Haematologica 2003 - Supplements

Haematologica 2003 - Supplements Haematologica 2003 - Supplements

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

P6.3 ENDOTHELIAL CELL-TUMOR CELL INTERACTIONS IN MULTIPLE MYELOMA. Ivan Van Riet 1 , Isabelle Vande Broek 1 , Kewal Asosingh 1 , Els Van Valckenborgh 1 , Liesbeth Hellebaut 1 , Xavier Leleu 2 , Thierry Facon 2 , Ben Van Camp 1 and Karin Vanderkerken 1 1 Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium; 2 Department of Hematology, Hopital Huriez, Lille, France. Endothelial cells (EC) represent, within the tumormicroenvironment, important interactive partners for multiple myeloma (MM) cells, since they are involved in different aspects of the paracrine network that underlies the pathogenesis of MM. Myeloma cells interact with bone marrow EC (BM-EC) during extravasation/homing and can directly activate EC resulting in neovascularization. Using the in vivo 5T2MM-mouse model our group previously demonstrated that the specific localization of myeloma cells in the bone marrow (BM) is a result of the combination of both a selective entry/adhesion and a selective survival/growth of the tumor cells in the BM (1). In the same murine model the selective entry in BM could be associated with a selective adhesion to BM-EC, involving CD44v10 (1, 2). After adhesion to BM-EC, myeloma cells will receive chemotactic signals that will stimulate their migration to the extravascular (medullar) compartment. We could demonstrate that the migration of both human and murine MM cells could be triggered by laminin-1 and MCP-1 (both produced by BM stromal cells, including EC). These migratory responses are mediated by the 67kD laminin-receptor (LR) and the CCR-2 chemokine-receptor, respectively (3, 4, 5). It was also found that 67kD LR can be upregulated in MM cells after contact with BM-EC and that this receptor is also involved in the bone marrow homing of 5T2MM cells in vivo (3). In order to cross bone marrow endothelium, MM cells also need to degrade the basement membrane. Using Matrigel ® invasion assays, we could demonstrate that human isolated (CD138 positive) MM cells are indeed invasive and that this invasive capacity could be enhanced by the presence of BM- EC. Moreover we found that the transendothelial invasion of MM cells involves MMP-9. In the 5T33MM model we could demonstrate an in vivo, bone marrow microenvironmentdependent, transcriptional upregulation of MMP-9 in the MM cells (6). In vitro experiments demonstrated that the production of this protease could be up-regulated in both murine (5T33MM) and human (CD138 positive) MM cells by interaction with BM- EC (6, 7). In human MM cells, this BM-EC-mediated upregulation of MMP-9 seems to involve hepatocyte growth factor (7). After extravasation, MM cells continue to interact with BM-EC, directly contributing to the formation of new blood vessels. We could demonstrate/confirm that murine (5T33MM) and human MM cells have angiogeneic-inducing potential and express different pro-angiogenic factors including VEGF-A, b-FGF and/or angiopoietin-1(8, 9). Moreover we found that the expression of some of these factors (VEGF and bFGF) in human MM cells is regulated by the BM-stroma. Comparing the functional activity of human MM-cell derived VEGF-A and bFGF in the in vitro proliferation and migration of EC, we found that VEGF plays a major but not exclusive role (9). Most recently we 3found that both human and murine MM cells also express transcripts for platelet-derived growth factor PDGF (A, B, C and D). This factor induces angiogeneic responses similar to VEGF, through interaction with receptor tyrosine kinases (RTK). We are currently investigating in the in vivo 5T33MM-mouse model the effect of a new potent RTK inhibitor, i.e. SU11657, that blocks the receptors for VEGF and PDGF. In conclusion, EC are involved in different aspects of tumor-host communication in MM and may therefore also represent, as important component of the stromal cell population, an interesting target for new therapeutical approaches. Future efforts, including the use of microarray analysis, should further clarify the molecular background of these MM cell-EC interactions K. Vanderkerken et al., Brit. J. Cancer, 82, 953-959, 2000. K. Asosingh et al., Cancer Research, 61, 2862-2865, 2001. I. Vande Broek et al., Brit. J. Cancer, 85, 1387-1395, 2001. K. Vanderkerken et al., Clin Exp Metastasis.;19, 87-90, 2002. I. Vande Broek et al., Brit. J. Cancer, in press, 2003. E. Van Valckenborgh et al., Int J Cancer. 101:512-8, 2002. I. Vande Broek et al., Blood, 100, 209a, 2002. E. Van Valckenborgh et al.,. Brit J Cancer, 86, 796-802, 2002. L. Hellebaut et al., submitted, 2003. P6.4 ANGIOGENIC ENDOTHELIAL CELLS WITHIN THE BONE MARROW OF MULTIPLE MYELOMA. A PHENOTYPIC AND FUNCTIONAL ASSESSMENT Angelo Vacca, Roberto Ria, Domenico Ribatti, Fabrizio Semeraro,Francesca Merchionne, Franco Dammacco Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, and Department of Human Anatomy and Histology, University of Bari Medical School, I-70124 Bari, Italy Endothelial cells of tumor vessels greatly differ from those of quiescent normal vessels due to their rapid proliferation which reflects enhanced angiogenesis associated with tumor progression (growth, invasion, metastasis) (1). They also differ in the profile and level of cell adhesion molecules because attachment to one another and to extracellular matrix during sprouting (that implies cell proliferation and migration) is remarkably reduced. Their survival is largely dependent on growth factors secreted by the tumor and its microenvironment (cells and matrix), and on their expression of specific receptors for these factors. Moreover, they are abnormal in shape and highly permeable due to fenestrae, vesicles, transcellular holes, widened intercellular junctions, a discontinuous basement membrane and scarce accessory stabilizing cells such as pericytes. They share the lining of new vessels with tumor cells able to mimic vessels. The fast growth of endothelial and tumor cells coupled with structural and functional abnormalities of endothelial cells make tumor vessels tortuous and dilated, with uneven diameter, excessive branching and shunts. Thus, tumor blood flow is chaotic and variable, and leads to hypoxic and acidic regions in the tumor that stimulate further angiogenesis. Bone marrow angiogenesis is mandatory for progression of multiple myeloma (MM) (2, 3), and is characterized by thin, tortuous and arborised vessels, and single or clustered endothelial cells (2). This process is sustained by vascular endothelial growth factor (VEGF) (4), basic fibroblast growth factor (bFGF), and matrix metalloproteinases (MMPs) (2) secreted by plasma cells. Besides their morphological picture, information on phenotype, function and structure of endothelial cells is circumstantial (2). In this presentation bone marrow endothelial cells of active MM are compared to normal quiescent endothelial cells in an attempt to add further information to the issue. Endothelial cells were extracted from bone marrow of 57 patients, and compared for the antigenic and genetic phenotype, functions and ultrastructural morphology with their normal S40

