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.

significant inhibition of cell proliferation. Further, pretreatment<br />

of the 8226/S and 8226/Dox40 cells with Rituximab followed by<br />

treatment with paclitaxel (10nM) for an additional 24h resulted in<br />

significant potentiation of cytotoxicity and synergy was achieved.<br />

The synergy in cytotoxicity was determined to be due to the<br />

induction of apoptosis. The mechanism by which Rituximab<br />

sensitized the 8226/Dox40 cells to paclitaxel-mediated apoptosis<br />

was investigated. Using western blot analysis it was found that<br />

treatment with Rituximab (20g/ml-24h) resulted in selective<br />

down-regulation of anti-apoptotic Bcl-xL expression with little<br />

effect on other gene products involved in apoptosis (c-IAP1, c-<br />

IAP2, XIAP, survivin, Mcl-1, Bcl-2). In addition, treatment with<br />

paclitaxel (10nM) arrested the cells at the G2/M phase of the cell<br />

cycle and resulted in the down-regulation of Mcl-1, Bcl-2, BclxL,<br />

c-IAP1 and survivin. These various gene modifications by<br />

Rituximab and paclitaxel resulted in functional complementation<br />

and the induction of the apoptotic signaling pathway. These<br />

findings suggest that signal I mediated by Rituximab and<br />

resulting in down regulation of anti-apoptotic Bcl-xL was<br />

important to circumvent the resistance of 8226/Dox40 cells to<br />

paclitaxel (signal II)-mediated apoptosis. Further, even though<br />

the expression of CD20 was low, nevertheless, the combination<br />

treatment resulted in significant percentage of cells undergoing<br />

apoptosis. The present findings also demonstrate that Rituximabmediated<br />

sensitization of the 8226/Dox40 cells to paclitaxelmediated<br />

apoptosis is independent of the MDR phenotype of the<br />

cells, as the functionality of the MDR pump was unaffected by<br />

single or combination treatments. These findings are of potential<br />

clinical significance. (Supported in part by the Jonsson<br />

Comprehensive Cancer Center At UCLA (A. J).<br />

386<br />

“An Antibody-avidin Fusion Protein Targeting the<br />

Human Transferrin Receptor Inhibits the Growth and<br />

Induces Apoptosis in Eight Human Malignant Plasma<br />

Cell Lines”<br />

Patrick P. Ng1, Xiao-Hu Gan1, Benjamin Bonavida1,<br />

Fuyuhiko Tamanoi1, Gary J. Schiller2, Alan K.<br />

Lichtenstein2, Dharminder Chauhan3, Kenneth C.<br />

Anderson3, Sherie L. Morrison1, Manuel L. Penichet1.<br />

1UCLA Department of Microbiology, Immunology and Molecular<br />

Genetics, 2UCLA Division of Hematology and Oncology,<br />

Department of Medicine, Los Angeles CA. 3Dana-Farber Cancer<br />

Institute, Harvard Medical School, Boston MA<br />

We have previously reported that an anti-rat transferrin receptor<br />

(TfR) IgG3-avidin fusion protein exhibits anti-proliferative/proapoptotic<br />

activity against the rat myeloma cell line Y3-Ag1.2.3<br />

and the rat T-cell lymphoma cell line C58(NT)D.1.G.OVAR.1.<br />

This activity was not observed in two rat cell lines of<br />

nonhematopoietic lineage (bladder carcinoma BC47 and<br />

gliosarcoma 9L) suggesting the potential use of anti-TfR IgG3-<br />

avidin for the treatment of hematopoietic malignancies. In<br />

addition, an anti-human TfR-IgG3-avidin fusion protein (antihTfR-Av)<br />

was shown to inhibit proliferation and induce apoptosis<br />

in human erythroleukemia cell line K562 (Ng et al., PNAS, 2002,<br />

99:10706). Recent studies demonstrated that anti-hTfR-Av also<br />

inhibits the growth and induces apoptosis in the human malignant<br />

plasma cell lines 8226/S, 8226/Dox40 [doxorubicin-resistant and<br />

also multidrug resistant], U266, MM.1S, OCI-My5, S6B45,<br />

ARH-77, and IM-9, although different levels of sensitivity were<br />

observed. Interesting, such anti-proliferative/pro-apoptotic<br />

activity seems not to be correlated with the level of TfR<br />

expressed on the surface of the cells. We are currently<br />

investigating the mechanism by which anti-hTfR-Av mediates its<br />

