Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2013 Mar;15(3):231–238. doi: 10.1593/neo.121962

The Role of Angiogenesis in Human Non-Hodgkin Lymphomas1

Domenico Ribatti *, Beatrice Nico *, Girolamo Ranieri , Giorgina Specchia , Angelo Vacca §
PMCID: PMC3593147  PMID: 23479502

Abstract

The role of angiogenesis in the growth of lymphomas and survival of patients with leukemias and other hematological malignancies has become evident since 1994. Angiogenic factors, such as vascular endothelial growth factor and its receptors together with other tumor microenvironment components, including myelo-monocytic cell, mast cells, endothelial progenitor cells, and circulating endothelial cells, have been shown to be important in the progression and maintenance of lymphoproliferative disorders. In this review article, we present an overview of the literature focusing on the relationship between angiogenesis and disease progression and the recent advantages in the antiangiogenic treatment in human non-Hodgkin lymphomas.

Introduction

In the evolution of tumor growth, the avascular phase is followed by a vascular one [1]. Assuming that such growth is dependent on angiogenesis and that this depends on the release of angiogenic factors, the acquisition of an angiogenic ability can be seen as an expression of progression from neoplastic transformation to tumor growth and metastasis. All solid tumors, including those of the colon, lung, breast, cervix, bladder, prostate, and pancreas, progress through these two phases [2]. The role of angiogenesis in the growth and survival of leukemias and other hematological malignancies has become evident since 1994 [3] in a series of demonstrations that the progression is clearly related to their degree of angiogenesis.

Lymphomas constitute a large group of more than 40 lymphoproliferative disorders, classified on the basis of morphologic, immunologic, genetic, and clinical criteria. The importance of angiogenesis in lymphoproliferative disorders has been studied in relation to their impact on the prognosis of patients, suggesting high relevance in different types of lymphomas [4–6]. Non-Hodgkin lymphomas (NHLs) are a heterogeneous group of lymphoproliferative malignancies with different patterns of behavior responses to treatment. B cell lymphomas represent approximately 88%, and T and natural killer (NK) cell lymphomas 12%, respectively, of all NHLs. Among B cell lymphomas, the incidence of diffuse large B cell lymphomas (DLBCLs) is 30%, of follicular lymphoma (FL) 25%, of extranodal marginal zone lymphoma of mucosa-associated lymphatic tissue 7%, of chronic lymphocytic leukemia (CLL) 7%, and of mantle cell lymphoma (MCL) 5%.

Lymphoid tumors are generally divided into one of two categories, namely, indolent lymphomas versus aggressive lymphomas, based on the characteristics of the disease at the time of presentation and the patients' life expectancy if the disease is left untreated. Generally, T cell lymphomas have a more aggressive clinical behavior than B cell lymphomas of comparable histology and patients with MCLs or anaplastic large lymphomas have a 5-year survival rate of approximately 30% and 80%, respectively [7].

In this review article, we present an overview of the literature focusing on the relationship between angiogenesis and disease progression and the recent advantages in the antiangiogenic treatment in human NHL.

In Vitro and Vivo Experimental Models

Conditioned media of lymphoma cells induced a five-fold increased proliferation of cultured endothelial cells, suggesting the release of a soluble proangiogenic factor [8]. Human lymphoid tumor cells constitutively produce significant amounts of the extracellular matrix degrading enzymes matrix metalloproteinase-2 (MMP-2) and MMP-9, as demonstrated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis gelatin zymography and in situ hybridization [9]. Moreover, human lymphoid tumor cells are able to interact with extracellular matrix components vitronectin and fibronectin and this interaction it is mediated by αvβ3 integrin, allowing them to adhere to the substratum and enhancing their proliferation and protease secretion [10].

Lymphoma cells are able to induce an angiogenic response when tested in vivo in the hamster check pouch model [11]. Similarly, lymphoma bioptic specimens, when implanted on the chick embryo chorioallantoic membrane (CAM), evoked a strong angiogenic response [12]. The angiogenic response did not correlate with either the malignancy grade or the immunologic phenotype of the tumors. Different human Burkitt's lymphoma cells when inoculated onto the CAM formed solid tumors [13]. However, Epstein-Barr virus-positive cells induced massive recruitment of chick leukocytes at the tumor border and the development of granulation tissue with large number of blood and lymphatic vessels, although all cell lines tested have almost identical vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) expression [13].

Angiogenesis in Normal Lymph Nodes

The lymph node microvasculature consists of arterioles, metarterioles, anastomosing capillaries, small venules, and high endothelial post-capillary venules. Dense plexuses of capillaries arise from arterioles in the medullary cords, in the periphery of the deep cortex units, and in the outermost stratum of the extrafollicular zone of the peripheral cortex. In contrast, the folliculo-nodules and center of the deep cortex units are little vascularized by a loose capillary network, while no vessels occur in the subsinus layer [14–16]. When tissue fragments from normal lymph nodes are grafted in vivo on the chick embryo CAM, stereomicroscopic observation of the area around the implant revealed little hyperemia and a small number of growing vessels [12].

Angiogenesis in Benign Lymphadenopathies

In both reactive lymph nodes and lymph nodes with FLs, microvascular density (MVD) is higher in the paracortex than in the follicles and that there is no difference in MVD between reactive germinal centers and neoplastic follicles [17]. Moreover, MVD in the paracortex in reactive lymph nodes is higher than in diffuse large lymphomas [17]. In FL, several studies have recognized an increase in MVD in reactive parts of affected lymph nodes outside the follicles, compared to the neoplastic follicles [18–21].

Other authors [22] have shown that MVD is higher in lymphomas than in reactive lymph nodes and in aggressive than indolent lymphomas or that MVD in reactive lymph nodes is comparable to that observed in lymphomas [21].

Angiogenesis in NHLs

As concerns the morphologic features of tumor blood vessels, two patterns of laminin and type IV collagen expression are recognizable in the perivascular stroma of B-NHL, classified accordingly to the working formulation in low-, intermediate- and high-grade tumors [22]. A granular, speckled, and low-intensity staining was expressed by laminin and more frequently associated with the intermediate- and high-grade tumors. A linear continuous staining was co-expressed by laminin and type IV collagen and was more frequently associated with low-grade tumors. The granular and linear patterns may correspond to different steps in the basement membrane deposition: the granular pattern to the first one, when endothelial proliferation takes place; the linear pattern to the second stage, when basement membrane is completely differentiated [23]. This hypothesis is in agreement with the evidence that endothelial sprouting is associated with a higher concentration of laminin than type IV collagen around the outgrowing capillaries [24].