quiescent counterpart, the human umbilical vein endothelial cells (HUVEC). MM endothelial cells express highly: a) vascular endothelial growth factor receptor-2 (VEGFR-2) and tyrosin kinase with Ig and EGF homology-2 (Tie2/Tek), suggesting their engagement in vessel sprouting, i.e. in angiogenesis; b) CD34 and CD133 (AC133), suggesting recruitment of endothelial progenitor cells into an ancillary vascular network, i.e. into embryonic vasculogenesis; c) basic fibroblast growth factor receptor-2 (bFGFR-2) and bFGFR-3, suggesting that they are prone to this growth factor secreted by plasma cells and stromal cells; d) endoglin, a marker of tumor vessels; d) E-selectin and β3 molecules, suggesting more opportunities of interactions with plasma cells; e) aquaporin 1, suggesting hyperpermeability. On the contrary, they poorly express vascular-endothelial (VE)- cadherin, as angiosarcoma cells. Fluorescent activated cell sorting (FACS) analysis of some antigens shows their heterogeneous expression, suggesting well defined subpopulations of cells. The main genetic markers are Tie-2/Tek, VEGFs, bFGFs and the corresponding receptors. MM endothelial cells rapidly form a close capillary network in vitro (matrigel assay), and generate on their turn numerous new vessels in vivo (chick embryo chorioallantoic membrane [CAM] assay). They secrete VEGF, bFGF, metalloproteinase-2 (MMP-2) and MMP-9, that are growth and invasive factors both for themselves and plasma cells. Ultrastructurally, they show vescicles, fenestrae, and hyperplasia of endoplasmic reticulum that are absent in HUVEC. Thalidomide interferes with their proliferative activity and capillarogenesis on matrigel. Our data suggest that both embryonic vasculogenesis and angiogenesis concur to the formation of vascular tree of MM bone marrow and disease progression. Because of the heterogeneous antigenic phenotype, a mixture (or sequence) of antiangiogenic agents coupled with thalidomide is envisaged as a possibler biologic therapy (5) of MM. Holmgren L, O'Reilly MS, Folkman J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1995;1:149-153. Vacca A, Ribatti D, Roncali L, Ranieri G, Serio G, Silvestris F, Dammacco F. Bone marrow angiogenesis and progression in multiple myeloma. Br J Haematol 1994;87:503-508. Rajkumar SV, Mesa RA, Fonseca R, Schroeder G, Plevak MF, Dispenzieri A, Lacy MQ, Lust JA, Witzig TE, Gertz MA, Kyle RA, Russel SJ, Greipp PR. Bone marrow angiogenesis in 400 patients with monoclonal gammopathy of undetermined significance, multiple myeloma, and primary amyloidosis. Clin Cancer Res 2002;8:2210-2216. Bellamy WT, Richter L, Frutiger Y, Grogan TM. Expression of vascular endothelial growth factor and its receptors in hematopoietic malignancies. Cancer Res 1999;59:728-733. Hideshima T and Anderson KC. Molecular mechanisms of novel therapeutic approaches for multiple myeloma. Nat Rev Cancer 2002;2:927-937. P6.5 GENOMIC AND PROTEOMIC CHANGES FOLLOWING MM CELL-MICROENVIRONMENTAL INTERACTION Constantine S. Mitsiades1,2, Nicholas S. Mitsiades1,2, Ciaran McMullan1,2, Galinos Fanourakis1,2, Reshma Shringarpure1,2, Nikhil C. Munshi1,2, Towia Liberman3, Kenneth C. Anderson1,2. 1. Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; 2. Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; 3. Harvard Institutes of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA. The response of multiple myeloma (MM) patients to conventional therapies is significantly affected by interactions of MM tumor cells with their local bone marrow (BM) microenvironment, including biologic sequelae induced by BM-derived cytokines, and adhesion to extracellular matrix proteins and BM stromal cells (BMSCs). Indeed, these interactions can confer protection to MM cells against pro-apoptotic therapies (e.g. dexamethasone or cytotoxic chemotherapy), with adverse implications for patient outcome. The need to develop rational strategies to target and abrogate this microenvironment-derived drug-resistance of MM cells has provided the impetus for comprehensive profiling of the molecular sequelae triggered by exposure of MM cells to microenvironmental stimuli, such as BM-derived cytokines (such as IL-6 and insulin-like growth factors (IGFs)) or co-culture with BMSCs. IL-6 is known for its role as a growth/survival factor for MM cells and an important regulator of osteoclastogenesis, while the MM-BMSCs interaction is known to trigger NF-êB-mediated IL-6 secretion by BMSCs. The major emphasis on IGFs is warranted by our recent studies showing that IGFs not only stimulate MM cell proliferation, survival and attenuated response to apoptosis-inducing agents (e.g. Dex or Apo2L/TRAIL), but are also expressed at high levels in serum of MM patients (endocrine IGF), as well as locally in the BM microenvironment by autocrine (MM cells) and paracrine (including BMSCs and osteoblsts) sources (CS Mitsiades et al. Blood 2002; 100, 170a). Importantly, we have recently shown that IGF-1 receptor (IGF- 1R/CD221) is expressed on all MM cell lines and patients cells tested and that its inhibition by several different strategies (including neutralizing antibodies, inhibitory peptides or small molecule Tyr kinase inhibitors) significantly suppresses MM cell proliferation, survival and resistance to other drugs, both in vitro and in vivo (CS Mitsiades et al. Blood 2002; 100, 170a). To characterize the molecular sequelae triggered by these microenvironemtal interactions of MM cells with their BM milieu, we performed gene expression profiling, using U133A Affymetrix oligonucleotide microarrays, and proteomic analyses of the signaling state of MM cells, using multiplex immunoblotting arrays, as recently described (N. Mitsiades et al. Blood 2003;101(6):2377 and CS Mitsiades et al. Semin Oncol, in press). These studies involved ex vivo stimulation of MM cells with pathophysiologically-relevant concentrations of IGF-1, IGF- 2 and IL-6, as well as incubation of MM cells in an ex vivo model of co-culture with BMSCs. In this model, MM-1S cells stably transfected with a construct for Green fluorescent protein (GFP) were co-cultured with BMSCs: the 2 cellular compartments were subsequently sorted by fluorescence activated cell sorting (FACS) on the basis of the GFP+ status of MM cells vs GFP- of BMSCs (thereby minimizing any potential background signaling and transcriptional changes that may be induced during mAb-based positive selection and maximizing the post-sort yield of tumor cells). Molecular profiles of co-cultured cells were compared with their respective profiles in isolated cultures, as well as with profiles generated by co-culture in the setting of treatment with novel anti-MM agents such as proteasome inhibitor PS-341, hsp90 inhibitor 17-AAG, histone deacetylase inhibitor SAHA, and anti- IGF-1R inhibitor. Analyses of these gene expression and proteomic data (using hierarchical clustering, functional clustering and relevenace networks algorithms, as well as subsequent confirmatory and mechanistic assays) showed that the distinct molecular signatures of MM cells treated with cytokine or co-cultured with BMSCs also feature overlapping patterns of activation of proliferative /anti-apoptotic signaling events. Indeed, BM-derived cytokines and co-culture with BMSCs triggered activation of PI-3K/Akt and Raf/MAPK signaling pathways in MM cells; upregulated the S41

