Ph-Negative Chronic Myeloproliferative Neoplasms – Population Analysis, a Single Center 10-years’ Experience

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5556-5556
Author(s):  
Vasily Shuvaev ◽  
Irina Martynkevich ◽  
Alla Abdulkadyrova ◽  
Vera Udaleva ◽  
Tatyana Zamotina ◽  
...  

Abstract Objectives and background. Nowadays chronic myeloproliferative neoplasms (MPN) other than chronic myelogenous leukemia undergo renaissance of interest. It results from advances in decryption of molecular mechanisms of pathogenesis and invention of target drugs. Epidemiological information is needed to assess potential effect and additional costs of new diagnostic and therapeutic techniques. The objective of our study was to review experience of MPN diagnostic and treatment in our center for past ten years. Methods. Our institution serves as primary hematological outpatient department for a half of Saint-Petersburg city with about 2 million inhabitants. We reviewed patients' charts to obtain information about incidence, symptoms, diagnostic test results, treatment options and relationship to prognostic factors. Statistical methods included descriptive statistics, nonparametric ANOVA for frequencies comparisons and Kaplan-Meyer method with log-rank test for survival comparisons in Statistica 7.0 package. Results. Since 2004 to 2013 there were 570 newly diagnosed MPN patients (pts) in our center. This group consisted of primary myelofibrosis (PMF) (203 pts; 126 female, 77 male; median age 63 years, range 16-83 years), essential thrombocythemia (ET) (201 pts; 146 female, 55 male; median age 58 years, range 23-78 years), polycythemia vera (PV) (166 pts; 96 female, 70 male; median age 57 years, range 20-85 years). The incidence rates were stable during study period: PMF incidence varied from 0.65 to 1.35 with mean of 1.01 new patient per 100 000 inhabitants per year; ET had incidence from 0.60 to 2.1 with mean of 1.00 and PV had incidence from 0.5 to 1.15 with mean of 0.83. The most prevalent symptoms of disease were: splenomegaly (65.5%), constitutional symptoms (fever, night sweats, weight loss) (31.0%), anemia (36.3%) thrombosis (24.1%) for PMF; fatigue (33.2%), headache and dizziness (25.6%), arthralgia (21.8%), erythromelalgia (15.8%) for ET; plethora (82.5%), headache and dizziness (52.4%), fatigue (31.3%) for PV. JAK2V617F was detected in 49.7% of PMF pts, 57.8% of ET pts and in 97.7% of PV pts. Thrombosis rates according WHO IPSET-thrombosis system risks` groups of ET and PV pts were: low-risk group 3.33% (3/90), intermediate-risk group 11.1% (13/117) and 39.4% (63/160) in high-risk group with highly significant (p<0.0001) differences between risks' groups. There were 169 lethal outcomes in the analysed group (102 PMF; 31 ET; 36 PV). Ten-years overall survival rates were 49.8% in PMF pts, 84.6% in ET pts and 78.3% in PV pts. (fig.1). Overall survival in PMF was significantly influenced by risk stratification as IPSS, DIPSS and DIPSS+. Survival curves according DIPSS+ groups are presented in fig.1. Conclusions. Patients with MPN are presented in substantial number; therefore need much finance for novel therapy introduction. Risk stratification systems has high predictive value. Innovative drugs treatment results should be evaluated in comparison with historical control. Figure1 Overall survival in PMF patients according to DIPPS+ stratification groups. Figure1. Overall survival in PMF patients according to DIPPS+ stratification groups. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 5054-5054
Author(s):  
Santiago Barrio ◽  
Miguel Gallardo ◽  
Alicia Arenas ◽  
Rosa M. Ayala ◽  
Inmaculada Rapado ◽  
...  

