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
Material
Language
  • 1
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 2047-2047
    Abstract: Background: The outcomes of alternative donor haematopoietic stem cell Transplantation(HSCT)for Inherited bone marrow failure syndromes(IBMFS) have not been well described,especially the conditioning regimens are major challenges ,each disease type has different characteristics, whether engraftment can be achieved with less toxicity. Method : With this background,we retrospectively analyzed alternative donor HSCT for 42 patients with IBMFS in our single center from November 2012 to August 2018. Results: 27 cases were Fanconia anemia(FA),7 cases were dyskeratosis congenital(DC), 8 cases were severe congenital neutropenias(SCN). The median age at diagnosis and transplantation were 4(1 to 25 ) years and 10(1.9 to 26)years respectively. Male to female was 28 :14. All patients were confirmed to have BMF and disease-specific pathogenesis-related gene mutations. 16 cases had disease specific congenital anomalies, 10 patients had family history. Chromosomal fragility test was positive in 8 cases of FA group. Indication of HSCT for FA and DC patients which were 30 patients had BMF or transfusion dependency at transplantation;4 cases had clonal disease (2 cases myelodysplasia, 2 cases acute myeloid leukaemia). Indication of HSCT for SCN patients were uncontrollable severe infection .FA received low dose Busulfan (Bu;total dose of 6.4 mg/kg, IV), Fludarabine (Flu; total dose of 120 mg/m2, IV) , Cyclophosphamide (Cy; total dose of 2.0 g/m2, IV) based-reduced intensity conditioning(RIC) ; DC patients received low dose TBI (total 300cGy, Special position, supine) , Flu(total dose of 120 mg/m2, IV) , Cy( total dose of 3.0 g/m2, IV) based-RIC, while SCN patients had Bu(total dose of 12.8 mg/kg, IV),Cy( total dose of 3.6 g/m2, IV) or Flu(total dose of 160 mg/m2, IV) based -myeloablative conditioning(MA); and all patients combinated either of 2 different rabbit ATG ,ATG-T , rabbit anti-human thymocyte immunoglobulin, total dose 5.0 -10 mg/kg in 26 cases or ATG-F ,rabbit anti-human lymphocyte immunoglobulin, total dose 20 mg/kg in 14 cases. Campath-1, Anti-CD52 mAb was accepted with total dose 1mg/kg in 2 cases. Donor types were matched unrelated donor(MUD) in 22 patients ,Haploidentical donor (HID) in 17 patients,unrelated cord blood (UCB) in three cases. Unmanipulated stem cells were used for all patients. The Haplo-HSCT cohort received granulocyte colony-stimulating factor (G-CSF)-primed BM combined with peripheral blood stem cells (PBSCs) , The MUD HSCT cohort only received G-CSF PBSCs. The UCB HSCT cohort received one unit CB . No primary graft failure was observed. The median myeloid engraftment time was 14 (range, 10 to 21) days.Survivor median follow-up time was 38 months (range, 9-63 months), the overall survival in all patients was 76.1% ,in FA,DC,SCN were 72.4% ,100%,53.0% respectively. Cumulative incidence of 100 days acute graft-versus-host disease(GVHD) was 48.1%,Cumulative incidence 1 year and 3 years of chronic GVHD were 35.0% and 69.3% respectively. The positive chromosomal fragility test was the only independent adverse prognostic factor in multivariate analysis for FA patients rather than age ,donor type and graft source. Main causes of death were GVHD (50%) and infection (20%).No secondary malignancies occurred after HSCT till the last follow up time. Conclusion: In our study, alternative donor and disease-specific conditioning regimen HSCT for IBMFS showed promising prognosis especially for DC patients. Chromosomal fragility test positive was the only independent adverse prognostic factor in HSCT for FA patients. 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: 2019
    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: Frontiers in Immunology, Frontiers Media SA, Vol. 13 ( 2023-1-4)
