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
  • Wang, Hui  (7)
  • Medicine  (7)
  • XA 33000  (7)
Material
Language
Years
Subjects(RVK)
  • Medicine  (7)
RVK
  • 1
    In: Blood, American Society of Hematology, Vol. 138, No. Supplement 1 ( 2021-11-05), p. 1735-1735
    Abstract: Introduction Multiple myeloma is a hematological malignancy that is prone to be companied by bone marrow destruction, renal impairment and extramedullary infiltration. Current treatments include proteasome inhibitor, immunomodulatory drug, hematopoietic stem cell transplantation, and monoclonal antibody targeted therapies. However, it is still clinically incurable. Chimeric antigen receptor (CAR) T-cell therapy is a new immune targeted therapeutic strategy. It is reported better clinical efficacy for relapsed/refractory multiple myeloma (r/r MM) treatment has been achieved by immunotherapies targeted B-cell maturation antigen (BCMA). Therefore, it is important to investigate the treatment of a novel human BCMA-specific CAR-T therapy for r/r MM. Objective The objective of this clinical study is to evaluate the safety and efficacy of the novel human BCMA-targeting CAR-T therapy in patients with r/r MM, especially patients who relapsed from prior CAR-T therapy. Method This work is a clinical study registered and investigated by our center. CD3+ T cells were negatively selected from patients' peripheral blood mononuclear cells, activated and modified by lentivirus to produce anti-BCMA CAR-T cells. The cells were administrated intravenously to patients after expanding for 7-13 days in vitro. 24 patients had enrolled in this study, with 13 males and 11 females. The median age was 53 years old (range,41-75), and patients with cytogenetically high risk factors accounting for 41.66% (10/24). 50% (12/24) was infiltrated with extramedullary lesions. 16.6% (4/24) of them had relapsed from other CAR-T therapies before this enrollment. 50% (12/24) had been previously conducted autologous hematopoietic stem cell transplantation (HSCT), whereas 4.16% (1/24) with allogeneic HSCT. Patients with the expression of BCMA in the plasma cells higher than 30%, accounted for 25% (6/24). 2-3 days after being administered the lymphodepleting chemotherapy regimen, CAR-T cells were infused intravenously. The indicators of patients' condition were detected, including inflammatory cytokine concentration, serum protein levels, CAR-T cell number copies, and the proportion of plasma cells by bone marrow biopsy. The improvement of patients, the occurrence of adverse reactions, the incidence and grade of cytokine release syndrome (CRS), was analyzed and evaluated. Result All patients received infusions of CAR-T positive cells at the average dose of 9.45×10 6/kg (5-17.5) and the median injection day is the 10th day (8th-13th day) after cell isolation. After infusion, 100% (24/24) of the patients had fever lasting for 48 hours, with 37.5% (9/24) of them showing low blood pressure and being treated with drug. Heart rate increase was found in 45.8% (11/24). Nausea, diarrhea and transient consciousness disorder occurred in 50% (12/24), 33.3% (8/24), and 12.5% (3/24) of them, respectively. 16.6% (4/24) was administrated with dexamethasone to relieve symptoms, with the total dose less than 20 mg, while nobody was treated with IL-6 receptor antagonist. CAR-T cells had expanded in all patients, reaching the peak at the 4th day after infusion (Figure). The levels of IL-6, IL-8 and IFN-γ in peripheral blood also increased significantly. The incidence of CRS is 100%, of which grade I, II and III is 62.5%, 33.3% and 4.2%, respectively. 