GET THE APP

Multiple Myeloma: The Role of Angiogenesis in Disease Progression
Journal of Bone Research

Journal of Bone Research
Open Access

ISSN: 2572-4916

+44 1478 350008

Research Article - (2013) Volume 1, Issue 2

Multiple Myeloma: The Role of Angiogenesis in Disease Progression

Roberto Ria*, Simona Berardi, Antonia Reale, Annunziata De Luisi, Ivana Catacchio, Vito Racanelli and Angelo Vacca
Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy
*Corresponding Author: Dr. Roberto Ria, Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Policlinico - Piazza Giulio Cesare, 11, I-70124 BARI, Italy, Tel: +39-080-559-31-06, Fax: +39-080-547.88.59 Email:

Abstract

Angiogenesis, the formation of new blood vessels from pre-existing ones, plays an important role in the biology of multiple myeloma and has a prognostic value in this disease. Multiple myeloma is a plasma cell malignancy that home to and expand in the bone marrow where actively interacts with stromal cells inducing neovascularization, a constant hallmark of disease progression. Myeloma-induced angiogenesis involves either the direct production of angiogenic molecules by myeloma cells and the recruitment and activation of bone marrow stromal cells. Indeed, the angiogenic factors released in the bone marrow microenvironment by multiple myeloma plasma cells stimulate stromal cells to secrete their own angiogenic factors and induce the acquisition of a phenotypic and functional adaptation by non-endothelial cells, such as macrophages, which contribute to the completion of the neovessel wall (vasculogenic mimicry). In this review we summarize recent data which give strong evidence for an increased angiogenic activity in the bone marrow microenvironment and support the hypothesis that angiogenesis is not only important for tumour growth but may also promote plasma cell growth in multiple myeloma.

<

Keywords: Multiple myeloma; Angiogenesis; Bone marrow microenvironment; Tumor progression; Vascular endothelial growth factor

Introduction

Multiple Myeloma (MM) is a malignancy of immunoglobulin (Ig)-synthesizing plasma cells, that home to and expand in the bone marrow [1]. Although presenting with the same histological features, MM is characterised by a high genomic heterogeneity [2,3]. There is a growing awareness that the interactions between MM plasma cells, stromal cells, hematopoietic cells, and the extracellular matrix (ECM) is as important as the genetic changes in the disease progression. Pathophysiological interactions of myeloma cells within the bone marrow microenvironment are highlighted by the progressionassociated bone disease and neovascularization, and are witnessed by autocrine/paracrine circuits that activate multiple signalling pathways and affect the most important aspects of the malignant phenotype, i.e., apoptosis/survival, proliferation, invasion, bone damage and angiogenesis [3,4].

Neovascularization, the formation of new vessels, represents one of the principal aspects of these interactions and a constant hallmark of disease progression [5].

This process is supported by angiogenic factors such as Vascular Endothelial Growth Factor (VEGF), Fibroblast Growth Factor-2 (FGF-2) and Hepatocyte Growth Factor (HGF) which are directly secreted by the tumour plasma cells and the stromal cells.

Angiogenesis in multiple myeloma progression

Angiogenesis is an essential event for progression disease (growth, invasion and metastasis) in solid and haematological tumours, including MM, and has a prognostic value [6]. It is a tightly regulated multistep process [7] which begins with the activation of resting endothelial cells, continues with the degradation of the ECM, proliferation and migration of endothelial cells toward the angiogenic stimulus, and ends with the constitution of new blood vessels enveloped by a basement membrane [8].

Angiogenesis is uncontrolled and unlimited in time, and essential for tumour growth, invasion and metastasis during the transition from the avascular to the vascular phase [9]. Angiogenesis occurs in step with the transition from Monoclonal Gammopathy of Undetermined Significance (MGUS) to non active MM (complete response, plateau phase) and to active MM. Vacca et al. [10] showed that angiogenesis was increased in BM sections of patients with active MM disease. They reported an increase in BM microvessel density (MVD), a marker for angiogenesis, in MM patients, especially in advanced disease, compared with MGUS patients, which suggests a vascular phase during active MM and a pre-vascular phase in non active MM and MGUS [11]. Rajkumar et al. [12] demonstrated in a large cohort of 400 patients that a progressive increase in bone marrow-MVD occurs across the whole spectrum of plasma cells disorders, including primary amyloidosis, MGUS, smoldering and active myeloma, which added further evidence to the hypothesis that angiogenesis is related to malignant plasma cells growth [12,13].

The pro-angiogenic bone marrow microenvironment

Recent advances in myeloma biology demonstrated the emerging role of the bone marrow microenvironment in promoting proliferation and survival of the malignant plasma cell clone [14]. Bone marrow angiogenesis is regulated by soluble pro- and anti-angiogenic factors which mediate the paracrine interactions between myeloma cells, endothelial cells and bone marrow stromal cells (BMSC) [15].