Abstract Abstract 5054 Objectives: In the present study we have determined the antitumor effects, molecular mechanisms of action and potential synergies between Ruxolitinib and the molecular targeted drugs Sorafenib, KNK437, Dasatinib and Perifosine, in Philadelphia negative chronic myeloproliferative neoplasms (MPN). Materials and methods: The cytotoxic and cytostatic effects of the different compounds were determined in the JAK2V617F positive cell lines HEL and Ba/F3 JAK2V617F EPOR, and in mononuclear and bone marrow CD34 positive cells from 19 MPN patients. The effects of different drugs at the molecular level were analyzed by flow cytometry and western blot. The IC50 and the synergy combination index (CI) were determined with GrafPath Prism software and Calcusyn software respectively. Results: Ruxolitinib (IC50PV=15nM), as well as Sorafenib (IC50PV=8uM), KNK437 (IC50PV=100uM) and Perifosine (IC50PV=15uM), was able to inhibit proliferation in cell line models and in cells from MPN patients. Dasatinib (IC50PV=12nM), however, was only effective in patient samples, with similar effects in both MPN patients and control donors. Moreover, Dasatinib, KNK437 and Sorafenib showed a strong synergistic effect in combination with Ruxolitinib (CIPV<0. 3, table 1). The Western blot analysis confirmed that Sorafenib inhibited the activation of ERK and P38, and, as a consequence, of STAT5. Ruxolitinib blocked the ERK and STAT5 activation, Dasatinib blocked SRC and STAT5, and KNK437 decreased the stability of the protein JAK2, reducing its expression. Conclusions: The blockage of JAK2 related proliferative pathways has the potential to inhibit cell proliferation in MPNs. Furthermore, the combination of Ruxolitinib with inhibitors targeted against these pathways has a strong synergistic effect. This may be related to a decrease in the activation of the final common effector STAT5. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Zhu Shen ◽  
Wenfei Du ◽  
Cecelia Perkins ◽  
Lenn Fechter ◽  
Vanita Natu ◽  
...  

Predicting disease natural history remains a particularly challenging endeavor in chronic degenerative disorders and cancer, thus limiting early detection, risk stratification, and preventive interventions. Here, profiling the spectrum of chronic myeloproliferative neoplasms (MPNs), as a model, we identify the blood platelet transcriptome as a generalizable strategy for highly sensitive progression biomarkers that also enable prediction via machine learning algorithms. Using RNA sequencing (RNA seq), we derive disease relevant gene expression and alternative splicing in purified platelets from 120 peripheral blood samples constituting two independently collected and mutually validating patient cohorts of the three MPN subtypes: essential thrombocythemia, ET (n=24), polycythemia vera, PV (n=33), and primary or post ET/PV secondary myelofibrosis, MF (n=42), as well as healthy donors (n=21). The MPN platelet transcriptome discriminates each clinical phenotype and reveals an incremental molecular reprogramming that is independent of patient driver mutation status or therapy. Leveraging this dataset, in particular the progressive expression gradient noted across MPN, we develop a machine learning model (Lasso-penalized regression) predictive of the advanced subtype MF at high accuracy (AUC-ROC of 0.95-0.96) with validation under two conditions: i) temporal, with training on the first cohort (n=71) and independent testing on the second (n=49) and ii) 10 fold cross validation on the entire dataset. Lasso-derived signatures offer a robust core set of < 10 MPN progression markers. Mechanistic insights from our data highlight impaired protein homeostasis as a prominent driver of MPN evolution, with persistent integrated stress response. We also identify JAK inhibitor-specific signatures and other interferon, proliferation, and proteostasis associated markers as putative targets for MPN-directed therapy. Our platelet transcriptome snapshot of chronic MPNs establishes a methodological foundation for deciphering disease risk stratification and progression beyond genetic data alone, thus presenting a promising avenue toward potential utility in a wide range of age-related disorders.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1522-1522
Author(s):  
Martina Crysandt ◽  
Edgar Jost ◽  
Susanne Isfort ◽  
Tim H Brümmendorf ◽  
Stefan Wilop

