4 had progressed of whom 3 received a salvage therapy before alloHSCT. Interestingly, 6 out of 7 patients who were not in CR at the time of transplantation (5 PR and 1 progressive disease) converted into a CR after alloHSCT. At the time of analysis, 14 patients were alive and 13 remained disease-free after a median follow-up of 30.4 months. One-year OS and PFS from alloHSCT were 82% and 76%, respectively. Among responding patients (i.e. in CR or PR) after nivolumab monotherapy, those who underwent subsequent alloHSCT (N=13) had a better outcome than those who were not consolidated with alloHSCT (N=35) (Figure 2). In the transplanted group, none of the patients relapsed whereas in the non-transplanted group 60% of the patients relapsed (p 〈 0.001). In the transplanted group, all patients experienced graft-versus-host disease (GVHD), acute (N=14) and/or chronic (N=7) GVHD, including 7 patients with grade III-IV GVHD. At the time of analysis, GVHD had resolved in 9 out of 13 patients. Two patients experienced non-infectious febrile syndrome which resolved with corticosteroids and one patient experienced a sinusoidal obstructive syndrome. Two patients died, one from steroid-refractory GVHD and encephalitis, one from unexplained hemoptysis after experienced steroid-refractory GVHD. Conclusions: Although patients who achieve a CR upon anti-PD1 therapy may experience prolonged remissions, most R/R HL patients treated with anti-PD1 antibody eventually progress or relapse. Our study demonstrates unprecedented disease-free survival in patients undergoing consolidation with alloHSCT after anti-PD1 therapy. Interestingly, alloHSCT post anti-PD1 can convert incomplete responses into CR in most cases. Despite expected toxicities, alloHSCT after anti-PD1 therapy appears manageable and safe in most patients. Our results suggest that consolidation with alloHSCT may represent a good option in patients treated with anti-PD1, notably in patients who are unable to achieve a CR. Disclosures Herbaux: Gilead Sciences, Inc.: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria; Janssen: Consultancy, Honoraria. Stamatoullas:Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Ltd, Cambridge, MA, USA: Consultancy. Brice:bristol myers squibb: Consultancy, Honoraria. Houot:bristol myers squibb: Consultancy, Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 209-209
    Abstract: Allogeneic stem cell transplantation (AlloSCT) is a curative option for acute myeloid leukemia (AML) patients. In first complete remission (CR1), young patients ( 〈 60 years) with intermediate or unfavorable ELN disease risk are usually considered for AlloSCT. In contrast, because of the expected higher toxicity in older patients, the benefit of this treatment remains a matter of debate after 60 years, especially in the intermediate ELN risk group. In this multicenter analysis from the French Innovative Leukemia Organization (FILO), we investigated whether AlloSCT was beneficial for AML patients over 60 years old in CR1. Inclusion criteria were: patients between 60 and 70 years of age diagnosed with AML from 2007 to 2017; CR1 after intensive chemotherapy; ELN-2010 intermediate or unfavorable risk group. AlloSCT was evaluated as a time dependent variable in survival calculations and in a multivariate Cox model adjusted on age, ELN group, transplantation period and stratified by transplantation center. We also used a multistate model as follow: initial state for all patients was "No Allo - CR" with time 0 at the time of CR. From initial state, patients can transit to "Allo-CR", or move through 2 absorbing states (non-relapse death (No Allo-NRM) or relapse (No Allo-Relapse). Similarly, once transplanted (i.e. in the state "Allo-CR"), patients can move to "Allo-Relapse" or "Allo - NRM" when such events occurred. The model allows the dynamic prediction of probability for a patient to be in a specific state considering specific initial state and time. We analyzed 521 consecutive patients in 6 centers who matched inclusion criteria. Median age was 65 years (range: 60-70). ELN-2010 risk was intermediate and unfavorable in 376 (72%) and 145 (28%) patients, respectively. While all patients had a theoretical