The increased angiogenic potential of active myeloma was first demonstrated by Vacca et al. who reported that serum-free conditioned media from bone marrow plasma cells of patients with active MM had a significantly higher angiogenic activity in vitro (matrigel capillarogenesis assay) and in vivo (chick chorioallantoic membrane assay) than samples from patients with non-active MM or MGUS [16]. Bone marrow plasma extracts contained higher levels of FGF-2 in samples from active patients versus those in non-active MM or MGUS. Moreover, the in vitro and in vivo angiogenic activity of the conditioned media could be reduced by FGF-2 antibodies [16]. A variety of growth factors, including VEGF, Hepatocyte Growth Factor (HGF), Plateletderived Growth Factor (PDGF), and Tumor Necrosis Factor-Alpha (TNF-alpha), are known to promote angiogenesis [17].

Tumour cells and inflammatory cells, such as mast cells and macrophages, release these factors which stimulate resting endothelial cells by means of cognate specific tyrosine-kinase receptors [18,19]. Angiogenic cytokines may represent a potential indicator of response to therapy in solid and hematologic tumors [20,21]. Receptor/cytokine binding activates endothelial cells and increases cell proliferation, modifies cell adhesion proteins and increases secretion of proteolytic enzymes, cell migration and invasion [22].

VEGF stimulates proliferation and chemotaxis in both endothelial cells and stromal cells [23]. Moreover, VEGF acts as an autocrine inducer of growth and chemotaxis via VEGFR-1 [24]. It increases IL-6 (a major growth and survival factor for MM plasma cells) production by bone marrow stromal cells via VEGFR-2 and thus forming a paracrine loop for tumor growth and angiogenesis [25]. Also, adhesion of plasma cells to bone marrow stromal cells increases VEGF secretion by both cell types enhancing angiogenesis [26]. VEGF production by plasma cells is also regulated by TNF-α of bone marrow stromal cells [27]. TNF-α mediates the upregulation of adhesion molecules of plasma cells and bone marrow stromal cells, and thus enhances heterotypic adhesion and activates IL-6 and VEGF secretion by bone marrow stromal cells [28]. This mechanism mediates MM cell homing and migration, as well as angiogenesis [29,30].

VEGF signalling also contributes to inhibit antiangiogenic signals such as Semaphorin3A (SEMA3A) whose autocrine loops usually activates to self-limit the physiologic angiogenesis [31].

FGF-2 is another important angiogenic growth factor, and it represent a potent activator of endothelial proliferation and can thus stimulate angiogenesis, promote stromal fibroblast proliferation and extracellular matrix formation leading to excessive bone marrow fibrosis and can directly affect neoplastic cells by acting on their high affinity to FGFRs [32].

FGF-2 increases IL-6 secretion; conversely IL-6 enhances FGF-2 expression and secretion by MM plasma cells, thus forming a paracrine IL-6/FGF-2 cross-talk between MM plasma cells and bone marrow stromal cells that triggers neovascularisation as well as MM cell growth and survival [33].

Endothelial cells

All the angiogenic stimuli present in the bone marrow microenvironment act on endothelial cells causing a series of modifications that in turn remain stabilized and irreversible. In fact, tumor endothelial cells differ greatly from those of quiescent healthy vessels [34]. They proliferate rapidly in keeping with the enhanced angiogenesis that accompanies tumor progression [35].

Their intercellular adhesion and to the ECM during sprouting (that implies cell proliferation and migration) is greatly reduced since they have different profile and level of cell adhesion molecules [36]. Their survival is markedly dependent on growth factors secreted by the tumor and its microenvironment, and on their expression of specific receptors for these factors [37]. They are abnormal in shape and highly permeable due to the presence of fenestrae, vesicles, transcellular holes, widened intercellular junctions, and a discontinuous basement membrane. They share the lining of new vessels with tumor cells able to mimic vessels [38,39].

The fast growth of endothelial and tumour cells, coupled with their structural and functional abnormalities, make tumour vessels thin, tortuous, and arborized.

As a consequence, tumor blood flow is chaotic and variable and leads to hypoxic and acidic environment that stimulate further angiogenesis [40].

MM endothelial cells intensely express markers of vivid angiogenesis such as VEGFR-2 and Tie/Tek. This implies synergistic activity of VEGF and Ang-2, produced by plasma cells, in the induction of sprouts from existing vessels [34]. MM endothelial cells sizably express CD133, a marker of the progenitor endothelial cells involved in pre-natal vasculogenesis [41]. It has been proved that some CD133+ hematopoietic stem and progenitor cells contribute to the formation of the vessel wall of newly forming blood vessels together with FVIIIRA+, VEGFR-2+, and VE-cadherin+ MM endothelial cells [42]. MM plasma cells and inflammatory cells secrete high levels of VEGF, FGF-2, and Insulin-like Growth Factor (IGF), which recruit bone marrow and circulating hematopoietic stem and progenitor cells into the tumour microenvironment, where they differentiate into MM endothelial cells and participate to the formation of the new vessel wall. High expression of β3-integrin, which prevents apoptosis of endothelial cells and favours their adhesion to the ECM, proliferation, migration, and capillarogenesis, also implies vivid neovascularisation [43,44]. Overexpression of endoglin by endothelial cells, that enhances the expression of the adhesion molecule CD31, which is the ligand of the plasma cell CD38, suggests enhanced opportunities for plasma cells to interact with the new-formed blood vessels enter circulation and disseminate [34].