Moreover, the expression of tenascin in the stroma of B-NHL has been investigated and related to histologic malignancy and angiogenesis [25]. It is well known that tenascin stimulates angiogenesis, and because it forms a long reticulum with long extensions directly from vessels, it could also provide a pathway that favors migration of endothelial cells [26]. The presence of an increased number of immature vessels in DLBCL compared with FL, classified accordingly to the ">World Health Organization (WHO) classification, has also been demonstrated [17].

At ultrastructural level, the presence of immature capillaries in the stroma of diffuse intermediate-grade and high-grade B-NHLs has been shown [22]. These capillaries lack the basement membrane and generally consisted of two endothelial cells arranged in parallel, with thickened cytoplasm, resulting in slit-like lumen. On the contrary, in follicular intermediate-grade and low-grade B-NHLs, differentiated fenestrated capillaries surrounded by a continuous basement membrane were recognizable. Moreover, a morphologic heterogeneity of tumor blood vessels between histologic subtypes of lymphomas has been shown together with different patterns of neovascularization in both low-grade and high-grade B-NHLs [27,28]. In low-grade B-NHL, the vessel lumen is formed either by endothelial cell body curving or, more frequently, by the fusion of intracellular vacuoles in poorly differentiated endothelial cells. In high-grade B-NHL, however, the prevalent neoangiogenic pattern is the formation of a slit-like lumen [27,28]. Both low-grade and high-grade tumors exhibited development of transluminal bridges, expression of intussusceptive microvascular growth, and alternative mode of tumor vessel growth [29].

The clinical significance of increased MVD is not clear and difficult to establish because most of the studies describe heterogeneous populations including a wide variety of histologic subtypes of NHL and different treatment regimens.

As concerns the evaluation of MVD in bioptic specimens, a correlation between MVD and histologic subtype in NHL has not been established [30], or differences in MVD in patients with chemotherapy-resistant DLBCL and those with chemosensitive lymphomas has not been found [31]. Other studies in NHL and in DLBCL found no correlation between MVD and VEGF expression [32–34]. On the contrary, an increased vascularity pretreatment predicted favorable outcome in terms of progression-free and overall survival in patients with FL who received chemotherapy in association with interferon-α2b [18], or in FL, a high MVD predicted progressive disease and overall survival and correlated with transformation to DLBCL [32].

MVD is highest in aggressive subtypes including Burkitt's lymphoma and peripheral T cell lymphoma, compared with intermediate in DLBCL and lower in indolent FL [22]. In DLBCL, the average MVD correlates with the intensity of VEGF tumor cell immunoreactivity [35]. On the contrary, another study of the same group on patients affected by DLBCL treated with anthracycline-based chemotherapy showed no correlation between increased MVD and lymphoma cell VEGF expression [36]. In cutaneous T cell and B cell lymphomas, MVD is higher than in normal skin of a benign cutaneous lymphoproliferative disorder [37–39].

Several studies have demonstrated that high levels of VEGF in lymphoma samples correlate with advanced tumor stage and higher risk for relapsed/refractory disease after standard chemotherapy.

In NHL, high pretreatment levels of serum VEGF was a prognostic factor for survival in multivariate analysis [40]. In both T and B cell lymphomas, a negative correlation between the overall survival rate, respectively, 5-year disease-free survival and the pretreatment serum level of VEGF has been established [41], while in patients with DLBCL treated with cyclophosphamide, doxorubicin, vincristine, and prednisolone, high serum level of VEGF was associated with adverse outcome, having lower values in survivors than in non-survivors [42].

VEGF expression was also demonstrated in peripheral T cell lymphoma, DLBCL, MCL, primary effusion lymphoma, and CLL/small lymphocytic lymphoma [43–46]. An adverse outcome associated with an increased VEGF tissue expression in aggressive and indolent lymphomas of B cell and T cell origin [47] has been demonstrated. In angioimmunoblastic T cell lymphoma, VEGF-A gene is overexpressed in both tumor and endothelial cells in comparison with reactive lymph nodes in association with a short survival time [48], and a high expression of VEGF-A in aggressive T cell lymphomas compared to indolent B cell lymphomas has been found [44]. In contrast, only a minority of indolent FLs show variable expression of VEGF-A [18,32], and transformation from indolent B cell lymphoma to aggressive DLBCL and poor prognostic subgroups within DLBCL are associated with increased VEGF expression [49].

In primary diffuse central nervous system lymphomas (PCNSLs), VEGF expression is correlated to MVD and VEGF expression is associated with a longer survival and blood-brain barrier alteration [50]. In 24 human diffuse large B cell PCNSL studied by means of immunocytochemistry and confocal laser microscopy, it has been demonstrated that 1) Aquaporin 4 (AQP4) expression was directly correlated with Ki-67 index, while AQP4 expression was low in tumor areas with a low Ki-67 index. 2) Different cells participated to vessel formation: CD20+ tumor cells and factor VIII+ endothelial cells; AQP4+ tumor cells and CD31+ endothelial cells; CD20+ and AQP4+ tumor cells; glial fibrillary acidic protein positive endothelial cells surrounded by glial fibrillary acidic protein positive tumor cells. Overall, these data suggest the importance of AQP4 in PCNSL due to its involvement in pathogenesis and resolution of cerebral edema. AQP4 is also involved in migration of tumor cells. It was also documented that tumor microvasculature in PCNSL is extremely heterogeneous, confirming the importance of neoangiogenesis in their pathogenesis [51].