2 patients showed grade I CRES, constituting 8.3% (2/24). All patients were assessed for the efficacy of CAR-T cells 2 weeks after infusion. ORR was 100%, with 4.2% (1/24) MR, 8.3% (2/24) PR, 62.5% (15/24) VGPR and 25% (6/24) CR. 18 patients were treated for more than 1 month, with 11.1% (2/18) PR, 44.4% (8/18) VGPR, 11.1% (2/18) CR, and 33.3% (6/18) sCR. 16 patients were infused before more than 2 months, with 25% (4/16) VGPR, 12.5% (2/16) CR, 50% (8/16) sCR, and 12.5% (2/16) PD. 6 patents were administrated more than 3 months ago, with 1 developing deep remission to sCR from VGPR. The others remain the same condition. Conclusion The novel human BCMA targeted CAR-T cell therapy of this study showed safety and efficacy in the treatment of r/r MM patients with extramedullary infiltration, high-risk cytogenetical factors as well as relapse with prior BCMA CAR exposures. Deep remission can be achieved. However, more observation need to be conducted. The CAR-T treatment of BCMA target still cannot prevent the disease progress of a small numbers of patients. The control after CAR-T therapy needs more investigation. Figure 1 Figure 1. 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: 2021
    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, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 10287-10287
    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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3224-3224
    Abstract: Introduction: With GIAC regimen, haploidentical blood and marrow transplantation (haplo-BMT) has achieved comparable outcomes with identical sibling transplant (Dao-Pei Lu et al., Blood 2006; 107:3065). Our previous study has shown that the third party cell co-infusion in haplo-BMT (GIAC-3 regimen) could significantly reduce aGVHD and transplant-related mortality (TRM). We have also demonstrated that individualized chemotherapy to decrease leukemia burden followed by conditioning could improve disease-free survival (DFS) in refractory/relapsed AML. Objective: To learn the outcomes of our haplo-BMT with these integrated approaches, all patients who received haplo-BMT for hematologic malignancies in our center were analyzed retrospectively. Methods: Between April 2012 and December 2014, consecutive 514 patients with hematologic malignancies who underwent haplo-BMT were included. The median age was 20 (1.8 to 64) years old. The diagnosis included AML 232 (45.1%), ALL 207 (40.3%), MDS 27(5.3%), CML 14 (2.7%), lymphoma 13 (2.5%) and others 21 (4.1%). Transplants at CR1, ≥CR2 or advanced disease were 216 (42.0%), 114 (22.2%), 184 (35.8%), respectively. All patients received unmanipulated bone marrow (BM) and peripheral blood stem cells as graft after myeloablative conditioning plus ATG. Majority of the patients with AML received BuCy-based conditioning, while most ALL patients received TBICy-based regimen. Fludarabine was substituted for cyclophosphamide in some patients due to impaired organ function or high tumor burden. For refractory/relapsed diseases, individualized chemotherapy followed by conditioning was administered. Cyclosporine/tacrolimus, short-term Methotrexate, and Mycophenolate mofetil were employed for GVHD prophylaxis. Either 1ml/kg (recipient's body weight) haploidentical BM from the second haploidentical donor or one unit of unrelated cord blood was infused right after haplo-BMT as the third party cells. Minimal residual disease (MRD) was monitored routinely by quantitative PCR or flow cytometry. The patients with persistent MRD were interfered by immunosuppressant withdrew, adoptive immunotherapy with cytokine induced killer or NK cells or donor lymphocyte infusion. Results: All patients but 5 achieved durable engraftment. The cumulative incidences of grade II to IV aGVHD and grade III to IV aGVHD were 32.2%, 19.8%, respectively. The cumulative incidences of cGVHD and extensive cGVHD were 48.3%, 18.4%, respectively. 100-day TRM and 2-year TRM were 4.1%, 14.9%, respectively. Two-year relapse rate was 22.8%. With the median follow up 17 (6 to 38) months, overall 2-year DFS rates in CR1, ≥CR2 and advanced disease were 75.6%, 70.9%, 49.2%, respectively. For AML, two-year DFS rates in CR1, ≥CR2 and advanced disease were 74.1%, 76.9%, 48.2% (CR1 vs. ≥CR2, p=0.84; CR vs. advanced disease, p=0.000). For ALL, two-year DFS rates in CR1, ≥CR2 and advanced disease were 78.9%, 56.6%, 38%, respectively (CR1 vs. ≥CR2, p=0.018; CR1 vs. NR, p=0.000; ≥CR2 vs. NR P=0.02 ). Conclusions: With our strategies, overall outcomes of haplo-BMT have been improved remarkably and very encouraging. Therefore, haplo-BMT should be an important way to save life for the patients with hematologic malignancies who need urgent BMT but without matched either sibling or unrelated donor. 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: 2015