anti-proliferative/pro-apoptotic activity. We will also determine if<br />

it can be used in combination with various agents currently used<br />

in the treatment of hematopoietic malignancies. Importantly, antihTfR-Av<br />

can be further loaded with biotinylated anti-cancer<br />

drugs, which may increase its intrinsic anti-cancer activity. These<br />

approaches may lead to more effective therapeutics for in vivo<br />

eradication of plasma cell malignancies such as multiple<br />

myeloma and ex vivo purging of plasma cancer cells in<br />

autologous transplantation.<br />

387<br />

A chimeric humanized anti-CD40 antibody renders<br />

human multiple myeloma (MM) cells refractory to the<br />

mitogenic and protective effects of IL-6<br />

Yu-Tzu Tai,1 Constantine S. Mitsiades,1 Leurence P.<br />

Catley,1 Renate Burger,1 Masaharu Akiyama,1 Klaus<br />

Podar,1 Reshma Shringarpure,1 Teru Hideshima,1<br />

Dharminder Chauhan,1 Nicholas Mitsiades,1 Nikhil C.<br />

Munshi,1 Paul Richardson,1 Steven P Treon, and Kenneth<br />

C. Anderson1<br />

1The Jerome Lipper Multiple Myeloma Center, Department of<br />

Medical Oncology, Dana-Farber Cancer Institute, Boston, Mass.,<br />

USA<br />

CD40 is expressed on B-cell malignancies including human<br />

multiple myeloma (MM) and a variety of carcinomas, and there is<br />

considerable interest in using CD40 activating agents as novel<br />

cancer therapies. Recently, a humanized anti-CD40 Ab (SGN-14)<br />

demonstrates a significant anti-tumor effect in SCID mice<br />

xenografted with human MM (Cancer Res. 60, 3225-3231, 2000).<br />

In this study, we examined the therapeutic impact of SGN-14 in<br />

human MM using MM.1S cells (CD138+++CD40+) and plasma<br />

cell leukemia patient cells. SGN-14 (0.01-100 µg/ml) did not<br />

significantly stimulate proliferation of CD40-expressing MM.1S<br />

and MM.1R or two patient plasma cell leukemia cells (p >0.1 for<br />

all samples tested). SGN-14 (5 µg/ml) neither significantly<br />

induce AKT nor NF-B activation in MM.1S cells, in contrast to<br />

5 µg/ml sCD40L-treated counterparts. ERK activation was<br />

induced by SGN-14, although to a lesser extent than sCD40L.<br />

SGN-14 did not block nor enhance AKT/NF-B activation<br />

induced by sCD40L at the same concentration (5 µg/ml).<br />

Notably, 24-hr pretreatment of cells with SGN-14 blocked<br />

sCD40L-mediated PI3K/AKT and ERK activation. In contrast,<br />

cells treated with sCD40L for 24 h still retained the ability to<br />

further activate downstream signaling pathways in response to<br />

sCD40L. Importantly, pretreatment of MM.1S cells with SGN-14<br />

rendered them refractory to further proliferation induced by IL-6<br />

(p < 0.05): the two-fold increase of DNA synthesis induced by<br />

IL-6 (50 ng/ml) was completely blocked by pretreatment with 10<br />

µg/ml SGN-14 for 48 hrs. Furthermore, IL-6 did not overcome<br />

the inhibition of DNA synthesis triggered by Dex and IMiDs<br />

(0.01-10 µM) in cells pretreated with SGN-14 for 24-48 hrs.<br />

Using oligonucleotide microarray analysis, we first found<br />

differential gene expression profile between SGN-14-treated vs<br />

sCD40L-treated MM.1S cells and identified an approximately<br />

three-fold reduction of interleukin 6 receptor (IL6R) expression<br />

in SGN-14-treated MM.1S cells over 6-24 hr. Currently, the<br />

mechanism by which SGN-14 down regulates IL6R and<br />

possibly CD40 receptor is under further investigation. In addition,<br />

VEGF secretion was inhibited in the presence of SGN-14 (0.01-<br />

10 µg/ml) in MM.1S and MM.1R cells, as well as two plasma<br />

cell leukemia patient cells, correlating with reduced baseline<br />

migration of plasma leukemia cells in the presence of SGN-14.<br />

SGN-14 also induces antibody-dependent cell-mediated<br />

cytotoxicity and enhanced tumor cell lysis by effectors treated<br />

S260

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

Saved successfully!

Ooh no, something went wrong!