The degree of VEGF expression correlated with the expression level of VEGFR-1 and VEGFR-2 in DLBCL lymphoma cells [35], and VEGFR-1, VEGFR-2, and VEGFR-3 are expressed in CLL, suggesting the possibility that VEGF acts as an autocrine/paracrine factor [52]. Moreover, VEGF prevents apoptosis and increases phosphorylation of VEGFR-1 and VEGFR-2, further supporting the existence of an autocrine prosurvival loop in CLL [53]. Blocking of VEGF and VEGFRs, by using neutralizing antibodies or tyrosine kinase inhibitors, resulted in decreased levels of p-STAT-3 and apoptosis of CLL cells [54]. High VEGF and VEGFR-1 expression identified a subgroup of patients affected by DLBCL with improved overall survival and progression-free survival when treated with anthracycline-based chemotherapy, suggesting that the autocrine signaling through VEGFR-1 may be susceptible to this therapeutic approach [36]. When immunodeficient mice engrafted with human DLBCL were treated with antibodies against human or murine VEGFR-1 or VEGFR-2, a significant tumor reduction of 50% was observed after treatment with human anti-VEGFR-1 but not with murine anti-VEGFR-1. By contrast, inhibition of murine VEGFR-2 resulted in a similar tumor reduction, but inhibition of human VEGFR-2 had no antitumor effect [55]. Anti-VEGFR-2 antibody was as effective as rituximab, and when combined, tumor volume was reduced even more to 75% [55]. A lesser expression of hypoxia-inducible factor-1 and hypoxia-inducible factor-2 and VEGF in indolent lymphomas, consisting mainly of FL, than in aggressive lymphomas has been observed [56]. Accordingly, only a minority of indolent lymphomas, showing histologic transformation to aggressive lymphoma, expressed VEGF-A in contrast to aggressive lymphomas [57].

Inhibition of autocrine or paracrine VEGFR-mediated loops with receptor-specific antibodies suppresses the growth of lymphomas by increasing tumor apoptosis and decreasing vascularization, respectively. These results confirm the role of VEGF in lymphomagenesis and support the targeting of VEGFRs as a therapeutic approach for aggressive lymphomas.

Other angiogenic growth factors may contribute to the angiogenic process and tumor progression in NHL. Among these, fibroblast growth factor-2 (FGF-2) is one of the best characterized proangiogenic cytokines. Because of its pleiotropic activity that may affect both tumor vasculature and tumor parenchyma, FGF-2 may contribute to cancer progression by inducing neovascularization, as well as by acting directly on tumor cells.

Various lymphoblastoid cell lines secrete FGF-2 [58]. Pazgal et al. [59] measured FGF-2 serum concentration in patients with NHL before and after treatment, conducted an immunohistochemical study to determine the expression of FGF-2 and FGF receptor-1 (FGFR-1) and MVD, and evaluated the prognostic significance of FGF-2 and FGFR-1 expression. They demonstrated that FGF-2 expression was correlated with poor survival and progression-free survival, while FGFR-1 expression was correlated with decreased rate of achievement of complete remission. Moreover, they did not detect a significant change in serum FGF-2 levels after two to three cycles of chemotherapy, nor they did find a correlation between MVD and NHL histology or grade or between MVD and prognosis. Moreover, in malignant lymphoma, high pretreatment levels of FGF-2 were a prognostic factor for survival in multivariate analysis, independently of other risk factors, including serum lactate dehydrogenase and number of extranodal sites [40]. Soluble VEGF, FGF-2, and platelet-derived growth factor-β levels decline after radiotherapy in NHL, suggesting that may have predictive significance for response to treatment and recurrence [60].

The Role of Myelo-monocytic Cells

At least three categories of proangiogenic bone marrow-derived circulating cells have been implicated in tumor angiogenesis: 1) cells that contribute directly to the structural components of angiogenesis, including endothelial progenitor cells (EPCs) and pericyte progenitors; 2) myeloid progenitor subsets that can differentiate into endothelial-like cells and incorporate into the tumor neovessels; 3) a large heterogeneous group of cells of monocytic lineage that functions as vascular modulators that are not physically part of the vasculature [61].

It is well established that neoangiogenesis and growth of murine lymphomas is dependent on the recruitment of bone marrow-derived proangiogenic hematopoietic cells [62]. Increased hematopoietic infiltration by myeloid progenitors CD68+ and VEGFR-1+ and producing VEGF-A has been correlated with histologic subtypes of lymphoma, suggesting their involvement in the development of a proangiogenic phenotype [19,34,63]. In aggressive subtypes of Burkitt's lymphoma and DLBCL, VEGF-A-producing CD68+ VEGFR1+ myelo-monocytic cells are closely associated to new-formed blood vessels [34]. Genetic depletion of this subpopulation of CD68+ VEGFR1+ myelo-monocytic cells was sufficient to inhibit angiogenesis in various tumor experimental models, including lymphoma [62].

The Role of Macrophages and Mast Cells

It is well known that, among inflammatory cells found in tumors, tumor-associated macrophages and mast cells support tumor growth and neovascularization by producing a wide array of angiogenic cytokines. Tumor-associated macrophages have profound influence on the regulation of tumor angiogenesis. In fact, the degree of macrophage infiltration is positively correlated with tumor stage and angiogenesis in several human tumors in which a relationship between MVD and tumor progression has been clearly demonstrated [64].

Angiogenesis extent and macrophage density increase simultaneously with pathologic progression in B cell NHL, suggesting that an increase number of macrophages may be recruited and activated locally by malignant B cells and that angiogenesis associated with B-NHL may be induced, at least, partly, by angiogenic factors secreted by macrophages [19]. A high number of intratumoral macrophages correlate with poor prognosis in FL treated with chemotherapy alone, and rituximab appears to circumvent the unfavorable prognosis associated with high number of macrophages [65,66].

The extent of angiogenesis has been correlated with the number of mast cells in B-NHL and both counts increase in step with the increase of malignancy grade [64,67]. Tryptase together with other angiogenic factors stored in mast cell secretory granules may contribute to angiogenesis in B-NHL [64,67]. In an ultrastructural study of samples of B cell NHL, the presence of a heterogeneous population of mast cells characterized by the presence of granules with semilunar aspect and containing scrolls has been demonstrated [28,68]. Semilunar granules are the expression of a slow but progressive release of angiogenic factors due to chronic and progressive stimulation of mast cell degranulation, while, in the granules containing scrolls is stored tryptase, an angiogenic factor [69]. In B cell CLL, there is a striking association between the number of mast cells and MVD in bone marrow and both increase as the disease progresses [70]. Moreover, the consistent decrease of bone marrow angiogenesis after sequential fludarabine induction and alemtuzumab consolidation therapy in advanced CLL parallels the reduction of mast cells [71].

The Role of EPCs and Circulating Endothelial Cells Circulating EPCs (CEPCs) have been detected within the blood flow during tumor growth and several evidences indicate that bone marrow-derived CEPCs contribute to tumor growth and tumor angiogenesis [72]. An increased number of CD133+ CD34+ VEGFR-2+ CEPC has been found in younger patients and those with aggressive NHL, and the levels of CEPC decreased following complete response to treatment [73]. Moreover, lymph node EPCs were detected in vascular structure and in the stroma and correlated with an increased angiogenesis in indolent lymphoma [73]. In angiogenesis-defective Idmutant mice, VEGFR-2+ EPCs constitute >90% of tumor vessels following wild-type bone marrow rescue in a murine xenograft model of aggressive B cell lymphoma [74].