    Abstract: We aimed to evaluate prognostic factors of a second allogeneic stem cell transplantation (allo-HSCT2) among hematological malignancy patients who have relapsed after the first allo-HSCT(allo-HSCT1). Methods We retrospectively analyzed 199 hematological malignancy patients who received allo-HSCT2 as a salvage treatment post allo-HSCT1 relapse between November 2012 and October 2021. Results The median age at allo-HSCT2 was 23 (range: 3-60) years. The median time to relapse after HSCT1 was 9 (range: 1-72) months. Prior to allo-HSCT2, patients had the following hematopoietic cell transplantation-comorbidity indexes (HCT-CI): 127 with a score of 0, 52 with a score of 1, and 20 with a score of 2 or greater. Fifty percent of patients received chimeric antigen receptor (CAR) T-cell therapy following HSCT1 relapse. Disease status was minimal residual disease (MRD)-negative complete remission (CR) among 119 patients, MRD-positive CR among 37 patients and non-remission (NR) for 43 patients prior to allo-HSCT2. Allo-HSCT2 was performed from a new donor in 194 patients (97.4%) and 134 patients (67.3%) received a graft with a new mismatched haplotype. The median follow-up time was 24 months (range: 6-98 months), and the 2-year OS and LFS were 43.8% ± 4.0% and 42.1% ± 4.1%, respectively. The 2-year cumulative incidence of relapse (CIR) and non-relapse mortality (NRM) was 30.0%±4.8% and 38.5%±3.8%, respectively. Cox regression multivariate analysis showed that disease statusof MRD-negative CR, HCT-CI score of 0 prior to allo-HSCT2, and new mismatched haplotype donor were predictive factors of improved OS and LFS compared to patients without these characteristics. Based on these three favorable factors, we developed a predictive scoring system for patients who received allo-HSCT2. Patients with a prognostic score of 3 who had the three factors showed a superior 2-year OS of 63.3% ± 6.7% and LFS of 63.3% ± 6.7% and a lower CIR of 5.5% ± 3.1% than patients with a prognostic score of 0. Allo-HSCT2 is feasible and patients with good prognostic features prior to allo-HSCT2 —disease status of CR/MRD- and HCT-CI score of 0 as well as a second donor with a new mismatched haplotype could have the maximal benefit from the second allo-HSCT. Conclusions Allo-HSCT2 is feasible and patients with good prognostic features prior to allo-HSCT2 —disease status of CR/MRD- and HCT-CI score of 0 as well as a second donor with a new mismatched haplotype could have the maximal benefit from the second allo-HSCT.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2606827-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 2660-2660
    Abstract: Introduction: CD19-CAR-T cells induce high rates of initial response among patients with refractory/relapsed and MRD positive high risk B-cell acute lymphoblastic leukemia (B-ALL). Afterwards allogeneic hematopoietic stem cell transplantation (HSCT) can further reduce relapse rate. Our previous results had shown that CR obtained by CD19 CAR-T had comparable significance to CR by chemotherapy before Allogeneic HSCT in B-ALL. Patients and Methods: Between July 2015 and Mar 2018, consecutive 135 patients with refractory/relapsed or high risk B-ALL obtained CR with CD19-CART therapy followed by allogeneic HSCT were retrospective analyzed. Median follow-up of survivors was 13 months (range, 3-32 months). Results: The median age was 11 (2-49) years. The median disease course before transplant was 21(4-143) months. The median time from CART therapy to HSCT was 69 (35-312) days. Disease status was 108 cases relapsed diseases, 11 cases refractory, and 16 persistent/recurrent measurable residual diseases (MRD). MRD pre-conditioning measured by flow cytometry and QT-PCR was positive in 20(14.8%) subjects. Donor source was haploidentical donors in 107(79.3%), identical sibling in 7(5.1%), and unrelated in 21(15.6%). Most subjects (87.4%) received conventional myeloablative