    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. 21 ( 2013-11-15), p. 2805-2805
    Abstract: EZH2 gene is located on chromosome 7q, a frequently affected genomic area in myeloid neoplasms including in MDS. The SET domain protein encoded by EZH2 is a component of the polycomb repressive complex-2 (PRC2) that has histone lysine 27 residue of histone 3 (H3K27) methyltransferase activity, whose methylation forms a repressive epigenetic marker for gene regulation. A spectrum of loss-of-function mutations of EZH2 have been identified in myeloid neoplasms including in MDS, underlying the biological and pathological importance of EZH2 gene product in MDS pathogenesis. To further characterize molecular alterations of EZH2 in the hematopoietic progenitor cell compartment of MDS, we have undertaken the study to assess EZH2 expression in bone marrow CD34+ cells of patients with MDS. We used Q-RTPCR to compare EZH2 RNA levels between the CD34+ cells that were isolated from patients with MDS (N=74) and healthy individuals (N=9). Q-RTPCR results indicate that 51% of the patients (N=38) had reduced EZH2 expression. Although the whole patient cohort did not show a significantly difference of EZH2 expression compared controls, the subset of patients bearing chromosome 7 (7-) or 7q deletion (7q-) (N=16) presented a significant reduction of EZH2 RNA expression (0.4 fold of control, p=0.04). Thirteen of the 16 patients (81%) of the 7-/7q- subset had an over 50% decrease of EZH2 RNA expression level in bone marrow CD34+ cells. These results suggest that haploinsufficiency plays a key role in the molecular regulation underlying reduced EZH2 expression in hematopoietic progenitor cells of MDS. In order to further evaluate molecular alterations of EZH2 in patients without 7/7q deletion, we subsequently performed capture deep sequencing to survey potential EZH2 mutations in the subset of diploid patients (N=32) of the main cohort using their bone marrow mononuclear cells (BM-MNNC). Sequencing of all coding exons of EZH2 gene reveals that three of the 32 diploid patients (9%) carry EZH2 mutations, including one missense (C590Y), one nonsense and one splicing mutation. We then took off these three patients with EZH2 mutations and analyzed potential sole impact of EZH2 RNA expression level in the diploid/EZH2-wildtype background in the remaining 29 patient subset. Fourteen (48%) of these patients have an over 50% reduction of EZH2 expression in bone marrow CD34+ cells. Furthermore, compared to other diploid patients that are without EZH2 reduction, patients with reduced EZH2 expression ( 〈 50%) have a significant increase for the expression of a panel of innate immune regulatory genes. These genes include JMJD3 (p=0.05), TLR2 (p=0.02), IL-8 (p=0.03), IL-1B (p=0.02), and S100A9 (p=0.02). Of importance, overexpression of these innate immune genes in bone marrow CD34+ cells have been demonstrated to be implicated in the pathogenesis of MDS (Wei et al. Leukemia 2013). Of interest, survival analysis revealed that patients with reduced EZH2 RNA expression in bone marrow CD34+ cells had significant longer survival in the diploid/EZH2-wildtype subset (N=29, 42 months v.s. 22 months p=0.04). The same result also applies to the whole patient cohort of this study (N=74, 33 months v.s. 15 months, p=0.004). In contrast to worse prognosis associated with EZH2 gene mutation, this data suggest that down-regulation of EZH2 expression may not have such impact on survival. Taken together, results of current study indicate that, besides genomic mutation, down-regulation of EZH2 expression in the hematopoietic progenitor cell compartment also occurs in a subset of patients with MDS, and more frequently in patients with 7/7q deletions. Furthermore, this study also suggests that in patients with diploid karyotype and wildtype EZH2 gene, reduced EZH2 expression and its co-occurrence with overexpression of innate immune genes may have potential pathogenic and prognostic implications. Further investigation of EZH2 gene regulation and its interaction with innate immune signals should be performed in MDS. 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: 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. 124, No. 21 ( 2014-12-06), p. 3241-3241
