MDM2 SNP309 and TP53 SNPArg72Pro Polymorphisms in Myelodysplastic Syndrome.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1745-1745
Author(s):  
João Agostinho Machado Neto ◽  
Fabiola Traina ◽  
Paula Melo Campos ◽  
Marilisia Andreoli ◽  
Irene Lorand Metze ◽  
...  

Abstract Abstract 1745 Poster Board I-771 Introduction Myelodysplastic syndrome (MDS) encompasses a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis, refractory cytopenia and a tendency to progress towards acute myeloid leukemia (AML). The progression of the disease may be associated with genetic or epigenetic alterations and a possible change in protein function. MDM2/P53 pathway plays an important role in the control of apoptotic and proliferation mechanisms. Single nucleotide polymorphisms (SNPs) were identified in the TP53 and MDM2 genes. MDM2 SNP309 results in higher levels of MDM2 and attenuates p53 pathway. The SNP in codon 72 of the TP53 gene results in either a C or G nucleotide and leads to either Proline (Pro) or Arginine (Arg), respectively. The Arg variant has been shown to be more potent in apoptosis induction and the Pro variant has been shown to be better in inducing cell-cycle arrest and to have a greater ability to repair damaged-DNA. The aim of the present study was to investigate the incidence of MDM2 and TP53 polymorphisms in MDS patients and to correlate the frequency of these SNPs with age, neutrophis and platelets at diagnosis, low risk versus high risk disease according to FAB (RA and RARS versus AREB and AREBt) and IPSS (Low and Int-1 versus Int-2 and high), cytogenetic risk (low versus intermediate and high risk), disease progression and overall survival. Patients and Methods We studied 103 healthy controls and 63 patients with MDS. According to FAB, patients were distributed as follows: 43 RA, 10 RARS, 7 RAEB, 1 RAEBt and 2 CMML. The median follow-up time was 40 months (range 2 – 159 months). Samples were obtained from peripheral blood or bone marrow and were screened for the presence of polymorphisms MDM2 SNP309 and TP53 SNPArg72Pro, by PCR analysis with specific primers and appropriate restriction enzyme. Appropriate statistical analyses were used for each test. Results The frequencies of genotypes for MDM2 SNP309 and TP53Pro7Arg were similar between MDS and healthy controls; MDM2 SNP309: 51% vs 53%, for TT, 38% vs 32% for TG, and 11% vs 15% for GG, TP53Pro7Arg: 47.5% vs 44%, for Arg/Arg, 47,5% vs 42% for Pro/Arg, and 5% vs 14% for Pro/Pro. No differences were observed between MDS patients with presence or absence of the polymorphisms in relation to age, neutrophis and platelets at diagnosis, low risk versus high risk disease according to FAB, IPSS and cytogenetic risk, disease progression and overall survival. Conclusions MDM2 and TP53 polymorphisms have been described to affect the risk for cancer, onset age and overall survival in solid tumors and leukemias. This was the first study to report these SNPs in MDS and leads to believe that these polymorphisms are not associated with risk for the disease and with clinical data. Keywords: MDM2, p53, myelodysplasia, polymorphisms Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4300-4300
Author(s):  
Hiroshi Handa ◽  
Takafumi Matsushima ◽  
Norifumi Tsukamoto ◽  
Masamitsu Karasawa ◽  
Hiroyuki Irisawa ◽  
...  

Abstract Telomerase activity has been found in most common cancers indicating that telomerase detection may be a useful marker in cancer diagnosis. For detection of telomerase activity and the expression of associated genes in cells, TRAP assay and RT-PCR are customarily used. Immunohistochemical detection of hTERT is useful to detect telomerase-positive cells in a background of non- cancerous cells. We developed a method for the detection of intra-nuclear hTERT protein, in a sub-population of hematopoietic cells, using concurrent staining cell surface antigen and multi color flow cytometry. Human leukemia and myeloma cell lines showed 100% positivity, whereas neutrophils of normal subjects showed 0% positivity, it is consistent with telomerase activity assessed by TRAP assay (r=0.71, p<0.0001) and previous observations. Then we applied this method to analyze hTERT expression in myelodysplastic syndrome (MDS). Forty MDS patients samples were obtained, 36 patients were diagnosed as low risk MDS (RA), 14 patients were diagnosed as high risk MDS (RAEB or RAEB-t) according to FAB classification. All samples were acquired after informed consent was obtained from the patients. Expression of hTERT protein was higher in CD34-positive blast-gated cells than CD34-negative blast-gated cells. The percentage of the CD34+ cells expressing hTERT ranged from 9.66% to 90.91% in low risk MDS patients, whereas from 50.46% to 97.68% in high risk MDS. The expression level was higher in the high risk group compared to that in the low risk group in MDS (p=0.0054, p=0.0084). This observation implied that telomerase up-regulation and hTERT expression were important for disease progression and could be a marker of more advanced disease. In subsets of MDS and AML bone marrow specimens obtained from these patients, we examined the hTERT expression in CD34+/CD38 high cells and CD34+/CD38 low cells containing stem cell fraction. Of interest, some of the patients showed higher expression of hTERT in CD34+/CD38 low cells than in CD34+/CD38 high cells. This observation is inconsistent with previous reports describing normal bone marrow hematopoietic cell findings. We speculated that this phenomenon could be a marker of MDS abnormality and that telomerase up-regulation may be initiated in the more primitive precursor fraction containing hematopoietic stem cells during the disease progression. Telomerase studies may be useful for definition of the risks associated with disease severity. Multi-parameter nature of flow cytometry and its ability to identify cellular sub-populations will facilitate a fuller understanding of the mechanisms of activation of telomerase.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3819-3819 ◽  
Author(s):  
Wei-gang Tong ◽  
Tapan Kadia ◽  
Gautam Borthakur ◽  
Elias Jabbour ◽  
Sherry Pierce ◽  
...  

