Incidence and Impact of Activating Mutations of the RAS-RAF-MEK-ERK Pathway in Patients with Philadelphia-Negative (Ph-negative) Myeloproliferative Neoplasms (MPNs) and Myelodysplastic/Myeloproliferative Disorders–Unclassified (MDS/MPD-U)

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3172-3172
Author(s):  
Fabio PS Santos ◽  
Bianca Lisboa ◽  
Tarcila S Datoguia ◽  
Ricardo Helman ◽  
Welbert Oliveira Pereira ◽  
...  

Abstract Introduction: Mutations that activate the RAS-RAF-MEK-ERK pathway have long been known to occur in patients with solid tumors and hematological malignancies. The most common mutations occur in the Ras family of GTPases (HRAS, NRAS, KRAS) and the Raf family of serine-threonine kinases (ARAF, BRAF, CRAF). In myeloid malignancies, RAS mutations have mainly been described in patients with acute myeloid leukemia, chronic myelomonocytic leukemia (CMML) and myelodysplastic syndrome. There are few studies describing the incidence of mutations of the RAS-RAF-MEK-ERK pathway in patients with MPNs other than CMML. Objective: To describe the incidence, clinical features and prognostic impact of Ras and Raf mutations in patients with Ph-negative MPNs and MPN/MDS-U Methods: Paired DNA (sorted CD66b-granulocytes/skin biopsy) from patients with MPNs or MPN/MDS was subjected to whole exome sequencing on a Illumina HiSeq 2000 platform using Agilent SureSelect kit (see our abstract “Whole Exome Sequencing of Myeloproliferative Neoplasms and Myelodysplastic/Myeloproliferative Disorders”). Tumor coverage was 150x and germline coverage was 60x. Somatic variants calls were generated by combining the output of Somatic Sniper (Washington University), Mutect (Broad Institute) and Pindel (Washington University), followed by in-house filters to reduce false positive calls. Statistical calculations were done in Stata, v11.0. Results: We found clonal activating mutations of the RAS-RAF-MEK-ERK pathway in 8 patients (6.7% of cases). Diagnosis included primary myelofibrosis (PMF; N=5), MDS/MPD-U (N=2) and essential thrombocythemia (ET; N=1). Their clinical features are summarized in Table 1 (three of these patients [UPIs #11, #13, #99] are also described in the abstract “Genomic Profile of Patients with Triple Negative (JAK2, CALR and MPL) Essential Thrombocythemia and Primary Myelofibrosis”). There were 7 NRAS mutations and 1 BRAF mutation. In 5 cases the variant allele fraction (VAF) of reads in the tumor sample indicated that the mutation was present in a subclone at the time of sequencing. We next compared the clinical features of these 8 patients with 79 patients (MF=43, ET=35, MDS/MPD=1) who did not harbor these mutations. Patients with NRAS/BRAF mutations had lower hemoglobin (8.3 vs. 11.8 g/dL, p=0.001), higher white blood cell counts (28.37 vs. 7.7 x109/L, p=0.008) and had higher lactate dehydrogenase (1041 vs. 685 IU/L, p=0.02). They also had worse overall survival compared to unmutated cases (Hazard ratio [HR]=11.57; p=0.001). Most patients with NRAS/BRAF mutations had a high number of concomitant driver mutatons (median 5 vs. 1; p<0.0001). When the number of driver mutations was analyzed together with NRAS/BRAF mutations in a Cox model, NRAS/BRAF mutations were no longer independent predictors of survival (HR=1.48; p=0.61). Conclusions: Activating mutations of the RAS-RAF-MEK-ERK pathway occur in 6-7% of patients with Ph-negative MPNs, and they tend to co-occur with a high number of concomitant driver mutations. In most cases the mutation was present in a subclone, suggesting that they are late occurring. Patients with NRAS/BRAF mutations had a trend for worse outcome, but that was mainly dependent on the total number of driver mutations. The activity of MEK and BRAF inhibitors needs to be explored in patients with Ph-negative MPNs who harbor activating mutations of the RAS-RAF-MEK-ERK pathway. Table 1. Clinical features of patients with NRAS/BRAF mutations UPI Diagnosis Mutation VAF Concomitant driver genes and Chromosomal abnormalities Outcomes 7 MF NRAS p.G12S 47% ASXL1, CALR, STAG2, U2AF1 Died from disease progression 11 MF NRAS p.G12R 5% ASXL1, CBL, CUX1 (double mutant), EZH2 Died from disease progression 13 MF NRAS p.G12D 48% ASXL1, DNMT3A, ETV6 (double mutant) JARID2, U2AF1 Died from disease progression 18 MF NRAS p.G13D 25% JAK2, Del(5q) Underwent allogeneic transplantation; disease relapsed day+80; alive 29 MDS/MPD-U BRAF p.D594G 25% JAK2, Del(5q) Transformed to AML; entered CR with induction chemotherapy; underwent allogeneic transplantation; disease relapsed day+35; alive 99 ET NRAS p.G12D 43% ASXL1, CSF3R, STAG2 Alive 109 MF NRAS p.Q61R 19% CALR, DNMT3A, ZRSR2 Alive 122 MDS/MPD-U NRAS p.G12S 7% ASXL1, EZH2 (double mutant), PTPN11, TET2 (double mutant) Transformed to AML; underwent allogeneic transplantation; died on day+58 Disclosures No relevant conflicts of interest to declare.

