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
  • Medicine  (7)
  • XA 33000  (7)
  • 1
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 10652-10653
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 4463-4463
    Abstract: Cytokine-induced killer (CIK) cells are activated T cells with natural killer (NK) properties that can be expanded in vitro in presence of recombinant human interleukin-2 (rhIL-2) starting from peripheral blood mononuclear cells stimulated by interferon-γ and anti-CD3 antibody. They express CD3 and CD56 as well as the NKG2D antigen and show major histocompatibility complex (MHC)–unrestricted cytotoxicity toward neoplastic but not normal targets. CIK cells express several chemokine receptors, and in vivo models suggest that they can migrate to the site of tumors after intravenous administration, there carrying out their cytotoxic potential and helping to control tumor growth. CIK cells have shown cytotoxic activity in vitro and in vivo against hematopoietic neoplastic cells, including AML (acute myeloid leukemia), CML (chronic myelogenous leukemia), and CLL (chronic lymphocytic leukemia). Their cytotoxicity against patient with B-NHL (B-cell non-Hodgkin lymphoma) , however, has not been fully investigated. The elderly population is susceptible to haematological malignancies, and these elderly patients are intolerant to cytotoxic drugs. Therefore, the exploration of a safe and reliable strategy reduse dose of chemotherapy is critical in improving the prognosis of elderly patients with haematological malignancies. To evaluate the effectiveness and safety of autologous cytokine-induced killer (CIK) cells for consolidation treatmemt in elderly patients with diffuse large B-cell lymphoma. Peripheral blood mononuclear cells (PBMC) were isolated from 20 elderly patients with diffuse large B-cell lymphoma. PBMCs were augmented by priming with interferon gamma (IFN-γ) followed by IL-2 and monoclonal antibody (mAb) against CD3. Autologous CIK cells (range 5 × 10(9)-1 × 10(10)) were then infused back to individual patients. The regimen was repeated every 4 weeks. The host cellular immune function, tumour-related biological parameters, imaging characteristics, disease condition, quality of life and survival time were assessed. Fourteen patients received 6 cycles of transfusion and 6 received 4 cycles. After treatment of CIK cells plus IL-2, the general conditions of 20 patients were to different extent improved No adverse effects were observed. The percentages of CD3(+), CD3(+) CD8(+) and CD3(+) CD56(+) cells were significantly increased (p 〈 0.05), and the levels of serum β2 microglobulin and lactate dehydrogenase (LDH) were markedly decreased (p 〈 0.05) after autologous CIK cell transfusion. Cancer-related symptoms were profoundly alleviated, as demonstrated by the improved quality of life (p 〈 0.01)., Complete remission(CR) observed in 11 patients before the treatmemt of CIK was still CR; Partial remission(PR) in 9 patients ,After the treatmemt of CIK, the transformation of disease state from partial remission to complete remission was seen in 4 patients. At the end of follow-up, the mean survival time was 24 months. Transfusion with autologous CIK cells is safe and effective for treating haematological malignancies in elderly 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: 2014
    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. 116, No. 21 ( 2010-11-19), p. 4893-4893
    Abstract: Abstract 4893 The addition of rituximab to cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) has been shown to improve the outcome in all age groups with newly diagnosed diffuse large B-cell lymphoma (DLBCL). We conducted a retrospective analysis to evaluate the impact on clinical outcomes of adding rituximab to cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) treatment for diffuse large B-cell lymphoma (DLBCL) patients in china. A propensity score method was used to compensate for the non-randomized study design. From January 2004 to December 2009, 68 patients were newly diagnosed with DLBCL Using Hans' algorithm based on CD10, BCL-6, and MUM1, the non-germinal center (N-GCB) subgroup 45(66.2%) and germinal center B-cell-like (GCB) 23(33.8%). 32 in the rituximab plus CHOP-based chemotherapy (R+) group, and 36 in the CHOP-based chemotherapy only (R-) group. The complete response rate was significantly higher in the R+ group than in the R- group (81.1 vs. 68.1%, P 〈 0.005,); The complete response rate of N-GCB and GCB in the R+ group was78.2% and 82.1%, p 〉 0.05 respectively. The complete response rate of N-GCB and GCB in the R- group was58.2% and 71.3 %, p P 〈 0.001. The rituximab can overcome poor outcomes for N-GCB subgroup of DLBCL. The progression-free survival (PFS) at 2 years was 62.4% in the R+ group and 57.0% in the R- group. The 2-year overall survival (OS) was 76.9% for the R+ group and 69.5% for the R- group, P 〈 0.001. The 2-year overall survival (OS) was 72% in N-GCB Subgroup and 78% in GCB Subgroup for the R+ group, and 48% in N-GCB Subgroup and 68% in GCB Subgroup for the R- group. A multivariate analysis revealed that the addition of rituximab was a strong independent prognostic factor for PFS (hazard ratio 0.64, 95% CI 0.43–0.96, P = 0.031). A subgroup analysis revealed that R+ particularly benefited N-GCB subgroup patients). IPI also showed significant impact for PFS (hazard ratio 1.72, 95% CI 1.34–2.14 for one score increase, P 〈 0.001 as well as OS P 〈 0.001. In summary, the addition of rituximab to CHOP-based chemotherapy results in better outcomes for DLBCL patients, particularly patients N-GCB subgroup of DLBCL. 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: 2010