Frequent interactions between plasma cells and new-formed blood vessels are also mediated by the high expression of E-selectin by endothelial cells [45]. Moreover, MM endothelial cells intensely express a water transporter, namely Aquaporin-1, which enhances vascular permeability, facilitates plasma extravasation, increases interstitial pressure, induces hypoxia, and upregulates Hypoxia Inducible Factor-1 Alpha (HIF-1α) and VEGF [46].

A paracrine loop for tumour angiogenesis (figure 1) and growth has been demonstrated in MM patients, mediated by VEGF-A and FGF-2 [47,48]. Plasma cells secrete VEGF-A and this induces endothelial cell proliferation and chemotaxis through VEGFR-2, prevalently expressed on these cells, which display constitutive autophosphorylation of VEGFR-2 and the associated kinase ERK-2 [24,49].

Another important role is played by the paracrine loop existing between MM endothelial cells and plasma cells involving CXCchemokines and their cognate receptors, which mediate plasma cell proliferation and chemotaxis [50].

Bone marrow endothelial cells express and secrete high amounts of the CXC-chemokines CXCL8/IL-8, CXCL11/interferon-inducible T-cell alpha chemoattractant (I-TAC), CXCL12/stromal cell-derived factor (SDF)-1α, and CCL2/monocyte chemotactic protein (MPC)-1 [50].

Several MM cell lines display a complex expression pattern of chemokine receptors (CXCR, CCR), some of which also mediate the interactions between plasma cells and stromal cells in the bone marrow microenvironment [51].

To summarize, MM endothelial cells show constitutively ultrastructural features of enhanced metabolic activation, an high expression of typical endothelial markers (Tie2/Tek, VEGFR-2, FGFR- 2, CD105-endoglin, and VE-cadherin), an high secretion of matrix metalloproteinases-2 and -9, and up-regulation of angiogenic genes (VEGF, FGF-2, Gro-α chemokine, transforming growth factor beta, Tie2/Tek, HIF-1α, ETS-1, and osteopontin) [34].

Vasculogenic Mimicry in Mm

In some aggressive tumours the vessel wall is lined with only cancer cells as a mosaic of cancer cells and endothelial cells. This phenomenon is called “vasculogenesis mimicry” [52].

In healthy subjects, cells of monocyte lineage (other mesodermalderived cells) can generate endothelial cell progenitors or act as pluripotent stem cells [41,53,54]. They can develop an endothelial cell phenotype, especially when stimulated by VEGF and/or bFGF, and produce a functional capillary-like mesh permeable by blood cells, hence recapitulating embryo vasculogenesis [54-57].

Bone marrow monocytes and macrophages of MM patients can be induced to assume a number of endothelial cell properties and form capillary-like structures in vitro through vasculogenesis. Moreover, macrophages contribute to build neovessels in MM through vasculogenic mimicry, and in MGUS they are prone to a vascular switch that marches in step with the progression toward MM [58].

In fact, MM bone marrow macrophages exposed to VEGF and bFGF develop a number of phenotypic properties similar to those of paired bone marrow endothelial cells, and form capillary-like structures morphologically mimicking those produced by MM endothelial cells. At the ultrastructural level, MM macrophages exhibit numerous cytoplasmic extroversions arranged in tube-like structures [58].

All these features are lacking or minimal in macrophages of patients with MGUS or with benign anaemia which, however, will become phenotypically and functionally similar to those of MM under angiogenic stimulation [58].

Bone marrow biopsies of MM, but not of MGUS, harbour ‘mosaic’ vessels since these are formed by MM endothelial cells, endothelial celllike macrophages and macrophages themselves [58].

Antiangiogenic strategies

The actual therapeutic strategies of MM consist of conventional chemotherapy in combination with biological-based therapies in various settings, targeting not only the MM plasma cells but also its microenvironment and new therapeutic targets are currently available [59].

The proteasome inhibitor bortezomib (Velcade, formerly PS-341), a boronic acid dipeptide, is a potent, highly selective, and reversible proteasome inhibitor that targets 26S proteasome complex and inhibits its function [60]. The 26S proteasome is an ATP-dependent multicatalytic protease mediating intracellular protein degradation [61].

Proteasomal degradation of misfolded or damaged proteins proceeds by recognition of polyubiquitinated proteins by the 19S regulatory subunit of the 26S protease and subsequently hydrolysis to small polypeptides [61].