2020 ◽  
Vol 2020 ◽  
pp. 1-4
Author(s):  
Stephen E. Langabeer ◽  
Lisa Lee Tokar ◽  
Laura Kearney ◽  
Cathal O’Brien ◽  
Kowshika Thavarajah ◽  
...  

Acquired, activating mutations of MPL W515 are recognised driver mutations of the myeloproliferative neoplasms (MPN), namely, essential thrombocythemia and primary myelofibrosis. The most common mutation at this codon is W515L with several other mutations also described at a lower frequency. Of these less common mutations, MPL W515S has only been reported sporadically with limited information on clinicopathological associations. We describe the case of an elderly man with persistent thrombocytosis presenting with an ischemic cerebral event. Bone marrow biopsy showed evidence of prefibrotic myelofibrosis with targeted sequencing demonstrating the presence of the rare MPL W515S mutation. Thrombolytic and cytoreductive therapies resulted in a favorable outcome and follow-up. This case provides additional, necessary, and phenotypic data for the rare MPN-associated MPL W515S mutation.


Blood ◽  
2017 ◽  
Vol 129 (6) ◽  
pp. 667-679 ◽  
Author(s):  
William Vainchenker ◽  
Robert Kralovics

Abstract The genetic landscape of classical myeloproliferative neoplasm (MPN) is in large part elucidated. The MPN-restricted driver mutations, including those in JAK2, calreticulin (CALR), and myeloproliferative leukemia virus (MPL), abnormally activate the cytokine receptor/JAK2 pathway and their downstream effectors, more particularly the STATs. The most frequent mutation, JAK2V617F, activates the 3 main myeloid cytokine receptors (erythropoietin receptor, granulocyte colony-stimulating factor receptor, and MPL) whereas CALR or MPL mutants are restricted to MPL activation. This explains why JAK2V617F is associated with polycythemia vera, essential thrombocythemia (ET), and primary myelofibrosis (PMF) whereas CALR and MPL mutants are found in ET and PMF. Other mutations in genes involved in epigenetic regulation, splicing, and signaling cooperate with the 3 MPN drivers and play a key role in the PMF pathogenesis. Mutations in epigenetic regulators TET2 and DNMT3A are involved in disease initiation and may precede the acquisition of JAK2V617F. Other mutations in epigenetic regulators such as EZH2 and ASXL1 also play a role in disease initiation and disease progression. Mutations in the splicing machinery are predominantly found in PMF and are implicated in the development of anemia or pancytopenia. Both heterogeneity of classical MPNs and prognosis are determined by a specific genomic landscape, that is, type of MPN driver mutations, association with other mutations, and their order of acquisition. However, factors other than somatic mutations play an important role in disease initiation as well as disease progression such as germ line predisposition, inflammation, and aging. Delineation of these environmental factors will be important to better understand the precise pathogenesis of MPN.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2888-2888 ◽  
Author(s):  
David G. Kent ◽  
Christina Ortmann ◽  
Yvonne Silber ◽  
Joanna Baxter ◽  
Beatriz Bellosillo ◽  
...  