Within immunodeficient mice engrafted with lymphoma cells, a significant increase in the number of circulating endothelial cells (CECs) was observed after a period of 21 days, they being correlated with tumor size and serum level of VEGF [75]. An increase in CEC in patients with lymphoma compared with the control cases has been reported [76]. In those patients achieving complete remission after chemotherapy, the number of CECs was similar to healthy controls [76]. Accordingly, an increased number of CEC has been recognized in younger patients and those with aggressive NHL and the levels of CEC decreased following complete response to treatment [73].

Genetically Modified Lymphoma Endothelial Cells

Chromosomal abnormalities involving all chromosomes may occur in lymphomas, and characterization of genetic abnormalities, while not an absolute requirement, can be essential to the diagnosis of many lymphomas [77].

The presence of lymphoma-specific chromosomal translocations in endothelial cells in B cell lymphomas has been demonstrated, suggesting that microvascular endothelial cells in B cell lymphomas are, in part, tumor related [78]. Moreover, 15% to 85% of microvascular endothelial cells harbored lymphoma-specific genetic alterations consisting not only of B cell-specific translocation of immunoglobulin heavy locus (IGH) but also secondary genetic alterations in FL [78]. As suggested by Streubel et et al. [78], four mechanisms may be involved: lymphoma cells and endothelial cells may be derived from a multipotent hemangioblastic precursor cell targeted by neoplastic transformation that can differentiate in tumor cells or endothelial cells sharing the same genetic abnormalities; the endothelial cells carrying the genetic alterations of the lymphoma may arise from a cell that was already committed to the lymphoid lineage; fusion of lymphoma cells and endothelial cells with formation of hybrid vessels or gene transfer by means of the uptake of apoptotic bodies from tumor cells by neighboring cells may be alternative mechanisms.

The expression of a transcript called T cell Ig and mucin-containing molecule 3 has been identified in microvessels of DLBCL but not in reactive lymph nodes, suggesting that the lymphoma endothelium may act as a functional barrier facilitating the establishment of lymphoma immune tolerance [79].

Antiangiogenesis in NHL

Antiangiogenesis is a promising therapeutic approach in cancer. Preclinical studies with various angiogenesis inhibitors have produced remarkable antitumor effects in animal models and inhibition of angiogenesis is a major area of therapeutic development for the treatment of hematological malignancies.

Endostatin

Endostatin is an endogenous inhibitor of angiogenesis, which inhibits endothelial cell proliferation and migration, induces apoptosis, and causes a G1 arrest of endothelial cells. Moreover, endostatin inhibits MMP-2 activity, blocks the binding of VEGF to VEGFR-2, and stabilizes cell-cell and cell-matrix adhesions, preventing the breakage of these junctions required during angiogenesis [80].

Various endogenous inhibitors of angiogenesis may be found in the bloodstream, and a circulating form of human endostatin has been identified [81]. In a subgroup of patients with large cell and immunoblastic lymphoma, patients with high serum endostatin levels had a significantly better survival as compared with those with lower levels [82]. In a mouse model of B cell lymphoma, a delay in tumor growth has been shown after administration of endostatin [83] and continuous infusion of endostatin inhibits tumor growth and the mobilization and differentiation of EPC in mice bearing an angiogenic human lymphoma [84].

Treatment of lymphoma-bearing mice with endostatin caused an increase in the frequency of apoptotic cells in the endothelial cell compartment and most of the CECs were apoptotic or dead, while cyclophosphamide had no such effect. This difference probably occurred because most of the circulating apoptotic cells were hematopoietic and not endothelial in nature [75]. Endostatin administration in advanced stages of tumor growth led to tumor regression even in cyclophosphamide- and rituximab-resistant cases [83]. This effect was induced by inhibition of proliferation and stimulation of apoptosis in endothelial cells.

Immunomodulatory Drugs

Thalidomide exerts its antiangiogenic action through the inhibition of various cytokines, including tumor necrosis factor-α (TNF-α) and VEGF [85]. Thalidomide as single agent demonstrated a low overall response rate in patients with relapsed/refractory indolent NHL [86] and in heavily pretreated patients with recurrent lymphoma [87]. In combination with fludarabine, thalidomide was associated with significant therapeutic efficacy in CLL [88].

Lenalidomide is a more potent analog of thalidomide with preferentially TNF-α inhibitory properties and weaker antiangiogenic effect. Lenalidomide has been used as monotherapy in the treatment of both aggressive (DLBCL and transformed) and indolent relapsed/refractory NHL, MCL, and angioimmunoblastic T cell lymphoma [89–94]. Lenalidomide has also been studied in relapsed/refractory CLL, inducing complete and partial remissions, and has considerable activity in both heavily pretreated CLL patients and patients with unfavorable prognostic factors [95,96]. Moreover, thalidomide has shown antitumor activity in combination with rituximab in patients with relapsed or refractory MCL [97].

Bortezomib, a proteasome inhibitor, exerts anticancer activity mainly by inhibiting nuclear factor-κB (NF-κB), which has a pivotal role in the synthesis of antiapoptotic and angiogenic factors [98]. Clinical studies using bortezomib in relapsed or refractory B cell NHL, MCL, or marginal zone B cell lymphoma have shown promising results [99–104].

Anti-VEGF Neutralizing Antibodies and VEGFR Inhibitors

Stimulation of VEGFRs and other receptor tyrosine kinases causes activation of signaling pathways in endothelial cells. Many of the processes involved in tumor growth, progression, and metastasis are mediated by signaling molecules acting downstream from activated receptor tyrosine kinases. The VEGF/VEGFR pathway is considered a key regulator of angiogenesis and most of the agents currently in preclinical and clinical development focus on the inhibition of this pathway.

Bevacizumab (Avastin), a recombinant humanized monoclonal antibody directed against VEGF-A, has been the first antiangiogenic agent to be approved by the US Food and Drug Administration. Bevacizumab inhibit tumor growth, either alone or in combination with chemotherapy in untreated DLBCL [33,105]. A long disease-free survival in patients with aggressive NHL subtypes treated with bevacizumab as single agent has been reported [105]. Anti-VEGF neutralizing antibodies and VEGFR inhibitors blocked the prosurvival effect of CD154 (CD40 ligand) on CLL cells and decreased the migration of CLL cells through the endothelium [106,107].