indication for AlloSCT in CR1, 199 (38%) were actually transplanted (129 (34%) and 70 (48%) in the intermediate and unfavorable risk group, respectively). In the whole cohort, AlloSCT as time-dependent variable significantly improved relapse-free survival ([RFS] at 5 years, No AlloSCT vs. AlloSCT: 14% vs. 47% p 〈 0.001) and overall survival ([OS] at 5 years, No AlloSCT vs. AlloSCT: 24% vs. 51% p 〈 0.001). In subgroup analysis based on ELN-2010 risk classification, AlloSCT significantly improved outcome of both ELN intermediate (No AlloSCT vs. AlloSCT: 5-y RFS: 16% vs. 50% p 〈 0.001; 5-y OS: 26% vs. 54% p 〈 0.001) and unfavorable (No AlloSCT vs. AlloSCT: 5-y RFS: 7% vs. 44% p 〈 0.001; 5-y OS: 17% vs. 46% p 〈 0.001) risk group patients (Figure A and B). By multivariate time-dependent Cox model, AlloSCT significantly decreased the risk of relapse (HR [95%CI]: 0.29 [0.20-0.41] p 〈 0.001) and increased the risk of NRM (HR [95%CI]: 2.61 [1.38-4.94] p = 0.003). This led to a significant advantage for AlloSCT in both RFS (HR [95%CI]: 0.48 [0.36-0.64] p 〈 0.001) and OS (HR [95%CI]: 0.60 [0.44-0.81] p = 0.001). This benefit was observed in both intermediate and unfavorable ELN-2010 risk groups, with lower risk of relapse (intermediate: HR [95%CI]: 0.30 [0.19-0.46] p 〈 0.001; unfavorable: HR [95%CI]: 0.37 [0.19-0.72] p = 0.004) and better OS (intermediate: HR [95%CI]: 0.67 [0.47-0.96] p = 0.028; unfavorable: HR [95%CI]: 0.51 [0.29-0.91] p = 0.022). Multistate model showed that 5 years after CR1, few patients were still alive in CR without AlloSCT (i.e. in the initial "No Allo-CR" state), whatever the ELN-2010 risk group (intermediate: 9%; unfavorable: 1% Figure C and D). Among patients who were transplanted, the probability for transiting to Allo-NRM state within 5 years post CR1 was 17% and 24% in intermediate and unfavorable ELN-2010 groups, respectively. Corresponding values for patients without AlloSCT transiting to No Allo-NRM state were 11% and 12%. Moreover, considering a landmark at 6 months after CR1, the multistate model showed that patients who received AlloSCT had lower probability of relapse at 5 years (22% and 33% in intermediate and unfavorable ELN-2010 groups, respectively) compared to those who did not (68% and 78% in intermediate and unfavorable groups, respectively). AlloSCT for CR1 AML patients over 60 years of age is routinely feasible and significantly improves outcome in both intermediate and unfavorable ELN-2010 risk groups. Less than 10% of patients are long term disease free survival without AlloSCT, even in intermediate risk group, supporting that AlloSCT remains the first curative option for these patients. Figure. Figure. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3261-3261
    Abstract: Background: Azacitidine (AZA) is the reference treatment for higher-risk MDS patients ineligible for intensive chemotherapy (IC) (Lancet Oncol 2009). It also improves overall survival (OS) in elderly AML patients with more than 30% marrow blasts ineligible for IC over conventional care (Dombret et al., EHA 2014). To date, no reliable biological marker predictive of AZA response has been reported. In a preliminary retrospective work in 32 patients, we found that quantification of BCL2L10 (an anti-apoptotic member of the Bcl-2 family of proteins) bone marrow mononuclear cells (BMMC) positive cells by flow cytometry (FCM) in HR-MDS patients represents a new potential biomarker for AZA response (Cluzeau et al. Oncotarget 2012). The aim of the present study was to validate those preliminary findings in a larger prospective multicenter cohort, analyzed blindly in 2 different laboratories. Methods: FCM was performed on fresh BMMC obtained at different times during AZA treatment: at treatment onset, after 3 or 6 cycles of AZA and at relapse, as previously described (Cluzeau et al., Oncotarget 2012) after several steps of fixation, permeabilization, and consecutive treatment with i) an anti-BCL2L10 antibody (Cell Signaling) and ii) a donkey anti-Rabbit FITC-antibody (Santa Cruz). All assays were performed in two different laboratories with two kinds of cytometers: Paris (Canto Becton Dickinson), Nice (Miltenyi Biotec). MDS and AML patients treated with AZA were prospectively included from 6 centers in this correlative study (clinicaltrial.gov: NCT 01210274). Response was assessed by IWG 2006 criteria for MDS or by Cheson et al (2003) for AML. Results: 75 MDS or AML patients were included. Median age was 73 years (range 35-91) and M/F was 37/38. 