Abstract Cancers evolve as a consequence of the stepwise accumulation of somatic lesions, with competition between subclones and sequential subclonal evolution. Some driver mutations arise either early or late in the evolution of different individual tumors, indicating that the final malignant properties of a subclone reflect the sum of mutations acquired rather than the order in which they arose. However virtually nothing is known about the cellular consequences of altering the order in which mutations are acquired. The myeloproliferative neoplasms (MPNs) readily permit clonal analysis and are chronic malignancies, thereby facilitating the dissection of disease evolution and intra-tumoral clonal architecture. In this study, we genotyped >7000 colonies from 24 MPN patients who harbored mutations in both JAK2 and TET2. We found that mutation of JAK2 and TET2 each occurred first in 12/24 patients, with TET2-first and JAK2-first patients observed in all 3 MPN subtypes. Patients who acquired a TET2 mutation first presented on average 12.3 years later than JAK2-first patients (p=0.0043) and had a lower proportion of JAK2V617F homozygous erythroid colonies (p=0.0001) suggesting that the order of acquisition affected both cellular composition and disease evolution. Moreover, compared to TET2-first patients, JAK2-first patients had an increased frequency of megakaryocyte/erythrocyte progenitors (p=0.0001) and decreased frequency of common myeloid progenitors (p=0.001). In order to determine whether disease evolution was also affected at the stem cell level, we isolated individual HSCs (lin-CD34+CD38+CD90-CD45RA+) and found that JAK2 single-mutant HSCs were significantly less prevalent than their JAK2/TET2 double-mutant counterparts. In marked contrast, TET2 single-mutant HSCs were significantly more prevalent than JAK2/TET2 double-mutant HSCs, suggesting that TET2 single-mutant HSCs exhibit a substantial self-renewal advantage and were not out-competed by their double-mutant progeny. Consistent with this interpretation, functional progenitor expansion from single cell cultures of individual HSCs (as assessed by secondary colony formation) was differentially altered by the second mutation acquired. Whereas the mean number of secondary colonies produced by double-mutant HSCs was increased when TET2 was the second mutation (p=0.012), the mean number of colonies was decreased when JAK2 was the second mutation (p=0.002) despite both HSCs bearing the same set of genetic lesions. Together our data indicate that acquisition of a JAK2 mutation reduces the competitiveness of TET2 single mutant HSCs, whereas acquisition of a TET2 mutation enhances that of JAK2 single mutant HSCs. These observations represent the first demonstration that the order of mutation acquisition influences stem and progenitor cell behaviour and clonal evolution in cancer. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e18561-e18561
Author(s):  
Gabriela Hobbs ◽  
Jennifer Lombardi Story ◽  
Maura A. Blaney ◽  
Philip C. Amrein ◽  
Amir Tahmasb Fathi ◽  
...  

e18561 Background: Post-myeloproliferative neoplasm (MPN) acute myeloid leukemia (AML) and accelerated phase disease (AP) are associated with poor outcomes; the optimal management of these patients at transformation is uncertain in the era of widely available molecular testing. Methods: We performed a retrospective analysis of adult patients with MPN that had transformed into AML (greater than 20% blasts) or AP (10-19% blasts and dysplastic features) from 2006-2016. Patients were 18 or older at AML/AP diagnosis. Outcomes were described by Kaplan and Meier and the Log rank test. Results: We identified 30 patients, with transformed MPN into AML (n = 23) or AP (n = 7), including one patient in AP at presentation. Most were male (73%). Disease evolved from polycythemia vera (n = 6), essential thrombocythemia (n = 11), primary myelofibrosis (n = 9) and other MPN diagnoses (MPN/MDS overlap, n = 3; MPN NOS, n = 1). The median age at MPN diagnosis was 67.5 yrs, and at transformation 72 yrs. The median time from MPN diagnosis to transformation was 5.6 yrs (range 0.3-36.0). 12 patients had JAK2 V617F testing both at MPN diagnosis and AML diagnosis; 6 had JAK2 mutations at both time points and 2 lost JAK2 at transformation. At AML/AP transformation, 11 patients had NGS mutation testing; the most common somatic mutations were NRAS (5/11), RUNX1 (2/11), and SRSF2 (2/11). 20 patients received treatment outside of supportive care. Of these, 8 achieved a CR or CRi (40%; 7/10 with induction and 1/8 with HMA). The only HMA response was on a trial of HMA+SGN33a. The median survival from AML/AP transformation was 5.8 mo. Of treated patients, median survival was 7.4 mo; 37% of treated patients were alive at 1 yr. Nine patients (31%; 7 with AML and 2 with AP) underwent allogeneic transplantation; 2 relapsed during follow-up. 74% of patients treated with allo-HCT were alive at 1 yr after AML/AP transformation. Conclusions: AML or accelerated phase disease arising out of MPN carries a dismal prognosis. HMA monotherapy had little efficacy in this group (0/7 achieved remission). Allogeneic transplantation offered the best chance of survival at one year, but fewer than a third of patients were able to proceed to transplant.


