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

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

formation (in total 28 samples). Time to AML progression was 2-40<br />

months (mean 13 months). The methodology utilized for CGH was a<br />

variation <strong>of</strong> <strong>the</strong> basic protocol described by Kallioniemi et al (Genes<br />

Chrom C 1994;10:231-43). DNA copy number changes were described<br />

according to <strong>the</strong> ISCN (2005) guidelines. Results. Average number <strong>of</strong><br />

chromosome imbalances per MDS sample was 6 (range 0-17). Average<br />

number <strong>of</strong> chromosome imbalances per AML sample was 12 (range 3-<br />

21). X and Y chromosomes were excluded from analysis. Unexpectently,<br />

a common pattern <strong>of</strong> chromosomal loci gains was observed in different<br />

MDS patients during AML transformation (Table 1) namely 20p11.2,<br />

21q11.2, 21q21, 4q12, 16p11.2, 7q11.2, 18p11.2, 5q11.2, 20q11.2,<br />

18q11.2, 14q13, 16q13. Chromosomal losses were less frequently detected<br />

in this series. Confirmation <strong>of</strong> Ch-CGH Results. In 2 patients with<br />

abnormal karyotypes (1 patient with trisomy 8 and 1 with monosomy<br />

7) Ch-CGH showed amplification/loss <strong>of</strong> <strong>the</strong> relevant chromosomal loci.<br />

In 2 patients with 5q31 deletion in Ch-CGH was verified by FISH analysis<br />

using a 5q31 probe hybridised to bone marrow smears. In a patient<br />

with amplification <strong>of</strong> 22q11.2 in Ch-CGH 3 signals were obsereved in<br />

44% <strong>of</strong> cell nuclei when hybridised with 22q11.2 probe. Ch-CGH results<br />

were also verified by array-CGH in a sample (Gene Chip Affymetrix).<br />

Discussions. The finding <strong>of</strong> <strong>the</strong> existence <strong>of</strong> common chromosomal loci<br />

gains who appear only during <strong>the</strong> transition <strong>of</strong> MDS to AML is extremely<br />

important, given <strong>the</strong> extreme clinical and genetic heterogeneity <strong>of</strong><br />

MDS patients. Identification <strong>of</strong> oncogene(s) located in <strong>the</strong> amplified<br />

chromosomal areas that may contribute to <strong>the</strong> transformation <strong>of</strong> MDS<br />

to AML will give new <strong>the</strong>rapeutic targets.<br />

Table 1.<br />

0629<br />

INCREASED BONE MARROW ENDOTHELIAL PROGENITOR CONTENT IN MDS PATIENTS<br />

CORRELATES WITH DISEASE STAGE, AND MAY BE INVOLVED IN THE ANGIOGENESIS<br />

RESPONSE LEADING TO ACUTE LEUKEMIA<br />

C. Osório, 1 A.P. Elias, 1 M. Gomes da Silva, 2 S. Dias1 1 IPO/IGC, LISBON; 2 Haematology Department, IPO, LISBON, Portugal<br />

Myelodysplastic syndromes (MDS) are bone marrow (BM) disorders<br />

considered as pre-leukemic stages; an increase in BM angiogenesis has<br />

been suggested to contribute towards leukaemia progression. However,<br />

<strong>the</strong> mechanisms that regulate <strong>the</strong> vascular increase in MDS are poorly<br />

understood. In <strong>the</strong> present study we hypo<strong>the</strong>sized changes within <strong>the</strong><br />

MDS BM microenvironment might contribute to increased BM angiogenesis,<br />

and eventually result in AML onset and progression. For this purpose,<br />

we determined <strong>the</strong> cellular content and cell apoptosis <strong>of</strong> MDS BM<br />

samples (at diagnosis), and compared <strong>the</strong>se to BM samples from patients<br />

with o<strong>the</strong>r BM diseases (lymphomas, CLL, o<strong>the</strong>rs). In parallel, we<br />

analysed <strong>the</strong> BM expression <strong>of</strong> factors that might contribute towards<br />

increased vascularisation and/or altered cell turnover, such as VEGF, PlGF,<br />