    Abstract: EZH2 encodes a key histone methylation regulatory molecule. Genetic mutations of EZH2 occur in ~10% patients with MDS and are associated with poor prognosis. However, the expression patterns of EZH2 are less well studied in MDS. To characterize these expression patterns, we assessed EZH2 mRNA expression in primary patient bone marrow CD34+ cells (n=78) (age: 33-91; IPSS: low 24%, Int-1 31%, Int-2 27%, High 14%; Karyotype: diploid 62%, 5q-/7q- 23%, others 15%). QRTPCR assays indicated that 47% (n=37) of patients had reduced EZH2 mRNA expression (less than 50% of controls), but this finding was not statistically significant (p= 0.159). Subtype analyses based on various karyotypes revealed that EZH2 is significantly underexpressed in patients bearing chromosome 7 or 7q deletions (mean=0.4 fold, p=0.006). Seventy-five percent of patients with 7 or 7q deletions have EZH2 expression less than 50% of that of controls. Chromosome 7 deletions were also associated with lower EZH2 expression than that seen in diploidy and other cytogenetic abnormalities (p=0.041). Wepreviously found that the overexpression of a group of innate immune genes contributes to MDS pathogenesis and is related to deregulation of histone methylation. Because EZH2 is a key regulator of histone methylation, we assessed the relationship between deregulation of these genes and EZH2 under-expression. To exclude the effects of cytogenetic defects and EZH2 mutations, we studied only the subset of patients with normal karyotypes and wildtype EZH2. We surveyed capture deep sequencing results of 32 of the diploid patients from the cohort that had sequencing data available. Three patients carry EZH2 mutations, including missense (nt148511133), nonsense (nt148524257), and splicing mutations (nt148524257). In the remaining 29 patients with normal karyotypes and wildtype EZH2, 14 (48%) had EZH2 under-expression. We then compared mRNA expression of 11 innate immune genes known to overexpress in MDS between the patients with EZH2 underexpression and others. We observed that mRNA levels of all 11 immune genes tested were higher in the EZH2 underexpression group and statistically significant (p 〈 0.05) for the genes JMJD3, IL-8, IL-1B, TLR-2, and S100-A9. We then performed survival analysis for EZH2 expression in MDS. Surprisingly, multivariate analysis in the whole cohort indicated that EZH2 underexpression is associated with better overall survival (OS) (HR 0.23, 95% CI (0.07-0.72); p=0.013). We also performed analysis in the subset without chromosome 7 deletion and observed a similar association (HR 0.18 (0.06-0.55) p=0.012). To investigate whether this result was related to responses to therapy, we reviewed treatment records and found that 61% of patients in the cohort (n=53) received hypomethylating agents (HMA). In this HMA treatment subset, non-responders (n=27) tended to have lower EZH2 expression than responders (n=26) (mean EZH2 of 0.497 vs 0.944, p=0.12). However, we noticed that in the subset of HMA responders, EZH2 expression was significantly lower (p=0.02) in patients who achieved longer responses (more than 12 months, n=15) than in those who progressed or relapsed within 12 months following treatment. We are currently investigating whether this impact of EZH2 underexpression on HMA responses contributes to its effect on OS. Taken together, the results of this study indicate that underexpression of EZH2 in the bone marrow hematopoietic progenitor cell compartment may have unique effects on the molecular pathogenesis, prognosis, and treatment of MDS and may do so through a unique mechanism that differs from that of previously characterized EZH2 mutations. Further investigations are also required to determine the relationships between EZH2, HMA-based treatments, and patient survival. Disclosures Garcia-Manero: Epizyme, Inc: Research Funding.