2018 ◽  
Vol 36 (6_suppl) ◽  
pp. 306-306 ◽  
Author(s):  
Malshundria Prophet ◽  
Kun Xiao ◽  
Theodore Stewart Gourdin ◽  
Rebecca J Nagy ◽  
Lesli Ann Kiedrowski ◽  
...  

306 Background: Activating BRAF fusion proteins are rare in prostate cancer (PCa) patients. Driver missense BRAF mutations have not been reported in detail in this population. Methods: We examined ctDNA-derived genomic profiles (Guardant 360) from 2,721 unique PCa patients, to identify BRAF genomic anomalies (SNVs, amplification). The ctDNA results were compared with PCa tissue-based genomics from the TCGA database (1,851 unique patients). Results: BRAF missense mutations were found in 76 ctDNA patients (2.8%) and were from all known mutation classes (I, II, III) as well as variants of unknown significance (VUSs). Only 4 patients had the V600E mutation. Multiple examples of known, autonomously active, non-canonical mutations were found (27), including K601E (12), G469A (5), D594G (2), and G466E (2). There were 45 VUSs. Mutations were primarily clonal but subclonal mutations were also found. In addition BRAF was commonly amplified, usually in the presence of multiple other amplified genes. BRAF missense mutations were more common with ctDNA than TCGA (2.8% vs 1.4%). Neither dataset identified frequent V600E mutations (ctDNA: 4/2,721; TCGA 1/1,851). However patients with the same non-canonical BRAF mutations were found in each dataset (K601E, G469A, G466E, D594G). Each dataset contained unique mutations found in only one patient. BRAF mutations potentially treatable with BRAF or MEK inhibitors (class I, II) were about half of all mutations (ctDNA 40.8%; TCGA 50%). We treated a PCa patient with a clonal BRAF(G469A) mutation with targeted therapy. The patient was resistant to multiple lines of hormonal and cytotoxic therapy. Trametinib produced a clinical and RECIST response. Conclusions: ctDNA-based genomic analysis identified multiple BRAF amplifications and missense SNVs in PCa patients. SNVs are largely non-canonical, but include known activating mutations that could act as drivers. The analysis also identified more BRAF missense mutations than did tissue genomic profiling, but the mutational landscape, overall frequency of mutations was similar with either method. ctDNA-based genomic profiling can identify actionable BRAF driver mutations that may respond to MEK and BRAF inhibitors.


2021 ◽  
Author(s):  
Maira da Costa Cacemiro ◽  
Juçara Gastaldi Cominal ◽  
Luiz Miguel Pereira ◽  
Maria Gabriela Berzoti-Coelho ◽  
Giovana Michelassi Berbel ◽  
...  

Abstract Myeloproliferative neoplasms (MPN) are hematological disorders characterized by increased proliferation of precursor and mature myeloid cells. MPN patients may present driver mutations in JAK2, MPL, and CALR genes, which are essential to describe the molecular mechanisms of MPN pathogenesis. Despite all the new knowledge on MPN pathogenesis, many questions remain to be answered to develop effective therapies to cure MPN or impair its progression to acute myeloid leukemia. The present study examined the expression levels of the Hippo signaling pathway members in patients with polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), as well as the role that they play in disease pathogenesis. The Hippo pathway is a tumor suppressor pathway that participates in the regulation of cell proliferation, differentiation, and death. Our main findings were: (i) expression of tumor suppressor genes from Hippo pathway were downregulated and seemed to be associated with cell resistance to apoptosis and increased proliferation rate; and (ii) Hippo pathway-related gene expression was associated with mutation status in ET and PMF patients. Therefore, the decreased expression of Hippo pathway-related genes may contribute to the malignant phenotype, apoptosis resistance, and cell proliferation in MPN pathogenesis.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 482-482
Author(s):  
Koki Ueda ◽  
Kazuhiko Ikeda ◽  
Kazuei Ogawa ◽  
Akiko Shichishima-Nakamura ◽  
Kotaro Shide ◽  
...  

