27.12.2012 Views

12th Congress of the European Hematology ... - Haematologica

12th Congress of the European Hematology ... - Haematologica

12th Congress of the European Hematology ... - Haematologica

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

12 th <strong>Congress</strong> <strong>of</strong> <strong>the</strong> <strong>European</strong> <strong>Hematology</strong> Association<br />

Myeloproliferative disorders - Biology<br />

0411<br />

ACQUIRED ISODISOMY IS COMMON IN ATYPICAL MYELOPROLIFERATIVE DISORDERS<br />

C. Curtis, 1 F.H. Grand, 1 S. Kreil, 1 D.G. Oscier, 2 A.G Hall, 3 N.C.P Cross 1<br />

1 Wessex Regional Genetics Lab, SALISBURY, United Kingdom; 2 Royal<br />

Bournemouth Hospital, BOURNEMOUTH, United Kingdom; 3 University <strong>of</strong><br />

Newcastle Upon Tyne, NEWCASTLE UPON TYNE, United Kingdom<br />

Recent evidence has indicated that acquired isodisomy is a novel<br />

mechanism by which mutations in cancer may be reduced to homozygosity.<br />

Typically, acquired isodisomy is associated with oncogenic<br />

changes ra<strong>the</strong>r than tumour suppressor genes, eg. <strong>the</strong> activating JAK2<br />

V617F mutation in patients with myeloproliferative disorders (MPD) is<br />

<strong>of</strong>ten associated with acquired isodisomy for chromosome 9p. We have<br />

undertaken a screen for regions <strong>of</strong> acquired isodisomy as a means to<br />

identify genomic regions that may harbour novel oncogenes in patients<br />

with atypical MPD (n=30), blast crisis <strong>of</strong> chronic myeloid leukaemia<br />

(CML-BC; n=20) and chronic lymphocytic leukaemia (CLL; n=20).<br />

Genomewide SNP analysis was performed using Affymetrix 50K XbaI<br />

arrays and analysed for copy number changes and length <strong>of</strong> homozygous<br />

tracts. Chromosome deletions and gains identified by cytogenetic<br />

analysis were also detected by SNP analysis. Large tracts <strong>of</strong> homozygosity<br />

(defined as >20Mb running to a telomere), strongly suggesting<br />

acquired isodisomy, were seen 12 (40%) aMPD patients, a single case<br />

(5%) <strong>of</strong> CML-BC but were not seen in CLL. The homozygous tracts<br />

encompassed diverse genomic regions in aMPD, but two common<br />

regions (3 cases for each region) were identified at 7q and 11q. Because<br />

<strong>of</strong> <strong>the</strong> recurrent involvement <strong>of</strong> tyrosine kinases in MPDs, we focused<br />

our initial screen on this class <strong>of</strong> gene. The entire coding regions <strong>of</strong><br />

EPHB6 and EPHA1 were sequenced in patients with 7q isodisomy but<br />

no sequence changes were identified. Similarly, no changes were found<br />

in selected regions <strong>of</strong> BRAF and PIK3CG. Ongoing sequence analysis is<br />

focused on o<strong>the</strong>r candidate genes within <strong>the</strong> two regions.<br />

0412<br />

JAK2V617F-NEGATIVE ET PATIENTS DO NOT DISPLAY CONSTITUTIVELY ACTIVE<br />

JAK/STAT SIGNALLING<br />

S. H. Schwemmers, 1 H. Pahl, 1 B. Will1 C. F. Waller, 1 K. Abdulkarim, 2<br />

P. L. Johansson, 2 B. Andreasson, 2 H. L. Pahl1 1 University Hospital Freiburg, FREIBURG, Germany; 2 Sahlgrenska University<br />

Hospital, GÖTEBORG, Sweden<br />

Background. Presence <strong>of</strong> <strong>the</strong> Jak2V617F mutation in only 40-60% <strong>of</strong><br />

patients with Essential Thrombocy<strong>the</strong>mia (ET) underscores <strong>the</strong> heterogeneity<br />