Abstract Abstract 1522 Introduction Dose calculation of chemotherapeutic agents is mainly based on body surface area (BSA). Historically, in many institutions, doses are not adapted to a BSA above 2 sqm, although recent data suggested that this relative dose reduction in patients above 2 sqm is associated with a worse outcome. Obesity is a widespread and increasing phenomenon in the developed countries and is mainly defined by the body mass index (BMI). It is known, that the risk to develop haematological or solid malignancies is increased in obese patients – and, additionally, weight influences outcome in several tumours. Therefore, in our study, we analyzed the prognostic impact of obesity in newly diagnosed AML regarding the response to the first cycle of induction therapy and overall survival. Methods We identified 145 patients with newly diagnosed AML who were treated with induction therapy containing cytarabine and an anthracycline in our institution. Clinical data including several laboratory parameters associated with nutritional status (cholesterol, triglycerides, protein, C-reactive protein, albumin, lymphocyte count, transferrin, pseudo-cholinesterase, fT3, b-type natriuretic peptide), long-term medication with statins, chemotherapy dosing as well as response and overall survival have been assessed retrospectively from the institution's database. In our institution, all patients with a high BSA received a chemotherapy-dose calculated with a cut-off of not more than 2 sqm. Results In our cohort, median BMI was 25.2 kg/sqm (range 17.0 – 48.1). Seventeen patients had a BMI above 31 kg/sqm, 128 below. The median BSA of all patients was 1.83 sqm (range 1.49 – 2.40). In 41 patients, chemotherapy doses have been adjusted owing to a BSA of more than 2 sqm. We included cytogenetic risk group, BMI, BSA above 2.0 sqm, weight, long-term medication with statins and laboratory parameters in our univariate and multivariate analysis. Only cytogenetic risk group (p=0.001), triglycerides (p=0.008) and the BMI (p=0.032) were independent risk factors for overall survival. Univariate analysis showed similar results. Patients with a BMI >31 showed a significantly worse response (PR + CR) on first induction therapy (47.1% vs. 74.8%, p=0.018) and a shorter median survival (11.2 vs. 25.1 months, p=0.004). Both BMI-groups showed the same distribution of well-known risk factors including age, cytogenetic risk groups and secondary AML. Discussion/Conclusion In our cohort, a high BMI was associated with poorer response and impaired overall survival, whereas BSA was not. This leads to the conclusion, that the adverse effect may be mediated by obesity itself and not caused by underdosing of chemotherapy. Although an effect of BMI is known from several solid tumours, the reason remains unclear. Possible explanations include altered metabolisation and production of growth factors in adipose tissue (possibly indicated by elevated triglycerides), impaired or accelerated hepatic drug activation and/or metabolisation or impact on immune function. This study is limited by the retrospective single-center design and the relatively small patient number. Nevertheless, the data clearly confirmed other well established risk factors like the cytogenetic risk group supporting the validity of this approach. The study results suggest BMI as an independent risk factor for AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4095-4095
Author(s):  
Edwin Chen ◽  
Lawrence J Breyfogle ◽  
Rebekka K. Schneider ◽  
Luke Poveromo ◽  
Ross L. Levine ◽  
...  

