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  (5)
Material
Publisher
  • American Society of Hematology  (5)
Language
Years
Subjects(RVK)
  • 1
    In: Blood Advances, American Society of Hematology, Vol. 5, No. 7 ( 2021-04-13), p. 1837-1847
    Abstract: Treatment refusal and death as a result of toxicity account for most treatment failures among children with acute myeloid leukemia (AML) in resource-constrained settings. We recently reported the results of treating children with AML with a combination of low-dose cytarabine and mitoxantrone or omacetaxine mepesuccinate with concurrent granulocyte colony-stimulating factor (G-CSF) (low-dose chemotherapy [LDC]) for remission induction followed by standard postremission strategies. We have now expanded the initial cohort and have provided long-term follow-up. Eighty-three patients with AML were treated with the LDC regimen. During the study period, another 100 children with AML received a standard-dose chemotherapy (SDC) regimen. Complete remission was attained in 88.8% and 86.4% of patients after induction in the LDC and SDC groups, respectively (P = .436). Twenty-two patients in the LDC group received SDC for the second induction course. Significantly more high-risk AML patients were treated with the SDC regimen (P = .035). There were no significant differences between the LDC and SDC groups in 5-year event-free survival (61.4% ± 8.7% vs 65.2% ± 7.4%, respectively; P = .462), overall survival (72.7% ± 6.9% vs 72.5% ± 6.2%, respectively; P = .933), and incidence of relapse (20.5% ± 4.5% vs 17.6% ± 3.9%, respectively; P = .484). Clearance of mutations based on the average variant allele frequency at complete remission in the LDC and SDC groups was 1.9% vs 0.6% (P & lt; .001) after induction I and 0.17% vs 0.078% (P = .052) after induction II. In conclusion, our study corroborated the high remission rate reported for children with AML who received at least 1 course of LDC. The results, although preliminary, also suggest that long-term survival of these children is comparable to that of children who receive SDC regimens.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 5253-5253
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 5253-5253
    Abstract: Background: Acute myeloid leukemia (AML) is the most common form of hematological malignant tumors that threatens human health. In the last decades, the rapid evolution in cytogenetics and molecular abnormalities made a breakthrough in the diagnosis and prognosis prediction of AML. However, there still has high heterogeneity among AML patients. Recently, researchers focused on long non-coding RNAs (lncRNAs), which once were addressed as products of "junk DNA", may play a key role in the initiation and progression of AML. Furthermore, a study from Ohio State's Comprehensive Cancer Center built a useful prognostic lncRNA score system for elder patients ( 〉 60 years) with cytogenetically normal AML by 48 lncRNAs [Garzon et al. PNAS 2014; 111:18679-84]. It can be expected that lncRNAs will promote the diagnosis and risk categories of AML in the near future. In this study, a risk scoring system was constructed upon 3 lncRNAs in de novo AML patients. We also sought to explore the functionality of these lncRNAs. Methods: By using Arraystar Human LncRNA Array V4.0, we obtained deregulated transcripts in AML, including 3,499 lncRNAs and 3,105 mRNAs (GSE103828). Expression patterns of all deregulated transcripts were extracted from 151 AML patients of The Cancer Genome Atlas (TCGA) RNA sequencing data. Through mathematical modeling, we identified 3 lncRNAs whose expression levels were independently associated with overall survival (OS). We then constructed a risk scoring system based on the 3 lncRNAs, age and 2008 WHO risk categories. Then the Receiver Operating Characteristic (ROC) was used to identify its test power and the best threshold score for 3-year survival status. Bone marrow samples from patients with AML and iron deficiency anemia (IDA) were collected. qRT-PCR was performed to verify the expression of lncRNAs in AML patients and IDA controls. Results: In the TCGA dataset, the area under ROC curve was 0.765, which indicated the risk scoring system has a good efficiency to predict 3-year survival status for AML patients, and the threshold score 1.639 was recommended to distinguish the high and low risk score groups (Figure A). Patients with higher risk score had a shorter OS (median 440.31 days vs. 886.16 days, p 〈 0.001, Figure B) than those with lower risk score. To promote the practicality of the risk scoring system, we constructed Nomogram predictive modeling. The patient with 1.639 risk score was shown in the Nomogram (Figure C). We then performed qRT-PCR to verify the expression of lncRNAs and the availability of the risk scoring system. All of the 3 lncRNAs, RP11-222K16.2, LINC00899 and RP11-305O6.3, showed significantly lower expression in AML (n=46) compared to IDA controls (n=17, P 〈 0.05), and the risk scoring system performed remarkable predicting effectiveness among AML patients (Figure D). Conclusion: We identified 3 deregulated lncRNAs in AML and constructed a risk scoring system based on the it, which provided distinct insights into the clinical and biological implications of lncRNAs expression in de novo AML patients. It may improve the risk stratification of newly-diagnosed AML patients. Further studies on the mechanisms of the lncRNAs are in process. 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 ...
