Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 45(5): 768-772, 2023 Oct.
Article in Chinese | MEDLINE | ID: mdl-37927018

ABSTRACT

Objective To compare the macular structure and microcirculation in both eyes of the patients with myopic anisometropia.Methods Optical coherence tomography angiography(OCTA)was employed to scan the macular areas in both eyes of 44 patients with myopic anisometropia.The patients were assigned into high and low groups based on the refractive diopter,and the parameters such as retinal thickness,choroidal thickness,vascular density,and perfusion density in the macular areas of both eyes were compared between the two groups.Results Other macular areas except the central and external nasal areas and the choroid of the fovea in the high group were thinner than those in the low group(all P<0.05).There was no statistically significant difference in retinal vascular density or perfusion density in different areas between the two groups(all P>0.05).Conclusion In the patients with myopic anisometropia,most areas of the retina in the case of high myopia is thinner than that in the case of low myopia,while there is no difference in retinal vascular density or perfusion density in both eyes.


Subject(s)
Anisometropia , Myopia , Humans , Choroid/blood supply , Microcirculation , Retina , Tomography, Optical Coherence/methods
2.
Tumori ; 106(5): 346-355, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32811340

ABSTRACT

Hereditary diffuse gastric cancer (HDGC), comprising 1%-3% of gastric malignances, has been associated with CDH1 variants. Accumulating evidence has demonstrated more than 100 germline CDH1 variant types. E-cadherin encoded by the CDH1 gene serves as a tumor suppressor protein. CDH1 promoter hypermethylation and other molecular mechanisms resulting in E-cadherin dysfunction are involved in the tumorigenesis of HDGC. Histopathology exhibits characteristic signet ring cells, and immunohistochemical staining may show negativity for E-cadherin and other signaling proteins. Early HDGC is difficult to detect by endoscopy due to the development of lesions beneath the mucosa. Prophylactic gastrectomy is the most recommended treatment for pathogenic CDH1 variant carriers. Recent studies have promoted the progression of promising molecular-targeted therapies and management strategies. This review summarizes recent advances in CDH1 variant types, tumorigenesis mechanisms, diagnosis, and therapy, as well as clinical implications for future gene therapies.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Carcinogenesis/genetics , Genetic Predisposition to Disease , Stomach Neoplasms/genetics , Gastrectomy , Germ-Line Mutation/genetics , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Stomach Neoplasms/therapy
3.
Proc Natl Acad Sci U S A ; 117(33): 20117-20126, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747558

ABSTRACT

t(8;21)(q22;q22) acute myelogenous leukemia (AML) is morphologically characterized by a continuum of heterogeneous leukemia cells from myeloblasts to differentiated myeloid elements. Thus, t(8;21) AML is an excellent model for studying heterogeneous cell populations and cellular evolution during disease progression. Using integrative analyses of immunophenotype, RNA-sequencing (RNA-seq), and single-cell RNA-sequencing (scRNA-seq), we identified three distinct intrapatient leukemic cell populations that were arrested at different stages of myeloid differentiation: CD34+CD117dim blasts, CD34+CD117bri blasts, and abnormal myeloid cells with partial maturation (AM). CD117 is also known as c-KIT protein. CD34+CD117dim cells were blocked in the G0/G1 phase at disease onset, presenting with the regular morphology of myeloblasts showing features of granulocyte-monocyte progenitors (GMP), and were drug-resistant to chemotherapy. Genes associated with cell migration and adhesion (LGALS1, EMP3, and ANXA2) were highly expressed in the CD34+CD117dim population. CD34+CD117bri blasts were blocked a bit later than the CD34+CD117dim population in the hematopoietic differentiation stage and displayed high proliferation ability. AM cells, which bear abnormal myelocyte morphology, especially overexpressed granule genes AZU1, ELANE, and PRTN3 and were sensitive to chemotherapy. scRNA-seq at different time points identified CD34+CD117dim blasts as an important leukemic cluster that expanded at postrelapse refractory stage after several cycles of chemotherapy. Patients with t(8;21) AML with a higher proportion of CD34+CD117dim cells had significantly worse clinical outcomes than those with a lower CD34+CD117dim proportion. Univariate and multivariate analyses identified CD34+CD117dim proportion as an independent factor for poor disease outcome. Our study provides evidence for the multidimensional heterogeneity of t(8;21)AML and may offer new tools for future disease stratification.