Abstract Myeloproliferative neoplasms (MPN) are characterized by chronic proliferation of myeloid cells, extramedullary hematopoiesis and occasional leukemic transformation. Mutations in JAK2, CALR and MPL have been established as drivers of myeloproliferative phenotype, but their roles in disease progression with clonal expansion remain unclear. In addition, studies have shown mutations in epigenetic modifiers including TET2, DNMT3A, ASXL1 and EZH2, and aberrant expressions of microRNAs in MPN, but downstream of these changes is also largely unknown. Recently, we showed high expression of HMGA2 mRNA partly correlated with reduced microRNA let-7 in granulocytes of patients with MPN, including 100% patients with primary myelofibrosis (MF) and 20% polycythemia vera and essential thrombocythemia (Harada-Shirado et al, Brit J Haematol, 2015). In mice, loss of epigenetic modifiers such as BMI1 and EZH2, along with the Arf/Ink4a knockout (Oguro et al, J Exp Med, 2012) or the JAK2 V617F (Sashida et al, ASH, 2013), leads to overexpression of HMGA2 with accelerating MPN. We have generated transgenic (Tg) mice of Hmga2 cDNA with truncated 3'UTR (ΔHmga2) lacking binding sites of let-7 thatrepresses expression of HMGA2 (Ikeda et al, Blood, 2011). Δ Hmga2 mice overexpress HMGA2 and develop MPN-like disease, and represent a clonal advantage in competitive repopulations with serial bone marrow (BM) transplants (BMT). Here, to clarify if HMGA2 affect JAK2 V617F+ hematopoiesis, we crossed Δ Hmga2+/- mice with JAK2 V617F+/- Tg mice (Shide et al, Leukemia, 2008). Δ Hmga2-/-JAK2 V617F-/- wild type (WT), Δ Hmga2+/-JAK2 V617F-/- (Δ Hmga2 -Tg), Δ Hmga2-/-JAK2 V617F+/- (JAK2 V617F-Tg) and Δ Hmga2+/-JAK2 V617F+/- (double-Tg) mice were born at expected Mendelian ratios and we could analyze 5 - 6 of each. At 3 months old, leukocytosis, thrombocytosis, anemia and splenomegaly were most severe in double-Tg compared with JAK2 V617F-Tg or Δ Hmga2 -Tg mice. Relative to WT, peripheral leukocyte and platelet counts were nearly 16- and 4-fold higher in double-Tg, while 3- and 2-fold higher in JAK2 V617F-Tg mice, respectively. Mean spleen weights were 0.067, 0.10, 0.83 and 2.8 g in WT, Δ Hmga2 -Tg, JAK2 V617F-Tg and double-Tg mice, while BM cell counts were 2.4, 2.8, 0.4 and 1.2 x 107/femur, respectively. However, JAK2 V617F-Tg and double-Tg equally showed MF whereas no MF was detected in WT and DHmga2-Tg, suggesting that HMGA2 partly recovers cellularity in fibrotic BM. In the absence and presence of JAK2 V617F, HMGA2 augments lineage- Sca1+ Kit+ cells (WT: Δ Hmga2-Tg: JAK2 V617F-Tg: double-Tg= 0.17%: 0.19%: 0.17%: 0.27% in BM cells), endogenous erythroid colonies (1: 11: 13: 21 CFU-E/104 BM cells) and CD71+ Ter119+ erythroblasts (23%: 29%: 5.7%: 10% in BM and 2.0%: 4.4%: 7.9%: 16% in spleen cells), indicating HMGA2 contributes to expansion of hematopoietic stem/progenitor cells (HSPC) and erythroid commitment in JAK2 V617F+ hematopoiesis. Most Δ Hmga2-Tg and JAK2 V617F-Tg survived for over one year, but all double-Tg mice died within 4 months after birth due to severe splenomegaly and MF with no acute leukemia. To study the effect of HMGA2 on JAK2 V617F+ HSPC activity, we performed BMT with 0.25 x 106 Ly5.2+Δ Hmga2-Tg, JAK2 V617F-Tg or double-Tg cells with 0.75 x 106 Ly5.1+ competitor WT cells to lethally irradiated Ly5.1+ WT mice. Proportions of Ly5.2+ cells were higher in recipients of Δ Hmga2 -Tg than double-Tg cells, while JAK2 V617F-Tg cells were almost rejected at 8 weeks after BMT. To confirm role of HMGA2 without let-7 repression in JAK2 V617F+ hematopoiesis, we performed another BMT with 1 x 104 KIT+ cells of JAK2 V617F-Tg mice transduced with retroviral vector of Hmga2 with each let-7 -site-mutated full-length 3'UTR (Hmga2-m7) to sublethally irradiated WT mice. Recipients of JAK2 V617F-Tg cells with Hmga2-m7 developed MPN-like disease, whereas donor cells were rejected in recipients of JAK2 V617F cells with empty vector. In conclusion, HMGA2 may play a crucial role in hematopoiesis harboring JAK2 V617F by expanding HSPC, leading to disease progression. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 2 (24) ◽  
pp. 3581-3589 ◽  
Author(s):  
Bridget K. Marcellino ◽  
Ronald Hoffman ◽  
Joseph Tripodi ◽  
Min Lu ◽  
Heidi Kosiorek ◽  
...  