  • 3
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 5179-5179
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 5179-5179
    Abstract: Introduction: Acute myeloid leukemia (AML) is a heterogeneous disease characterized by the clonal proliferation of immature myeloid progenitor cells in the bone marrow, compressing normal blood cell production and leading to bone marrow failure ultimately. Overwhelming evidence has established that non-coding RNAs (ncRNAs), such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) have great role in AML pathogenesis. Circular RNAs (circRNAs) that occupy gene expression at the transcriptional or post-transcriptional level have great potential to be biomarker for types of cancers. We have screened one altered circRNA named circ-ANAPC7 in AML before. In this study, we aimed to validate its expression by enlarging sample size and illuminating the diagnostic and monitoring value of circ-ANAPC7 in AML. Methods: Real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was supposed to confirm the expression of circ-ANAPC7 of AML patients. The sequences of circ-ANAPC7 primers were as follows: 5′- GGGAGCAGCACTTAGGAACAT -3′ (sense) and 5′-AAAGCTGGTACTTCTGAGGTGG-3′ (antisense). Receiver operating characteristic (ROC) curve was carried out to evaluate the diagnostic value. Overall survival rate and event-free survival rate were estimated by the Kplan-Meier analysis and compared using the log-rank test. All tests were two-sided, and P 〈 0.05 was defined as a significant difference. Results: The expression level of circ-ANAPC7 in newly diagnosed AML was significantly higher than CR patients and iron deficiency anemia (IDA) control group (P 〈 0.001) (Figure 1A). Furthermore, we chose 24 AML patients who undergo the condition of newly diagnosed AML, CR and relapse to dynamical monitor the expression of circ-ANAPC7. We discovered that circ-ANAPC7 expression level changed accompanied with the disease condition transformation. It was high expressed in newly diagnosed and relapsed AML patients. When patients got CR, the expression level of circ-ANAPC7 decreased (P 〈 0.05). In the continuous CR patients, it remained in a minimal level (Figure 1C). ROC curve analysis revealed that circ-ANAPC7 has significant value of AML diagnosis (AUC = 0.915, P 〈 0.001) (Figure 1B). Moreover, we conducted survival analysis to explore long-term effect of circ-ANAPC7 expression in AML patients. The result revealed that circ-ANAPC7 expression was not related to overall survival (OS) and disease-free survival (DFS) of AML patients (P 〉 0.05) (Figure 1D). Conclusions: We validated that circ-ANAPC7 was upregulated in AML patients. The clinical analysis revealed that circ-ANAPC7 may be a predictive index for diagnosing and supervising early recurrence of AML. What's more, additional molecular mechanisms and biological functions of circ-ANAPC7 merit deeper investigation. 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: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 132, No. Supplement 1 ( 2018-11-29), p. 5254-5254
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 5254-5254
    Abstract: Introduction: Acute myeloid leukemia (AML) is a malignant clonal proliferative disease originating from hematopoietic stem cells, characterized by anemia, hemorrhage, infection and organ infiltration. Most of the patients had poor prognosis and the molecular mechanism of AML is not clear. Therefore, it is urgent to find new molecular markers and therapeutic targets for AML. Long noncoding RNAs (lncRNAs) are a class of RNAs, which are longer than 200nt and without the ability of encoding functional proteins [A. Fatica et al., 2014] . Accumulating studies have found that lncRNAs play an important role in the development of hematological tumors, especially in AML [Huang J et al., 2014; Xing C-y et al., 2015]. In order to investigate the mechanism of lncRNAs in AML, we screened the differentially expressed RNAs in 5 AML patients and 5 IDA controls by microarray (GSE103828), and lncRNA AC092580.4 was verified markedly low expressed in AML (fold change=15.25, P= 0.0002). Then, We further explored the relationship between the expression levels of AC092580.4 and the clinical characteristics and prognosis of AML. Methods: Bone marrow (BM) samples from patients with AML and iron deficiency anemia (IDA) were collected and qRT-PCR was performed to verify the expression levels of AC092580.4 in AML patients and controls. The Receiver Operating Characteristic (ROC) was established to analyze whether AC092580.4 could be used as a bio-marker for newly diagnosed AML patients. The clinical data, including age, gender, percentage of blast cells in BM, chromosome and gene abnormalities, risk stratification, white blood cell counts and overall survival were collected. The correlation between the expression levels of AC092580.4 and clinical characteristics were analyzed by SPSS22.0. Results: The expression levels of AC092580.4 in newly diagnosed AML (n=83) was significantly lower (Figure A) than IDA (n=24) (p 〈 0.0001). The ROC (Figure B) was established and the area under curve was 0.868 (p 〈 0.001, CI: 0.780 - 0.957), which indicated that AC092580.4 could be used as an effective index to distinguish the newly diagnosed AML from the controls. Clinical data were collected from 83 newly diagnosed AML patients, 67% was the mean value of the percentage of blast cells in BM . It was found that the