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.

with a p value <strong>of</strong> ≥ 0.10. Sets <strong>of</strong> measurement data were compared for<br />

statistical significance using <strong>the</strong> Student’s t test or Mann-Whitney U<br />

test. Sets <strong>of</strong> enumeration data were compared by χ 2 and Fisher’s exact<br />

tests. Two-sided tests with an a level <strong>of</strong> 0.05 were used in all analyses.<br />

Results. 74% <strong>of</strong> informative cases had stage A disease, 43% had unmutated<br />

IgVH genes, 30% had high CD38 expression and 20% had adverse<br />

cytogenetic abnormalities (loss <strong>of</strong> p53 or ATM). The median time from<br />

diagnosis to blood sampling was 6.5 months and <strong>the</strong> median follow-up<br />

time from diagnosis 35.6 months. Low p53 or p21CIP1 index values<br />

were present in one third <strong>of</strong> patients and were associated with a significantly<br />

shorter overall survival and treatment-free interval (Figure 1).<br />

Although p53 dysfunction was strongly associated with loss <strong>of</strong> p53 or<br />

ATM, <strong>the</strong> majority <strong>of</strong> patients with p53 dysfunction did not have ei<strong>the</strong>r<br />

<strong>of</strong> <strong>the</strong>se adverse chromosomal abnormalities. Sub-group analysis<br />

revealed p53 dysfunction to be a powerful predictor <strong>of</strong> adverse outcome<br />

among patients classified as good risk by clinical staging, chromosomal<br />

analysis, IgVH mutation analysis or CD38 expression. In multivariate<br />

analysis, p53 dysfunction was <strong>the</strong> only independent predictor <strong>of</strong><br />

adverse outcome in patients with early-stage disease, those without<br />

adverse chromosomal abnormalities, and those with mutated IgVH<br />

genes (hazard ratio 7.8, 3.1 and 14.6 respectively). Summary/Conclusions.<br />

These findings identify p53 dysfunction as a novel independent prognostic<br />

factor in CLL. In addition, <strong>the</strong>y provide <strong>the</strong> first demonstration<br />

in human cancer that clinical outcome can be predicted by functional<br />

probing <strong>of</strong> a DNA-damage response pathway.<br />

Figure 1. TFI and OS <strong>of</strong> <strong>the</strong> two groups <strong>of</strong> CLL patients.<br />

0911<br />

THE ANTITUMOR ACTIVITY OF LUMILIXIMAB IS MEDIATED THROUGH THE INDUCTION<br />

OF CASPASE-9DEPENDENT APOPTOSIS AND IS SYNERGISTIC WITH RITUXIMAB AND<br />

FLUDARABINE IN CD23 + CLL AND LYMPHOMA CELLS<br />

N. Pathan, 1 J.C. Byrd, 2 K. Hariharan, 1 P. Chu, 1 A. Molina1 1 2 Biogen Idec, SAN DIEGO, USA; Ohio State University, COLUMBUS,<br />

USA<br />

Background. The effectiveness <strong>of</strong> monoclonal antibody <strong>the</strong>rapy in treating<br />

patients with chronic lymphocytic leukemia (CLL) has prompted<br />

interest in o<strong>the</strong>r antibodies specific for alternate B-cell antigens. The CD23<br />

antigen, a transmembrane glycoprotein that functions as a low-affinity<br />

receptor for immunoglobulin E, is highly expressed on most CLL cells and<br />

represents a potential <strong>the</strong>rapeutic target. Aims. We examined <strong>the</strong> ability<br />

<strong>of</strong> lumiliximab, an IgG1 chimeric monoclonal antibody against CD23, to<br />

mediate cytotoxicity via direct apoptosis, antibody-dependent cellular<br />

cytotoxicity (ADCC), and complement-dependent cytotoxicity (CDC)<br />

in CD23 + malignant B-cell lines and CLL cells. We also evaluated <strong>the</strong> antitumor<br />

activity <strong>of</strong> lumiliximab in a disseminated human lymphoma model.<br />

Methods. Cytotoxicity was determined using flow cytometry to assess<br />

<strong>the</strong> induction <strong>of</strong> apoptosis via caspase-3, 51CR-release to determine<br />

ADCC, and flow cytometry analysis <strong>of</strong> propidium iodide staining to<br />

quantify CDC. The activation <strong>of</strong> caspase-8 and -9 and expression <strong>of</strong> proand<br />

anti-apoptotic proteins were assessed before and after treatment with<br />

lumiliximab using Western blot analysis. The anti-tumor activity <strong>of</strong> lumiliximab<br />

was evaluated in vivo using <strong>the</strong> CD23 + SKW6.4 human lymphoma/SCID<br />

mouse model. Results. In CD23 + B-cell lines, lumiliximab<br />

induced apoptosis, ADCC, and CDC. Lumiliximab induced apoptosis<br />

and ADCC in CLL cells; however, <strong>the</strong> degree <strong>of</strong> ADCC was low to moderate<br />

compared with that seen in CD23 + B-cell lines. There was no evidence<br />

<strong>of</strong> CDC induction in primary CLL cells after exposure to lumiliximab.<br />

Treatment with lumiliximab in <strong>the</strong> presence <strong>of</strong> an anti-human IgG<br />

Fc crosslinking antibody resulted in increased staining <strong>of</strong> activated caspase-3<br />

in CLL cells’<strong>the</strong> median percentage <strong>of</strong> apoptosis was 46% compared<br />

with 16% with <strong>the</strong> isotype control (p

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

Saved successfully!

Ooh no, something went wrong!