Subject(s)
Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/pathology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/metabolism , Adult , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/metabolism , Transcriptome
4.
FEBS Open Bio ; 10(8): 1655-1667, 2020 08.
Article in English | MEDLINE | ID: mdl-32594651

ABSTRACT

Paclitaxel (PTX) has previously been used to treat tumours of various tissue origins, such as lung, breast, ovarian, prostate cancers and leukemia. PTX-induced apoptosis is associated with p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinase (ERK), nuclear factor-kappa B (NF-κB) and c-Jun N-terminal kinase or stress-activated protein kinase (JNK/ SAPK) pathways. Transforming growth factor-beta-activated kinase 1 (TAK1) and TAK1-binding protein 1 (TAB1) play an important role in cell apoptosis through the p38, ERK, NF-κB and JNK signal transduction pathways. To investigate the role of TAK1 in PTX-induced cell apoptosis, we treated HEK293 and 8305C cells with 0-20 µM PTX for 6, 12 or 24 h. To investigate whether TAK1 can cooperate with PTX for cancer treatment, we transfected cells with TAK1, TAB1 or control plasmid and treated them with PTX (3-10 µM) for 9-24 h. Apoptosis rates were analysed by flow cytometry (Annexin V/PI). Endogenous TAK1 and TAB1, caspase-7 cleavage, poly ADP-ribose polymerase (PARP) cleavage, Bcl-xL level, phospho-p44/42, phospho-JNK and phospho-p38 were detected by western blot. We show that in HEK293 and 8305C cells, PTX enhanced the endogenous TAK1/TAB1 level and induced cell apoptosis in a dose- and time-dependent manner. Upon TAK1 overexpression in HEK293 cells treated with PTX, apoptosis rate, JNK phosphorylation and PARP cleavage increased contrary to heat-shocked or untreated cells. CRISPR editing of the tak1 gene upon PTX treatment resulted in lower phospho-JNK and PARP cleavage levels than in cells transfected with the control or the TAK1- or TAB1 + TAK1-containing plasmids. TAK1-K63A could not induce JNK phosphorylation or PARP cleavage. We conclude that PTX induces HEK293 and 8305C cell apoptosis through the TAK1-JNK activation pathway, potentially highlighting TAK1's role in chemosensitivity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Paclitaxel/pharmacology , Cells, Cultured , Humans , Signal Transduction/drug effects
5.
Stem Cells Dev ; 26(20): 1460-1467, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28810819

ABSTRACT

Recent reports state that C-type lectin-like molecule-1 (CLL-1) in acute myeloid leukemia (AML) is expressed primarily on myeloid cells, but there is still no investigation about its prognostic significance on leukemic blast compartment. Hence, this study aimed to evaluate the prognostic value of CLL-1 in 123 patients with de novo CD34+ Non-M3 AML. Multiparameter flow cytometry was used to assess the expression of CLL-1 on immature compartment in AML and control groups. We found that CLL-1 expression level on blast compartment was closely linked to clinical characteristics, treatment response, and survival outcome of patients. Decreased expression of CLL-1 was observed on immature compartment from AML patients as compared with controls (62.6% vs. 86.5%, P < 0.05). Logistic model exhibited that CLL-1low independently predicted low complete remission rate with an odds ratio of 4.57 (2.53-6.61, P < 0.05). Additionally, CLL-1 expression level at diagnosis was inversely correlated to the residual blast cells (residual leukemia cell) after induction chemotherapy (r = -0.423, P < 0.05). Furthermore, multivariate Cox regression model demonstrated that CLL-1low was still an independent adverse predictor (P < 0.05 for event-free survival, P < 0.05 for overall survival). Notably, CLL-1low was able to discriminate poor survival patients from intermediate- and favorable-risk groups. Taken together, CLL-1 is a novel prognostic predictor that could be exploited to supplement the current AML prognostic risk stratification system, and potentially optimize the clinical management of AML.


Subject(s)
Antigens, CD34/metabolism , Lectins, C-Type/metabolism , Leukemia, Myeloid, Acute/metabolism , Adolescent , Adult , Aged , Disease-Free Survival , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Multivariate Analysis , Prognosis , Remission Induction , Young Adult
6.
J Hematol Oncol ; 8: 48, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25957890

