13.11.2014 Views

Haematologica 2003 - Supplements

Haematologica 2003 - Supplements

Haematologica 2003 - Supplements

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

Scientific sessions<br />

1. Development of normal and malignant<br />

plasma cell<br />

001<br />

The Essential Role of Oncogenic FGFR3 in<br />

Maintenance of t(4:14) Myeloma.<br />

Suzanne Trudel*, Scott Ely#, Yildiz Farooqui*, Davide F.<br />

Robbiani*, Maurizio Affer*, Marta Chesi*, Peter L.<br />

Bergsagel*.<br />

Medicine, Division of Hematology/Oncology*, Cornell Medical<br />

College, New York, NY, USA; Pathology#, Cornell Medical<br />

College, New York, NY, USA.<br />

Chromosomal translocations to the immunoglobulin heavy-chain<br />

locus on chromosome 14q32 are present in the majority of<br />

multiple myeloma (MM) patients and may represent the first and<br />

defining genetic event that leads to the development of MM. The<br />

t(4;14) translocation which occurs in approximately 15% of<br />

patients results in the dysregulated expression of fibroblast<br />

growth factor receptor 3 (FGFR3) and MMSET. Wild-type<br />

FGFR3 appears to be weakly transforming in a hematopoietic<br />

murine model. The subsequent acquisition of FGFR3 activating<br />

mutations is associated with disease progression and is strongly<br />

transforming in experimental models. These findings suggest a<br />

pathogenic correlation between FGFR3 expression and myeloma<br />

however it remains to be proven how dysregulation of FGFR3<br />

mediates an early oncogenic process in MM and whether FGFR3<br />

is required for tumor maintenance. We have used<br />

pharmacological inactivation of FGFR3 to address this question<br />

directly in human MM.<br />

We have developed 3 screening assays for identification of<br />

FGFR3 inhibitors and have used these to establish PD173074 as a<br />

selective inhibitor of FGFR3. Using this inhibitor we confirmed<br />

that inactivation of FGFR3 blocks its oncogenic potential. We<br />

have previously shown that activated forms of FGFR3 induce<br />

transformation of NIH 3T3. Using this same assay we tested the<br />

ability of PD173074 to inhibit the Y373C-FGFR3 induced<br />

transformation of NIH 3T3 cells. Although it had no effect on<br />

Ras-induced transformation, it completely inhibited foci<br />

formation induced by activated FGFR3. Similarly, PD173074<br />

prevented in vivo growth of Y373C-FGFR3 transfected NIH 3T3<br />

cells in nude mice but had no inhibitory effect on growth of Ras<br />

V12 expressing cells.<br />

To establish that FGFR3 activation provides a critical and nonredundant<br />

pro-proliferative and anti-apoptotic signal in MM we<br />

exposed FGFR3 expressing myeloma cell lines to PD173074.<br />

PD173074 inhibited cell proliferation of FGFR3 expressing<br />

KMS11 and KMS18 cells with an IC50 of 12.5 nM and 20 nM,<br />

respectively. 8226 cells, which lack FGFR3 expression, displayed<br />

no growth inhibition demonstrating that PD173074 exhibits<br />

minimal nonspecific cytotoxicity. Further characterization of this<br />

finding demonstrated that inhibition of cell growth is related to<br />

G0/G1 cell cycle arrest. PD173074 also induced delayed, doseresponsive<br />

apoptosis of these cells. Immunohistochemical<br />

analysis demonstrated an increase in cleaved caspase 3 positivity,<br />

suggesting that FGFR3 activation protects MM cells from<br />

caspase-dependent cell death. To explain the marked delay in<br />

apoptosis we speculated that inhibition of FGFR3 in these cells<br />

induces cell cycle arrest and differentiation. Inhibition of FGFR3<br />

resulted in the differentiation of KMS11 and KM18 cells from a<br />

plasmablast-like phenotype to a more mature plasma cell<br />

characterized morphologically and by the induction of CD31<br />