Histone Deacetylase Inhibitors

Acetylation and deacetylation of histone proteins are important mechanisms for the regulation of gene expression. The interest in histone deacetylases (HDACs) as antineoplastic drugs originated with the observation that these agents could reverse the malignant phenotype of transformed cells [108]. HDACs represent an emerging class of therapeutic agents effective in hematological malignancies [109] that induce tumor cell cytostasis, differentiation, and apoptosis, in part due to an angiostatic effect, through an inhibition of VEGFR expression in endothelial cells [110,111].

Vorinostat, panobinostat, and MGCD0103 have been evaluated in NHL [112,113]. Successful therapy with vorinostat was associated with a reduced MVD and an increase of the antiangiogenic molecule thrombospondin-1 following treatment [111]. Panobinostat was evaluated in cutaneous T cell lymphoma (CTCL), and microarray analysis of skin biopsies showed a consistent down-regulation of proangiogenic gene guanylate cyclase 1A3 and angiopoietin-1 [114]. Two HDAC inhibitors, sodium butyrate and suberoylanilide hydroxamic acid, reduced VEGF production and induced growth suppression and apoptosis in human MCL cell lines [115].

Concluding Remarks

Over the last 20 years, the importance of angiogenesis in human lymphoma is now well recognized, and several factors involved in its control are being identified. Within the different types of B cell NHL, angiogenesis may be prominent in aggressive rather than indolent subtypes. In addition to the demonstration that lymph node bioptic specimens involved with NHL contain high number of MVD and high number of inflammatory cells secreting angiogenic cytokines, several studies reported high levels of soluble angiogenic factors in sera of patients with NHL.

The antiangiogenic therapy is an important tool for the treatment of human lymphoma. However, a significant number of patients are resistant, whereas those who respond have minimal benefits. A tumor resistance and also significant side effects including toxicity can occur.

Further research should provide new useful therapeutic approaches and increase options for patients with resistant or refractory disease.

Acknowledgments

We are grateful to M.V.C. Pragnell for linguistic revision.

Footnotes

1

The research leading to these results has received funding from the European Union Seventh Framework Programme (FP7/2007-2013) under grant 278570 to D.R. and grant 278706 to A.V.