ABSTRACT

BACKGROUND: Minimal residual disease detection in the bone marrow is usually performed in patients with acute myeloid leukemia undergoing one course of induction chemotherapy. To optimize the chemotherapy strategies, more practical and sensitive markers are needed to monitor the early treatment response during induction. For instance, peripheral blood (PB) blast clearance rate may be considered as such a monitoring marker. METHODS: PB blasts were monitored through multiparameter flow cytometry (MFC). Absolute counts were determined before treatment (D0) and at specified time points of induction chemotherapy (D3, D5, D7, and D9). The cut-off value of D5 peripheral blast clearance rate (D5-PBCR) was defined through receiver operating characteristic (ROC) analysis. Prognostic effects were compared among different patient groups according to D5-PBCR cut-off value. RESULTS: D5-PBCR cut-off value was determined as 99.55%. Prognostic analysis showed that patients with D5-PBCR ≥99.55% more likely achieved complete remission (94.6% vs. 56.1%, P < 0.001) and maintained a relapse-free status than other patients (80.56% vs. 57.14%, P = 0.027). Survival analysis revealed that relapse-free survival (RFS) and overall survival (OS) were longer in patients with D5-PBCR ≥99.55% than in other patients (two-year OS: 71.0% vs. 38.7%, P = 0.011; two-year RFS: 69.4% vs. 30.7%, P = 0.026). In cytogenetic-molecular intermediate-risk group, a subgroup with worse outcome could be distinguished on the basis of D5-PBCR (<99.55%; OS: P = 0.033, RFS: P = 0.086). CONCLUSIONS: An effective evaluation method of early treatment response was established by monitoring PB blasts through MFC. D5-PBCR cut-off value (99.55%) can be a reliable reference to predict treatment response and outcome in early stages of chemotherapy. The proposed marker may be used in induction regimen modification and help optimize cytogenetic-molecular prognostic risk stratification.


Subject(s)
Blast Crisis/drug therapy , Flow Cytometry/methods , Leukemia, Myeloid, Acute/drug therapy , Neoplasm, Residual/diagnosis , Adolescent , Adult , Aged , Area Under Curve , Blast Crisis/mortality , Female , Humans , Induction Chemotherapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis , ROC Curve , Remission Induction , Survival Analysis , Young Adult
8.
Proc Natl Acad Sci U S A ; 111(7): 2620-5, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24497509

ABSTRACT

The gene encoding DNA methyltransferase 3A (DNMT3A) is mutated in ∼20% of acute myeloid leukemia cases, with Arg882 (R882) as the hotspot. Here, we addressed the transformation ability of the DNMT3A-Arg882His (R882H) mutant by using a retroviral transduction and bone marrow transplantation (BMT) approach and found that the mutant gene can induce aberrant proliferation of hematopoietic stem/progenitor cells. At 12 mo post-BMT, all mice developed chronic myelomonocytic leukemia with thrombocytosis. RNA microarray analysis revealed abnormal expressions of some hematopoiesis-related genes, and the DNA methylation assay identified corresponding changes in methylation patterns in gene body regions. Moreover, DNMT3A-R882H increased the CDK1 protein level and enhanced cell-cycle activity, thereby contributing to leukemogenesis.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , Leukemia, Myelomonocytic, Chronic/genetics , Animals , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , DNA Methyltransferase 3A , Flow Cytometry , Gene Expression Profiling , Humans , Immunoblotting , Immunophenotyping , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Inbred BALB C , Microarray Analysis , Mutagenesis, Site-Directed , Mutation, Missense/genetics
9.
Blood ; 119(12): 2873-82, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22318203

ABSTRACT

The BCR-ABL fusion protein generated by t(9;22)(q34;q11) in chronic myeloid leukemia (CML) plays an essential role in the pathogenesis of the myeloproliferative disorder status at the chronic phase of the disease, but progression from the chronic phase to blast crisis (BC) is believed to require additional mutations. To explore the underlying mechanisms for BC, which is characterized by a blockage of blood cell differentiation, we screened several genes crucial to hematopoiesis and identified 10 types of mutations in RUNX1 among 11 of 85 (12.9%) patients with acute transformation of CML. Most of the mutations occurred in the runt homology domain, including H78Q, W79C, R139G, D171G, R174Q, L71fs-ter94, and V91fs-ter94. Further studies indicated that RUNX1 mutants not only exhibited decreased transactivation activity but also had an inhibitory effect on the WT RUNX1. To investigate the leukemogenic effect of mutated RUNX1, H78Q and V91fs-ter94 were transduced into 32D cells or BCR-ABL-harboring murine cells, respectively. Consistent with the myeloblastic features of advanced CML patients with RUNX1 mutations, H78Q and V91fs-ter94 disturbed myeloid differentiation and induced a BC or accelerated phase-like phenotype in mice. These results suggest that RUNX1 abnormalities may promote acute myeloid leukemic transformation in a subset of CML patients.