<strong>of</strong> this myeloproliferative disorder (MPD). Several distinct mutations,<br />

ei<strong>the</strong>r in Jak2 (exon 12) or in c-Mpl (W515L) have been described<br />

in subsets <strong>of</strong> o<strong>the</strong>r Jak2V617F-negative MPDs, Polycy<strong>the</strong>mia vera (PV)<br />

and Idiopathic Myel<strong>of</strong>ibrosis (IMF). Analogous to Jak2V617F, <strong>the</strong>se<br />

mutations cause constitutive Jak2 and STAT activation. It has <strong>the</strong>refore<br />

been proposed that constitutive activation <strong>of</strong> <strong>the</strong> Jak/STAT pathway<br />

underlies <strong>the</strong> molecular etiology <strong>of</strong> all MPDs. Aims. We investigated <strong>the</strong><br />

alternative hypo<strong>the</strong>sis that distinct alterations, separate from <strong>the</strong><br />

Jak/STAT signal transduction pathway, underlie a subset <strong>of</strong> Jak2V617Fnegative<br />

ET. We <strong>the</strong>refore compared gene expression pr<strong>of</strong>iles <strong>of</strong> ET<br />

patients with and without <strong>the</strong> Jak2V617F mutation. Methods. 32 patients<br />

with ET, diagnosed according to <strong>the</strong> PVSG criteria, were included in <strong>the</strong><br />

study after giving informed consent. Jak2V617F allele expression was<br />

quantified in purified granulocytes by real time qRT-PCR. cDNA<br />

microarrays were processed according to <strong>the</strong> protocol <strong>of</strong> Eisen and<br />

Brown using <strong>the</strong> Lowess (Locally weighted scatter plot smoo<strong>the</strong>r) subgrid<br />

normalization method for comparison across slides. Statistical significance<br />

<strong>of</strong> differences was determined after correction for <strong>the</strong> false<br />

discovery rate by <strong>the</strong> method <strong>of</strong> Benjamini and Hochberg. Differences<br />

in gene expression were verified with qRT-PCR and protein phosphorylation<br />

detected by Western Blotting. Results. Unsupervised clustering<br />

<strong>of</strong> gene expression patterns in ET patients revealed two distinct subclasses<br />

<strong>of</strong> patients. These subclasses differed in presence or absence <strong>of</strong> <strong>the</strong><br />

Jak2V617F mutation. Patients lacking <strong>the</strong> Jak2V617F mutation displayed<br />

significantly lower expression <strong>of</strong> <strong>the</strong> STAT target genes Pim-1 and<br />

SOCS2. In addition, Jak2V617F-negative patients showed lower levels<br />

<strong>of</strong> STAT phosphorylation. Conclusions. These data demonstrate that a<br />

large proportion <strong>of</strong> Jak2V617F-negative ET patients do not display constitutive<br />

Jak/STAT signaling. Hence, we propose that alterations in dif-<br />

152 | haematologica/<strong>the</strong> hematology journal | 2007; 92(s1)<br />

ferent signal transduction pathways can lead to <strong>the</strong> clinical phenotype<br />

<strong>of</strong> ET. Elucidation <strong>of</strong> novel ET-inducing changes will facilitate both a<br />

molecular classification <strong>of</strong> ET and <strong>the</strong> development <strong>of</strong> rationally<br />

designed <strong>the</strong>rapies.<br />

0413<br />

MPL MUTATIONS IN PRIMITIVE MYELOFIBROSIS ARE NOT RESTRICTED TO MPL 515<br />

MUTATIONS BUT ONLY MPL 515 MUTATIONS ARE ONCOGENIC EVENTS PRESENT AT THE<br />

STEM CELL LEVEL<br />

S. Giraudier, 1 R. Chaligné, 2 C. James, 3 C. Tonetti, 4 J.P. Le Couédic, 2<br />

R. Besancenot, 2 I. Godin, 2 L. Lordier, 2 K. Malloum, 5 P. Mossuz, 6<br />

M.C. Le Bousse-Kerdilès, 7 W. Vainchenker, 2 S. Giraudier1 1 INSERM, U790 and <strong>Hematology</strong> Laboratory H, VILLEJUIF AND CRÉTEIL;<br />

2 INSERM, U790, VILLEJUIF; 3 INSERM, U217, BORDEAUX; 4 Faculté de<br />

Médecine Paris XII, CRÉTEIL; 5 AP-HP, Laboratoire d'hématologie, La pit,<br />

PARIS; 6 Laboratoire d'Hématologie, GRENOBLE; 7 INSERM, U602, VILLE-<br />

JUIF, France<br />

Background. The MPL 515 mutations have recently been described in<br />

10% <strong>of</strong> primitive myel<strong>of</strong>ibrosis (PMF) as an essential oncogenic event.<br />

