Haematologica 2003 - Supplements

Haematologica 2003 - Supplements Haematologica 2003 - Supplements

supplements.haematologica.org
from supplements.haematologica.org More from this publisher
13.11.2014 Views

130 mTOR, a novel therapeutic target for multiple myeloma? F. Bakker, A. Wohlfarth, W. Baum, R. Burger, A. Guenther, and M. Gramatzki University of Erlangen-Nuremberg, Department of Medicine III, Division of Hemato-/Oncology Recently, it was reported that the phospatidyl inositol 3-kinase (PI3-K) signaling pathway may play an important role for the growth and survival of malignant plasma cells in multiple myeloma (MM). The PI3-K and its downstream effector Akt, activated by interleukin(IL)-6 or by insulin-like growth factor(IGF)-1, were shown to mediate proliferative signals and to inhibit dexamethasone induced apoptosis. In addition, it could be shown that multiple downstream targets are phosphorylated in MM cells upon activation of the PI-3K. Rapamycin is a potent and highly selective inhibitor of the mammalian target of rapamycin (mTOR), a downstream effector of PI3-K which mediates activation of p70S6K and 4E-BP1 by Akt. Rapamycin is a naturally occuring product isolated from Streptomyces hygroscopicus, which has strong immunosuppressive properties. In addition to its immunosuppressive properties, rapamycin was found to inhibit the growth of normal and malignant B- and T- lineage cells. In myeloma cells, rapamycin was shown to be able to revert IL-6 and IGF-1 induced phosphorylation of p70S6K and 4E-BP1 as well as IL-6 and IGF-1 induced cell growth under serum-free conditions. However, the overall effect of rapamycin on myeloma cell growth was relatively modest in these experiments. From other systems, it is known that serum is a potent activator of the PI3-K signaling pathway. For example, serum albumin was recently identified as the major plasma component responsible for Akt activation in B-CLL cells (…). Therefore, experiments conducted under serum-free conditions may not reveal the relevance of PI3-K signaling for myeloma cell growth and the potential role of PI3-K signaling pathway inhibitors in the treatment of multiple myeloma. By Western Blot analysis, we were able to show that fetal calf serum (FCS) is a far more potent activator of the p70S6K than either IL-6 or IGF-1 in all five human MM cell lines tested. Subsequently, we studied the effect of rapamycin treatment on the growth of these MM cell lines in the presence of FCS. Rapamycin was highly effective at inhibiting the growth of all five MM cell lines tested, as determined by [3H]-Thymidine incorporation (IC50 ranging from 0.24 to 1.8 ng/ml, growth inhibition at 100 ng/ml ranging from 74 to 97 %). Furthermore, we could show that this growth inhibition is mainly mediated by a G1 cell-cycle arrest. Next, the in vivo efficacy of rapamycin was evaluated in a SCID mouse xenograft model for human plasmacytoma, based on the IL-6 dependent human myeloma cell line INA-6. Starting on day 2 after tumor inoculation, a total of 14 mice received an oral dose of 2 mg/kg rapamycin 3 times per week for a total of two weeks. This dose was comparable to the maximum tolerated dose determined in studies with patients with advanced solid tumors. Treatment with rapamycin significantly delayed tumor development and prolonged survival of mice compared to untreated controls (median survival: 108 days vs. 58 days). The in vitro and preclinical in vivo efficacy of rapamycin indicates that inhibition of mTOR may provide an interesting treatment option for patients with malignant plasma cell tumors. 131 Inhibition of Protein Geranylgeranylation Induces Apoptosis in Myeloma Cells by Reducing Mcl-1 Protein Levels Niels W.C.J. van de Donk1, Marloes M.J. Kamphuis1, Berris van Kessel2, Henk M. Lokhorst2 and Andries C. Bloem1 Departments of Immunology1 and Hematology2, University Medical Center Utrecht, Utrecht, The Netherlands Prenylation is a class of lipid modification involving covalent attachment of hydrophobic isoprenoid molecules to target proteins including small GTP-binding proteins such as Ras and members of the Rac and Rho families. Farnesyl transferase (FTase) and geranylgeranyl transferase (GGTase) catalyze the transfer and binding of farnesyl and geranylgeranyl moieties from farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP), respectively. Prenylation is essential for membrane attachment and the subsequent participation of prenylated proteins in diverse signaling pathways regulating growth and survival. Lovastatin inhibits 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, which is the rate-limiting enzyme of the mevalonate pathway. This pathway leads to formation of cholesterol and isoprenoids like FPP and GGPP. Treatment of plasma cell lines and ex vivo purified tumor cells from myeloma patients with lovastatin resulted in the induction of apoptosis. Co-treatment of cells with lovastatin and mevalonate or GGPP, but not FPP, abrogated lovastatin-induced apoptosis. Furthermore, induction of apoptosis and reduction of cell viability by inhibition of FTase were less pronounced when compared to inhibition of GGTase I. This implies that geranylgeranylation of proteins is critical for the regulation of survival of myeloma cells. Apoptosis triggered by inhibition of geranylgeranylation was associated with reduction of Mcl-1 protein expression, which, in turn, resulted in the collapse of the mitochondrial transmembrane potential, cytochrome c release from mitochondria into the cytosol, and stimulation of caspase-3 activity. Lovastatin enhanced the cytotoxic effects of dexamethasone and doxorubicin in a synergistic fashion. This synergism may be due to the lovastatin-mediated Mcl-1 downregulation. These results show that geranylgeranylation of proteins is a key event in the regulation of myeloma tumor cell survival, probably through the regulation of Mcl-1 expression. Based on these findings, we started a Phase I/II trial to evaluate the combination of dose-escalating lovastatin and CHOP or VAD chemotherapy in refractory non-Hodgkin’s lymphoma and multiple myeloma patients. At the moment, 5 patients have been included in this trial. 132 Targeting the IGF-I Receptor Survival Pathways in Multiple Myeloma Using Selective IGF-I Receptor Tyrosine Kinase Inhibitors T. Strömberg, L. Dimberg, O. Larsson, M. Axelsson, K. Nilsson, H. Jernberg-Wiklund Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala, Sweden; Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden The insulin-like growth factor I receptor (IGF-IR) is strongly suggested to play a key role in malignant transformation. In tumor cells from multiple myeloma (MM), IGF-I has been established as an important survival factor, and we and others have previously shown that a blocking anti-IGF-IR antibody (IR3) augments apoptosis induced by serum-starvation, Fas- S145