    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. 124, No. 21 ( 2014-12-06), p. 5221-5221
    Abstract: Introduction Tumor necrosis factor related apoptosis inducing ligand (TRAIL) can induce the apoptosis of many human leukemia cells while sparing of normal cells, but its resistance is also universal. Our previous study on apoptosis of t(8;21) positive acute myeloid leukemia cell line Kasumi-1 induced by rhTRAIL showed that the survival rate no longer decreased significantly when rsTRAIL reached a certain concentration which implied Kasumi-1 cells might have a resistant tendency to TRAIL. Then, we established a TRAIL-resistant Kasumi-1 cell line (Kasumi-1 TR) by intermittently escalating rsTRAIL concentration in culture media, and compared the mRNA expression profile with the original Kasumi-1 cell line by using Affymetrix Human Genome U133 Plus 2.0 Array. Methods Kasumi-1 TR cell line was established by intermittently treated Kasumi-1 cells with progressively escalating rsTRAIL concentration. Proliferation of leukemia cells were measured by CCK-8 assay, and rsTRAIL IC50 of cells and resistance index were calculated according to proliferation of cells treated with rsTRAIL at different concentrations. TRAIL and TRAIL receptors 1-4 on cells surface were detected by flow cytometry. Expression profiles of Kasumi-1 cells and Kasumi-1 TR cells were analyzed by Affymetrix Human Genome U133 Plus 2.0 Array to identify differentially expressed genes, and the search of genes possibly related with TRAIL-resistance were using by GO functional analysis and pathway enrichment analysis. Results 1) Kasumi-1 TR cells proliferation was faster than that of Kasumi-1 cells(Fig 1A); 2) IC50 of 24 hours for Kasumi-1 cells was 756.833ng/ml (logIC50 2.879 ± 0.148), IC50 of 24 hours for Kasumi-1 TR cells was 1634646.005ng/ ml (logIC50 6.213 ± 0.637), the RI of 24h was 2159 (Fig 1B); IC50 of 48 hours for Kasumi-1 cells was 345.390ng/ml (logIC50 2.538 ± 0.153), IC50 of 48 hours for Kasumi-1 TR cells was 33642.641ng/ml (logIC50 is 4.257 ± 0.317), the RI for 48h was 97 (Fig 1C); 3) Cell surface expression of TRAIL and its receptors 1-4 had no difference between two cell lines(Fig 1D). 4) There were 1537 genes up regulated by more than 2 times while 487 genes down regulated by more than 2 times in Kasumi-1 TR cells compared with the original Kasumi-1 cells (Fig 1E). Of which BCL-2 family antiapoptotic gene BCL2 is increased by 3.153 times and BCL2A1 increased by 18.23 times, IFNAR1 involved in JAK/STAT pathway increased by 12.841 times and TRAIL death receptor TNFRSF10A down regulated by 3.256 times(Fig 1F). Conclusions: The Kasumi-1 cell line with rsTRAIL resistance (Kasumi-1 TR) is established, and its resistance may be associated with the up expression of BCL2, BCL2A1, IFNAR1 and down regulated expression of DR4. Acknowledgment This work was supported by grants from NSFC (30672415) and STCSM (054119528). 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: 2014
    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. 1413-1413
    Abstract: Introduction: Chimeric antigen receptor (CAR) T-cell therapy has displayed potent anti-leukemia activity in refractory/relapsed acute lymphocytic leukemia (ALL). However, the influence of CAR-T therapy on host systemic and local immunity has not been well examined. We therefore applied high-throughput T cell receptor β chain sequencing to track dynamic change of T-cell repertoire in vivo induced by CAR-T therapy in B-cell acute lymphocytic leukemia patients. Patients and Methods: Six patients with 45 samples were under observation. The samples obtained to be tested were the peripheral blood mononuclear cell (PBMC) samples and bone marrow mononuclear cell (BMMC) samples before and after CAR-T administration, as well as the CAR-transduced autologous T cell samples on the day when they were to be infused to patients. The information of samples and patients was summarized in Table 1. The TCR full length mRNAs of these samples were deeply sequenced using the ImmunHubTM TCR profiling system (ImmunQuad Biotech). Briefly, a 5'RACE unbiased amplification protocol was used. An algorithm was applied to raw sequencing data for PCR and sequencing errors correction and V, D, J, C gene segments mapping with IMGT®. The inverse Simpson index and the clonality index was calculated to estimate TCRβ clonotype diversity and the state of clonal proliferation of T cells. The donut chart and clone tracking heat map were generated by R. Result: Compared to preinfusion TCR diversity, we