pretransplant conditioning with total body radiation (TBI), the rest with busulfan (Bu). Antithymocyte globulin was used in haploidentical and unrelated transplants. Cyclosporine, short-term methotrexate, and mycophenolate mofetil were employed for GVHD prophylaxis. There were no cases of graft-failure except one early death on Day 0 for septic shock. The median time to neutrophil engraftment was 14 days (10, 26 days), and median time to platelet engraftment 14 days (5, 70 days). The incidences of non-relapse mortality within 100 days were 4.4% (0.8, 7.9%) The incidence of grades II-IV acute graft-versus-host disease (GvHD) were 32.1% (24.3, 39.9%) and grades III-IV GvHD 10.5% (5.4, 15.6%). Chronic GvHD and extensive chronic GvHD were 69.7% (60.7, 78.7%) and17.6% (10.7, 24.5%). Cumulative incidence of relapses (CIRs) at 2-year was 11.1% (5.4, 16.8%). There were totally 14 subjects relapsed after HSCT, among which 8 were CD19 negative relapse, 5 CD19 positive and 1 partial CD19 positive. And among the 8 CD19 negative relapse after transplant, 4 subjects had CD19 negative MRD before conditioning. Leukemia-free survival (LFS) was 76.5% (64.2, 88.8%) and overall survival (OS) was 80.8% (72.6, 89.0%) at two years after transplant. In multivariate analysis subjects who were MRD- positive pre-transplant had a higher 2-year CIR (43.5% [18.4, 68.6%] vs. 5.9% [1.2, 10.6%] ; p=0.000) and worse 2-year OS (61.5% [35.6, 87.4%] vs. 83.6% [75, 92.2%] ; p=0.034). Conclusions: Our clinical results showed that CART therapy followed by allogeneic HSCT was a promising modality for refractory/relapsed B-ALL. CD19 negative relapse accounted for most relapse after allogeneic HSCT. 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 ...
  • 4
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 12 ( 2021-5-7)
    Abstract: Patients often undergo consolidation allogeneic hematopoietic stem cell transplantation (allo-HSCT) to maintain long-term remission following chimeric antigen receptor (CAR) T-cell therapy. Comparisons of safety and efficacy of allo-HSCT following complete remission (CR) achieved by CAR-T therapy  versus by chemotherapy for B-cell acute lymphoblastic leukemia (B-ALL) has not been reported. We performed a parallel comparison of transplant outcomes in 105 consecutive B-ALL patients who received allo-HSCT after achieving CR with CAR-T therapy (n=27) or with chemotherapy (n=78). The CAR-T-allo-HSCT group had more patients in second CR compared to the chemotherapy-allo-HSCT group (78%  vs.  37%; p & lt;0.01) and more with complex cytogenetics (44%  vs. 6%; p & lt;0.001) but the proportion of patients with pre-transplant minimal residual disease (MRD) was similar. The median follow-up time was 49 months (range: 25-54 months). The CAR-T cohort had a higher incidence of Grade II-IV acute graft- versus -host disease (aGVHD 48.1% [95% CI: 46.1-50.1%] vs. 25.6% [95%CI: 25.2-26.0%]; p=0.016). The incidence of Grade III-IV aGVHD was similar in both groups (11.1%  vs. 11.5%, p=0.945). The overall incidence of chronic GVHD in the CAR-T group was higher compared to the chemotherapy group (73.3% [95%CI: 71.3-75.3%] vs. 55.0% [95%CI: 54.2-55.8%], p=0.107), but the rate of extensive chronic GVHD was similar (11.1% vs. 11.9%, p=0.964). Efficacy measures 4 years following transplant were all similar in the CAR-T vs. the chemotherapy groups: cumulative incidences of relapse (CIR; 11.1% vs.12.8%; p=0.84), cumulative incidences of non-relapse mortality (NRM; 18.7% vs. 23.1%; p=0.641) leukemia-free survival (LFS; 70.2% vs. 64.1%; p=0.63) and overall survival (OS; 70.2% vs. 65.4%; p=0.681). We found that pre-transplant MRD-negative CR predicted a lower CIR and a higher LFS compared with MRD-positive CR. In conclusion, our data indicate that, in B-ALL patients, similar clinical safety outcomes could be achieved with either CD19 CAR T-cell therapy followed by allo-HSCT or chemotherapy followed by allo-HSCT. Despite the inclusion of more patients with advanced diseases in the CAR-T group, the 4-year LFS and OS achieved with CAR T-cells followed by allo-HSCT were as remarkable as those achieved with chemotherapy followed by allo-HSCT. Further confirmation of these results requires larger, randomized clinical trials.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2606827-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1830-1830