Abstract Abstract 3819 Poster Board III-755 Background Myelodysplastic syndrome (MDS) is a heterogeneous group of bone marrow disorders characterized by dysplastic changes in the myeloid lineages, ineffective hematopoiesis, and an increased risk of transformation to acute myeloid leukemia (AML). In most cases, bone marrow is hyperceulluar but in 10 to 20% of cases, bone marrow can be hypocellular (defined as < 30% cellularity in patients < 70 years, or < 20% cellularity in patients 70 years or older), a condition that overlaps and is difficult to differentiate from aplastic anemia (AA). Currently, there are no good prognostic model for patients with hypocellular MDS. Methods In order to improve the prognostic assessment and to better understand the natural history of hypoplastic MDS, we analyzed the associations between disease characteristics and survival in 253 cases of hypocellular MDS presented to MDACC between 1993 and 2007. This is the largest study so far on patients with hypocellular MDS. We also compared the presenting characteristic and survival between these patients and a group of patients with hyper/normocelluar MDS (n=1725) during the same time period. Results Patients with hypocellular MDS usually presented with more significant thrombocytopenia (p< 0.019), neutropenia (p< 0.001), low β-2 microglobulin (p< 0.001), more transfusion dependency (p< 0.001), and more intermediate-2/high risk disease (57% vs. 42%, p= 0.02) compared to their hyper/normocelluar counterparts. There was no difference in overall survival between the hypocellular and the hyper/normocellular groups (p= 0.312). We divided the patients randomly into study and test group, and a multivariate analysis of prognostic factor identified the following adverse, independent factors (p < 0.001): poor performance status (ECOG 2-4), poor bone marrow cytogenetics (chromosome 7 or complex), anemia (< 10 g/dl), increased bone marrow blasts (≥ 5%) and high serum LDH (> 600 IU/l). In this model, each characteristic has a score of 1. A new prognostic model based on these factors could classify this group of patients into three risk categories independent of IPSS score. Patients with low risk (n= 66; scores 0-1) had a median survival of 30 months, and 2-year/3-year survival of 62%/44%. Patients with intermediate risk (n=44; score 2) had a median survival of 19.4 months, and 2-year/3-year survival of 43%/20%. Patients with high risk disease (n= 59; scores 3-5) had a median survival of 7.3 months, and 2-year/3-year survival of 12%/6%. When this new prognostic model was applied to test group (n=84), the median survival was 55.7, 13.5 and 8.6 months (p< 0.0001) for patients in low, intermediate and high risk groups, respectively. Patients that received immunotherapy (ATG/cyclosporine) had a better median survival and overall survival than patients treated with supportive care, hypomethylating agents, or induction chemotherapy (p< 0.0001). Conclusions Here, we proposed a new simple prognostic model that allows to predict prognosis in patients with hypocellular MDS. Analysis of prognostic factors in patients with hypocellular MDS may help us understand the biology of the disease, and develop risk-adapted therapies for this group of patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2822-2822
Author(s):  
Renata Scopim-Ribeiro ◽  
Joao Machado-Neto ◽  
Paula de Melo Campos ◽  
Patricia Favaro ◽  
Adriana S. S. Duarte ◽  
...  