Abstract The Philadelphia chromosome–negative myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and the prefibrotic form of primary myelofibrosis (PMF), frequently progress to more overt forms of MF and a type of acute leukemia termed MPN-accelerated phase/blast phase (MPN-AP/BP). Recent evidence indicates that dysregulation of the tumor suppressor tumor protein p53 (TP53) commonly occurs in the MPNs. The proteins MDM2 and MDM4 alter the cellular levels of TP53. We investigated in 1,294 patients whether abnormalities involving chromosomes 1 and 12, which harbor the genes for MDM4 and MDM2, respectively, and chromosome 17, where the gene for TP53 is located, are associated with MPN disease progression. Gain of 1q occurred not only in individuals with MPN-BP but also in patients with PV and ET, who, with further follow-up, eventually evolve to either MF and/or MPN-BP. These gains of 1q were most prevalent in patients with a history of PV and those who possessed the JAK2V617F driver mutation. The gains of 1q were accompanied by increased transcript levels of MDM4. In contrast, 12q chromosomal abnormalities were exclusively detected in patients who presented with MF or MPN-BP, but were not accompanied by further increases in MDM2/MDM4 transcript levels. Furthermore, all patients with a loss of 17p13, which leads to a deletion of TP53, had either MF or MPN-AP/BP. These findings suggest that gain of 1q, as well as deletions of 17p, are associated with perturbations of the TP53 pathway, which contribute to MPN disease progression.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1724-1724
Author(s):  
Line Nederby ◽  
Laura Laine Larsen ◽  
Peter Hokland ◽  
Anne Stidsholt Roug

Abstract Abstract 1724 Introduction: BCR-ABL negative myeloproliferative neoplasms (MPNs) (polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF)) have preponderance to progress to hematopoietic insufficiency with fibrosis and/or blast transformation. This progression is often difficult to detect, but can be preceded by a stage of variable duration commonly termed acceleration, which is not homogeneously defined, but characterized by worsening cytopenias and constitutional symptoms. The monitoration of this process is clearly important, but at the present time biomarkers for this are lacking. We and others have described the human myeloid inhibitory C-type lectin-like receptor (hMICL) (also known as CLL-1) to be a very stable marker for the malignant cells in acute myeloid leukemia (AML), even in CD34- cases (van Rhenen et al. Leukemia 2007, Roug et al. Cytometry 2011). Given the need for new markers in MPN we hypothesized that determining the level of circulating hMICL expressing stem cells could be predictive of disease progression in MPN. Materials and Methods: Peripheral blood (PB) was obtained from 37 PV-, 19 ET-, and 8 PMF patients. A total of 57 (36 PV, 16 ET, and 5 PMF) patients were in stable phase while 7 (1 PV, 3 ET, and 3 PMF) were defined as being in an accelerated phase. Samples were prepared for five-color flow cytometry protocols using the antibodies anti-CD34, anti-CD38, anti-CD45, anti-CD14, and anti-hMICL. Acquisition was performed on a BD FACSCanto II and data analyses were carried out in FlowJo software. We focused on the following subsets: CD45lowSSClowCD14-CD34+hMICL+ and CD45lowSSClowCD14-CD34+CD38-hMICL+. Gates defining hMICL positivity were established on the basis of hMICL- lymphocytes as internal controls. One million cells were analyzed per sample and subsets of interest were calculated as percentage of these. Results: We found a percentage of CD34+hMICL+ (mean 1.176%, SD 0.908) and CD34+CD38-hMICL+ (mean 0.256%, SD 0.329) in PMF patients, which was statistically different from both PV- (mean 0.038%, SD 0.101, and mean 0.005%, SD 0.018, respectively) (p=0.0002 and p<0.0001, respectively) and ET patients (mean 0.116%, SD 0.252, and mean 0.009%, SD 0.024, respectively) (p=0.001 and p=0.0007, respectively). Somewhat surprisingly, compared to PV patients the CD34+hMICL+ compartment in ET patients was significantly higher (p=0.036). Importantly, the stem cell compartment in the accelerated phase patients was significantly higher than in the stable phase ones: CD34+hMICL+ (stable phase: mean 0.108%, SD 0.31, and accelerated phase: mean 0.983%, SD 0.917, respectively) (p=0.0002) and CD34+CD38-hMICL+ (stable phase: mean 0.017%, SD 0.081, and accelerated phase: mean 0.202%, SD 0.325, respectively)(p=0.0003). Conclusion: We have identified circulating CD34+hMICL+ and CD34+CD38-hMICL+ cells as an interesting biomarker for evaluating the disease state in MPN patients. The addition of hMICL discriminates between normal and neoplastic stem- and progenitor cells in MPN patients and reliably assigns disease stages in these patients superior to the mere evaluation of circulating CD34+ and CD34+CD38- cells. Importantly, hMICL+ cells, though rare, were dependably identified in PB. Thus, this study provides possible options for flow cytometric monitoring of MPN patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5473-5473
Author(s):  
Marianna De Muro ◽  
Ambra Di Veroli ◽  
Marco Montanaro ◽  
Roberto Latagliata ◽  
Cristina Santoro ◽  
...  