Besides eliminating damaged/misfolded proteins, the proteasome also regulates key cellular processes, including modulation of transcription factors, such as NF-κB, cell cycle progression, inflammation, immune surveillance, growth arrest, and apoptosis [62].

Bortezomib has inhibitory effects on the NF-κB activity in MM cells. NF-κB is a major transcriptional factor which mediates expression of many protein including cytokines, chemokines, cell adhesion molecules, as well as those involved in anti-apoptosis and cellular growth control [62]. Its activity is regulated by association with IκB family proteins [63]. Varius stimuli, including cytokines, such as TNFα and IL-1β, trigger phosphorylation of IκB protein by IκB kinase [64]. Phosphorylated IκB is subsequently polyubiquitinated by specific enzymes and degradated by the 26S proteasome, which allows p50/p65 NF-κB nuclear translocation and binding to consensus mtifits in the promoter region of target genes [62,64].

Expression of adhesion molecules, such as ICAM-1 and VCAM-1, on both MM cells and bone marrow stromal cells are also regulated by NF-κB [65,66]. Thus inhibition of NF-κB by bortezomib downregulates these adhesion molecules, thereby enhancing susceptibility of MM cells to therapeutic agents in the context of the bone marrow milieu [62]. Another important aspect is that induction of IL-6 transcription and secretion by bone marrow stromal cells is mediated via NF-κB activation which, in turn, increases secretion of other cytokines, such as VEGF, from MM plasma cells [67].

Furthermore, MM cell adherence to bone marrow stromal cells triggers IL-6 secretion via NF-κB activation, associated with an increased MM cell growth and leads to a reduction of VEGF secretion. Bortezomib significantly blocks the MM cell adhesion induced by IL-6 secretion from bone marrow stromal cells [62].

Bortezomib is also directly cytotoxic, triggering stress response and apoptotic signalling via multiple pathways [62].

As the result of inhibition of proteasome activity, it causes the accumulation of misfolded polyubiquinated proteins, resulting in endoplasmic reticulum stress which triggers caspase-4 and downstream signalling [68]. Bortezomib also induces ROS which play a critical role in the initiation of the apoptotic cascades by disruption of membrane potential and the release of cytochrome c from mitochondria, followed by caspase-9 activation [69]. Proteasome inhibitors have a potent activity against mitotic endothelial cells, so they target aberrant blood vessel development associated with tumor growth, in fact, bortezomib inhibits the proliferation of MM endothelial cells associated with downregulation of VEGF, IL-6, IGF-I, Ang-1 and Ang-2 [62]. Moreover, bortezomib inhibits DNA repair activity by cleavage of DNA dependent protein kinase catalytic subunit (DNA-PKcs), thereby restoring sensitivity to DNA-damaging chemotherapeutic agents, such as doxorubicin and melphalan [70].

Bortezomib also down-regulates caveolin-1 tyrosine phosphorylation, which is required for VEGF-mediated MM cell migration, and also blocks the caveolin-1 phosphorylation induced by VEGF (transcriptional target of NF-κB) in endothelial cells, thereby inhibiting ERK-dependent cell proliferation. It inhibits the transcription of important adhesion molecules such as ICAM-1, VCAM1 and E-selectin [71].

All these biological activity are responsible of the higher and rapid therapeutic efficacy of this drug as well as of its potential persistence of antiangiogenic activity after the term of patients treatment [72,73].

Thalidomide has a direct tumoricidal activity, an antiangiogenic effect by modulation of various angiogenic genes [74].

It modulates TNF-α signalling through direct and/or indirect effects on the tumour microenvironment, reduces FGF-2, VEGF and IL-6 secretion in bone marrow stromal cells and by MM cells [75-77]. It also stimulates the activation and expansion of T cells and augments NK-cell –mediated cytotoxicity through its direct effect on T cells with a consequent increase in IL-2 and interferon gamma (IFN-γ) secretion, and interferes with NF-κB activity by blocking its ability to bind to DNA or suppresses IκB kinase activity, thus abrogating normal inflammatory cytokine production [78,79]. Thalidomide also disrupts the host marrow-MM cell interaction by selective modulation of the density of cell surface adhesion molecules [80].

Treatment with thalidomide is associated with sedation, fatigue, constipation, rash, deep-vein thrombosis, and peripheral neuropathy [81].

Lenalidomide, a derivative of thalidomide, is less toxic and more potent than the parent drug [82]. In patients with relapsed or refractory MM, lenalidomide can overcome resistance not only to conventional chemotherapy but also to thalidomide [83,84].

The bisphosphonates are other compounds that, although originally used to reduce bone loss in MM due to an anti-osteoclast activity, have also been shown to have a direct effect on MM cells.