    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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2296-2296
    Abstract: Introduction: Cytogenetic abnormality is considered to be an independent prognostic factor in newly diagnosed acute myeloid leukemia (AML). However, recent studies have demonstrated that acquired gene mutations also play an important role in the pathogenesis and prognosis of AML. It has been well known that minimal residual disease (MRD) pre-conditioning has remarkable impact on disease-free survival (DFS) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in acute lymphoblastic leukemia, but the effect of MRD pre-transplant on allo-HSCT in AML is still unclear. Objective: In present study, the effect of NCCN risk stratification which has integrated gene mutations into cytogenetics as well as MRD pre-transplant on DFS after allo-HSCT in AML was studied in order to learn whether risk-directed conditioning and prevention of relapse are needed. Methods: Between April 2012 and March 2015, consecutive 258 patients with AML in complete remission (CR) (186 cases in CR1 and 72 cases in CR2) who underwent allo-HSCT in our hospital were analyzed retrospectively. The median age was 25 (1.8-64) years. Male (M) to female (F) was 147:111. The median disease course was 6 (1-51) months. According to 2015-NCCN risk stratification, 63 (24.4%) cases were in low risk, 112 (43.4%) cases in intermediated risk, and 83 (32.2%) cases in high risk. MRD in bone marrow pre-conditioning was detected by eight-color flow cytometry. Results: With the median follow up 18 (5-41) months, overall 2-year DFS was78.0%. No significant difference in DFS was found among low-risk (78.6%), intermediated-risk (76.0%) and high-risk (80.3%) patients (P=0.886). 205 (79.5%) cases were MRD- and 53 (20.5%) cases were MRD+ before conditioning. DFS after transplant in MRD+ patients was significant lower than that in MRD- patients(65.0% vs. 81.4%, P=0.003). Univariate analysis showed that DFS was not associated with patient age (≤14years vs. 〉 14years, P=0.292), disease course before HSCT (≤6 months vs. 〉 6months, P=0.532), WBC counts at diagnosis (≤50×109/L vs. 〉 50×109/L, P=0.120), CBC recovery pre-HSCT (yes vs. no, P=0.664), disease status (CR1 vs. CR2, P=0.201), extramedullary leukemia before transplant (yes vs. no, P=0.532), conditioning regimen (BUCy/Flu-based vs. TBICy/Flu-based, P=0.753), donor type (identical sibling vs. unrelated vs. haploidentical, P=0.743), donor-recipient gender (M-M vs. M-F vs. F-M vs. F-F, P=0.245), donor-recipient blood type (compatibility vs. major incompatibility vs. minor incompatibility vs. major and minor incompatibility, P=0.402), mononuclear cells infused (≤8×108/kg vs. 〉 8×108/kg, P=0.583), CD34+ cells infused (≤4×106/kg vs. 〉 4×106/kg, P=0.946), and CD3+ cells infused (≤1.6×108/kg vs. 〉 1.6×108/kg, P=0.143). DFS was significant lower in the patients with secondary AML (79.4% in primary AML vs. 53.5% in secondary AML, P=0.006) and MRD+ cases before transplant (81.4% in MRD- vs. 65.0% in MRD+, P=0.003). Accumulative non-relapse mortality (NRM) was significant higher in secondary AML (11.7% in primary AML vs. 33.3% in secondary AML, P=0.004) and MRD+ patients (10.5% in MRD- vs. 21.9% in MRD+, P=0.010). Accumulative relapse rate was significant higher in CR2 cases (8.0% in CR1 vs. 17.5% in CR2, P=0.046). Multivariate analysis showed that MRD pre-HSCT was the only impact factor on DFS and NRM with higher DFS (P=0.020) and lower NRM (P=0.045) in MRD- cases. Conclusions: Allo-HSCT has attenuated the influence of cytogenetics and gene mutations on DFS in AML. Secondary AML has lower DFS and higher NRM. Although disease status (CR1 vs. CR2) has no significant influence on DFS, relapse rate in CR2 is higher than that in CR1. MRD pre-conditioning was a key impact factor on DFS after allo-HSCT in AML but not conditioning regimen and donor type. 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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 312-312