    Abstract: Background: C-CAR088, an anti-BCMA CAR T-cell therapy, is a novel 2nd generation 4-1BB chimeric antigen receptor T (CAR-T) cell therapy targeting BCMA. Previously presented results from an ongoing study of C-CAR088 in R/R MM (NCT03751293, NCT03815383, NCT04322292, NCT04295018) included a 95.7% overall response rate (ORR) for the dose of 1.0~6.0x10 6 CAR-T cells/kg with a favorable safety profile (Lu, 2020 ASH Oral Presentation #182). Here we present the updated results of the study, with more patients and longer follow up time. Methods: Dose escalation and expansion studies were conducted at four medical centers in China to evaluate the safety and efficacy of C-CAR088 in patients with R/R MM who were previously treated with at least 2 lines of therapy, including proteasome inhibitors (PIs) and IMiDs. C-CAR088 was administered to patients as a single infusion after lymphodepletion with fludarabine (30 mg/m 2) and cyclophosphamide (300 mg/m 2) daily for 3 days. The primary endpoint was the incidence of adverse events (AEs), including dose-limiting toxicities (DLTs), and the secondary endpoints included overall response rate(ORR), duration of response (DOR), and progression-free survival (PFS) by IMWG Uniform Response Criteria. Results: As of July 2nd, 2021, 31 patients had been infused with C-CAR088. The median vein-to-vein time was 18 days. The manufacturing success rate was 100%. 4, 13 and 14 patients were infused with 1.0, 3.0 and 4.5~6.0 x10 6 CAR+ T cells/kg respectively. The median follow-up time for all patients was 8.0 months (0.1-24.2). The median age of patients was 61 years (45-74). The median number of prior lines of therapy was 4 (2-13). There were 25 (80.6%) patients with at least one high risk cytogenetic abnormality and 17 (54.8%) patients with at least two high risk cytogenetic abnormalities. 7 patients (22.6%) received bridging therapy before C-CAR088 therapy. Cytokine release syndrome (CRS) developed in 29/31 (93.5%) patients, grade 1 in 18/31 (58.1%), grade 2 in 8/31 (25.8%) and grade 3 in 3/31 (9.7%) respectively. The median time to the first onset of CRS was 6 days (1-11) and the median duration of CRS was 5 days (2-14). 9/31 (29%) patients used tocilizumab and 6/31 (19.4%) patients used corticosteroids to manage CRS. Only one patient developed a grade 1 neurotoxicity. No DLTs were observed and all adverse events were reversible. One patient died of septic shock on day 2 after receiving C-CAR088. Clinical efficacy was assessed in 28 patients with ≥ 1 month of follow up. Among the 28 patients, 3, 11 and 14 patients were infused with the dose of 1.0 x 10 6 CAR+ T cells/kg 3 x10 6 CAR+ T cells/kg, and 4.5~6x10 6 CAR+ T cells/kg respectively. The ORR was 27/28 (96.4%): 4 (14.3%) achieved CR, 12 (42.9%) achieved sCR and 9 (32.1%) achieved very good partial response (VGPR). At the dose level of 1.0 x10 6 CAR+ T cells/kg, 3(100%) patients achieved VGPR. The median DOR was 3.7 months (1.8-5.8), and the median PFS was 4.6 months (2.7-6.2). The CR rate was 54.5% (6/11) and 71.4% (10/14) in the 3.0 and 4.5~6.0 x10 6 CAR+ T cells/kg cohorts respectively. The median time to CR was 2.0 (0.5-9.5) months. Minimal residual disease (MRD) was testedbyEuroFlow-based flow cytometric analysis in 16 patients who had CR, 15/16 (93.7%) patients were MRD negative with the sensitivity of 10 -5. With a median follow-up of 9.5 months (1.9-24.2) in ≥ 3.0x10 6 CAR+ T cells/kg cohorts, the median DOR and PFS had not been reached. The Kaplan-Meier estimation of PFS at 6 and 12 months was 81.1% (95% CI:65.9% ~99.8%) and 69.5 % (95% CI:51.6 % ~93.6%) respectively. 