Abstract Abstract 2822 Introduction: Acquired mutations in TET2 and DNMT3A have been found in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML), and may predict a worse survival in these diseases. TET2 mutations are considered to be a loss-of-function mutation and results in decreased 5-hydroxymethylcitosine (5-hmc) levels. In normal CD34+ cells, TET2 silencing skews progenitor differentiation towards the granulomonocytic lineage at the expense of lymphoid and erythroid lineages. Dnmt3a participates in the epigenetic silencing of hematopoietic stem cell regulatory genes, enabling efficient differentiation. Here, we attempted to evaluate the expression of TET2 and DNMT3A in total bone marrow cells from normal donors, patients with MDS and AML, and in CD34+ cells from MDS and normal controls during erythroid differentiation. Materials and Methods: The study included normal donors (n = 21), patients with MDS (n = 43) and AML (n = 42) at diagnosis. All normal donors and patients provided informed written consent and the study was approved by the ethics committee of the Institution. MDS patients were stratified into low and high-risk according to WHO classification (RCUD/RCMD/RARS=31 and RAEB1/RAEB2=12). TET2 and DNMT3A mRNA expression was assessed by quantitative PCR. CD34+ cells from normal donors (n = 9) and low-risk MDS patients (n = 7) were submitted to erythroid differentiation. Cells were collected and submitted to immunophenotyping for GPA and CD71 (days 6 and 12) and q-PCR for TET2 and DNMT3A expression (days 6, 8 and 12). Results of gene expression in normal donors and patients are presented as median, minimum-maximum, and were compared using Mann-Whitney test. Student t test was used for comparison of gene expression during CD34+ erythroid diferentiation. Overall survival was defined from the time of sampling to the date of death or last seen. Univariate analysis for overall survival was conducted with the Cox proportional hazards model. Results: TET2 expression was significantly reduced in both AML (0.62; 0.01–32.69) and MDS (1.46; 0.17–21.30) compared to normal donors (2.72; 0.43–31.49); P<0.0001 and P=0.01, respectively. TET2 expression was also significantly reduced in AML compared to MDS (P=0.0007). MDS patients were stratified into low and high-risk disease, and we still observed a significant reduction in TET2 expression in high-risk (0.73, 0.17–7.25) when compared to low-risk (1.58; 0.48–21.30; P=0.02) patients, but no difference was noted between normal donors vs. low-risk MDS, and high-risk MDS vs. AML. In MDS cohort, the median overall survival was 14 months (range 1–83), increased TET2 expression was associated with a longer survival (HR, 0.44; 95% CI, 0.21–0.91, P=0.03), and, as expected, WHO high-risk disease was associated with a shorter survival (HR, 10.16; 95% CI, 3.06–33.72, P<0.001), even though the confidence interval (CI) was large. TET2 expression did not impact survival in our cohort of AML patients. The erythroid differentiation was effective in cells from normal donors and MDS patients, as demonstrated by the flow cytometry analyses of GPA and CD71. TET2 expression was significantly increased on day 12 of erythroid differentiation, P<0.05. On the other hand, DNMT3A expression was similar between normal donors (0.74; 0.22–1.53), MDS (0.78; 0.26–3.46) and AML (0.95, 0.15–6.46), and during erythroid differentiation, with no impact on survival. Conclusion: These data suggest that decreased TET2 expression may participate in leukemogenesis, and supports the participation of TET2 in the erythroid differentiation of MDS. DNMT3A was not differentially expressed in AML and MDS, indicating that the presence of mutations in this gene may be the predominant mechanism of changes in protein function. We thus suggest that decreased TET2 expression may explain the reduced levels of 5-hmc found in TET2 wild type patients, and may become a predictive marker for outcomes in MDS and other myeloid diseases. Further studies would be necessary to better elucidate the clinical relevance and biologic significance of our findings, and whether the decreased TET2 expression results in hypermethylation in these diseases. Disclosures: Maciejewski: NIH: Research Funding; Aplastic Anemia&MDS International Foundation: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2824-2824
Author(s):  
Elias J. Jabbour ◽  
Naval G. Daver ◽  
Tina (Xiao Qin) Dong ◽  
Tapan M. Kadia ◽  
Susan O'Brien ◽  
...  