P6.3<br />

ENDOTHELIAL CELL-TUMOR CELL INTERACTIONS IN<br />

MULTIPLE MYELOMA.<br />

Ivan Van Riet 1 , Isabelle Vande Broek 1 , Kewal Asosingh 1 ,<br />

Els Van Valckenborgh 1 , Liesbeth Hellebaut 1 , Xavier Leleu 2 ,<br />

Thierry Facon 2 , Ben Van Camp 1 and Karin Vanderkerken 1<br />

1<br />

Department of Hematology and Immunology, Vrije Universiteit<br />

Brussel (VUB), Brussels, Belgium; 2 Department of Hematology,<br />

Hopital Huriez, Lille, France.<br />

Endothelial cells (EC) represent, within the tumormicroenvironment,<br />

important interactive partners for multiple<br />

myeloma (MM) cells, since they are involved in different aspects<br />

of the paracrine network that underlies the pathogenesis of MM.<br />

Myeloma cells interact with bone marrow EC (BM-EC) during<br />

extravasation/homing and can directly activate EC resulting in<br />

neovascularization. Using the in vivo 5T2MM-mouse model our<br />

group previously demonstrated that the specific localization of<br />

myeloma cells in the bone marrow (BM) is a result of the<br />

combination of both a selective entry/adhesion and a selective<br />

survival/growth of the tumor cells in the BM (1). In the same<br />

murine model the selective entry in BM could be associated with<br />

a selective adhesion to BM-EC, involving CD44v10 (1, 2). After<br />

adhesion to BM-EC, myeloma cells will receive chemotactic<br />

signals that will stimulate their migration to the extravascular<br />

(medullar) compartment. We could demonstrate that the<br />

migration of both human and murine MM cells could be triggered<br />

by laminin-1 and MCP-1 (both produced by BM stromal cells,<br />

including EC). These migratory responses are mediated by the<br />

67kD laminin-receptor (LR) and the CCR-2 chemokine-receptor,<br />

respectively (3, 4, 5). It was also found that 67kD LR can be upregulated<br />

in MM cells after contact with BM-EC and that this<br />

receptor is also involved in the bone marrow homing of 5T2MM<br />

cells in vivo (3). In order to cross bone marrow endothelium, MM<br />

cells also need to degrade the basement membrane. Using<br />

Matrigel ® invasion assays, we could demonstrate that human<br />

isolated (CD138 positive) MM cells are indeed invasive and that<br />

this invasive capacity could be enhanced by the presence of BM-<br />

EC. Moreover we found that the transendothelial invasion of MM<br />

cells involves MMP-9. In the 5T33MM model we could<br />

demonstrate an in vivo, bone marrow microenvironmentdependent,<br />

transcriptional upregulation of MMP-9 in the MM<br />

cells (6). In vitro experiments demonstrated that the production of<br />

this protease could be up-regulated in both murine (5T33MM)<br />

and human (CD138 positive) MM cells by interaction with BM-<br />

EC (6, 7). In human MM cells, this BM-EC-mediated upregulation<br />

of MMP-9 seems to involve hepatocyte growth factor<br />

(7).<br />

After extravasation, MM cells continue to interact with BM-EC,<br />

directly contributing to the formation of new blood vessels. We<br />

could demonstrate/confirm that murine (5T33MM) and human<br />

MM cells have angiogeneic-inducing potential and express<br />

different pro-angiogenic factors including VEGF-A, b-FGF<br />

and/or angiopoietin-1(8, 9). Moreover we found that the<br />

expression of some of these factors (VEGF and bFGF) in human<br />

MM cells is regulated by the BM-stroma. Comparing the<br />

functional activity of human MM-cell derived VEGF-A and<br />

bFGF in the in vitro proliferation and migration of EC, we found<br />

that VEGF plays a major but not exclusive role (9). Most recently<br />

we 3found that both human and murine MM cells also express<br />

transcripts for platelet-derived growth factor PDGF (A, B, C and<br />

D). This factor induces angiogeneic responses similar to VEGF,<br />

through interaction with receptor tyrosine kinases (RTK). We are<br />

currently investigating in the in vivo 5T33MM-mouse model the<br />