8 patients in the ≥ 3.0x10 6 CAR+ T cells/kg cohorts discontinued the study. 7 discontinued due to disease progression (PD), and 1 discontinued for other anticancer therapy. 4 progressed within 6 months, 2 progressed within 6-12 months, and 1 progressed within 12-24 months. C-CAR088 proliferated and expanded well in patients' blood. The median C max was 734,868 copies/μg gDNA. The median AUC 0~28day was 7,468,779 day·copies/μg gDNA. The median T max was 14 days. The median T last was 84 days. 71% (95% CI: 42%~92%) of patients with C max equal to or greater than the median C max achieved CR/sCR. Conclusion: C-CAR088 has a manageable safety profile, which includes low neurotoxicity rates (with no gr ≥3 events). Deep and durable responses were observed in ≥ 3.0x10 6 CAR-T cells/kg cohorts. Doses of 3.0 and 6.0×10 6 CAR T cells/kg were selected for further study. Figure 1 Figure 1. Disclosures Zhu: CBMG: Current Employment. Huang: CBMG: Current Employment. Li: CBMG: Current Employment. Lan: CBMG: Current Employment. Chen: CBMG: Current Employment. Humphries: CBMG Ltd: Current Employment. Yao: CBMG: Current Employment, Current holder of stock options in a privately-held company.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2005
    In:  Environmental Geology Vol. 49, No. 1 ( 2005-11), p. 82-89
    In: Environmental Geology, Springer Science and Business Media LLC, Vol. 49, No. 1 ( 2005-11), p. 82-89
    Type of Medium: Online Resource
    ISSN: 0943-0105 , 1432-0495
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2005
    detail.hit.zdb_id: 190352-4
    detail.hit.zdb_id: 1459034-7
    detail.hit.zdb_id: 882719-9
    SSG: 13
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Frontiers Media SA ; 2023
    In:  Frontiers in Genetics Vol. 14 ( 2023-3-16)
    In: Frontiers in Genetics, Frontiers Media SA, Vol. 14 ( 2023-3-16)
    Abstract: Objective: To assess the causal effect of systemic iron status by using four biomarkers (serum iron; transferrin saturation; ferritin; total iron-binding capacity) on knee osteoarthritis (OA), hip OA, total knee replacement, and total hip replacement using 2-sample Mendelian randomization (MR) design. Methods: Three instrument sets were used to construct the genetic instruments for the iron status: Liberal instruments (variants associated with one of the iron biomarkers), sensitivity instruments (liberal instruments exclude variants associated with potential confounders), and conservative instruments (variants associated with all four iron biomarkers). Summary-level data for four OA phenotypes, including knee OA, hip OA, total knee replacement, and total hip replacement were obtained from the largest genome-wide meta-analysis with 826,690 individuals. Inverse-variance weighted based on the random-effect model as the main approach was conducted. Weighted median, MR-Egger, and Mendelian randomization pleiotropy residual sum and outlier methods were used as sensitivity MR approaches. Results: Based on liberal instruments, genetically predicted serum iron and transferrin saturation were significantly associated with hip OA and total hip replacement, but not with knee OA and total knee replacement. Statistical evidence of heterogeneity across the MR estimates indicated that mutation rs1800562 was the SNP significantly associated with hip OA in serum iron (odds ratio, OR = 1.48), transferrin saturation (OR = 1.57), ferritin (OR = 2.24), and total-iron binding capacity (OR = 0.79), and hip replacement in serum iron (OR = 1.45), transferrin saturation (OR = 1.25), ferritin (OR = 1.37), and total-iron binding capacity (OR = 0.80). Conclusion: Our study suggests that high iron status might be a causal factor of hip OA and total hip replacement where rs1800562 is the main contributor.