observed inverse Simpson's index of 4 of 6 patients' PBMC samples (Fig. 1A) and of 3 of 4 patients' BMMC samples (Fig. 1B) had been increasing to day 7 after CAR-T treatment. As time went by, a decline of TCR diversity from day 7 to day 28 was detected in both PBMC and BMMC samples (Fig. 1). Of note, a relative rising tide of TCR diversity was observed after the decline in PBMC samples (Fig. 1A). The decreased TCR diversity leaded us to test the change of TCR clonality. As shown by Fig. 2A and 2B, in comparison with the pre-treatment and the CAR-T samples, we detected highly clonal proliferation of T cells in both blood and bone marrow. Next, we applied the clonality index to quantitatively define T cell clonal expansion (Fig. 2C and 2D). The clonality index was raised from 0.16±0.07 and 0.14±0.05 before treatment to 0.27±0.09 and 0.3±0.15 at last time point post infusion in PBMCs and BMMCs, respectively (mean ± SD, student's t test, P=0.03 and P=0.17, respectively).CAR-T-induced T-cell clonal expansion triggered us to trace back the original clonal source. For tracking clonal evolution, we selected the 100 most prevalent T cell clonotypes found at the last time point in both PBMCs and BMMCs and tracked their frequencies at earlier time points. As was displayed by the heat map, the top 100 T cell clones dominating in PBMCs and BMMCs at the last time point were barely found in CAR-T cell pool and were the low-frequency clonotypes in preinfusion samples (Fig. 3A and 3B). In order to quantitatively describe this phenomenon, we compared the total frequency of top 10 T cell clones in PBMC and BMMC samples at the last time point to their corresponding total frequency in preinfusion samples and the infused CAR-T pool (Fig. 3C, 3D, 3E and 3F). The total frequency of the top 10 T cell clones was 23%±11% and 27%±14% at last time point, 4%±6% and 8%±7% before treatment and 0.05%±0.09% and 0.01%±0.01% in the infused CAR-T samples in PBMCs and BMMCs, respectively (mean ± SD, Fig. 3C, 3D, 3E and 3F; student's t test, P=0.01, P=0.01, P=0.08 and P=0.03, respectively). Only one patient's (patient 6's) top 10 T cell clones in PBMCs at last time point were not found in both CAR-T pool and preinfusion sample, which may be caused by the limitation of sequencing detection (Fig. 3C). Conclusion: For the first time, we demonstrated CAR-T therapy could stimulate the clonal proliferation of endogenous non-CAR T cells in patients. Along with other groups' animal results (Barber A et al. J Immunol. 2009) indicating that CAR-T therapy could facilitate the infiltrating of tumor antigen-specific T cells, these expanded T cell clones of patients in our trial are most likely tumor antigen specific, which could provide synergistic anti-tumor effect following CAR-T adoptive transfer. The trial was registered in Chinese Clinical Trial Registry and the registration number is ChiCTR-OCC-15007008. 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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 4943-4943
    Abstract: Abstract 4943 The clinical safety of intravenous itraconazole use exceeding 14 days is unknown. However, many cases need further treatment, but the absorption of itraconazole oral in severely ill patients is variable and unpredictable. Gastric acidity and food influence the absorption of oral formulation. The intravenous formulation, on the other hand, achieves adequate blood levels more rapidly and its bioavailability is predictable and invariable compared to the oral formulation. We performed this study to investigate the clinical safety of long-term intravenous itraconazole for the treatment of invasive fungal infection (IFI) in severely ill patients with hematological malignancies. 20 patients (median age, 70 years; range, 38–82 years) with hematological malignancies who had met the IFI inclusion criteria were enrolled. All the infection sites were the lung. These patients were given intravenous itraconazole for 14 days. There were significant effective but the lesions were not fully absorbed and no severe adverse events were found. So the long-term intravenous itraconazole therapy was enabled. Clinical efficacy and adverse events were recorded. Itraconazole 200 mg via intravenous infusion (administered over 1 hour) twice a day for 4 doses followed by 200 mg once a day until the lesions was fully absorbed. The treatment median time was 27(18-58) days. During this period, closely monitor changes in vital signs, chest CT, liver enzymes, renal function, electrocardiogram and electrolytes were assessed. We found that the patients achieved composite endpoints including survival, defervescence and undetectable lesions, except one patient died of primary disease - acute myeloid leukemia. Treatment-related adverse events were found in 7 patients (35%) during the study and none of them were severe adverse events. Such as hypokalemia (10.0%), gastrointestinal disorders (10.0%), elevation of liver enzymes (10.0%) and pleural effusion (5.0%). All the adverse events occurred during the first two weeks of the treatment and no significant exacerbation in the following days. In conclusion, long-term intravenous itraconazole therapy was effective for IFI and the drug-related adverse events have been shown to be generally predictable and manageable. 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: 2010