Abstract TET2 mutations are early somatic events in the pathogenesis of acute myeloid leukemia (AML), myelodysplastic syndrome (MDS) and myeloproliferative neoplasms (MPN) and are one of the most common genetic lesions found in these diseases. In MPN, TET2 mutations are enriched within more advanced disease phenotypes such as myelofibrosis and leukemic transformation and often co-occur with the JAK2V617F mutation, which is present in the majority of MPN patients. We have developed and characterized a Jak2V617F conditional knockin mouse (Jak2VF/+), the phenotype of which closely recapitulates the features of human MPN. To determine the impact of Tet2 loss on Jak2V617F-mediated MPN, we crossed Tet2 conditional knockout mice with Jak2VF/+ knockin and Vav-Cre transgenic mice and backcrossed the compound mutant animals. We then characterized the effects of heterozygous and homozygous loss of Tet2 on the phenotype of Jak2VF/+ mice. We assessed peripheral blood counts, histopathology, hematopoietic differentiation using flow cytometry, colony formation and re-plating capacity. We also evaluated the effects of Tet2 loss on the transcriptome of the HSC compartment using gene expression microarrays and on HSC function using competitive bone marrow transplantation assays. Similar to Jak2VF/+/VavCre+ mice, Tet2+/-/Jak2VF/+/VavCre+ and Tet2-/-/Jak2VF/+/VavCre+ mice develop leukocytosis, elevated hematocrits (HCT) and thrombocytosis. Tet2-/-/Jak2VF/+/VavCre+ mice demonstrate enhanced leukocytosis and splenomegaly compared to the other groups. All groups demonstrate myeloid expansion, erythroid hyperplasia and megakaryocytic abnormalities consistent with MPN in the bone marrow and spleen, while more prominent myeloid expansion and megakaryocytic morphological abnormalities are observed in Tet2-/-/Jak2VF/+/VavCre+ mice as compared to the other groups. Notably, we do not see the development of acute myelogenous leukemia (AML) in Tet2-/-/Jak2VF/+/VavCre+ mice at 6 months. We see enhanced expansion of lineagelowSca1+cKithigh (LSK) cells (enriched for HSC) most prominently in the spleens of Tet2+/-/Jak2VF/+/VavCre+ and Tet2-/-/Jak2VF/+/VavCre+ mice as compared to Jak2VF/+/VavCre+ mice. In colony forming assays, we find that Tet2-/-/Jak2VF/+/VavCre+ LSK cells have enhanced re-plating activity compared to Jak2VF/+/VavCre+ LSK cells and that Tet2-/-/Jak2VF/+/VavCre+ LSK cells form more colonies that Tet2-/-/Jak2+/+/VavCre+ cells. Gene expression analysis demonstrates enrichment of a HSC self-renewal signature inTet2-/-/Jak2VF/+/VavCre+ LSK cells. Concordant with this, we find that Tet2-/-/Jak2VF/+/VavCre+ LSK cells have enhanced competitive repopulation at 16 weeks as compared to Jak2VF/+/VavCre+ and Tet2+/-/Jak2VF/+/VavCre+ LSK cells. In aggregate these findings demonstrate that Tet2 loss promotes disease progression in MPN but is insufficient to drive full leukemic transformation. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 32 (4_suppl) ◽  
pp. 373-373
Author(s):  
Andrea Necchi ◽  
Salvatore Lo Vullo ◽  
Patrizia Giannatempo ◽  
Elena Farè ◽  
Daniele Raggi ◽  
...  

373 Background: Mediastinal GCTs and PMNSGCTs poorly benefit from CT and half of patients (pts) still die for disease. Enhancing the risk stratification may result in tailoring a personalized treatment strategy since diagnosis. Methods: Between 1985 and 2012, 87 pts with PM-GCT were treated at our center. Of them, pure seminomatous histology was excluded. Cox proportional hazards regression analysis was conducted to examine the prognostic impact of these candidate factors on overall survival (OS): type of 1st-line CT (high [HDCT] vs conventional dose [CDCT]), post-CT surgery, type of baseline elevated serum tumor marker (STM), presence of lung or liver-bone-brain metastases (LBB), STM response (still elevated vs normal or normalized), and histology (viable cancer [VC] vs necrosis/teratoma [NT]). OS curves were compared by Kaplan Meier method with the log-rank test. Results: The study included 68 cases with PMNSGCT. Median age was 28.5 yrs (IQR: 23-35). 12 pts (17.7%) presented with mediastinal syndrome, 23 (33.8%) had lung and 7 (10.3%) LBB metastases. 12 pts received upfront HDCT and 45 pts (66.2%) underwent post-CT surgery. The final model of poor prognostic factors included no surgery (HR: 8.74, 95%CI, 1.77-43.01), surgery + VC (HR: 6.97, 95%CI, 1.46-33.30), and lung metastases (HR: 2.92, 95%CI, 0.99-8.64). The model demonstrated moderate discriminatory ability for OS (c-statistic=0.68). A risk stratification model based on the combination of these factors and accounting for a 50% five-year survival cutoff identified 2 groups (poor prognosis, N=28 vs good prognosis, N=26) with distinct overall survival curves (p<0.001). Pre-operative STM and final histology were not associated (p=0.574 at Chi squared test). 5-yr OS after receiving 2nd line CT (n=25) was 18.7% (95%CI, 7.9-44.5). Results are limited by small numbers. Conclusions: Pts with PMNSGCT classified as having a good prognosis in this model had a fairly high survival estimate, while a strategy of consolidation CT for pts with poorest risk features warrants investigation, once the model is confirmed. The effect of surgery on survival was independent of post-CT STM, which also poorly predicted final histology.