Abstract Background: MPNs including Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF), are clonal hematopoietic diseases in which the discovery of molecular driver mutations (JAK2, CALR, MPL) has deeply modified diagnostic approach in recent years. To date available data on epidemiology of MPNs and perspective analysis are rare. Our aim is to study the incidence of MPN Ph negative in a specific region of Italy named Latium and its variability across five years. Moreover we prospectively report the general features of our population. Method: We present here the prospective epidemiologic analysis of 1116 adult patients affected by MPNs (PV=289, ET=550, PMF=209) diagnosed according to 2008 WHO criteria, from January 2011 to December 2015 in 15 hematological Centers (5 academic and 10 community-based Hospitals) in Latium. A total of 289 PV, 550 ET and 209PMF were identified. The overall incidence rate of 289PV was 1.0/105 in 2011 and 2012, 1.1/105 in 2013, 0.9/105 in 2014 and 2015. The overall incidence rate of 550ET was 2.0/105 in 2011, 2.4/105 in 2012, 2.2/105 in 2013, 1.8/105 in 2014 and 1,2/105 in 2015 and the overall incidence rate of 209PMF was 0.7/105 in 2011 and 2012, 1.0/105 in 2013, 0.7/105 in 2014 and 0.5/105 in 2015. We have observed also 63 cases of MPNu (36M/32F) and the incidence rate was 0.3/105 in 2011 and 2012, 0.14/105 in 2013, 0.24/105 in 2014 and 0.22/105 in 2015. Baseline features of PV, ET and PMF patients are summarized in table 1. We have also analyzed the presence of comorbidities including obesity, arhythmia and neoplasia observed at the diagnosis in 1.6, 6.2 and 4% of all population, respectively; thirty-five percent of 1116 pts presented other comorbidities such as diabetes, inflammatory bowel disease, renal and liver failure. As thrombotic risk factors we considered diabetes, dislipidemia, smoke, essential hypertension and thrombophilia observed in 11,8, 16,2, 13,2, 51,7 and 3% of total pts, respectively. Conclusions: We confirm in our prospective observational protocol the overall incidence of MPN Ph negative, previously reported in the literature and the major incidence of male gender in PV and PMF, female in of ET. The annual incidence from 2011-2015 in Latium is remained substantially the same during the observation period. The decreasing trend observed in 2015 is probably due to the different update of some Centers that was done in October 2015 not including patients diagnosed in the last two months. Disclosures Latagliata: Novartis: Consultancy, Honoraria; Bristol Myers Squibb: Honoraria; Celgene: Honoraria; Janssen: Consultancy, Honoraria; Shire: Honoraria. Breccia:Pfizer: Honoraria; Novartis: Consultancy, Honoraria; Bristol Myers Squibb: Honoraria; Celgene: Honoraria; Ariad: Honoraria. Cimino:Celgene: Honoraria; Bristol-Mayer: Honoraria.


Sign in / Sign up

Export Citation Format

Share Document