Abstract Abstract 2824 Background: HMA are standard of care in pts with high-risk MDS and commonly used in pts with lower risk. Pts with high-risk disease post HMA failure have a poor prognosis with a median survival of 4 months (Jabbour et al. Cancer. 2010;116:3830–4). The prognosis of patients with low and intermediate-1 risk MDS after HMA failure is not known. Aims: To assess survival in patients with low and intermediate-1 risk disease at the time of HMA failure that might benefit from specific strategies or investigational agents. Methods: Data from 699 patients with MDS (n=397) and chronic myelomonocytic leukemia (n=302) treated with HMA at one institution between 2000 and 2011 were reviewed. 126 (18%) of them were of low and intermediate-1 risk disease by IPSS score. Results: A total of 30 (24%) patients with MDS (n=26) and CMML (n=4) who had failed HMA therapy and remained of low and intermediate-1 risk disease were analyzed. These included 6 patients with low- and 24 intermediate-1 risk disease. Eleven patients had secondary disease. At the start of HMA, 22 patients were low-risk and 8 were intermediate-1 risk disease. Seventeen patients had a diploid cytogenetic analysis. Seven and 23 patients received azacitidine and decitabine, respectively. These patients had discontinued HMA because of primary resistance in all patients. The characteristics of the study group are shown in Table 1. Upon HMA failure, 24 (80%) were low-risk disease and 6 (20%) intermediate-1-risk disease. A total of five (17%) patients transformed subsequently into acute myeloid leukemia. After a median of 18 months from HMA failure, 12 (40%) patients remained alive. The median overall survival was 22 months with estimated 1- and 2-year overall survival rates of 65% and 45%, respectively. Considering overall survival from the start of HMA therapy, the median survival was 29.5 months with estimated 1- and 2-year overall survival rates of 80% and 60%, respectively. The 30 patients with HMA failure were subsequently assessed by the lower-risk MDACC risk model (Garcia-Manero et al. Leukemia. 2008;22:538–43): 8 (27%) had low-risk, 8 (27%) intermediate-risk, and 14 (46%) high-risk disease. Their 1-year survival rates were 66%, 73%, and 86%, respectively. Considering survival from the time of the initiation of HMA therapy, the estimated 1-year survival rates were 60%, 70%, and 100%, respectively, for patients with high-risk, intermediate-risk, and low-risk disease according to the MDACC risk model. After HMA failure, 11 (37%) patients received salvage investigational therapy, of whom 3 responded with 2 achieving hematologic improvement for a median of 12 months (range, 5–19) and one patient achieving a complete remission for 14 months that was lost thereafter, 1 (3%) received immunotherapy, 1 (3%) received growth factors only, and 17 (57%) elected not to receive any further treatment. No response was observed in the 2 patients who received subsequent immunotherapy or growth factors. Conclusion: The outcome of patients with low and intermediate-1 risk MDS after HMA failure is poor and appears to be predictable. Disclosures: Ravandi: Genzyme/Sanofi: Honoraria. Faderl:Genzyme: Advisory Board Other, Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2815-2815
Author(s):  
Asmita Mishra ◽  
Jeffrey E Lancet ◽  
Najla H Al Ali ◽  
Eric Padron ◽  
Viet Q. Ho ◽  
...  

Abstract Abstract 2815 Background: Azacitidine has emerged as the standard of care for treatment of higher risk MDS based upon results of the AZA-001 study. Several groups reported poor outcomes after AZA failure in patients with int-2 or high risk International Prognostic Scoring System (IPSS) risk groups with a median overall survival (OS) ranging from 4–8 months (mo). In the USA, AZA is approved for all FAB types and risk groups and is as first or second line therapy for anemia after erythroid stimulating agents in low /int-1 risk non-deletion 5q MDS and is the treatment of choice for thrombocytopenia. The outcome of patients with lower risk myelodysplastic syndrome (MDS) after AZA failure has not been characterized. We report our experience in a large cohort of low/int-1 (lower risk) MDS patients after AZA failure. Methods: Patients were identified through the Moffitt Cancer Center (MCC) MDS database. Individual charts were reviewed and relevant clinical data was extracted. Patients with low or intermediate-1 (int-1) risk disease as defined by IPSS who had received AZA treatment were identified. These patients were also risk stratified based on Global MD Anderson Score (MDAS). The primary objective was to estimate OS in these patients after AZA failure. AZA failure was defined as failure to respond after 4 or more treatment courses, loss of response, or disease progression while on therapy. All responses were defined according to the International Working Group (IWG) 2006 criteria. The Kaplan–Meier method was used to estimate median overall survival. Results: Two hundred eighty MDS patients with low/int-1 IPSS risk who had received AZA treatment were identified. Most patients (81%) were greater than 60 years of age (median, 69 years), and 90% of AZA treated patients were RBC transfusion dependent. Refractory cytopenia with multilineage dysplasia (RCMD) was the most common WHO subtype (44%), and 81% of patients had good risk cytogenetics (Table-1). The median time from MDS diagnosis to AZA treatment was 12.3 months; median number of AZA cycles received was six. At the time of AZA treatment, 241 patients (86 %) were risk stratified as int-1 versus 39 patients (14 %) who were stratified as low risk IPSS. The IWG 2006 responses to AZA treatment included 4% CR (n=10 ), 1% marrow CR (n=2), 4% PR (n=10), 27% Hematological improvement (HI) (n=75), whereas 52% (n=146) had stable disease with no HI (n=146) and 10% had progressive disease (n=10); 6 patients (2%) died on therapy, and responses were missing in 2 patients (<1%). The overall best response (HI or better) was 36%. The median OS for the entire cohort after AZA failure was 18.5 months (95% CI [13.5–23.5 mo], Figure 1A). The median OS for patients with low risk IPSS disease from time of AZA failure was 46 months versus 15 mo for int-1 patients (p<0.005, Figure 1B). When utilizing MDAS, median OS was 33.3 months for low risk patients, 21 months for int-1, 11 months for int-2, and 7.5 months for poor risk patients (p=0.005). Conclusions: To our knowledge this is the first report describing the outcome of lower risk MDS patients after AZA treatment failure. Outcome is particularly poor for those patients with int-1 risk MDS, with a median OS of 15 mo. Global MDAS identified patients upstaged to int-2 or high risk with less than one year OS. There is unmet need for effective novel therapies for lower risk MDS patients after AZA failure. Disclosures: List: Celgene: Consultancy. Komrokji:Celgene: Speakers Bureau.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2847-2847
Author(s):  
Fernanda Marconi Roversi ◽  
Nathalia Moreno Cury ◽  
Matheus Rodrigues Lopes ◽  
Fernando Vieira Pericole ◽  
Marisa Claudia Alvarez Prax ◽  
...  