20%, 19%, 36% and 25% patients had RA, RAEB-1, RAEB-2 and AML respectively. IPSS was low, int-1, int-2 and high in 2%, 26%, 35% and 37% respectively. IPSS-R was very low, low, int, high and very high in 3%, 2%, 16%, 20% and 59% respectively. Patients were treated by AZA (75mg/m²/day, 7 days every 4 weeks) for a median number of 6 cycles (range 1-50). Overall response rate (ORR) was 60%, including 28% CR, 17% marrow (m) CR, 7% PR and 8% stable disease (SD) with hematologic improvement (HI). In MDS, the ORR was 57% (33 % CR, 11% mCR, 7% PR and 6% SD with HI). In AML, the ORR was 62% (23% CR, 23% mCR, 8% PR and 8% and SD with HI, based on MDS criteria). The median % of BCL2L10 positive cells was 9.5% (range 0-95) and no correlation was observed between % of BCL2L10 positive cells and marrow blasts. The median % of BCL2L10 positive cells was 30% (range 0-95) in non-responders and 10% (range 0-56) in responders (p=0.01). The response rate was 7% and 64% in patients with ≥ 50% vs 〈 50% BCL2L10 positive cells, respectively (p 〈 0.0001). Median OS after FCM analysis performed before or during AZA treatment was 5.8 months in the 11 patients with more than 50% versus 11.7 months in the 64 patients with less than 50% of BCL2L10 positive cells (p=0.03). In 8 patients studied sequentially before, during AZA treatment and at relapse, the % of BCL2L10 positive cells remained stable below 50% and increased above 50% few months before relapse. The best prognostic cut off value for BCL2L10 positive cells was 50%. Flow cytometry results were reproducible in the two laboratories, with two different cytometers. Conclusion: We confirmed in this larger prospective multicenter cohort that the percentage of BCL2L10 positive cells, analyzed in 2 different labs, is inversely correlated with response and survival after AZA treatment in both MDS and AML patients, the best prognostic cut-off value for BCL2L10 positive cells being 50%. Our flow cytometry assay is reproducible in different laboratories and can be performed routinely at diagnosis and during AZA treatment. A multivariate analysis including other prognostic factors of response and OS with AZA will be presented. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3470-3470
    Abstract: INTRODUCTION Allogeneic hematopoietic cell transplantation (allo-HCT) is the only curative option for high-risk myelodysplastic syndrome (MDS). Yet, allo-HCT is associated with potentially life-threatening complications such as conditioning-regimen toxicity, graft-versus-host disease (GVHD) and relapse. The ever-rising number of patients undergoing allo-HCT yields an increase of those requiring admission to an intensive care unit (ICU). Despite ICU outcome improvement, ICU admission is not the solution for all patients. Although ICU triage policies are intended to identify patients more likely to recover from life-threatening complications, they may lack of specificity as they often include very heterogeneous cohorts of patients with regard of underlying disease, patient's characteristics and transplantation modalities. It is therefore crucial to more accurately identify prognostic factors that affect the overall survival (OS) of patients treated with allo-HCT. We hereby aimed to establish a prognostic scoring system for OS inclusive of early post-transplant complications suitable to guide clinicians when ICU admission is pondered. PATIENTS AND METHODS The SFGM-TC database (PROMISE) was used to retrieve data from patients who underwent allo-HCT for MDS. A derivation cohort comprised data from January 1999 to December 2009. A validation cohort comprised data from January 2010 to December 2013. We included patients above 18 years of age, receiving a first sibling or HLA-matched unrelated allo-HCT at the allele level (so-called 10/10) and surviving more than 100 days after HCT. Patients could receive either bone marrow or peripheral blood