TNF-α and TGF-β, respectively. Our criteria for BM collection and analysis,<br />

restricted to MDS BM samples collected at diagnosis (before any<br />

<strong>the</strong>rapeutic intervention), allowed us to study <strong>the</strong>se parameters in 12<br />

MDS patients and in 10 samples from patients with o<strong>the</strong>r BM diseases.<br />

Flow cytometry analysis <strong>of</strong> BM biopsies revealed a significant increase<br />

in <strong>the</strong> percentage <strong>of</strong> CD34 + , CD117 + and AC133 + progenitor cells which<br />

were related with MDS stages. Although MDS BM had globally a low-<br />

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

er BM cellular content, <strong>the</strong> proportion <strong>of</strong> o<strong>the</strong>r cell lineages remained<br />

largely unchanged between MDS BM (at diagnosis) and o<strong>the</strong>r diseases.<br />

In detail,AC133 + and KDR + cells percentage was significantly higher in<br />

MDS patients (5,6 and 1,7-fold respectively), suggesting an increase in<br />

endo<strong>the</strong>lial cells or progenitors (EPC) BM content. Accordingly, <strong>the</strong> apoptotic<br />

index <strong>of</strong> AC133 + cells was lower in MDS marrows (8% apoptotic<br />

AC133 + cells in MDS versus 30% in o<strong>the</strong>r bone marrow malignancies).<br />

To understand how <strong>the</strong> MDS BM microenvironment might contribute<br />

to towards an increase in BM vasculature, <strong>the</strong> total level <strong>of</strong> BM VEGF<br />

was determined (by ELISA), and <strong>the</strong> proportion <strong>of</strong> <strong>the</strong> different VEGF<br />

is<strong>of</strong>orms was assessed (by RQ-PCR). Although <strong>the</strong> bone marrow<br />

mononuclear cell number was lower in MDS BM, VEGF levels per cell<br />

were 2-fold higher in MDS marrows than in <strong>the</strong> o<strong>the</strong>r disases group.<br />

Interestingly, besides a clear increase in total VEGF, it was also observed<br />

that <strong>the</strong> proportion between <strong>the</strong> VEGF is<strong>of</strong>orms changed: is<strong>of</strong>orms<br />

VEGF145 and VEGF189 increased significantly in MDS BM, and with<br />

MDS progression. The quantification <strong>of</strong> factors that might be involved<br />

in <strong>the</strong> changes in cell turnover within <strong>the</strong> MDS BM samples, favouring<br />

endo<strong>the</strong>lial cell and EPC expansion in relation to o<strong>the</strong>r lineages, revealed<br />

that TNF-α expression was lower in MDS patients while TGF-β levels<br />

were similar in both groups, and PlGF was marginally expressed.<br />

Although obtained from a restricted group <strong>of</strong> patients, <strong>the</strong>se findings<br />

suggest that an increase in <strong>the</strong> EPC pool is directly related with MDS<br />

stages, may contribute to <strong>the</strong> angiogenic pr<strong>of</strong>ile in MDS, which in turn<br />

may lead to disrupted BM homeostasis, eventually resulting in leukemia<br />

onset and disease progression. Myelodysplastic syndromes (MDS) are<br />

bone marrow (BM) disorders considered as pre-leukemic stages; an<br />

increase in BM angiogenesis has been suggested to contribute towards<br />

leukaemia progression. However, <strong>the</strong> mechanisms that regulate <strong>the</strong> vascular<br />

increase in MDS are poorly understood. In <strong>the</strong> present study we<br />

hypo<strong>the</strong>sized changes within <strong>the</strong> MDS BM microenvironment might<br />

contribute to increased BM angiogenesis, and eventually result in AML<br />

onset and progression. For this purpose, we determined <strong>the</strong> cellular content<br />

and cell apoptosis <strong>of</strong> MDS BM samples (at diagnosis), and compared<br />