Abstract In recent years, the role of tumor microenvironment in neoplasm initiation and malignant evolution has been increasingly recognized. However, the bone marrow mesenchymal stem/stromal cell (BMMSC) contribution to disease progression remains poorly explored. We had previously performed a microarray analysis of myelodysplastic syndrome (MDS) patient-derived BMMSC (MDS-BMMSC) and found an underexpression of HAI-2/SPINT2, an endogenous inhibitor of the hepatocyte growth factor (HGF) activator. This gene has been described as methylated in various cancer types and has been associated with disease progression. Despite of being related to the pathogenesis of several neoplasms, the role of HAI-2/SPINT2 has not yet been fully elucidated in hematological diseases, such as MDS and acute myeloid leukemia (AML). Thus, the aim of this study was to evaluate HAI-2/SPINT2 expression in derived BMMSC and total bone marrow (BM) of healthy donors (HD), MDS and AML patients as well as in BMMSC treated with 5-Azacitidine (Aza), a DNA methyltransferase (DNMT) inhibitor. To achieve this, we collected BM hematopoietic cells and plastic-adherent BMMSC from aspirates of HD, MDS and AML patients. BMMSC were expanded to passage 4 and defined as CD73+/CD90+/CD105+/CD45-/CD34-/CD31-/HLA-DR-. A total of 29 HD and 121 patients at diagnosis (MDS=72 [low-risk=46, high-risk=26], AML with myelodysplastic related changes (AML-MRC)=17 and de novo AML=32) were included. HAI-2/SPINT2 mRNA was significantly decreased in MDS- (0.34[0.01-2.06];P <.01) compared to HD-BMMSC (0.89[0.46-1.59]). When patients were stratified according to WHO classification, HAI-2/SPINT2 expression was lower in both low-risk (0.31[0.01-1.33]) and high-risk (0.43[0.01-2.06]) MDS-BMMSC. Similar results were found in total BM: HAI-2/SPINT2 transcripts were significantly decreased in MDS (0.41[0.01-2.53];P <.01), AML-MRC (0.38[0.014-0.84];P <.01) and AML patients (0.33[0.01-2.07];P <.001) compared to HD (0.91[0.19-4.79]). To investigate whether this loss of expression was due to HAI-2/SPINT2 methylation, BMMSC were treated with Aza (1µM or IC50 value) for 48h. In MDS- and AML-BMMSC, Aza treatment resulted in a pronounced upregulation of HAI-2/SPINT2 mRNA and protein levels. Moreover, Aza treatment of HD-BMMSC did not improve the HAI-2/SPINT2 mRNA and protein levels as much as the observed in MDS- and AML-BMMSC. To better understand the role of HAI-2/SPINT2 downregulation in BMMSC physiology, its expression was inhibited in a BM stromal cell line (HS5). As previously reported, HAI-2/SPINT2 silencing resulted in an increased secretion of HGF, known to be overexpressed in plasma of MDS patients and considered a prognostic factor in MDS and AML patients (Matsuda et al., Leukemia, 2004). Moreover, after co-culture, HAI-2/SPINT2 knockdown improved survival of blasts isolated from AML-MRC and AML patients. We also observed an increased adhesion of CD34+ hematopoietic stem cells (HSC) to HAI-2/SPINT2 silenced HS5 cells. This prompted us to analyze the expression of cell adhesion molecules in MDS- and AML-BMMSC. We observed a significant augment in the expression of CD49b and CD49d integrins in MDS- and AML-compared to HD-BMMSC. Taken together, SPINT2 inhibition improves HGF secretion, consequently with alteration in molecule receptor adhesion, resulting in an increased expression of integrins (CD49b and CD49d) responsible for cell-to-cell adhesion. Thus, reactivation of HAI-2/SPINT2 levels after Aza treatment indicates that this gene is probably epigenetically silenced by methylation in MDS and AML, and is possibly a tumor suppressor gene. Interestingly, nowadays, epigenetic therapy by Aza is the first-line treatment for MDS patients, and induces prolonged survival and delayed AML evolution. Likewise, our results suggest that HAI-2/SPINT2 may play a role in deregulation of HGF cytokine secretion with consequently alteration in HSC adhesion and growth/survival. Tumor microenvironment niche is currently known to play a critical role in cancer initiation and progression, thus HAI-2/SPINT2 may contributes to functional and morphological abnormalities of microenvironment niche and with the stem/progenitor cancer cell progression. Hence, downregulation in HAI-2/SPINT2 gene expression, due to methylation in MDS- and AML-BMMSC, provides novel insights into the pathogenic role of the leukemic bone marrow microenvironment. Disclosures No relevant conflicts of interest to declare.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 5033-5033 ◽  
Author(s):  
T. Powles ◽  
A. Young ◽  
D. Short ◽  
P. Savage ◽  
C. Pappin ◽  
...  