In fact, zoledronic acid has a direct cytotoxic activity on tumor cells and suppresses angiogenesis, inhibits FGF-2- and VEGF-dependent proliferation of endothelial cells and inhibits VEGFR-2 in an autocrine loop [83,84]. Neridronate exerts its antiangiogenic activity through both a direct effect on endothelial cell proliferative activity and inhibitory effect on the responsivity of the endothelial cells to the proliferative stimuli mediated by angiogenic cytokines [85].

Conclusion

The bone marrow microenvironment plays a crucial role in the pathophysiology of MM. It is involved in the crosstalk between plasma cells and bone marrow stromal cells, which increases the survival, proliferation and migration of tumor cells themselves, and represents the substrate for angiogenesis which favour to disease progression. Due to interaction with active microenvironment, MM plasma cells also acquire drug resistance giving less opportunity to therapy response.

Many research studies have tried to better understand the biological mechanisms and the genetic basis of all the interactions between MM cells and bone marrow stromal cells. VEGF, FGF-2, IL-6, macrophages, mast cells, and many others cells and molecules, play the most important role in this process.

All these observations are indicative that: i) angiogenic stimuli provided by VEGF, FGF-2 and IGF release in the bone marrow microenvironment of MM patients by myeloma plasma cells are sufficient to recruit haematopoietic stem cells and macrophages to the tumor bed and induce their differentiation into endothelial cells contributing to the tumor vasculature; ii) haematopoietic stem cells and macrophages may be a source of endothelial cells in the bone marrow of MM patients during disease progression; iii) vasculogenic mimicry, contributing together with angiogenesis in MM, is induced by cytokines secreted by myeloma plasma cells.

Several studies have focused their investigation on novel drugs targeting the MM plasma cells and the microenvironment cells. Good results have been already done but MM still remain an incurable malignancy, indicating that the role of bone marrow microenvironment is important in MM progression, but its role is still not completely clear.

The goal for MM therapy remain the simultaneous block of plasma cell proliferation and survival, plasma cells/bone marrow stromal cells interaction, and bone marrow stromal cells activity by the combination of biological target drugs.

Funding

This work was supported by Associazione Italiana per la Ricerca sul Cancro (AIRC), Investigator Grant and Special Program Molecular Clinical Oncology 5 per mille n. 9965, Milan, Italy.

Acknowledgements

The authors would like to thank Prof. C. Perillo for editing the English version of this manuscript.