    Type of Medium: Online Resource
    ISSN: 1664-8021
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2023
    detail.hit.zdb_id: 2606823-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: SSRN Electronic Journal, Elsevier BV
    Type of Medium: Online Resource
    ISSN: 1556-5068
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 40-41
    Abstract: Introduction Current available treatments are limited once patients with B-ALL relapse following allogeneic hematopoietic stem cell transplantation (allo-HSCT). While chimeric antigen receptor (CAR) T-cell therapy offers a chance of remission, long-term outcomes for these patients remain poor. The benefit of bridging into a second transplant after CAR T-cell therapy remains inconclusive and available data are limited. Here, we report the long-term outcomes of 23 B-ALL patients who chose to undergo a second allo-HSCT after achieving complete remission (CR) from CAR T-cell therapy. Methods From April 2017 to April 2020, 23 R/R B-ALL patients (median age of 20 years, ranging from 3 to 58 years) who relapsed after first allo-HSCT received CAR T-cell therapy. The data were aggregated from seven different clinical trials (www.clinicaltrials.gov NCT03173417, NCT02546739 NCT03825718, NCT03825731, NCT03952923, NCT04100187 and www.chictr.org.cn ChiCTR1800016541). Patients' first transplant sources were HLA-identical sibling (n=5), matched-unrelated donor (MUD) (n=1), and haploidentical donors (haplo) (n=17). Eight of the 23 patients had disease relapse within 6 months following the first transplant. The median time from first transplant to CAR T-cell infusion was 261days (range: 117~2181 days). Before CAR T-cell infusion, patients' median bone marrow (BM) blasts by morphology were about 72.5% (1.5%-94.5%) including 12 patients with BM blasts & gt;70% (5 with BM blasts & gt;90%). Three of the 23 patients (13%) had received at least one prior donor lymphocyte infusion. No patients had active graft-versus-host disease (GVHD) prior to CAR T-cell therapy. Second generation CAR T-cells were generated by using purified T-cells from transplant donors (n=15) or patients (n=8). Twenty-two patients received T-cells modified with CD19-targeting CAR T-cells containing either a 4-1BB (n=18) or a CD28 co-stimulatory domain (n=4), and one patient received CD19-CD22 dual specificity CAR T-cells. All patients received a conditioning regimen of IV fludarabine (30mg/m2/d) and cyclophosphamide (250mg/m2/d) for 3 days followed by a single CAR T-cell infusion with a median dose of 3×105 cells/kg (1×105-6×105 cells/kg) in 21 patients. Two patients received a second CAR T-cell infusion in 2-3 months (1/3×105 cells/kg dose). Post CAR-T therapy, all patients bridged into a consolidation second transplantation with conventional myeloablative pre-transplantation conditioning regimens including 15 patients who received total body irradiation-based and 7 patients that received a busulfan-based conditioning regimen. Cyclosporin A, short-term methotrexate, and mycophenolate mofetil were used for GVHD prophylaxis. Results Patients' characteristics are shown in Table 1. On Day 30 post CAR-T-cell infusion, 23/23 (100%) patients achieved minimal residual disease (MRD)-negative CR. A total of 16/23 (69.6%) patients developed cytokine release syndrome (CRS) of which 14/23 (60.9%) had Grade I-II and 2/23 (8.7%) had Grade III CRS. Two patients had Grade III neurotoxicity. All patients with MRD-negative status subsequently bridged into a second transplant (2 from MUD and 21 from haplo donors) with a median interval time of 67 days (39- 329 days) from CAR T-cell therapy to a second transplant. At a median follow-up time of 258 days (84-978 days), no patients relapsed, which was encouraging. Five of 23 patients (21.7%) died from transplant-related mortality (TRM) at a median time of 295 days (103-372 days) (1 from GVHD and 4 from infection). The 1-year overall survival (OS) was 68.0% and 2-year OS was 54.4% (Fig.1). While there was a trend towards a more efficacious OS for patients whose CAR T-cells were derived from donors rather than from patients themselves but the number are too small to reach statistical significance (1-year OS 83.9% vs. 64.3%, 2-year OS 83.9% vs. 42.9%, P=0.739. Fig.2). After the 2nd transplant, four patients developed GVHD. Conclusions Our study demonstrates that even for R/R B-ALL patients who have relapsed following a first allo-HSCT , an MRD-negative CR status can still be achieved through CAR T-cell cell therapy without increasing CRS or neurotoxicity, making consolidation second allo-HSCT feasible for these patients. CAR T-cell therapy combined with a consolidation second HSCT are effective for these heavily pre-treated patients with an encouraging prospect for long-term survival. 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: 2020