ligation or dexamethasone (Georgii-Hemming et al 1996, Nilsson et al 1998). However, the biological significance of individual anti-apoptotic elements downstream the IGF-IR are not fully elucidated. Upon ligand interaction with the IGF-R -subunit, tyrosine residues in the intracellular, membrane-bound ß-subunit are autophosphorylated. This enables phosphorylation and docking of the insulin receptor substrates (IRS), thereby activating two important pathways mediating proliferation and survival, i.e. the phosphatidylinositol 3-kinase (PI 3-K)/Akt and the mitogenactivated protein kinase (MAPK) pathways. Thus, interfering with signaling at the level of the receptor tyrosine kinase (RTK) might represent an attractive strategy to sensitize MM cells to apoptosis. To characterize the effects of the IGF-I RTK inhibitors in MM, we used a panel of authentic MM cell lines as well as freshly purified CD138+ tumor cells from MM patients. The IGF-I RTK inhibitors proved to be effective in the MM cell lines and in the primary MM cells providing 50-90% cell death within 48 h incubation during standard, serum-containing culture conditions. The two drug resistant subclones of the MM cell line RPMI8226, RPMI8226/Dox 40 (doxorubicin) and RPMI8226/LR5 (melphalan), were also sensitive to the RTK inhibitors showing similar IC50s as the parental cell line. Additionally, the IGF-I RTK inhibitors were studied in combination with conventional cytotoxic drugs, e.g. dexamethasone. Analysis of the IGF-I RTK activity using immunoprecipitation of the IGF-IR -subunit and Western blotting shows that the IGF-I RTK inhibitors downregulate both basal and ligand-induced RTK activity. Furthermore, the downstream consequences of the perturbed IGF-IR signaling were investigated using phosphorylation site-specific antibodies directed against signaling molecules and substrates of the PI 3-kinase/Akt and the MAPK pathways. To identify target genes crucial for the IGF-IR mediated survival, gene expression of candidate regulators of apoptosis, e.g. the Bcl-2 family of genes, were analysed by ribonuclease-protection assay and Western blotting. 133 Inhibition of Protein Kinase C delta negatively regulates the Akt signaling pathway and induces apoptosis of myeloma cells Nizar J Bahlis, Christopher Maisel, Stanton Gerson Case Western Reserve University, Ireland Comprehensive Cancer Center, Cleveland, OH 44106 Multiple myeloma (MM) is an invariably fatal disease of terminally differentiated B-lineage or plasma cells. Several growth factors (IL-6, IGF-I, VEGF) provide proliferative and oncogenic signals to these cells, contributing to their resistant phenotype. These signals are often transduced by a family of phospholipid-regulated, serine / threonine kinases known as Protein Kinase C. The expression of these PKC isoenzymes is cell specific and each isoenzyme is reported to regulate a specific cellular function. We first determined that several myeloma cell lines (8226s, U266, MM1S and MM1R) expressed classical PKC isoforms (α,β,γ) as well as the novel isoform, PKC δ. Treatment of myeloma cells with the classical and novel PKC inhibitor, N- benzoyl-staurosporine (PKC412, Novartis) (500-1000nM) induced apoptosis in dexamethasone sensitive (MM1S) as well as dexamethasone (MM1R) and chemo-resistant (U266) cells. Cotreatment with IL-6 (100ng/ml) did not inhibit PKC412 induced cell death while the caspase inhibitor Z-VAD-fmk (100µM) significantly