References

  1. Dimopoulos MA, Kastritis E, Anagnostopoulos A (2006) Hematological malignancies: myeloma. Ann Oncol 17: x137-143.
  2. Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, et al. (2004) Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 64: 1546-1558.
  3. Zhan F, Huang Y, Colla S, Stewart JP, Hanamura I, et al. (2006) The molecular classification of multiple myeloma. Blood 108: 2020-2028.
  4. Bommert K, Bargou RC, Stühmer T (2006) Signalling and survival pathways in multiple myeloma. Eur J Cancer 42: 1574-1580.
  5. Ribatti D, Vacca A (2008) The role of microenvironment in tumor angiogenesis. Genes Nutr 3: 29-34.
  6. Folkman J (2001) Angiogenesis-dependent diseases. Semin Oncol 28: 536-542.
  7. Stasi R, Amadori S (2002) The role of angiogenesis in hematologic malignancies. J Hematother Stem Cell Res 11: 49-68.
  8. Carmeliet P (2000) Mechanisms of angiogenesis and arteriogenesis. Nat Med 6: 389-395.
  9. Vacca A, Ribatti D (2006) Bone marrow angiogenesis in multiple myeloma. Leukemia 20: 193-199.
  10. Vacca A, Ribatti D, Roncali L, Ranieri G, Serio G, et al. (1994) Bone marrow angiogenesis and progression in multiple myeloma. Br J Haematol 87: 503-508.
  11. Asosingh K, De Raeve H, Menu E, Van Riet I, Van Marck E, et al. (2004) Angiogenic switch during 5T2MM murine myeloma tumorigenesis: role of CD45 heterogeneity. Blood 103: 3131-3137.
  12. Rajkumar SV, Mesa RA, Fonseca R, Schroeder G, Plevak MF, et al. (2002) Bone marrow angiogenesis in 400 patients with monoclonal gammopathy of undetermined significance, multiple myeloma, and primary amyloidosis. Clin Cancer Res 8: 2210-2216.
  13. Niemöller K, Jakob C, Heider U, Zavrski I, Eucker J, et al. (2003) Bone marrow angiogenesis and its correlation with other disease characteristics in multiple myeloma in stage I versus stage II-III. J Cancer Res Clin Oncol 129: 234-238.
  14. De Raeve HR, Vanderkerken K (2005) The role of the bone marrow microenvironment in multiple myeloma. Histol Histopathol 20: 1227-1250.
  15. Giuliani N, Colla S, Rizzoli V (2004) Angiogenic switch in multiple myeloma. Hematology 9: 377-381.
  16. Vacca A, Ribatti D, Presta M, Minischetti M, Iurlaro M, et al. (1999) Bone marrow neovascularization, plasma cell angiogenic potential, and matrix metalloproteinase-2 secretion parallel progression of human multiple myeloma. Blood 93: 3064-3073.
  17. Carmeliet P, Moons L, Luttun A (2011) "Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions," Nature Medicine, vol. 7, pp. 575–583
  18. Ribatti D, Vacca A, Nico B, Crivellato E, Roncali L, et al. (2001) The role of mast cells in tumour angiogenesis. Br J Haematol 115: 514-521.
  19. Yu JL, Rak JW (2003) Host microenvironment in breast cancer development: inflammatory and immune cells in tumour angiogenesis and arteriogenesis. Breast Cancer Res 5: 83-88.
  20. Ria R, Portaluri M, Russo F, Cirulli T, Di Pietro G, et al. (2004) Serum levels of angiogenic cytokines decrease after antineoplastic radiotherapy. Cancer Lett 216: 103-107.
  21. Ria R, Cirulli T, Giannini T, Bambace S, Serio G, et al. (2008) Serum levels of angiogenic cytokines decrease after radiotherapy in non-Hodgkin lymphomas. Clin Exp Med 8: 141-145.
  22. Kuwano M, Fukushi J, Okamoto M, Nishie A, Goto H, et al. (2001) Angiogenesis factors. Intern Med 40: 565-572.
  23. Leek RD, Landers RJ, Harris AL, Lewis CE (1999) Necrosis correlates with high vascular density and focal macrophage infiltration in invasive carcinoma of the breast. Br J Cancer 79: 991-995.
  24. Ria R, Vacca A, Russo F, Cirulli T, Massaia M, et al. (2004) A VEGF-dependent autocrine loop mediates proliferation and capillarogenesis in bone marrow endothelial cells of patients with multiple myeloma. Thromb Haemost 92: 1438-1445.
  25. Dankbar B, Padró T, Leo R, Feldmann B, Kropff M, et al. (2000) Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood 95: 2630-2636.
  26. Hideshima T, Podar K, Chauhan D, Anderson KC (2005) Cytokines and signal transduction. Best Pract Res Clin Haematol 18: 509-524.
  27. Ria R, Roccaro AM, Merchionne F, Vacca A, Dammacco F, et al. (2003) Vascular endothelial growth factor and its receptors in multiple myeloma. Leukemia 17: 1961-1966.
  28. Hideshima T, Nakamura N, Chauhan D, Anderson KC (2001) Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma. Oncogene 20: 5991-6000.
  29. Hideshima T, Chauhan D, Hayashi T, Podar K, Akiyama M, et al. (2002) The biological sequelae of stromal cell-derived factor-1alpha in multiple myeloma. Mol Cancer Ther 1: 539-544.
  30. Tai YT, Podar K, Gupta D, Lin B, Young G, et al. (2002) CD40 activation induces p53-dependent vascular endothelial growth factor secretion in human multiple myeloma cells. Blood 99: 1419-1427.
  31. Vacca A, Scavelli C, Serini G, Di Pietro G, Cirulli T, et al. (2006) Loss of inhibitory semaphorin 3A (SEMA3A) autocrine loops in bone marrow endothelial cells of patients with multiple myeloma. Blood 108: 1661-1667.
  32. Ribatti D, Vacca A, Rusnati M, Presta M (2007) The discovery of basic fibroblast growth factor/fibroblast growth factor-2 and its role in haematological malignancies. Cytokine Growth Factor Rev 18: 327-334.