5033 Background: Women requiring chemotherapy for GTN are stratified into low risk and high risk disease and treated accordingly. The vast majority of patients are cured with chemotherapy. Despite this a small number relapse after completing treatment. The prognostic factors predictive of outcome for these relapsed patients are unclear and are investigated here. Methods: Clinical data at presentation and at the time relapse on patients with relapsed GTN was collected from a prospective data base at the Charing Cross Hospital. This included stage of disease, time to relapse, treatment details and rate of HCG rise. Statistical analysis was performed using the Kaplan Meier method. Univariant and multivariant analysis was performed on the data. Results: Between 1980 and 2004 1708 patients were treated with chemotherapy for GTN. Sixty of these patents have relapsed. The median age of these 60 patients was 29 years (range 30–51) and the median follow up was 10 years. The overall 5 year survival for patients with relapsed GTN was 91% (84–99%). All deaths were tumour related. Patients who initially presented with low risk disease (n = 27) had an overall survival of 100% while those who presented with high risk disease had an overall survival of 91% (66–96%) (p < 0.05). Five patients progressed during treatment for relapsed disease, all of these patients died. Ten patients relapsed for a second time. All of these patients are currently alive and disease free with a median of 11 years of follow up. Significant prognostic factors included stage of disease at presentation, the presence of metastatic disease at relapse and the rate of rise of HCG at relapse. Conclusions: The outcome for patients with relapsed GTN is good especially those who relapse after initially having low risk disease. Failure to obtain a complete remission on 2nd line chemotherapy is a poor prognostic indicator. No significant financial relationships to disclose.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2629-2629
Author(s):  
Shang-Ju Wu ◽  
Wen-Chien Chou ◽  
Ming Yao ◽  
Yo-Chia Yeh ◽  
Hwei-Fang Tien

Abstract The suppressor of cytokine signaling-1 (SOCS1) protein is a tumor suppressor. Hypermethylation of SOCS1, resulting in transcriptional silencing, is suggested to play an important role in the development of cancers. We sought to characterize SOCS1 methylation in primary myelodysplastic syndrome (MDS) and to clarify its clinical implications. We analyzed the methylation status of SOCS1 by methylation specific polymerase chain reaction in 114 patients with primary MDS and performed serial studies in 29 of them. SOCS1 methylation occurred in 54 patients (47.4%), more frequently in patients with high-risk subtypes of MDS than in those with low-risk ones (52.6% vs. 25.8%, p = 0.011). SOCS1 methylation was closely associated with N-RAS gene mutation (p = 0.010) and inversely associated with good-risk karyotype (p = 0.021). With a median follow-up of 17 months (range, 1 to 231 months), two patients, who did not have SOCS1 methylation at diagnosis, acquired it during disease progression. SOCS1 methylation disappeared after hematopoietic stem cell transplantation in two patients who had it initially. The patients with SOCS1 methylation had a higher cumulative risk of leukemic transformation than the others (55.8% vs. 27.7% at 3 years, p = 0.004). This difference remained significant within the subgroup of patients with high-risk subtypes of MDS (67.3% vs 45.1% at 3 years, p = 0.045). This is the first report to demonstrate the clinical relevance of SOCS1 methylation in MDS. It may play an important role in the pathogenesis of MDS, especially among patients with high-risk subtypes. Correlation of SOCS1 methylation with clinical characteristics, cytogenetics and RAS mutations Total(n=114) SOCS1 methylated(n=54) SOCS1 unmethylated(n=60) p value Age 64(7~68) 64(7~84) 65(19~86) 0.231 Sex(M/F) 81/33 36/18 45/15 0.327 WBC(1000/ml) 4675(220~227200) 4710(440~227200) 4640(1570~87420) 0.913 Hemoglobin(g/dl) 8.5(3.9~14.4) 8.9(4.4~12.9) 7.8(3.9~14.4) 0.348 Platelet(1000/ml) 88.5(2~607) 102(2~400) 74(3~607) 0.453 BM blast(%) 5.8(0~54.8) 7.2(0~54.8) 4.4(0~25.4) 0.033 Cytogenetics 0.021 Poor 19 13(68.4%) 6(31.6%)     -7/7q- 11 8(72.7%) 3(27.3%)     complex 10 6(60%) 4(40%) Intermediate 15 10(66.7%) 5(33.3%) Good 70 27(38.6%) 43(61.4%) FAB subtypes 0.011 Low-risk 31 8(25.2%) 23(74.2%)     RA 21 4(19.0%) 17(81.0)     RARS 10 