References

  • 1.Ribatti D, Nico B, Crivellato E, Roccaro AM, Vacca A. The history of the angiogenic switch concept. Leukemia. 2007;21:44–52. doi: 10.1038/sj.leu.2404402. [DOI] [PubMed] [Google Scholar]
  • 2.Ribatti D, Vacca A, Dammacco F. The role of the vascular phase in solid tumor growth: a historical review. Neoplasia. 1999;1:293–302. doi: 10.1038/sj.neo.7900038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.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. doi: 10.1111/j.1365-2141.1994.tb08304.x. [DOI] [PubMed] [Google Scholar]
  • 4.Kini AR. Angiogenesis in leukemia and lymphoma. Cancer Treat Res. 2004;121:221–238. doi: 10.1007/1-4020-7920-6_9. [DOI] [PubMed] [Google Scholar]
  • 5.Koster A, Raemaekers JMM. Angiogenesis in malignant lymphoma. Curr Opin Oncol. 2005;17:611–616. doi: 10.1097/01.cco.0000181404.83084.b5. [DOI] [PubMed] [Google Scholar]
  • 6.Ruan J, Hajjar K, Rafii S, Leonard JP. Angiogenesis and antiangiogenic therapy in non-Hodgkin's lymphoma. Ann Oncol. 2009;20:413–424. doi: 10.1093/annonc/mdn666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Fisher RI, Miller TP, Grogan TM. New REAL clinical entities. Cancer J Sci Am. 1998;4(suppl 2):S5–S12. [PubMed] [Google Scholar]
  • 8.Brandvold KA, Neiman P, Ruddell A. Angiogenesis is an early event in the generation of myc-induced lymphomas. Oncogene. 2000;19:2780–2785. doi: 10.1038/sj.onc.1203589. [DOI] [PubMed] [Google Scholar]
  • 9.Vacca A, Ribatti D, Ria R, Pellegrino A, Bruno M, Merchionne F, Dammacco F. Proteolytic activity of human lymphoid tumor cells. Correlation with tumor progression. Dev Immunol. 2000;7:77–88. doi: 10.1155/2000/74372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Vacca A, Ria R, Presta M, Ribatti D, Iurlaro M, Merchionne F, Tanghetti E, Dammacco F. αvβ3 integrin engagement modulates cell adhesion, proliferation, and protease secretion in human lymphoid tumor cells. Exp Hematol. 2001;29:993–1003. doi: 10.1016/s0301-472x(01)00674-9. [DOI] [PubMed] [Google Scholar]
  • 11.Wolf JE, Hubler WR. Tumour angiogenic factor associated with subcutaneous lymphoma. Br J Dermatol. 1975;92:273–277. doi: 10.1111/j.1365-2133.1975.tb03076.x. [DOI] [PubMed] [Google Scholar]
  • 12.Ribatti D, Vacca A, Bertossi M, De Benedictis G, Roncali L, Dammacco F. Angiogenesis induced by B-cell non-Hodgkin's lymphomas. Lack of correlation with tumor malignancy and immunologic phenotype. Anticancer Res. 1990;10:401–406. [PubMed] [Google Scholar]
  • 13.Becker J, Covelo-Fernandez A, von Bonin F, Kule D, Wilting J. Specific tumor-stroma interactions of EBV-positive Burkitt's lymphoma cells in the chick chorioallantoic membrane. Vasc Cell. 2012;4:3. doi: 10.1186/2045-824X-4-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Davidson JW, Hobbs BB, Fletch AL. The microcirculatory unit of the mammalian lymph node. Bibl Anat. 1973;11:423–427. [PubMed] [Google Scholar]
  • 15.Anderson AO, Anderson ND. Studies on the structure and permeability of the microvasculature in normal rat lymph nodes. Am J Pathol. 1975;80:387–418. [PMC free article] [PubMed] [Google Scholar]
  • 16.Herman PG. Microcirculation of organized lymphoid tissues. Monogr Allergy. 1980;16:126–142. [PubMed] [Google Scholar]
  • 17.Passalidou E, Stewart M, Trivella M, Steers G, Pillai G, Dogan A, Leigh I, Hatton C, Harris A, Gatte K, et al. Vascular patterns in reactive lymphoid tissue and in non-Hodgkin's lymphoma. Br J Cancer. 2003;88:553–559. doi: 10.1038/sj.bjc.6600742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Koster A, van Krieken JH, Mackenzie MA, Schraders M, Borm GF, van der Laak JA, Leenders W, Hebeda K, Raemaekers JM. Increased vascularization predicts favorable outcome in follicular lymphoma. Clin Cancer Res. 2005;11:154–161. [PubMed] [Google Scholar]
  • 19.Vacca A, Ribatti D, Ruco L, Giacchetta F, Nico B, Quondamatteo F, Ria R, Iurlaro M, Dammacco F. Angiogenesis extent and macrophage density increase simultaneously with pathological progression in B-cell non-Hodgkin's lymphomas. Br J Cancer. 1999;79:965–970. doi: 10.1038/sj.bjc.6690154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ridell B, Norrby K. Intratumoral microvascular density in malignant lymphomas of B-cell origin. APMIS. 2001;109:66–72. doi: 10.1111/j.1600-0463.2001.tb00015.x. [DOI] [PubMed] [Google Scholar]
  • 21.Arias V, Soares FA. Vascular density (tumor angiogenesis) in non-Hodgkin's lymphomas and florid follicular hyperplasia: a morphometric study. Leuk Lymphoma. 2000;40:157–166. doi: 10.3109/10428190009054893. [DOI] [PubMed] [Google Scholar]
  • 22.Ribatti D, Vacca A, Nico B, Fanelli M, Roncali L, Dammacco F. Angiogenesis spectrum in the stroma of B-cell non-Hodgkin's lymphomas. An immunohistochemical and ultrastructural study. Eur J Haematol. 1996;56:45–53. doi: 10.1111/j.1600-0609.1996.tb00293.x. [DOI] [PubMed] [Google Scholar]
  • 23.Paku S, Paweletz N. First steps of tumor-related angiogenesis. Lab Invest. 1991;65:334–346. [PubMed] [Google Scholar]
  • 24.Ausprunk DH, Folkman J. Migration and proliferation of endothelial cells in preformed and newly formed blood vessels during tumor angiogenesis. Microvasc Res. 1977;14:53–65. doi: 10.1016/0026-2862(77)90141-8. [DOI] [PubMed] [Google Scholar]
  • 25.Vacca A, Ribatti D, Fanelli M, Costantino F, Nico B, Di Stefano, R, Serio G, Dammacco F. Expression of tenascin is related to histologic malignancy and angiogenesis in B-cell non-Hodgkin's lymphomas. Leuk Lymphoma. 1996;22:473–481. doi: 10.3109/10428199609054786. [DOI] [PubMed] [Google Scholar]
  • 26.Rettig WJ, Erickson HP, Albino AP, Garin-Chesa P. Induction of human tenascin (neuronectin) by growth factors and cytokines: cell type-specific signals and signalling pathways. J Cell Sci. 1994;107:487–497. [PubMed] [Google Scholar]
  • 27.Crivellato E, Nico B, Vacca A, Ribatti D. B-cell non-Hodgkin's lymphomas express heterogeneous patterns of neovascularization. Haematologica. 2003;88:671–678. [PubMed] [Google Scholar]
  • 28.Crivellato E, Nico B, Vacca A, Ribatti D. Ultrastructural analysis of mast cell recovery after secretion by piecemeal degranulation in B-cell non-Hodgkin's lymphoma. Leuk Lymphoma. 2003;44:517–521. doi: 10.1080/1042819021000047001. [DOI] [PubMed] [Google Scholar]