  33. Mitsiades C. S, Mitsiades N. S, Munshi N. C, Richardson P. G, and Anderson K. C (2006) "The role of the bone marrow microenvironment in the pathophysiology and therapeutic management of multiple myeloma: interplay of growth factors, their receptors and stromal interactions," European Journal of Cancer, vol. 42, n. 11, pp. 1564-1573
  34. Vacca A, Ria R, Semeraro F, Merchionne F, Coluccia M, et al. (2003) Endothelial cells in the bone marrow of patients with multiple myeloma. Blood 102: 3340-3348.
  35. Holmgren L, O'Reilly MS, Folkman J (1995) Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1: 149-153.
  36. Dejana E (1996) Endothelial adherens junctions: implications in the control of vascular permeability and angiogenesis. J Clin Invest 98: 1949-1953.
  37. Ausprunk DH, Falterman K, Folkman J (1978) The sequence of events in the regression of corneal capillaries. Lab Invest 38: 284-294.
  38. Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G, et al. (2000) Openings between defective endothelial cells explain tumor vessel leakiness. Am J Pathol 156: 1363-1380.
  39. Folberg R, Hendrix MJ, Maniotis AJ (2000) Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 156: 361-381.
  40. Helmlinger G, Yuan F, Dellian M, Jain RK (1997) Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation. Nat Med 3: 177-182.
  41. Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, et al. (2000) Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood 95: 952-958.
  42. Ria R, Piccoli C, Cirulli T, Falzetti F, Mangialardi G, et al. (2008) Endothelial differentiation of hematopoietic stem and progenitor cells from patients with multiple myeloma. Clin Cancer Res 14: 1678-1685.
  43. Ribatti D, Nico B, Vacca A (2006) Importance of the bone marrow microenvironment in inducing the angiogenic response in multiple myeloma. Oncogene 25: 4257-4266.
  44. Hynes RO (2002) A reevaluation of integrins as regulators of angiogenesis. Nat Med 8: 918-921.
  45. Carlos T, Kovach N, Schwartz B, Rosa M, Newman B, et al. (1991) Human monocytes bind to two cytokine-induced adhesive ligands on cultured human endothelial cells: endothelial-leukocyte adhesion molecule-1 and vascular cell adhesion molecule-1. Blood 77: 2266-2271.
  46. Vacca A, Frigeri A, Ribatti D, Nicchia GP, Nico B, et al. (2001) Microvessel overexpression of aquaporin 1 parallels bone marrow angiogenesis in patients with active multiple myeloma. Br J Haematol 113: 415-421.
  47. Vacca A, Ria R, Ribatti D, Semeraro F, Djonov V, et al. (2003) A paracrine loop in the vascular endothelial growth factor pathway triggers tumor angiogenesis and growth in multiple myeloma. Haematologica 88: 176-185.
  48. Ribatti D, Nico B, Morbidelli L (2011) "Cell-mediated delivery of fibroblast growth factor-2 and vascular endothelial growth factor onto the chick chorioallantoic membrane: endothelial fenestration and angiogenesis," Journal of Vascular Research, vol. 38: 389-397
  49. Pellegrino A, Ria R, Di Pietro G, Cirulli T, Surico G, et al. (2005) Bone marrow endothelial cells in multiple myeloma secrete CXC-chemokines that mediate interactions with plasma cells. Br J Haematol 129: 248-256.
  50. Vande Broek I, Asosingh K, Vanderkerken K, et al. (2003) "Chemokine receptor CCR2 is expressed by human multiple myeloma cells and mediates migration to bone marrow stromal cell-produced monocyte chemotactic proteins MCP-1, -2, -3," British Journal of Cancer 88: 855-862
  51. Döme B, Hendrix MJ, Paku S, Tóvári J, Tímár J (2007) Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. Am J Pathol 170: 1-15.
  52. Rehman J, Li J, Orschell CM, March KL (2003) Peripheral blood "endothelial progenitor cells" are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation 107: 1164-1169.
  53. Zhao Y, Glesne D, Huberman E (2003) A human peripheral blood monocyte-derived subset acts as pluripotent stem cells. Proc Natl Acad Sci U S A 100: 2426-2431.
  54. Fernandez Pujol B, Lucibello FC, Gehling UM, Lindemann K, Weidner N, et al. (2000) Endothelial-like cells derived from human CD14 positive monocytes. Differentiation 65: 287-300.
  55. Risau W, Sariola H, Zerwes HG, Sasse J, Ekblom P, et al. (1988) Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development 102: 471-478.
  56. Scavelli C, Nico B, Cirulli T, Ria R, Di Pietro G, et al. (2008) Vasculogenic mimicry by bone marrow macrophages in patients with multiple myeloma. Oncogene 27: 663-674.
  57. Sirohi B, Powles R, Mehta J (2001) "The implication of compromised renal function at presentation in myeloma: similar outcome in patients who receive high-dose therapy: a singlecenter study of 251 previously untreated patients," Medical oncology (Northwood, London, England) 18: 39-50
  58. Roccaro AM, Hideshima T, Raje N, Kumar S, Ishitsuka K, et al. (2006) Bortezomib mediates antiangiogenesis in multiple myeloma via direct and indirect effects on endothelial cells. Cancer Res 66: 184-191.