2021 ◽  
Vol 11 ◽  
Author(s):  
Zhen Tan ◽  
Yubin Lei ◽  
Bo Zhang ◽  
Si Shi ◽  
Jiang Liu ◽  
...  

BackgroundPancreatic ductal adenocarcinoma (PDAC) is one of the most invasive solid malignancies. Immunotherapy and targeted therapy confirmed an existing certain curative effect in treating PDAC. The aim of this study was to develop an immune-related molecular marker to enhance the ability to predict Stages III and IV PDAC patients.MethodIn this study, weighted gene co-expression network (WGCNA) analysis and a deconvolution algorithm (CIBERSORT) that evaluated the cellular constituent of immune cells were used to evaluate PDAC expression data from the GEO (Gene Expression Omnibus) datasets, and identify modules related to CD4+ T cells. LASSO Cox regression analysis and Kaplan–Meier curve were applied to select and build prognostic multi-gene signature in TCGA Stages III and IV PDAC patients (N = 126). This was followed by independent Stages III and IV validation of the gene signature in the International Cancer Genome Consortium (ICGC, N = 62) and the Fudan University Shanghai Cancer Center (FUSCC, N = 42) cohort. Inherited germline mutations and tumor immunity exploration were applied to elucidate the molecular mechanisms in PDAC. Univariate and Multivariate Cox regression analyses were applied to verify the independent prognostic factors. Finally, a prognostic nomogram was created according to the TCGA-PDAC dataset.ResultsA four-gene signature comprising NAPSB, ZNF831, CXCL9 and PYHIN1 was established to predict overall survival of PDAC. This signature also robustly predicted survival in two independent validation cohorts. The four-gene signature could divide patients into high and low-risk groups with disparity overall survival verified by a Log-rank test. Expression of four genes positively correlated with immunosuppression activity (PD-L1 and PD1). Immune-related genes nomogram and corresponding calibration curves showed significant performance for predicting 3-year survival in TCGA-PDAC dataset.ConclusionWe constructed a novel four-gene signature to predict the prognosis of Stages III and IV PDAC patients by applying WGCNA and CIBERSORT algorithm scoring to transcriptome data different from traditional methods of filtrating for differential genes in cancer and healthy tissues. The findings may provide reference to predict survival and was beneficial to individualized management for advanced PDAC patients.


Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2216
Author(s):  
Marie Hautin ◽  
Clélia Mornet ◽  
Aurélie Chauveau ◽  
Delphine Bernard ◽  
Laurent Corcos ◽  
...  

Since the discovery of spliceosome mutations in myeloid malignancies, abnormal pre-mRNA splicing, which has been well studied in various cancers, has attracted novel interest in hematology. However, despite the common occurrence of spliceosome mutations in myelo-proliferative neoplasms (MPN), not much is known regarding the characterization and mechanisms of splicing anomalies in MPN. In this article, we review the current scientific literature regarding “splicing and myeloproliferative neoplasms”. We first analyse the clinical series reporting spliceosome mutations in MPN and their clinical correlates. We then present the current knowledge about molecular mechanisms by which these mutations participate in the pathogenesis of MPN or other myeloid malignancies. Beside spliceosome mutations, splicing anomalies have been described in myeloproliferative neoplasms, as well as in acute myeloid leukemias, a dreadful complication of these chronic diseases. Based on splicing anomalies reported in chronic myelogenous leukemia as well as in acute leukemia, and the mechanisms presiding splicing deregulation, we propose that abnormal splicing plays a major role in the evolution of myeloproliferative neoplasms and may be the target of specific therapeutic strategies.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 27-27
Author(s):  
Iyad Sultan ◽  
Osama Alsmadi ◽  
Anas Al Okaily ◽  
Razan Abu Khashabeh ◽  
Mayada Abu Shanap ◽  
...  