    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. 134, No. Supplement_1 ( 2019-11-13), p. 965-965
    Abstract: In China, it is estimated that there are over 50,000 patients with β-thalassemia major, of which the standard care requires regular red blood cell transfusions along with iron chelation, often associated with iron overload and organ damage. The only curative therapy, allogeneic hematopoietic stem cells (HSCs) transplantation, is primarily available to a fraction of young patients with HLA-matched sibling donors. A study has estimated that 1% of patients with β-thalassemia major in China expect to live beyond age 20, representing severe unmet needs. Recently, transplantation of genetically modified autologous HSCs is emerging as an attractive therapeutic alternative for β-thalassemia. Epidemiology data have shown that in some patients with β-thalassemia, manifested clinical symptoms could be ameliorated by elevated fetal hemoglobin (HbF). As BCL11A is one of the main transcription factors known to play critical roles to repress expression of γ-globin, a critical element of HbF, we are developing ET-01, in which autologous CD34+ cells are edited by CRISPR/Cas9 to disrupt the BCL11A erythroid enhancer and reactivate the developmentally silenced γ-globin, to treat patients with β-thalassemia. Our data have shown that in ET-01, the BCL11A enhancer can be edited with high precision and efficiency, resulting in HbF levels above a potential therapeutic threshold. We have developed and scaled up the manufacturing process under the cGMP standard. IND-enabling preclinical studies, including GLP studies using ET-01 manufactured at the clinical scale, are on-going. Overall our data support continuing development of ET-01 as a potentially safe and efficacious therapy for patients with β-thalassemia major To disrupt the BCL11A enhancer, Cas9 mRNA and synthetic sgRNA were delivered into CD34+ HSCs from healthy and β0/β0 donors using electroporation. Indels frequencies (i.e., gene editing efficiency) were approximately 80%, consistent for both types of donors. Efficiency of in vitro erythroid differentiation was essentially identical between ET-01 and unedited CD34+ HSCs. HbF+ cells and γ-globin mRNA levels were both significantly elevated in ET-01 compared to unedited CD34+ HSCs. Importantly, in ET-01 made from cells of β0/β0 donors, the level of γ-globin, plus that of β-globin, reached above a threshold with potentially clinically meaningful effect. Meanwhile, data from the colony-forming unit assay have shown colony types and total numbers of colonies were comparable between those of ET-01 and unedited CD34+ HSCs, suggesting that the colony-forming capability was not significantly affected in ET-01. To evaluate the specificity of gene-editing methodology used in ET-01, we generated a signature panel consisting of 42 sites with highest probability of off-target editing by in silico prediction and unbiased DiGenome-seq. These sites were deeply interrogated via targeted PCR amplification and next generation sequencing analysis. The data showed no significant off-target events at these sites, suggesting minimal off-target effects for ET-01. Moreover, to evaluate the in vivo engraftment potential of ET-01, we transplanted ET-01 and unedited CD34+ HSCs into immunodeficient NPG mice. Both types of cells produced similarly rapid and efficient engraftment in all mice, and similar multi-lineage reconstitution of human cells in multiple hematopoietic and immune organs. Importantly, the Indels efficiencies of ET-01 were maintained at similar levels in NPG mice after 4 months transplantation and after 2nd transplantation. These findings suggest that ET-01 retained in vivo long-term hematopoietic repopulating and differentiation capabilities. No tumorigenicity was observed in these in vivo studies. ET-01 manufacturing process has been successfully scaled up to 9.69x108 CD34+ HSC produced using mobilized leukopak from healthy donors. This upper bound is limited by the amount of starting materials obtained, not the actual process capacity. High cell viability, CD34+ purity and Indels frequencies were maintained throughout the manufacturing process and achieved in all batches. In vitro erythroid differentiation efficiency, HbF+ cells and γ-globin mRNA levels were similar to those observed at research scale. IND-enabling preclinical studies, including GLP studies using ET-01 manufactured at the clinical scale, are ongoing. Disclosures Fang: EdiGene Inc.: Employment. Yuan:EdiGene, Inc.: Employment. Yu:EdiGene, Inc.: Employment. Yang:EdiGene, Inc.: Employment. Liu:EdiGene Guangzhou Inc.: Employment. Shi:EdiGene Guangzhou Inc.: Employment. Zhang:EdiGene Inc.: Employment. Zhang:EdiGene Inc.: Employment. Zhao:EdiGene Inc.: Employment. Li:EdiGene Inc.: Employment. Wei:EdiGene, Inc.: Employment.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...