  • 29.Ribatti D, Djonov V. Intussusceptive microvascular growth in tumors. Cancer Lett. 2012;316:126–131. doi: 10.1016/j.canlet.2011.10.040. [DOI] [PubMed] [Google Scholar]
  • 30.Hazar B, Paydas S, Zorludemir S, Sahin B, Tuncer I. Prognostic significance of microvessel density and vascular endothelial growth factor (VEGF) expression in non-Hodgkin's lymphoma. Leuk Lymphoma. 2003;44:2089–2093. doi: 10.1080/1042819031000123519. [DOI] [PubMed] [Google Scholar]
  • 31.Bairey O, Zimra Y, Kaganovsky E, Shaklai M, Okon E, Rabizadeh E. Microvessel density in chemosensitive and chemoresistant diffuse large B-cell lymphomas. Med Oncol. 2000;17:314–318. doi: 10.1007/BF02782197. [DOI] [PubMed] [Google Scholar]
  • 32.Jørgensen JM, Sorensen FB, Bendix K, Nielsen JL, Olsen ML, Funder AM, D'Amore F. Angiogenesis in non-Hodgkin's lymphoma: clinicopathological correlations and prognostic significance in specific subtypes. Leuk Lymphoma. 2007;48:584–595. doi: 10.1080/10428190601083241. [DOI] [PubMed] [Google Scholar]
  • 33.Ganjoo KN, An CS, Robertson MJ, Gordon LI, Sen JA, Weisenbach J, Li S, Weller EA, Orazi A, Horning SJ. Rituximab, bevacizumab and CHOP (RA-CHOP) in untreated diffuse large B-cell lymphoma: safety, biomarker and pharmacokinetic analysis. Leuk Lymphoma. 2006;47:998–1005. doi: 10.1080/10428190600563821. [DOI] [PubMed] [Google Scholar]
  • 34.Ruan J, Hyjek E, Kermani P, Christos PJ, Hooper AT, Coleman M, Hempstead B, Leonard JP, Chadburn A, Rafii S. Magnitude of stromal hemangiogenesis correlates with histologic subtype of non-Hodgkin's lymphoma. Clin Cancer Res. 2006;12:5622–5631. doi: 10.1158/1078-0432.CCR-06-1204. [DOI] [PubMed] [Google Scholar]
  • 35.Gratzinger D, Zhao S, Marinelli RJ, Kapp AV, Tibshirani RJ, Hammer AS, Hamilton-Dutoit S, Natkunam Y. Microvessel density and expression of vascular endothelial growth factor and its receptors in diffuse large B-cell lymphoma subtypes. Am J Pathol. 2007;170:1362–1369. doi: 10.2353/ajpath.2007.060901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gratzinger D, Zhao S, Tibshirani RJ, His ED, Hans CP, Pohlman B, Bast M, Avigdor A, Schiby G, Nagler A, et al. Prognostic significance of VEGF, VEGF receptors, and microvessel density in diffuse large B cell lymphoma treated with anthracycline-based chemotherapy. Lab Invest. 2008;88:38–47. doi: 10.1038/labinvest.3700697. [DOI] [PubMed] [Google Scholar]
  • 37.Vacca A, Moretti S, Ribatti D, Pellegrino A, Pimpinelli N, Bianchi B, Bonifazi E, Ria R, Serio G, Dammacco F. Progression of mycosis fungoides is associated with changes in angiogenesis and expression of the matrix metalloproteinases 2 and 9. Eur J Cancer. 1997;33:1685–1992. doi: 10.1016/s0959-8049(97)00186-x. [DOI] [PubMed] [Google Scholar]
  • 38.Schaerer L, Schmid MH, Mueller B, Dummer RG, Burg G, Kempf W. Angiogenesis in cutaneous lymphoproliferative disorders: microvessel density discriminates between cutaneous B-cell lymphomas and B-cell pseudolymphomas. Am J Dermatopathol. 2000;22:140–143. doi: 10.1097/00000372-200004000-00009. [DOI] [PubMed] [Google Scholar]
  • 39.Mazur G, WoŸniak Z, Wrobel T, Maj J, Kuliczkowski K. Increased angiogenesis in cutaneous T-cell lymphomas. Pathol Oncol Res. 2004;10:34–36. doi: 10.1007/BF02893406. [DOI] [PubMed] [Google Scholar]
  • 40.Salven P, Orpana A, Teerenhovi L, Joensuu H. Simultaneous elevation in the serum concentrations of the angiogenic growth factors VEGF and bFGF is an independent predictor of poor prognosis in non-Hodgkin lymphoma: a single-institution study of 200 patients. Blood. 2000;96:3712–3718. [PubMed] [Google Scholar]
  • 41.Niitsu N, Okamato M, Nakamine H, Yoshino T, Tamaru J, Nakamura S, Higashihara M, Hirano M. Simultaneous elevation of the serum concentrations of vascular endothelial growth factor and interleukin-6 as independent predictors of prognosis in aggressive non-Hodgkin's lymphoma. Eur J Haematol. 2002;68:91–100. doi: 10.1034/j.1600-0609.2002.01609.x. [DOI] [PubMed] [Google Scholar]
  • 42.Aref S, Mabed M, Zalata K, Sakrana M, El Askalany H. The interplay between c-Myc oncogene expression and circulating vascular endothelial growth factor (sVEGF), its antagonist receptor, soluble Flt-1 in diffuse large B cell lymphoma (DLBCL): relationship to patient outcome. Leuk Lymphoma. 2004;45:499–506. doi: 10.1080/10428190310001607151. [DOI] [PubMed] [Google Scholar]
  • 43.Doussis-Anagnostopoulou IA, Talks KL, Turley H, Debnam P, Tan DC, Mariatos G, Gorgoulis V, Kittas C, Gatter KC. Vascular endothelial growth factor (VEGF) is expressed by neoplastic Hodgkin-Reed-Sternberg cells in Hodgkin's disease. J Pathol. 2002;197:677–683. doi: 10.1002/path.1151. [DOI] [PubMed] [Google Scholar]
  • 44.Foss HD, Araujo I, Demel G, Klotzbach H, Hummel M, Stein H. Expression of vascular endothelial growth factor in lymphomas and Castleman's disease. J Pathol. 1997;183:44–50. doi: 10.1002/(SICI)1096-9896(199709)183:1<44::AID-PATH1103>3.0.CO;2-I. [DOI] [PubMed] [Google Scholar]
  • 45.Chen H, Treweeke AT, West DC, Till KJ, Cawley JC, Zuzel M, Toh CH. In vitro and in vivo production of vascular endothelial growth factor by chronic lymphocytic leukemia cells. Blood. 2000;96:3181–3187. [PubMed] [Google Scholar]
  • 46.Kay NE, Bone ND, Tschumper RC, Howell KH, Geyer SM, Dewald GW, Hanson CA, Jelinek DF. B-CLL cells are capable of synthesis and secretion of both pro- and anti-angiogenic molecules. Leukemia. 2002;16:911–919. doi: 10.1038/sj.leu.2402467. [DOI] [PubMed] [Google Scholar]
  • 47.Kuramoto K, Sakai A, Shigemasa K, Takimoto Y, Asaoku H, Tsujimoto T, Oda K, Kimura A, Uesaka T, Watanabe H, et al. High expression of MCL1 gene related to vascular endothelial growth factor is associated with poor outcome in non-Hodgkin's lymphoma. Br J Haematol. 2002;116:158–161. doi: 10.1046/j.1365-2141.2002.03253.x. [DOI] [PubMed] [Google Scholar]
  • 48.Zhao WL, Mourah S, Mounier N, Leboeuf C, Daneshpouy ME, Legrès L, Meignin V, Oksenhendler E, Maignin CL, Calvo F, et al. Vascular endothelial growth factor-A is expressed both on lymphoma cells and endothelial cells in angioimmunoblastic T-cell lymphoma and related to lymphoma progression. Lab Invest. 2004;84:1512–1519. doi: 10.1038/labinvest.3700145. [DOI] [PubMed] [Google Scholar]