<strong>the</strong>se to BM samples from patients with o<strong>the</strong>r BM diseases (lymphomas,<br />

CLL, o<strong>the</strong>rs). In parallel, we analysed <strong>the</strong> BM expression <strong>of</strong> factors<br />

that might contribute towards increased vascularisation and/or<br />

altered cell turnover, such as VEGF, PlGF, TNF-α and TGF-β, respectively.<br />

Our criteria for BM collection and analysis, restricted to MDS BM<br />

samples collected at diagnosis (before any <strong>the</strong>rapeutic intervention),<br />

allowed us to study <strong>the</strong>se parameters in 12 MDS patients and in 10 samples<br />

from patients with o<strong>the</strong>r BM diseases. Flow cytometry analysis <strong>of</strong><br />

BM biopsies revealed a significant increase in <strong>the</strong> percentage <strong>of</strong> CD34 + ,<br />

CD117 + and AC133 + progenitor cells which were related with MDS<br />

stages. Although MDS BM had globally a lower BM cellular content,<br />

<strong>the</strong> proportion <strong>of</strong> o<strong>the</strong>r cell lineages remained largely unchanged<br />

between MDS BM (at diagnosis) and o<strong>the</strong>r diseases. In detail, AC133 +<br />

and KDR + cells percentage was significantly higher in MDS patients (5,6<br />

and 1,7-fold respectively), suggesting an increase in endo<strong>the</strong>lial cells or<br />

progenitors (EPC) BM content. Accordingly, <strong>the</strong> apoptotic index <strong>of</strong><br />

AC133 + cells was lower in MDS marrows (8% apoptotic AC133 + cells<br />

in MDS versus 30% in o<strong>the</strong>r bone marrow malignancies). To understand<br />

how <strong>the</strong> MDS BM microenvironment might contribute to towards<br />

an increase in BM vasculature, <strong>the</strong> total level <strong>of</strong> BM VEGF was determined<br />

(by ELISA), and <strong>the</strong> proportion <strong>of</strong> <strong>the</strong> different VEGF is<strong>of</strong>orms<br />

was assessed (by RQ-PCR). Although <strong>the</strong> bone marrow mononuclear cell<br />

number was lower in MDS BM, VEGF levels per cell were 2-fold higher<br />

in MDS marrows than in <strong>the</strong> o<strong>the</strong>r disases group. Interestingly, besides<br />

a clear increase in total VEGF, it was also observed that <strong>the</strong> proportion<br />

between <strong>the</strong> VEGF is<strong>of</strong>orms changed: is<strong>of</strong>orms VEGF145 and VEGF189<br />

increased significantly in MDS BM, and with MDS progression. The<br />

quantification <strong>of</strong> factors that might be involved in <strong>the</strong> changes in cell<br />

turnover within <strong>the</strong> MDS BM samples, favouring endo<strong>the</strong>lial cell and<br />

EPC expansion in relation to o<strong>the</strong>r lineages, revealed that TNF-α expression<br />

was lower in MDS patients while TGF-β levels were similar in both<br />

groups, and PlGF was marginally expressed. Although obtained from a<br />

restricted group <strong>of</strong> patients, <strong>the</strong>se findings suggest that an increase in <strong>the</strong><br />

EPC pool is directly related with MDS stages, may contribute to <strong>the</strong><br />

angiogenic pr<strong>of</strong>ile in MDS, which in turn may lead to disrupted BM<br />

homeostasis, eventually resulting in leukemia onset and disease progression.<br />

haematologica/<strong>the</strong> hematology journal | 2007; 92(s1) | 235

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

Saved successfully!

Ooh no, something went wrong!