stem cells. We excluded patients who received allo-HCT from an HLA-mismatched, haplo-identical donor, or ex-vivo T-cell depleted graft. To identify prognostic factors of 3-year OS, disease characteristics, donor and patients characteristics, transplantation modalities and early post-transplant complications were analyzed using a multivariable Cox model. Discrimination and calibration performance of the modelwas assessed by calculating c-index and comparing predicted and observed survivals. Finally, to favor daily use in clinical routine, we turned our prediction model into a point scoring system, in which each predictable variable was weighted by the nearest approximation of hazard ratio. RESULTS The derivation cohort included 393 patients and the validation cohort included 391 patients. The median follow-up from transplantationwas 3.8 years (range, 0.3 to 11.8 years) and 2.9 years (range, 0.4 to 5.5 years), respectively. The backward stepwise regression analysis revealed 3 independent predictors of 3-year OS: (i) the grade of acute GVHD (0/I vs. II vs. III/IV), (ii) the relapse before day 100 and (iii) the lack of platelet recovery before day 100 (Table 1). After over-optimism correction, the discrimination of the selected prognostic model was 0.67 (95%CI, 0.63-0.71) with a shrinkage factor of 0.903. A similar discrimination value was found in the validation cohort 0.65 (95%CI, 0.61-0.69) The point scoring system ranged from 0 to 8, discriminating low- (0), intermediate- (1 to 3), and high-risk (4 to 8) patients, according to survival prognosis (Table 2). The observed 3-year OS after transplantation in patients with low, intermediate and high scores was 70% (95%CI, 63% to 76%), 46% (95%CI, 38% to 55%) and 6% (95%CI, 2% to 16%) respectively (Figure 1). CONCLUSION We created then validated the first triage prognostic score based on early post-transplant complications, to quickly and simply estimate the survival probability after day 100 when ICU is to be considered. Our findings support the robustness, the reliability and the reproducibility of this scoring system. Additional studies are required to assess whether this scoring system may be suitable for hematologic malignancies other than MDS. Calibration of survival probability for the continuous prognostic model in the derivation cohort. Calibration of survival probability for the continuous prognostic model in the derivation cohort. Figure Figure. Disclosures Michallet: Bristol-Myers Squibb: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Astellas Pharma: Consultancy, Honoraria; MSD: Consultancy, Honoraria; Genzyme: Consultancy, Honoraria. Peffault De Latour:Novartis: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria, Research Funding; Alexion: Consultancy, Honoraria, Research Funding; Amgen: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 140, No. 3 ( 2022-07-21), p. 253-261
    Abstract: Splenectomy is effective in ∼70% to 80% of pediatric chronic immune thrombocytopenia (cITP) cases, and few data exist about it in autoimmune hemolytic anemia (AIHA) and Evans syndrome (ES). Because of the irreversibility of the procedure and the lack of predictions regarding long-term outcomes, the decision to undertake splenectomy is difficult in children. We report here factors associated with splenectomy outcomes from the OBS’CEREVANCE cohort, which prospectively includes French children with autoimmune cytopenia (AIC) since 2004. The primary outcome was failure-free survival (FFS), defined as the time from splenectomy to the initiation of a second-line treatment (other than steroids and intravenous immunoglobulins) or death. We included 161 patients (cITP, n = 120; AIHA, n = 19; ES, n = 22) with a median (minimum-maximum) follow-up of 6.8 years (1.0-33.3) after splenectomy. AIC subtype was not associated with FFS. We found that immunopathological manifestations (IMs) were strongly associated with unfavorable outcomes. Diagnosis of an IM before splenectomy was associated with a lower FFS (hazard ratio [HR], 0.39; 95% confidence interval [CI] , 0.21-0.72, P = .003, adjusted for AIC subtype). Diagnosis of an IM at any timepoint during follow-up was associated with an even lower FFS (HR, 0.22; 95% CI, 0.12-0.39; P = 2.8 × 10−7, adjusted for AIC subtype) as well as with higher risk of recurrent or severe bacterial infections and thrombosis. In conclusion, our results support the search for associated IMs when considering a splenectomy to refine the risk-benefit ratio. After the procedure, monitoring IMs helps to identify patients with higher risk of unfavorable outcomes.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3035-3035