abrogated its effect. PKC412 induced activation of caspase 9, caspase 3 and cleavage of PARP. Most importantly PKC412 reduced ser 473 phosphorylation of Akt and its downstream phosphorylated substrates GSK3-β, FKHR and Bad. Similarly to PKC412, the specific PKC δ inhibitor, Rottlerin (3µM), reduced Akt phosphorylation, induced caspase 3 and PARP cleavage and apoptosis of treated myeloma cells as well. However the classical PKC (α, β, γ) inhibitor GÖ6976 did not significantly affect their viability. Finally PKC412 (1µM) and Rottlerin (3µM) induced apoptosis in primary myeloma cells (CD138 high) while they had no effect on CD138 low bone marrow mononuclear cells. Our studies demonstrate that inhibition of PKCδ negatively regulates Akt in myeloma cells and activates the downstream apoptotic machinery providing a rationale for targeting PKC isoforms, especially PKCδ in multiple myeloma. 134 MOLECULAR PROFILE OF THE ANTI-MYELOMA ACTIVITY OF HISTONE DEACETYLASE (HDAC) INHIBITORS: BIOLOGICAL AND THERAPEUTIC IMPLICATIONS. Constantine S. Mitsiades1,2, Nicholas S. Mitsiades1,2, Ciaran J. McMullan1,2, Vassiliki Poulaki3, Reshma Shringarpure1,2, Teru Hideshima1,2, Masaharu Akiyama1,2, Dharminder Chauhan1,2, Nikhil Munshi1,2, Xuesong Gu4, Charles Bailey4, Marie Joseph4, Towia A. Libermann4, Victoria M. Richon5, Paul A. Marks5,6, Kenneth C. Anderson1,2. 1. Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115; 2. Department of Medicine, Harvard Medical School, Boston, MA 02115; 3. Massachusetts Eye and Ear Infirmary, Harvard Medical School, 234 Charles Street, Boston, MA 02114; 4. Genomics Center, Beth Israel Deaconess Medical Center, Harvard Institutes of Medicine, Boston, MA 02115; 5. Aton Pharma, Inc. Tarrytown, NY; 6. Memorial Sloan–Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA. Histone deacetylases (HDACs) affect cell differentiation and survival at the transcriptional level, by regulating the acetylation status of nucleosomal histones and the function of transcription factor complexes. HDAC inhibition induces differentiation and/or apoptosis in transformed cells. We recently showed (Blood, in press) that HDAC inhibitors, such as the prototypic hydroxamic acid-based HDAC inhibitor suberoylanilide hydroxamic acid (SAHA), potently induce cell death (through caspase-independent/calpain-dependent mechanism) of human multiple myeloma (MM) cells, including cell lines and MM patient-derived tumor cells, either sensitive or resistant to conventional or novel anti-tumor agents. SAHA also sensitized MM cells to death receptor (e.g. Fas or TRAIL receptor)-mediated apoptosis and inhibited IL-6 secretion in cocultures of bone marrow stromal cells (BMSCs) with MM cells. These comprehensive effects of SAHA, both on MM cells directly and on their microenvironmental interactions, prompted further investigation of the molecular sequelae of this class of agents, with particular focus on the transcriptional profile of SAHA-treatment, since HDAC inhibition exerts its anti-tumor activity by targeting predominantly the regulation of gene expression. HDAC inhibition was originally pursued with intent to induce differentiation of malignant (e.g. leukemic) cells, by derepressing transcriptional programs of cellular differentiation. Interestingly, however, our gene expression profiling (using U133A Affymetrix oligonucleotide microarrays) and subsequent confirmatory mechanistic and functional assays, indicate that S146