Subject(s)
Blast Crisis/genetics , Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation , Adolescent , Adult , Animals , Cell Differentiation/genetics , Female , Humans , Immunoprecipitation , Male , Mice , Middle Aged , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Young Adult
10.
Proc Natl Acad Sci U S A ; 108(6): 2450-5, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21262832

ABSTRACT

The full-length AML1-ETO (AE) fusion gene resulting from t(8;21)(q22;q22) in human acute myeloid leukemia (AML) is not sufficient to induce leukemia in animals, suggesting that additional mutations are required for leukemogenesis. We and others have identified activating mutations of C-KIT in nearly half of patients with t(8;21) AML. To test the hypothesis that activating C-KIT mutations cooperate with AE to cause overt AML, we generated a murine transduction and transplantation model with both mutated C-KIT and AE. To overcome the intracellular transport block of human C-KIT in murine cells, we engineered hybrid C-KIT (HyC-KIT) by fusing the extracellular and transmembrane domains of the murine c-Kit in-frame to the intracellular signaling domain of human C-KIT. We showed that tyrosine kinase domain mutants HyC-KIT N822K and D816V, as well as juxtamembrane mutants HyC-KIT 571+14 and 557-558Del, could transform murine 32D cells to cytokine-independent growth. The protein tyrosine kinase inhibitor dasatinib inhibited the proliferation of 32D cells expressing these C-KIT mutants, with potency in the low nanomolar range. In mice, HyC-KIT N822K induced a myeloproliferative disease, whereas HyC-KIT 571+14 induces both myeloproliferative disease and lymphocytic leukemia. Interestingly, coexpression of AE and HyC-KIT N822K led to fatal AML. Our data have further enriched the two-hit model that abnormalities of both transcription factor and membrane/cytosolic signaling molecule are required in AML pathogenesis. Furthermore, dasatinib prolonged lifespan of mice bearing AE and HyC-KIT N822K-coexpressing leukemic cells and exerted synergic effects while combined with cytarabine, thus providing a potential therapeutic for t(8;21) leukemia.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit , Leukemia, Myeloid, Acute , Mutation , Oncogene Proteins, Fusion , Proto-Oncogene Proteins c-kit , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Cytarabine/pharmacology , Humans , Leukemia, Lymphoid/genetics , Leukemia, Lymphoid/metabolism , Leukemia, Lymphoid/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Transgenic , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RUNX1 Translocation Partner 1 Protein , Translocation, Genetic/genetics
11.
Clin Cancer Res ; 12(10): 3043-9, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707600

ABSTRACT

PURPOSE: NOTCH signaling pathway is essential in T-cell development and NOTCH1 mutations are frequently present in T-cell acute lymphoblastic leukemia (T-ALL). To gain insight into its clinical significance, NOTCH1 mutation was investigated in 77 patients with T-ALL. EXPERIMENTAL DESIGN: Detection of NOTCH1 mutation was done using reverse transcription-PCR amplification and direct sequencing, and thereby compared according to the clinical/biological data of the patients. RESULTS: Thirty-two mutations were identified in 29 patients (with dual mutations in 3 cases), involving not only the heterodimerization and proline/glutamic acid/serine/threonine domains as previously reported but also the transcription activation and ankyrin repeat domains revealed for the first time. These mutations were significantly associated with elevated WBC count at diagnosis and independently linked to short survival time. Interestingly, the statistically significant difference of survival according to NOTCH1 mutations was only observed in adult patients (>18 years) but not in pediatric patients (< or = 18 years), possibly due to the relatively good overall response of childhood T-ALL to the current chemotherapy. NOTCH1 mutations could coexist with HOX11, HOX11L2, or SIL-TAL1 expression. The negative effect of NOTCH1 mutation on prognosis was potentiated by HOX11L2 but was attenuated by HOX11. CONCLUSION: NOTCH1 mutation is an important prognostic marker in T-ALL and its predictive value could be even further increased if coevaluated with other T-cell-related regulatory genes. NOTCH pathway thus acts combinatorially with oncogenic transcriptional factors on T-ALL pathogenesis.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/physiopathology , Receptor, Notch1/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , DNA Mutational Analysis , Female , Humans , Male , Molecular Sequence Data , Predictive Value of Tests , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Analysis
12.
Proc Natl Acad Sci U S A ; 102(4): 1104-9, 2005 Jan 25.
Article in English | MEDLINE | ID: mdl-15650049