effect of a new potent RTK inhibitor, i.e. SU11657, that blocks<br />

the receptors for VEGF and PDGF.<br />

In conclusion, EC are involved in different aspects of tumor-host<br />

communication in MM and may therefore also represent, as<br />

important component of the stromal cell population, an<br />

interesting target for new therapeutical approaches. Future<br />

efforts, including the use of microarray analysis, should further<br />

clarify the molecular background of these MM cell-EC<br />

interactions<br />

K. Vanderkerken et al., Brit. J. Cancer, 82, 953-959, 2000.<br />

K. Asosingh et al., Cancer Research, 61, 2862-2865, 2001.<br />

I. Vande Broek et al., Brit. J. Cancer, 85, 1387-1395, 2001.<br />

K. Vanderkerken et al., Clin Exp Metastasis.;19, 87-90, 2002.<br />

I. Vande Broek et al., Brit. J. Cancer, in press, <strong>2003</strong>.<br />

E. Van Valckenborgh et al., Int J Cancer. 101:512-8, 2002.<br />

I. Vande Broek et al., Blood, 100, 209a, 2002.<br />

E. Van Valckenborgh et al.,. Brit J Cancer, 86, 796-802, 2002.<br />

L. Hellebaut et al., submitted, <strong>2003</strong>.<br />

P6.4<br />

ANGIOGENIC ENDOTHELIAL CELLS WITHIN THE<br />

BONE MARROW OF MULTIPLE MYELOMA. A<br />

PHENOTYPIC AND FUNCTIONAL ASSESSMENT<br />

Angelo Vacca, Roberto Ria, Domenico Ribatti, Fabrizio<br />

Semeraro,Francesca Merchionne, Franco Dammacco<br />

Department of Biomedical Sciences and Human Oncology, Section<br />

of Internal Medicine and Clinical Oncology, and Department of<br />

Human Anatomy and Histology, University of Bari Medical School,<br />

I-70124 Bari, Italy<br />

Endothelial cells of tumor vessels greatly differ from those of<br />

quiescent normal vessels due to their rapid proliferation which<br />

reflects enhanced angiogenesis associated with tumor progression<br />

(growth, invasion, metastasis) (1). They also differ in the profile<br />

and level of cell adhesion molecules because attachment to one<br />

another and to extracellular matrix during sprouting (that implies<br />

cell proliferation and migration) is remarkably reduced. Their<br />

survival is largely dependent on growth factors secreted by the<br />

tumor and its microenvironment (cells and matrix), and on their<br />

expression of specific receptors for these factors. Moreover, they<br />

are abnormal in shape and highly permeable due to fenestrae,<br />

vesicles, transcellular holes, widened intercellular junctions, a<br />

discontinuous basement membrane and scarce accessory<br />

stabilizing cells such as pericytes. They share the lining of new<br />

vessels with tumor cells able to mimic vessels. The fast growth of<br />

endothelial and tumor cells coupled with structural and functional<br />

abnormalities of endothelial cells make tumor vessels tortuous<br />

and dilated, with uneven diameter, excessive branching and<br />

shunts. Thus, tumor blood flow is chaotic and variable, and leads<br />

to hypoxic and acidic regions in the tumor that stimulate further<br />

angiogenesis.<br />

Bone marrow angiogenesis is mandatory for progression of<br />

multiple myeloma (MM) (2, 3), and is characterized by thin,<br />

tortuous and arborised vessels, and single or clustered endothelial<br />

cells (2). This process is sustained by vascular endothelial growth<br />

factor (VEGF) (4), basic fibroblast growth factor (bFGF), and<br />

matrix metalloproteinases (MMPs) (2) secreted by plasma cells.<br />

Besides their morphological picture, information on phenotype,<br />

function and structure of endothelial cells is circumstantial (2). In<br />

this presentation bone marrow endothelial cells of active MM are<br />

compared to normal quiescent endothelial cells in an attempt to<br />

add further information to the issue.<br />

Endothelial cells were extracted from bone marrow of 57<br />

patients, and compared for the antigenic and genetic phenotype,<br />

functions and ultrastructural morphology with their normal<br />

S40

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

Saved successfully!

Ooh no, something went wrong!