130<br />

mTOR, a novel therapeutic target for multiple<br />

myeloma?<br />

F. Bakker, A. Wohlfarth, W. Baum, R. Burger, A. Guenther,<br />

and M. Gramatzki<br />

University of Erlangen-Nuremberg, Department of Medicine III,<br />

Division of Hemato-/Oncology<br />

Recently, it was reported that the phospatidyl inositol 3-kinase<br />

(PI3-K) signaling pathway may play an important role for the<br />

growth and survival of malignant plasma cells in multiple<br />

myeloma (MM). The PI3-K and its downstream effector Akt,<br />

activated by interleukin(IL)-6 or by insulin-like growth<br />

factor(IGF)-1, were shown to mediate proliferative signals and to<br />

inhibit dexamethasone induced apoptosis. In addition, it could be<br />

shown that multiple downstream targets are phosphorylated in<br />

MM cells upon activation of the PI-3K. Rapamycin is a potent<br />

and highly selective inhibitor of the mammalian target of<br />

rapamycin (mTOR), a downstream effector of PI3-K which<br />

mediates activation of p70S6K and 4E-BP1 by Akt. Rapamycin is<br />

a naturally occuring product isolated from Streptomyces<br />

hygroscopicus, which has strong immunosuppressive properties.<br />

In addition to its immunosuppressive properties, rapamycin was<br />

found to inhibit the growth of normal and malignant B- and T-<br />

lineage cells. In myeloma cells, rapamycin was shown to be able<br />

to revert IL-6 and IGF-1 induced phosphorylation of p70S6K and<br />

4E-BP1 as well as IL-6 and IGF-1 induced cell growth under<br />

serum-free conditions. However, the overall effect of rapamycin<br />

on myeloma cell growth was relatively modest in these<br />

experiments. From other systems, it is known that serum is a<br />

potent activator of the PI3-K signaling pathway. For example,<br />

serum albumin was recently identified as the major plasma<br />

component responsible for Akt activation in B-CLL cells (…).<br />

Therefore, experiments conducted under serum-free conditions<br />

may not reveal the relevance of PI3-K signaling for myeloma cell<br />

growth and the potential role of PI3-K signaling pathway<br />

inhibitors in the treatment of multiple myeloma. By Western Blot<br />

analysis, we were able to show that fetal calf serum (FCS) is a far<br />

more potent activator of the p70S6K than either IL-6 or IGF-1 in<br />

all five human MM cell lines tested. Subsequently, we studied the<br />

effect of rapamycin treatment on the growth of these MM cell<br />

lines in the presence of FCS. Rapamycin was highly effective at<br />

inhibiting the growth of all five MM cell lines tested, as<br />

determined by [3H]-Thymidine incorporation (IC50 ranging from<br />

0.24 to 1.8 ng/ml, growth inhibition at 100 ng/ml ranging from 74<br />

to 97 %). Furthermore, we could show that this growth inhibition<br />

is mainly mediated by a G1 cell-cycle arrest. Next, the in vivo<br />

efficacy of rapamycin was evaluated in a SCID mouse xenograft<br />

model for human plasmacytoma, based on the IL-6 dependent<br />

human myeloma cell line INA-6. Starting on day 2 after tumor<br />

inoculation, a total of 14 mice received an oral dose of 2 mg/kg<br />

rapamycin 3 times per week for a total of two weeks. This dose<br />

was comparable to the maximum tolerated dose determined in<br />

studies with patients with advanced solid tumors. Treatment with<br />

rapamycin significantly delayed tumor development and<br />

prolonged survival of mice compared to untreated controls<br />

(median survival: 108 days vs. 58 days). The in vitro and<br />

preclinical in vivo efficacy of rapamycin indicates that inhibition<br />

of mTOR may provide an interesting treatment option for patients<br />

with malignant plasma cell tumors.<br />

131<br />