    Abstract: Introduction: Relapse remains the main cause of failure of hematopoietic stem cell transplantation (HSCT) in acute leukemia. NK cells have the property of killing leukemia cells without GVHD aggravation theoretically. Moreover, in some cases, leukemia cells may lost HLA-I and/HLA-II antigens which would result in poor response to the immunotherapy except NK-based adoptive effectors. Objective: In present study, the safety and efficacy of donor-derived ex-vivo activated NK cells in management of relapse after allogeneic HSCT in high-risk acute leukemia were examined. Patients and methods: Between July 2012 and July 2014, 29 patients with acute leukemia who received NK cell infusion after HSCT were analyzed retrospectively. Some cases failed to chemotherapy combined with donor lymphocyte infusion (DLI) before NK cell therapy. The diagnosis were ALL (10 cases), AML (18 cases) and mixed acute leukemia (1 case). All patients were high-risk leukemia. The disease status before transplant was CR1 in 8 cases, CR2 in 7, CR3 in 1 and non-remission in 13. The types of donor included identical sibling (5 cases), haploientical family member (21 cases) and unrelated donor (3 cases). The conditioning and GVHD prophylactic regimens were reported previously (Lu DP et al., Blood 2006; 107:3065). Minimal residual disease (MRD) was detected by either quantitative RT-PCR for fusion genes or flow cytometry or both. The expression of HLA-I and HLA-II antigens in leukemia cells was evaluated by flow cytometry. Donor-derived either peripheral blood stem cells or lymphocytes were cultured for 6 days using original culture system (AIM-V medium with IL-2, IL-12, IL-15 and IL-21) or modified culture system (SCGM medium with IL-2, IL-12, IL-15, IL-18 and IL-21). Escalated dosage of NK cells were infused starting with 1×105 cells/kg (recipient’s body weight) with or without IL-2 injection. Nine patients were in prevention group and 20 cases were in treatment group. The patients with hematologic relapse received NK cells 3 days later after chemotherapy. Results: Compared with our original culture system, the modified culture system enhanced approximately 10% to 20% of the purity and 4 to 8 fold in number of NK cells by day 6. Furthermore, our modified culture system elevated the expression of function phenotype including TRAIL, NKG2D and CD62L on NK cells in approximately 8 to 10 folds at day 6 and simultaneously stimulated higher level of IFN-γ. One to 4 NK cell infusions were given in each case with two week interval. Two of 29 cases developed mild skin GVHD. No transfusion-related side effects were noted. In prevention group, four of 9 cases remain complete remission, and the other 5 patients became MRD positive or relapse. In treatment group, seven of 20 cases have response to NK cell therapy, and two out of 7 cases who response to NK cells had failed to chemotherapy plus DLI before. Among 11 patients who had response to NK cells, eight of them are AML, and the remaining 3 patients are ALL. Higher response rate (10/23 cases) was seen with NK cell therapy by our modified culture system compared with the one (1/6 cases) by our original culture system. Conclusions: Our preliminary results have demonstrated that donor-derived ex-vivo activated NK cells are safe and effective modality in the management of relapse after allogeneic HSCT in high-risk acute leukemia even failed to chemotherapy combined with DLI. Optimal culture system has improved not only NK cell’s purity, number and function phenotype but also clinical efficacy. 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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...