expression and increase in light chain secretion. In addition,<br />

FGFR3 inactivation had similar affects in vivo inducing growth<br />

arrest, apoptosis and differentiation of KMS11 tumors in a<br />

xenograph mouse model. Most importantly the reversion of the<br />

malignant phenotype was associated with delayed tumor<br />

progression and enhanced overall survival of PD173074 treated<br />

mice. These results provide evidence that FGFR3 is important<br />

for genesis and maintenance of myeloma. Further, they validate<br />

FGFR3 as a therapeutic target for a subset of MM patients.<br />

002<br />

The role of MMSET in t(4;14) myeloma<br />

Marta Chesi, Davide F. Robbiani, Maurizio Affer, Suzanne<br />

Trudel, W. Michael Kuehl and P. Leif Bergsagel<br />

Weill Medical College of Cornell University and Genetics<br />

Department, NCI<br />

The t(4;14)(p16;q32) translocation, that occurs in about 15-20%<br />

of multiple myeloma (MM), causes the concomitant<br />

dysregulation of two genes by their juxtaposition to the two<br />

immunoglobulin enhancers. FGFR3, is brought on the der(14)<br />

under the control of the IgH 3’ enhancer and the intronic<br />

enhancer, Emu, is translocated on der(4) where it dysregulates<br />

MMSET expression. The breakpoints are clustered in two groups:<br />

one falls in the 5’ UTR of MMSET, outside the coding sequence;<br />

the other falls into the 5’ coding exons, resulting into a N-term<br />

truncation of MMSET protein. Although the acquisition by the<br />

tumor cells of FGFR3 activating mutations indicates a role for<br />

FGFR3 in tumor progression in the few informative cases, we<br />

and other investigators reported the loss of der(14) or FGFR3<br />

expression in about 20% of t(4;14) myelomas. On the contrary,<br />

there is only one example of a t(4;14) MM that has lost der(4)<br />

and does not contain Ig/MMSET hybrid transcripts. Therefore<br />

MMSET seems to be the crucial gene in t(4;14) myeloma.<br />

MMSET belongs to the trithorax family of nuclear proteins<br />

characterized by the presence of a SET domain and several PHDtype<br />

zinc fingers and involved in chromatin remodeling. One of<br />

them, MLL, is located on 11q23 and translocated in acute<br />

leukemia. MMSET, also known as Nsd2, Trx5 and WHSC1, is<br />

the gene deleted in Wolf Hirsch Syndrome. Highly related genes,<br />

Nsd1 and Nsd3 have also been implicated in neoplastic<br />

transformation and found translocated in AML. There are two<br />

classes of MMSET mRNA transcripts, based on alternative<br />

splicing: MMSET type I encodes for a 674 aa protein, MMSET<br />

type II extends at the 3’ end of the type I and encodes for a 1365<br />

aa protein. Although we have found that the type I protein can<br />

block transformation of NIH3T3 fibroblasts by a variety of<br />

oncogenes, there is no direct evidence that MMSET can function<br />

as an oncogene. To study the oncogenic contribution of MMSET<br />

dysregulation in myeloma we followed two approaches 1) We<br />

generated retroviral vectors carrying EGFP, MMSETI and<br />

MMSETII fused to an IRES-neomycin cassette, and infected<br />

human myeloma cell lines (HMCL) that do not have a t(4;14)<br />

translocation and do not express MMSET; 2) we generated<br />

transgenic mice in which MMSETI and –II are under the control<br />

of the lck minimal promoter and Emu – a strategy that had<br />

previously enabled us and others to generate mice expressing<br />

transgenes in B and T cell lineages. Although we obtained<br />

HMCL neomycin resistant clones expressing MMSET mRNA,<br />

we were unable to detect MMSET protein expression. Under the<br />

same condition, however, MMSET exogenous protein was<br />

S88

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

Saved successfully!

Ooh no, something went wrong!