  59. Kisselev AF, Goldberg AL (2001) Proteasome inhibitors: from research tools to drug candidates. Chem Biol 8: 739-758.
  60. Roccaro AM, Hideshima T, Richardson PG, Russo D, Ribatti D, et al. (2006) Bortezomib as an antitumor agent. Curr Pharm Biotechnol 7: 441-448.
  61. Beg AA, Baldwin AS Jr (1993) The I kappa B proteins: multifunctional regulators of Rel/NF-kappa B transcription factors. Genes Dev 7: 2064-2070.
  62. Zandi E, Chen Y, Karin M (1998) Direct phosphorylation of IkappaB by IKKalpha and IKKbeta: discrimination between free and NF-kappaB-bound substrate. Science 281: 1360-1363.
  63. Hideshima T, Chauhan D, Schlossman R, Richardson P, Anderson KC (2001) The role of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma: therapeutic applications. Oncogene 20: 4519-4527.
  64. Hideshima T, Chauhan D, Richardson P, Mitsiades C, Mitsiades N, et al. (2002) NF-kappa B as a therapeutic target in multiple myeloma. J Biol Chem 277: 16639-16647.
  65. Nawrocki ST, Carew JS, Dunner K Jr, Boise LH, Chiao PJ, et al. (2005) Bortezomib inhibits PKR-like endoplasmic reticulum (ER) kinase and induces apoptosis via ER stress in human pancreatic cancer cells. Cancer Res 65: 11510-11519.
  66. Ling Y. H, Liebes L, Zou Y and Perez-Soler R  (2003) "Reactive oxygen species generation and mitochondrial dysfunction in the apoptotic response to Bortezomib, a novel proteasome inhibitor, in human H460 non-small cell lung cancer cells. Journal of Biological Chemistry 278: 33714-33723
  67. Mitsiades N, Mitsiades CS, Richardson PG, Poulaki V, Tai YT, et al. (2003) The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood 101: 2377-2380.
  68. Pajonk F, McBride WH (2001) The proteasome in cancer biology and treatment. Radiat Res 156: 447-459.
  69. San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, et al. (2008) Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med 359: 906-917.
  70. Ria R, Vacca A, Mangialardi G, Dammacco F (2008) Delayed complete remission in a patient with multiple myeloma. Eur J Clin Invest 38: 966-968.
  71. Vacca A, Scavelli C, Montefusco V, Di Pietro G, Neri A, et al. (2005) Thalidomide downregulates angiogenic genes in bone marrow endothelial cells of patients with active multiple myeloma. J Clin Oncol 23: 5334-5346.
  72. Sonneveld P (2001) "Segeren intensified chemotherapy in untreated multiple myeloma: a prospective randomized Phase III," Blood 98: 815
  73. D'Amato RJ, Loughnan MS, Flynn E, Folkman J (1994) Thalidomide is an inhibitor of angiogenesis. Proc Natl Acad Sci U S A 91: 4082-4085.
  74. Ribatti D and Vacca A (2005) "Novel Therapeutic Approches Targeting Vascular Endothelial Growth Factor and its receptors in haematological malignancies," Current cancer drug targets 5: 573-578
  75. Davies FE, Raje N, Hideshima T, Lentzsch S, Young G, et al. (2001) Thalidomide and immunomodulatory derivatives augment natural killer cell cytotoxicity in multiple myeloma. Blood 98: 210-216.
  76. Juliusson G, Celsing F, Turesson I, Lenhoff S, Adriansson M, et al. (2000) Frequent good partial remissions from thalidomide including best response ever in patients with advanced refractory and relapsed myeloma. Br J Haematol 109: 89-96.
  77. Gupta D, Treon S. P, Shima Y (2001) "Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factoor secretion: therapeutic application," Leukemia 15: 1950-1961
  78. Dimopoulos M, Spencer A, Attal M, Prince HM, Harousseau JL, et al. (2007) Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 357: 2123-2132.
  79. García-Sanz R, González-Porras JR, Hernández JM, Polo-Zarzuela M, Sureda A, et al. (2004) The oral combination of thalidomide, cyclophosphamide and dexamethasone (ThaCyDex) is effective in relapsed/refractory multiple myeloma. Leukemia 18: 856-863.
  80. Richardson PG, Schlossman RL, Weller E, Hideshima T, Mitsiades C, et al. (2002) Immunomodulatory drug CC-5013 overcomes drug resistance and is well tolerated in patients with relapsed multiple myeloma. Blood 100: 3063-3067.
  81. Richardson PG, Blood E, Mitsiades CS, Jagannath S, Zeldenrust SR, et al. (2006) A randomized phase 2 study of lenalidomide therapy for patients with relapsed or relapsed and refractory multiple myeloma. Blood 108: 3458-3464.
  82. Scavelli C, Di Pietro G, Cirulli T, Coluccia M, Boccarelli A, et al. (2007) Zoledronic acid affects over-angiogenic phenotype of endothelial cells in patients with multiple myeloma. Mol Cancer Ther 6: 3256-3262.
  83. Clézardin P (2002) The antitumor potential of bisphosphonates. Semin Oncol 29: 33-42.
  84. Wood J, Bonjean K, Ruetz S, Bellahcène A, Devy L, et al. (2002) Novel antiangiogenic effects of the bisphosphonate compound zoledronic acid. J Pharmacol Exp Ther 302: 1055-1061.
  85. Ribatti D, Nico B, Mangieri D, Maruotti N, Longo V, et al. (2007) Neridronate inhibits angiogenesis in vitro and in vivo. Clin Rheumatol 26: 1094-1098.
Citation: Ria R, Berardi S, Reale A, Luisi AD, Catacchio I, et al. (2013) Multiple Myeloma: The Role of Angiogenesis in Disease Progression. J Bone Marrow Res 1:117.

Copyright: © 2013 Ria R, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top