    Abstract: Abstract 3035 Background: due to a risk of relapse of underlying disease in patients transplanted with progressive malignancy, the use of antithymocyte globulins (ATG), incorporated within the conditioning regimen prior to allogeneic stem cell transplantation (allo-SCT), is still controversial. We report here on a study of 245 consecutive patients transplanted between January 1999 and December 2009 in 26 French and Belgian centers for progressive MDS, defined as stable, untreated, relapsed or refractory disease. Patients and Methods: Inclusion criteria included patients aged over 18 who received allo-SCT from either a sibling (n=153) or HLA-A, -B, -C, -DRB1 and -DQB1 allele matched unrelated donor (10/10) (n=86) for MDS or AML/RAEB-t (with 20–30% BM blasts). Data quality was ensured using computerized discrepancy errors and vigorous on-site data verification of every single file. A qualified research technicien has been appointed by the University-Hospital of Lille to assist on-site centers that couldn't meet data quality requirements. HLA matching was double-checked by the French Bone Marrow Donor Registry. Results: The first 239 files analyzed until now are presented, including 154 males and 85 females. According to the WHO classification at diagnosis, 85 patients had RA/RARS/RCMD, 86 RAEB1, 62 REAB2 and 6 RAEB-t/AML. Sixty-six patients had progressed to a more advanced disease before allo-SCT. At diagnosis, 102 patients had an IPSS int-2 or higher. Cytogenetic IPSS was recorded as favorable (n=109), intermediate (n=61), unfavorable (n=63) and missing (n=6). Disease status at transplant was established as follows: relapsed or refractory disease (n=106) and untreated or stable disease without hematological improvement (n=133). Median age at transplantation was 53 years (range, 20–70). Patients received myeloablative conditioning (n=105) and nonmyeloablative (n=134) including busulfan-based regimens (n=127), TBI-based regimens (n=92) or other alkylating-agent-based regimens (n=20). In this series, 95 patients (40%) received ATG as part of conditioning ('ATG' group), whereas 144 did not ('no-ATG' group). The analysis reference date of April 1st 2011, median follow-up in survivors was 50 months (IQR, 33–92) with 59 patients having died of relapse and 77 of TRM. The estimated 3-year OS and EFS was respectively 42.3%, and 32.4%. The probability of relapse, overall and event-free survival at 3 years was not significantly different between the two groups. In contrast, the cumulative incidence of grade 2–4 acute GVHD was 48% in the no-ATG group and 30% ATG group (P 〈 .001) and the cumulative incidence of grade 3–4 acute GVHD was 24% and 11% respectively (P 〈 .001). Although the cumulative incidence of chronic GVHD was similar in the no-ATG and ATG groups (64% vs 46%, p=.15), a trend for a lower TRM was observed in the ATG group (22% vs 31%, p=.06). In multivariate analysis, the absence of use of ATG was the strongest parameter associated with an increased risk of acute grade 2–4 [HR = 2.28, 95% CI: 1.39–3.74, p=.001] and grade 3–4 GVHD [HR = 2.19, 95% CI: 1.04–4.61, p=.035] . In conclusion, the addition of ATG to the conditioning regimen resulted in a decreased incidence of acute GVHD without increasing relapse rates and compromising patient survival undergoing allo-SCT for progressive MDS. Disclosures: Yakoub-Agha: Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Research Funding; Fresinus: Honoraria, Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria. Michallet:Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees; Fresinus: Honoraria, Membership on an entity's Board of Directors or advisory committees. Deconinck:Celgene: Honoraria. Mohty:Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...