    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. 136, No. Supplement 1 ( 2020-11-5), p. 1-2
    Abstract: Introduction Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 has demonstrated promising efficacy in patients with relapsed and refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). However, relapse after CART remains a major issue. Here, we analyzed the factors related to long-term efficacy, including overall survival (OS), leukemia-free survival (LFS) and cumulative relapse rate (CRR), following CAR-T therapy in 231 R/R B-ALL patients who achieved complete remission (CR) within one month after CAR T-cell therapy. Patients and Methods From April 2017 to March 2019, 254 patients with R/R B-ALL were enrolled onto one of five different clinical trials (NCT03173417; ChiCTR-ONC-17012829; NCT02546739; ChiCTR1800016541; and NCT03671460) at our center and received a second generation CD19+ CAR T-cell infusion. The median infused CAR T-cell dose was 3×105/kg (range: 0.2-10×105/kg). The CAR-T/T-cell ratio and the CD19+ B lymphocyte percentage in PBLC samples from 159 of the patients were analyzed using flow cytometry on day 0, 4, 7, 11, 14, 21, and 30 following CAR T-cell infusion. We performed single continuous variate factors analysis on the influence of the CAR-T/T-cell ratio and the percentage of CD19 + B-lymphocytes in day 30 post-infusion PBLC samples on the OS, LFS, and CRR. We also analyzed the impact of patient age, BM blast count, CAR-T-cell dose, and the interval time between CAR-T-cell therapy and consolidation allogeneic hematopoietic stem cell transplantation (allo-HSCT) on OS and LFS. Results Among 254 patients, 231 cases achieved CR within one month after CART therapy. A total of 211 CR patients had long-term follow-up of more than 30 days with a median follow-up of 12 months (1 to 29 months). On day 30 post CAR T-cell infusion, the median CAR-T/T-cell ratio in PBLC samples was 0.51% (range: 0%-44.8%), with 59 of 169 patients (34.9%) having a CAR-T/T-cell ratio of ≥1% and 85 of 169 patients (50.3%) with a CAR-T/T-cell ratio of ≥0.5%. The median percentage of CD19+ B lymphocytes in PBLC on day 30 was 0.0% (range: 0.0%-9.4%), of which 157 of 169 patients (92.9%) had & lt;0.5% CD19+ B-cell lymphocytes, and 137 of 169 patients (81.1%) had & lt;0.1% CD19+ B lymphocytes on day 30. Using a single continuous variate factors analysis, we found that increasing BM blasts and percentage of CD19+ B-lymphocytes in PBLC samples on day 30 correlated with a worse OS and LFS (Table 1). BM blasts of ≥70% were statistically significantly correlated with a worse OS and LFS when compared to BM blasts of & lt;70% (2-year OS of 52.6% vs. 65.0%, p=0.041; 2-year LFS of 43.3% vs. 58.6%, p=0.023). Unlike the BM blast data, for the CD19+ B-lymphocytes percentage in PBLC samples on day 30, we not identify a cut-off threshold. The CAR-T/T-cell ratio in PBLC samples on day 30 had no influence on OS or LFS. Unfortunately, the CAR-T/T- cell ratio and CD19+ B-lymphocyte percentage data beyond day 30 following CAR T-cell therapy was lacking for most patients and further analysis could not be performed to understand the impact of these factors on long-term survival. In our analysis, CAR T-cell dose, the interval time between CAR T-cell infusion and allo-HSCT did not significantly correlate with OS, LFS, or relapse. The remaining 184 patients in CR received a consolidation allo-HSCT after a median interval time of 67 days post CAR T-cell therapy (range: 30-334 days). Thirty-two of these patients (17%) relapsed with a median time to relapse of 221 days (57-490 days). The remaining 27 patients received CAR T-cell therapy only and 11 (41%) relapsed with a median time to relapse of 100 days (53-398 days). None of the four factors above had an influence on the CRR in either the bridging into allo-HSCT group or the CAR-T only group (Table 2). Conclusions Using a single continuous variate factors analysis, we found that a high BM blast count and the percentage of CD19+ B-lymphocytes in PBLC samples from R/R ALL patients on day 30 predicted a worse OS and LFS while age, the CAR-T/T-cell ratio on day 30, CAR-T cell dose, and the interval time between CAR-T cell infusion and allo-HSCT had no clear impact on long-term outcomes. 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: 2020
    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...