Background: Acute myelogenous leukemia (AML) is characterized by recurrent variants that have implications in diagnosis, prognosis and management. We analyzed all variants that were detected in our patients since the inauguration of our Next-Generation-Sequencing (NGS) facility. Methods: This is a retrospective study that analyzed all variants that were reported at our institution from Jan 2018 to Dec 2019 in patients with de-novo AML. NGS was performed using illumina miseq platform utilizing an amplicon panel that includes 50 genes known to be altered in AML. Variant Call Format (VCF) files were analyzed in a weekly meeting. For the purpose of this study, all VCFs were concatenated into a single MAF file. The Bioconductor package maftools was utilized to analyze this MAF file. Results: 78 patients (24 children, 48 males, median-age=31 yrs) were studied. 193 variants were detected in 63 samples (81% of patients). The median number of variants-per-sample were 2 (range, 1 to 6). Most commonly mutated genes wereTET2(26%),NPM1(21%), FLT3(17%), DNMT3A(15%),NRAS(13%)CBLC, CEBPA, KIT(8%). The most commonly found nucleotide alteration was C&gt;T transition which represented 50% of all mutations. Enrichment analysis showed the following significant associations (p&lt;0.05):FLT3rearrangement associated withNPM1andDNMT3Aalterations, andRUNX1T1/RUNX1rearrangement associated withKITalterations. Utilizing the drug gene interaction database, 72 mutations were categorized as druggable, the most frequently listed categories were clinically actionable variants (N=17), kinase-dependent (N=8) and druggable genome (N=7). Using log-rank test,NRASandDNMT3Avariants were found to be associated with significantly worse EFS (p=0.017 and 0.038, respectively), whileNRASvariants were associated with significantly worse OS (p=0.015). The pediatric age group had some noticeable difference; NPM1andTET2were significantly more altered in adults whileKITwas significantly more altered in children. Finally, we compared our results to The Cancer Genome Atlas (TCGA) de-novo AML data for 192 patients that was published previously. The TCGA raw data is available publicly and were used run through our pipeline for comparison. A striking similarity in the frequency of alterations (81% vs. 82%) and the top mostly altered genes. The following genes were significantly more altered in our cohort:TET2,GATA2, andCSF3R;whileIDH2was more altered in the TCGA cohort. Interestingly,CBLC(n=7) andZRSR2(n=5) variants were identified in KHCC patients but none in the TCGA cohort. Conclusion: We reported variants in 81% of our patients. Prognosis is significantly altered in patients withDNMT3AandNRASvariants. A wealth of actionable mutations were detected. Our results carry many similarities to the TCGA cohort, but significant difference in alterations involving 6 genes were reported. Our study lay the foundation for future studies that can evaluate big-data results obtained from our growing NGS facility. Figure Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2076-2076
Author(s):  
Santosh Saraf ◽  
Ardaman Shergill ◽  
Pritesh R. Patel ◽  
John G. Quigley ◽  
David Peace ◽  
...  