Here we report o<strong>the</strong>r MPL mutations found in PMF patients. These new<br />

mutations raise <strong>the</strong> question whe<strong>the</strong>r <strong>the</strong>se molecular events occur in a<br />

true stem cell (ie: lymphoid/myeloid progenitor cell), as it has already<br />

been shown for JAK2 V617F occurrence in PMF and PV. Aims. The aim<br />

<strong>of</strong> this work was to study <strong>the</strong> presence <strong>of</strong> MPL exon 10 mutations in a<br />

cohort <strong>of</strong> 100 PMF, determine <strong>the</strong> functions <strong>of</strong> <strong>the</strong> different mutations,<br />

and if <strong>the</strong>se mutations occurred in both myeloid and lymphoid lineages<br />

in JAK2 V617F negative PMF. Methods and Results. We <strong>the</strong>refore<br />

sequenced MPL exon 10 in 100 PMF patients. Six different mutations<br />

were found. In order to determine whe<strong>the</strong>r <strong>the</strong>se mutations were only<br />

genetic polymorphism or real gain <strong>of</strong> function mutations, we introduced<br />

all <strong>the</strong> mutations in Baf3 cells. We found that only <strong>the</strong> recently described<br />

MPL 515 mutations and a 506+519 mutant induced factor independence<br />

growth, activation <strong>of</strong> JAK/STAT, RAS/MAPK, and PI3K transduction<br />

pathways as well as P21 overexpression, a spontaneous cell cycling<br />

with an increase in S and G2M phases, and tumorigenesis in NUDE<br />

mice. Then we looked for <strong>the</strong> MPL 515 mutations in stem cells compartments:<br />

First in mature myeloid and lymphoid cells and second in lymphoid/myeloid<br />

progenitors progeny after CD34 + and CD34 + CD38 – cell<br />

isolation from peripheral blood. Three PMF patients harboring MPL515<br />

mutations were studied. Peripheral blood granulocytes and platelets<br />

were purified by standard methods and B, T, NK cells and monocytes<br />

were isolated by combined immunomagnetic and flow cytometric procedures.<br />

The same techniques were used to sort CD34+ and<br />

CD34 + CD38 – subpopulations from peripheral blood. Clonal<br />

B/NK/Myeloid differentiation from CD34 + CD38 – cells and T cell differentiation<br />

from CD34 + cells were performed respectively onto a MS5<br />

layer in <strong>the</strong> presence <strong>of</strong> cytokines and in Fetal Thymic Organ Cultures<br />

(FTOC). Genotyping <strong>of</strong> mature cell populations, B/NK/Myeloid clones<br />

and CD34 + derived T cells were performed by direct sequencing. Moreover,<br />

CD34 + cells were cultured in a one cell per well experiment to<br />

determine <strong>the</strong> sensitivilty <strong>of</strong> <strong>the</strong>se patient’s cells to low dose TPO. The<br />

MPL515 mutations were present in granulocytes and platelets from all<br />

patients, and in monocytes in one. We detected <strong>the</strong> mutation in NK<br />

cells in two cases. The MPL 515L and MPL 515K mutations could be subsequently<br />

detected in CD34 + cells and in B/NK/Myeloid and/or<br />

NK/myeloid CD34 + CD38 – derived clones from all IMF patients. Interestingly,<br />

MPL 515L homozygous clones were detected in B/NK/Myeloid<br />

and/or NK/Myeloid clones from 1 patient. However, using <strong>the</strong> FTOC<br />

assay, <strong>the</strong> mutations were not detected in T cell fractions derived from<br />

CD34 + cells. At least we found that MPL mutations induce a spontaneous<br />

megakaryocytic growth in cell culture but also a hypersensitivity<br />

to low dose TPO. Summary. These results demonstrate that <strong>the</strong> MPL<br />

515 mutations occur in a lymphoid/myeloid progenitor cell and give<br />

rise to a hypersensitivity to TPO. Thus, <strong>the</strong>se MPD take <strong>the</strong>ir origin in<br />

a true lymphoid/myeloid progenitor cell. In accordance with mouse<br />

model, this represent a good argument for a causative role <strong>of</strong> MPL mutations<br />

in PMF.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!