ABSTRACT

To explore the genetic abnormalities that cooperate with AML1-ETO (AE) fusion gene to cause acute myeloid leukemia (AML) with t(8;21), we screened a number of candidate genes and identified 11 types of mutations in C-KIT gene (mC-KIT), including 6 previously undescribed ones among 26 of 54 (48.1%) cases with t(8;21). To address a possible chronological order between AE and mC-KIT, we showed that, among patients with AE and mC-KIT, most leukemic cells at disease presentation harbored both genetic alteration, whereas in three such cases investigated during complete remission, only AE, but not mC-KIT, could be detected by allele-specific PCR. Therefore, mC-KIT should be a subsequent event on the basis of t(8;21). Furthermore, induced expression of AE in U937-A/E cells significantly up-regulated mRNA and protein levels of C-KIT. This may lead to an alternative way of C-KIT activation and may explain the significantly higher C-KIT expression in 81.3% of patients with t(8;21) than in patients with other leukemias. These data strongly suggest that t(8;21) AML follows a stepwise model in leukemogenesis, i.e., AE represents the first, fundamental genetic hit to initiate the disease, whereas activation of the C-KIT pathway may be a second but also crucial hit for the development of a full-blown leukemia. Additionally, Gleevec suppressed the C-KIT activity and induced proliferation inhibition and apoptosis in cells bearing C-KIT N822K mutation or overexpression, but not in cells with D816 mC-KIT. Gleevec also exerted a synergic effect in apoptosis induction with cytarabine, thus providing a potential therapeutic for t(8;21) leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Leukemia, Myeloid, Acute/genetics , Mutation , Oncogene Proteins, Fusion/genetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Transcription Factors/genetics , Translocation, Genetic , Adolescent , Adult , Apoptosis/drug effects , Benzamides , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit , Female , Humans , Imatinib Mesylate , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged , RUNX1 Translocation Partner 1 Protein
13.
Hematol J ; 3(3): 137-44, 2002.
Article in English | MEDLINE | ID: mdl-12111649

ABSTRACT

INTRODUCTION: Although retrospective analysis were frequently undertaken, and many prognostic systems for myelodysplastic syndromes (MDS) have been proposed worldwide, few such studies have been performed and the effectiveness of different scoring systems have not yet been verified in independent patient populations in China. The aim of this single center study was to evaluate the prognostic factors and compare the prognostic scoring systems in Chinese patients with MDS. MATERIALS AND METHODS: One hundred and twenty-eight patients diagnosed as primary MDS in our Institution were studied retrospectively to identify significant prognostic factors and to assess the predictive value of 11 previously described prognostic systems, including French-American-British (FAB) classification, World Health Organization (WHO) classification, Mufti, Sanz, Morra, Aul, Oguma, Toyama, Morel and international prognostic scoring system (IPSS). RESULTS: The median age of the patients was 50 years (range 13-82). The 2- and 5-year survival rate of the patients were 55.22+/-4.90% and 26.09+/-6.36% respectively, with a median survival of 31 months (range 1-127 months). Fifty patients (39.1%) had progressed to acute leukemia (AL) with a median time of 8 months (range 1-43 months). Major independent variables indicated by multivariate analysis were the percentage of bone marrow (BM) blast cells and complex karyotype aberrations for survival (P=0.042 and 0.042, respectively) and only the percentage of BM blast cells for AL transformation (P=0.023). All the systems except Mufti scores successfully discriminated risk groups concerning both survival and AL evolution, especially in the high risk group, ranging from 10 to 20 months and from 4 to 7 months, respectively. The FAB and WHO classification, as well as Sanz, Oguma, Morel and IPSS possessed lower P value (P<0.0001) than that of the rest scoring systems. CONCLUSION: The patients in our study were younger than these of the Western population, whereas the survival and AL transformation ratio were comparable to these previous studies. The BM blast proportion and complex chromosomal defects were highly significant for predicting outcome in MDS patients. Most investigated systems effectively stratified patients into groups with different life expectancies and identified a subset of patients with poor clinical outcome. The FAB, WHO classification, as well as Sanz, Oguma, Morel and IPSS scoring systems were more applicable for predicting survival and leukemia progression.


Subject(s)
Myelodysplastic Syndromes/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Blast Crisis , Cell Transformation, Neoplastic , China , Chromosome Aberrations , Cytogenetic Analysis , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/mortality , Prognosis , Retrospective Studies , Severity of Illness Index , Survival Analysis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...