expression levels of AC092580.4 were significantly different between the high (≥67%) and low( 〈 67%) percentage of blast cells (p=0.001), high white blood cell group (WBC≥100×109/L) and non high white blood cell group (WBC 〈 100×109/L) (p=0.021). There was no significant difference of the expression levels in the patients with favourable, intermediate and adverse according to Diagnosis and management of AML in adults 2017 ELN recommendations from an international expert panel (p=0.328). The overall survival rate (Figure C) analysis showed that AML patients with high expression levels of AC092580.4 have a better prognosis than low expression group (p=0.018), which indicated that AC092580.4 may be used as an effective index to predict prognosis. To explore the mechanism of AC092580.4 in AML, we obtained sequencing data of 151 adult AML patients from The Cancer Genome Atlas (TCGA) (https://cancergenome.nih.gov/abouttcga/). The correlation coefficient between AC092580.4 and coding gene was estimated by Pearson correlation method. The results showed that the correlation coefficient between GATA3 and AC092580.4 (r = 0.679, P 〈 0.05) was the most significant one among 27338 transcripts (Figure D). Therefore, we hypothesize that AC092580.4 may play an important role in the pathogenesis of AML by interacting with GATA3. GATA3 is a transcription factor of GATA family. While promoting the development of Th2 cells, it can also inhibit the Th1 polarization of Th cells, and eventually lead to higher expression of Th2, which may lead to the occurrence of tumor. Conclusions: Our study sheds light on a novel lncRNA AC092580.4 in AML, and provides its possible functional mechanism preliminary. Whether it can be used as a new powerful therapeutic target for AML treatment remains to be further studied. 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 ...
  • 5
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 4470-4470
    Abstract: Introduction: Multiple myeloma (MM) is a fatal B-cell malignancy characterized by abnormal proliferation of plasma cells (PCs) in bone marrow (BM). Despite the remarkable improvements have been done in knowledge on the pathobiology of MM, treatment and patients care, MM is still an incurable disease. Long non-coding RNAs (lncRNAs) are a new sort of noncoding RNA longer than 200 nucleotides and without the function of translating into canonical functional proteins. Increasing evidence suggests that lncRNAs have huge impact on adjusting gene expression through the processes of transcription and post-transcription regulation, genomic imprinting, and chromatin modification. We have validated several dysregulated lncRNAs in MM by microarray and bioinformatic analysis. Among them, we focused on the function and mechanism of ST3 beta-galactoside alpha-2,3-sialyltransferase 6 antisense RNA 1(ST3GAL6-AS1), which was upregulated markedly in MM patients. In our previous study, we verified the upregulation level and the co-expression relationship of ST3GAL6-AS1 and ST3 beta-galactoside alpha-2,3-sialyltransferase 6 (ST3GAL6) in bone marrow samples from MM patients. Methods: MM cell lines MM1.S and RPMI-8226 were confirmed to have high expression levels of ST3GAL6-AS1 at RNA level compared with healthy controls. Knockdown of ST3GAL6-AS1 was conducted in MM1.S and RPMI-8226 cells using lncRNA Smart Silencer from Ribobio (Guangzhou, China). ST3GAL6-AS1 knockdown and ST3GAL6 expression were confirmed by quantitative real-time PCR (qRT-PCR) at RNA level. We next evaluated the effects of ST3GAL6-AS1 knockdown on MM cell adhesion to human umbilical vein endothelial cell (HUVEC), fibronectin and collagen I coated plates after MM1.S and RPMI-8226 was labeled by CFSE. Migration and invasion to complete medium were assessed using transwell plates comparing ST3GAL6-AS1 knockdown cells to NC controls. Matrigel matrix was coated at transwell plates in invasion analysis. Cell count in adhesion analysis was conducted by Image J software, while migration and invasion analysis were evaluated by flow cytometer. Results: ST3GAL6-AS1 (Genebank number: NR_046683.1) was a 769bp antisense RNA, which located in chromosome 3. Its expression level was decreased dramatically after transfection of ST3GAL6-AS1 smart silencer or control siRNA into MM1.S and RPMI-8226 cells (Figure 1A). There was a significant reduction in the ability of knockdown MM1.S and RPMI-8226 cells to adhere to HUVEC, fibronectin and collagen I in vitro compared to NC controls (Figure 1B). Migration and invasion ability of MM cells transfected with lncRNA smart silencer in response to complete medium were also decreased obviously (Figure 1C). However, there was no significant difference in the ability of proliferation and apoptosis between ST3GAL6-AS1 knockdown group and NC controls. Furthermore, the expression level of ST3GAL6 was decreased in ST3GAL6-AS1 knockdown MM cells (Figure 1D). Conclusions: Knockdown of ST3GAL6-AS1 in MM cells significantly inhibits adhesion, migration and invasion in vitro, indicating that ST3GAL6-AS1 may play an important role in the malignant behavior of MM cells. The co-expression between ST3GAL6-AS1 and gene ST3GAL6 has been demonstrated in our previous study, which was further confirmed in present study. Researches are ongoing to address the potential mechanism among them in MM. 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: 2018
    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...