Inhibition of Protein Geranylgeranylation Induces<br />

Apoptosis in Myeloma Cells by Reducing Mcl-1 Protein<br />

Levels<br />

Niels W.C.J. van de Donk1, Marloes M.J. Kamphuis1,<br />

Berris van Kessel2, Henk M. Lokhorst2 and Andries C.<br />

Bloem1<br />

Departments of Immunology1 and Hematology2, University<br />

Medical Center Utrecht, Utrecht, The Netherlands<br />

Prenylation is a class of lipid modification involving covalent<br />

attachment of hydrophobic isoprenoid molecules to target<br />

proteins including small GTP-binding proteins such as Ras and<br />

members of the Rac and Rho families. Farnesyl transferase<br />

(FTase) and geranylgeranyl transferase (GGTase) catalyze the<br />

transfer and binding of farnesyl and geranylgeranyl moieties from<br />

farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate<br />

(GGPP), respectively. Prenylation is essential for membrane<br />

attachment and the subsequent participation of prenylated<br />

proteins in diverse signaling pathways regulating growth and<br />

survival. Lovastatin inhibits 3-hydroxy-3-methylglutaryl<br />

coenzyme A (HMG-CoA) reductase, which is the rate-limiting<br />

enzyme of the mevalonate pathway. This pathway leads to<br />

formation of cholesterol and isoprenoids like FPP and GGPP.<br />

Treatment of plasma cell lines and ex vivo purified tumor cells<br />

from myeloma patients with lovastatin resulted in the induction<br />

of apoptosis. Co-treatment of cells with lovastatin and<br />

mevalonate or GGPP, but not FPP, abrogated lovastatin-induced<br />

apoptosis. Furthermore, induction of apoptosis and reduction of<br />

cell viability by inhibition of FTase were less pronounced when<br />

compared to inhibition of GGTase I. This implies that<br />

geranylgeranylation of proteins is critical for the regulation of<br />

survival of myeloma cells. Apoptosis triggered by inhibition of<br />

geranylgeranylation was associated with reduction of Mcl-1<br />

protein expression, which, in turn, resulted in the collapse of the<br />

mitochondrial transmembrane potential, cytochrome c release<br />

from mitochondria into the cytosol, and stimulation of caspase-3<br />

activity. Lovastatin enhanced the cytotoxic effects of<br />

dexamethasone and doxorubicin in a synergistic fashion. This<br />

synergism may be due to the lovastatin-mediated Mcl-1<br />

downregulation. These results show that geranylgeranylation of<br />

proteins is a key event in the regulation of myeloma tumor cell<br />

survival, probably through the regulation of Mcl-1 expression.<br />

Based on these findings, we started a Phase I/II trial to evaluate<br />

the combination of dose-escalating lovastatin and CHOP or VAD<br />

chemotherapy in refractory non-Hodgkin’s lymphoma and<br />

multiple myeloma patients. At the moment, 5 patients have been<br />

included in this trial.<br />

132<br />

Targeting the IGF-I Receptor Survival Pathways in<br />

Multiple Myeloma Using Selective IGF-I Receptor<br />

Tyrosine Kinase Inhibitors<br />

T. Strömberg, L. Dimberg, O. Larsson, M. Axelsson, K.<br />

Nilsson, H. Jernberg-Wiklund<br />

Department of Genetics and Pathology, Rudbeck Laboratory,<br />

Uppsala, Sweden; Cancer Center Karolinska, Karolinska Institute,<br />

Stockholm, Sweden<br />

The insulin-like growth factor I receptor (IGF-IR) is strongly<br />

suggested to play a key role in malignant transformation. In<br />

tumor cells from multiple myeloma (MM), IGF-I has been<br />

established as an important survival factor, and we and others<br />

have previously shown that a blocking anti-IGF-IR antibody<br />

(IR3) augments apoptosis induced by serum-starvation, Fas-<br />

S145

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

Saved successfully!

Ooh no, something went wrong!