Abstract Abstract 2076 Myeloproliferative neoplasms (MPN) have similar incidence rates across different ethnicities in the United States. However, reported clinical studies rarely address the outcome in patients with different ethnicities. In this study, we retrospectively analyzed 127 patients with a diagnosis of MPN followed at the University of Illinois Hospital between January 2005 and July 2011. Of these patients, 69 were Caucasian and 58 Non-Caucasian (mostly African-American and Hispanic). The median age was 50 years (range: 20–85) in non-Caucasians and 51 years (range: 22–89) in Caucasians. Among 127 patients, 53 had polycythemia vera (PV), 52 essential thrombocythemia (ET), and 22 primary myelofibrosis (PMF). In each disease group, the following parameters were compared at diagnosis between Caucasian and non-Caucasian patients: age/gender, JAK2 V617F mutation status, cytogenetic abnormalities, family history of MPNs, constitutional symptoms, white blood cell (WBC) and platelet count, hemoglobin level, and spleen size. In addition, the two groups were compared for thrombotic or hemorrhagic events, cardiovascular complications, progression of disease, secondary cancer and overall survival. Among 53 PV patients, Caucasians (n=33) had higher WBC counts (11.7 vs. 8.1 ×103 cells/μL, p=0.03) and a greater spleen size (14 vs. 10 cm longitudinal diameter, p=0.03) at diagnosis compared to non-Caucasians (n=20). However, more frequent hemorrhagic (38.9% vs. 6.1%, p=0.003) and cardiovascular (27.8% vs. 3%, p=0.009) complications were seen in the non-Caucasian cohort of patients compared to the Caucasian cohort. Use of hydroxyurea (55% vs. 57%), aspirin (88% vs. 76%), anagrelide (10% vs. 24%), interferon (18% vs. 5%) and anticoagulation (21% vs. 20%) were not significantly different between Caucasians and non-Caucasians. In the ET patients (non-Caucasian: n=27; Caucasian: n=25), there were no significant differences in presenting characteristics, type of therapies, or clinical outcomes between the two groups. The 22 patients with PMF were classified according to the international prognostic scoring system (IPSS) at diagnosis. In 11 non-Caucasian and 11 Caucasian patients, differences of patients at low risk (18% vs. 9%), intermediate-1 (55% vs. 27%), intermediate-2 (27% vs. 55%), or high risk (9% vs. 0%) were not statistically significant. In a multivariate logistic regression analysis, we demonstrated that prior history of thrombosis and older age are independent prognostic factors for subsequent thrombotic or hemorrhagic events (p=0.03 and p=0.003, respectively). Neither WBC at diagnosis nor ethnicity were independent prognostic indicators in this series of patients. With a median follow-up of eight years (range 1–23 years), median overall survival was not reached for PV, ET, or PMF and no significant differences were detected in Caucasian or non-Caucasian groups. Our observations suggest that when given equal access to care, similar clinical outcomes are achieved in Caucasian and non-Caucasian patients with MPNs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. SCI-15-SCI-15
Author(s):  
Iannis Aifantis

Abstract Acute myeloid leukemia (AML) is the most common adult leukemia characterized by excessive proliferation of abnormal myeloid progenitors. AML continues to have a dismal survival rate amongst all subtypes of leukemia (<50% five-year overall survival rate), which can largely be attributed to limited advances in treatment regimens that, for the last decades, have relied on the use of two non-targeted cytotoxic drugs: cytarabine and anthracycline. Large-scale sequencing efforts have shed new light on genetic and epigenetic determinants of AML. Interestingly, these studies identified a frequent co-occurrence of somatic mutation between genes encoding cohesin complex subunits (such as STAG2, SMC1A, RAD21 and SMC3) and well-characterized AML oncogenic triggers, such as FLT3-ITD, TET2, and NPM1. Recent work has demonstrated an important role for the cohesin complex in normal stem/progenitor self-renewal and differentiation, gene regulation, and suppression of myeloproliferative neoplasms and AML, despite the precise mechanisms underlying these functions remaining poorly understood. It is believed that cohesin may suppress tumor formation by regulating chromatin looping at loci critical for self-renewal and myeloid progenitor differentiation. Utilizing established models of murine and human AML, this we will focus on the molecular mechanisms of cohesin-dependent myeloid tumor-suppression, with an emphasis on understanding novel treatment approaches that can exploit these functions. Using established protocols for identifying genome-wide changes in chromatin topology and gene expression, we propose undertaking an extensive characterization of cohesin-regulated chromatin changes driving AML. Furthermore, we investigate the application of targeted agents in cohesin-deficient AML whilst extensively mapping the mechanisms-of-action underlying these specific treatments. Ultimately, our work aims to generate novel, pre-clinical disease models of cohesin-mutated AML with strong mechanistic insights into the tumor-suppressive function of this complex. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document