4(40%) 6(60%) High-Risk 78 41(52.6%) 37(47.4%)     RAEB 33 20(60.6%) 13(39.4%)     RAEB-T 20 11(55.0%) 9(45.0%)     CMML 25 10(40.0%) 15(60.0%) AML 5 5(100%) 0 IPSS 0.008 Low-risk 57 21(36.8%) 36(63.2%)     Low 21 4(19.0%) 17(81.0%)     Int-1 10 4(40.0%) 6(60.0%) High-risk 46 29(63.0%) 17(37.0%)     Int-2 33 29(63.0%) 17(37.0%)     High 20 11(55.0%) 9(45.0%) N-RAS 0.010     Mutated 11 9(81.8%) 2(18.2%)     Wild 96 38(39.6%) 58(60.4%) K-RAS 1.000     Mutated 4 2(50%) 2(50%)     Wild 101 44(43.6%) 57(56.4%) Cumulative risk of leukemic transformation in MDS patients. A. Comparison for all patients with and without SOCS1 methylation. B. Comparison for patients of high-risk subtypes of MDS (RAEB, RAEB-T, CMML) with and without SOCS1 methylation Cumulative risk of leukemic transformation in MDS patients. A. Comparison for all patients with and without SOCS1 methylation. B. Comparison for patients of high-risk subtypes of MDS (RAEB, RAEB-T, CMML) with and without SOCS1 methylation


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 5049-5049
Author(s):  
EiriniIOANNIS Konstantinidou ◽  
Nicholas Zoumbos

Abstract Abstract 5049 Expression of Proteins P16INK 4a, P53 and Bmi-1 in Hematopoietic Stem Cells of Patients with MDS. The Role of Cellular Senescence. Irene Constandinidou1, Eleni Kalivioti1,Vasilios Fertakis1, Costas Dallas1, Polyxeni Lampropoulou1, Evangelia Tzouvara1, Panagiotis Zoumboulis2, Nicholas Zoumbos.1 1Hematology Division, University of Patras, Medical School, Patras, Greece 2Orthopaedic Division, University Hospital of Patras, Patras, Greece Introduction The myelodysplastic syndromes (MDS) comprise a spectrum of heterogeneous clonal stem/progenitor cell disorders characterized by marrow failure. One potential reason, apart from apoptosis, for hematopoietic stem cells (HSCs) functional failure is cellular senescence, which is believed to have evolved as a tumor suppressor mechanism capable of arresting growth to reduce risk of malignancy. It can be activated by both telomere-dependent (telomere shortening) and telomere-independent pathways (DNA damage, oncogenic or oxidative stress). There are two widely recognized major tumor suppressor pathways p16/pRb and p53-p14/ARF that regulate cellular senescence. Activation of either pathway is profoundly associated with cellular senescence. Bmi-1, on the other hand, is a transcriptional repressor that plays an essential role in the self-renewal of HSCs and leukemic stem cells as well. One of its major targets is the INK/ARF locus, which encodes p16 and p53 independently. Aim To study the role of cellular senescence in the pathogenesis of MDS through the expression of p16, p53, phospho p53 (activated form of p53, phosphorylated at Ser-15) and bmi-1. Patients and methods We examined the expression level of p16INK4a, Bmi-1, p53 and phospho p53 by flow cytometry (direct and indirect staining) in CD34+ bone marrow mononuclear cells (BMMNCs). Furthermore, we analyzed the relative telomere length (RTL) in BMMNCs by quantitative fluorescence in situ hybridization assay using flow cytometry (flow-FISH).We verified the presence of p16 by SDS PAGE western blotting. We studied 36 samples of MDS patients (11 RA, 6 RARS, 3 RCMD, 4 5q- and 12 RAEB I-II), 17 samples of age-matched healthy controls after informed consent and 8 cord blood samples. All MDS diagnoses were histologically confirmed by bone marrow examination and categorized into low and high risk according to the international prognostic scoring system (IPSS). Results Expression of p16 is significantly increased in low risk MDS (mean± SD=8,7±12) when compared to healthy controls (p=0,02) and high risk patients(p=0,004). Patients with 5q- syndrome express lower level of p16 in comparison with the other low risk group patients. Increased level of bmi-1 expression is noticed in high risk group (mean± SD=30,3 ±33,9) when compared to low risk group (mean± SD=25±24,6) and healthy controls (mean± SD=18 ±16,4), while 5q- syndrome patients appear to express higher level of bmi-1 than any other risk group. Significantly increased expression of p53 (p=0,03) is noticed in high risk group (mean ±SD=64,3±27) when compared to low risk (mean±SD=34,2±28,8). Patients with 5q- syndrome express increased level of p53 (mean± SD=57,4±26,3) similar to that of high risk group. Expression of phospho p53 seams increased (p=0,055) in low risk group(mean± SD=7,1± 13,9) in comparison to high risk group (mean± SD=0,09± 0,18) and healthy controls (mean± SD=0,19± 0,63). Phospho p53 is expressed in lower level in 5q- syndrome patients. Regarding the RTL there is a significant difference (p=0,0018) between MDS (mean± SD=9,7± 2,7) and cord blood samples (mean± SD=13,8± 1,9). Conclusions Increased levels of p16 and phospho p53 in low risk MDS suggest that cellular senescence may contribute to the ineffective hematopoiesis of MDS and probably in a telomere independent way. Low expression level of of phospho p53 in high risk MDS raises the question of genetic integrity of p53 in this group of patients and when combined with high levels of bmi-1 and low expression of p16 may play a role in the disease progression to AML. Further investigation of bmi-1, especially in the 5q- group - and other markers of cellular senescence (SA- b galactosidase, SAHF) is needed in order to clarify the impact of cellular senescence in different risk groups of MDS. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5026-5026 ◽  
Author(s):  
Julia Montoro ◽  
Teresa Vallespi ◽  
Esther Sancho ◽  
Olga Salamero ◽  
Laura Lopez-Andreoni ◽  
...  

Abstract Abstract 5026 Myelodysplastic syndromes (MDS) are clonal disorders of hematopoietic stem cell with low life expectancy due to several blood cytopenias and high risk of acute myeloid leukemia transformation (AML). Classically, evolution to AML, infection, and hemorrhage are reported as the main causes of death. However, there are few reports analyzing other causes of death in MDS patients, particularly the prevalence of non-MDS-related causes in large series from single-centers. We present here the analysis of causes of death of 200 patients (median age 75yr, range 16–96, 59% male) diagnosed of MDS in our institution between 2000 and 2010. Patients were diagnosed and classified according to the FAB criteria, WHO 2008 classification, IPSS (International Prognostic Scoring System) and SPI (Spanish Prognostic Index). Overall survival (OS) and survival of different MDS subtypes were analyzed. Two prognostic subgroups were defined: low-risk subgroup, composed by patients with low or intermediate-1 IPSS and low SPI; and high-risk subgroup, that included patients with intermediate-2 and high IPSS and intermediate and high SPI. Infection, hemorrhage, disease progression and transformation to AML were considered MDS-related deaths. All other causes of death were classified as non-MDS-related. Median follow-up of the series was 1.8 years (range: 0–11 years). MDS subtypes distribution was as follows: RA, RARS and 5q– 19%; RCMD and RCMD-RS 32%; RAEB-1 and RAEB-2 27%; hypoplastic and unclassified MDS 8%; and CMML 13%. One hundred twenty-nine patients (64.5%) belonged to the low-risk subgroup, whereas 65 patients (32.5%) to the high-risk subgroup. Only 6 patients (3%) could not be classified. Median OS of the whole series was 2 years, being of 3.7 years in the low-risk subgroup and of 0.9 years in the high-risk subgroup (P<0.001). At the time of the analysis, 141 patients (70.5%) had died: 78 (60.4%) of the low-risk subgroup and 59 (91%) of the high-risk subgroup. The cause of death was identified in 97 of 141 (68%) patients and it was related to MDS in 75 patients (77%) and non-MDS-related in 22 patients (23%). In the low-risk subgroup, causes of death were: evolution to AML 14, disease progression 5, infection 14 and bleeding 2. Non-MDS-related causes were: ischemic stroke 3, second malignancy 5, acute myocardial infarction 3, congestive heart failure 3 and others 3. In the high-risk subgroup the causes of death were: evolution to AML 18, disease progression 3, infection 13 and bleeding 5. Non-MDS-related causes were: second malignancy 1, congestive heart failure 1, and accidental 1 (Figure 1). The percentage of patients who died of unrelated-MDS causes was significantly higher in the low-risk subgroup than in the high-risk subgroup (32% vs. 7%, respectively; P=0.003). In conclusion, the most frequent cause of death in both low-risk and high-risk subgroups was related to the MDS (evolution to AML). Non-MDS-related deaths were more common in low-risk subgroup, being the most frequent causes heart failures and second malignancies. Causes of death in MDS should be taking into consideration when analyzing response and survival in clinical trials, particularly when applied to patients belonging to low-risk subgroups. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document