  • 49.Shipp MA, Ross KN, Tamayo P, Weng AP, Kutok JL, Aguiar RC, Gaasenbeek M, Angelo M, Reich M, Pinkus GS, et al. Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat Med. 2002;8:68–74. doi: 10.1038/nm0102-68. [DOI] [PubMed] [Google Scholar]
  • 50.Takeuchi H, Matsuda K, Kitai R, Sato K, Kubota T. Angiogenesis in primary central nervous system lymphoma (PCNSL) J Neurooncol. 2007;84:141–145. doi: 10.1007/s11060-007-9363-x. [DOI] [PubMed] [Google Scholar]
  • 51.Nico B, Annese T, Tamma R, Longo V, Ruggieri S, Senetta B, Cassoni P, Specchia G, Vacca A, Ribatti D. Aquaporin-4 expression in primary human central nervous system lymphomas correlates with tumour cell proliferation and phenotypic heterogeneity of the vessel wall. Eur J Cancer. 2012;48:772–781. doi: 10.1016/j.ejca.2011.10.022. [DOI] [PubMed] [Google Scholar]
  • 52.Bairey O, Boycov O, Kaganovsky E, Zimra Y, Shaklai M, Rabizadeh E. All three receptors for vascular endothelial growth factor (VEGF) are expressed on B-chronic lymphocytic leukemia (CLL) cells. Leuk Res. 2004;28:243–248. doi: 10.1016/s0145-2126(03)00256-x. [DOI] [PubMed] [Google Scholar]
  • 53.Lee YK, Bone ND, Strege AK, Shanafelt TD, Jelinek DF, Kay NE. VEGF receptor phosphorylation status and apoptosis is modulated by a green tea component, epigallocatechin-3-gallate (EGCG), in B-cell chronic lymphocytic leukemia. Blood. 2004;104:788–794. doi: 10.1182/blood-2003-08-2763. [DOI] [PubMed] [Google Scholar]
  • 54.Lee YK, Shanafelts TD, Bone ND, Strege AK, Jelinek DK, Kay NE. VEGF receptors on chronic lymphocytic leukemia (CLL) B cells interact with STAT 1 and 3: implication for apoptosis resistance. Leukemia. 2005;19:513–523. doi: 10.1038/sj.leu.2403667. [DOI] [PubMed] [Google Scholar]
  • 55.Wang ES, Teruya-Feldstein J, Wu Y, Zhu Z, Hicklin DJ, Moore MA. Targeting autocrine and paracrine VEGF receptor pathways inhibits human lymphoma xenografts in vivo. Blood. 2004;104:2893–2902. doi: 10.1182/blood-2004-01-0226. [DOI] [PubMed] [Google Scholar]
  • 56.Stewart M, Talks K, Leek R, Turley H, Pezzella F, Harris A, Gatter K. Expression of angiogenic factors and hypoxia inducible factors HIF 1, HIF 2 and CA IX in non-Hodgkin's lymphoma. Histopathology. 2002;40:253–260. doi: 10.1046/j.1365-2559.2002.01357.x. [DOI] [PubMed] [Google Scholar]
  • 57.Ho CL, Sheu LF, Li CY. Immunohistochemical expression of basic fibroblast growth factor, vascular endothelial growth factor, and their receptors in stage IV non-Hodgkin lymphoma. Appl Immunohistochem Mol Morphol. 2002;10:316–321. doi: 10.1097/00129039-200212000-00005. [DOI] [PubMed] [Google Scholar]
  • 58.Vacca A, Ribatti D, Iurlaro M, Albini A, Minischetti M, Bussolino F, Pellegrino A, Ria R, Rusnati M, Presta M, et al. Human lymphoblastoid cells produce extracellular matrix-degrading enzymes and induce endothelial cell proliferation, migration, morphogenesis, and angiogenesis. Int J Clin Lab Res. 1998;2:55–68. doi: 10.1007/s005990050018. [DOI] [PubMed] [Google Scholar]
  • 59.Pazgal I, Zimr Y, Tzabar C, Okon E, Rabizadeh E, Shaklai M, Bairey O. Expression of basic fibroblast growth factor is associated with poor outcome in non-Hodgkin's lymphoma. Br J Cancer. 2002;86:1770–1775. doi: 10.1038/sj.bjc.6600330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ria R, Cirulli T, Giannini T, Bambace S, Serio G, Portaluri M, Ribatti D, Vacca A, Dammacco F. Serum levels of angiogenic cytokines decrease after radiotherapy in non-Hodgkin lymphomas. Clin Exp Med. 2008;8:141–145. doi: 10.1007/s10238-008-0170-2. [DOI] [PubMed] [Google Scholar]
  • 61.Ribatti D. The paracrine role of Tie-2-expressing monocytes in tumor angiogenesis. Stem Cells Dev. 2009;18:703–706. doi: 10.1089/scd.2008.0385. [DOI] [PubMed] [Google Scholar]
  • 62.De Palma M, Venneri MA, Roca C, Naldini L. Targeting exogenous genes to tumor angiogenesis by transplantation of genetically modified hematopoietic stem cells. Nat Med. 2003;9:785–795. doi: 10.1038/nm871. [DOI] [PubMed] [Google Scholar]
  • 63.Farinha P, Masoudi H, Skinnider BF, Shumansky K, Spinelli JJ, Gill K, Klasa R, Voss N, Connors JM, Gascoyne RD. Analysis of multiple biomarkers shows that lymphoma-associated macrophage (LAM) content is an independent predictor of survival in follicular lymphoma (FL) Blood. 2005;106:2169–2174. doi: 10.1182/blood-2005-04-1565. [DOI] [PubMed] [Google Scholar]
  • 64.Ribatti D, Vacca A, Marzullo A, Nico B, Ria R, Roncali L, Dammacco F. Angiogenesis and mast cell density with tryptase activity increase simultaneously with pathological progression in B-cell non-Hodgkin's lymphomas. Int J Cancer. 2000;85:171–175. [PubMed] [Google Scholar]
  • 65.Canioni D, Salles G, Mounier N, Brousse N, Keuppens M, Morchhauser F, Lamy T, Sonet A, Rousselet MC, Foussard C, et al. High numbers of tumor-associated macrophages have an adverse prognostic value that can be circumvented by rituximab in patients with follicular lymphoma enrolled onto the GELA-GOELAMS FL-2000 trial. J Clin Oncol. 2008;26:440–446. doi: 10.1200/JCO.2007.12.8298. [DOI] [PubMed] [Google Scholar]
  • 66.Taskinen M, Karjalainen-Lindsberg ML, Nyman H, Eerola LM, Leppä S. A high tumor-associated macrophage content predicts favorable outcome in follicular lymphoma patients treated with rituximab and cyclophosphamidedoxorubicin- vincristine-prednisone. Clin Cancer Res. 2007;13:5784–5789. doi: 10.1158/1078-0432.CCR-07-0778. [DOI] [PubMed] [Google Scholar]
  • 67.Ribatti D, Nico B, Vacca A, Marzullo A, Calvi N, Roncali L, Dammacco F. Do mast cells help to induce angiogenesis in B-cell non-Hodgkin's lymphomas? Br J Cancer. 1998;77:1900–1906. doi: 10.1038/bjc.1998.316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Crivellato E, Nico B, Vacca A, Dammacco F, Ribatti D. Mast cell heterogeneity in B-cell non-Hodgkin's lymphomas: an ultrastructural study. Leuk Lymphoma. 2002;43:2201–2205. doi: 10.1080/1042819021000016159. [DOI] [PubMed] [Google Scholar]
  • 69.Ribatti D, Crivellato E. Mast cells, angiogenesis, and tumour growth. Biochim Biophys Acta. 2012;1822:2–8. doi: 10.1016/j.bbadis.2010.11.010. [DOI] [PubMed] [Google Scholar]
  • 70.Ribatti D, Molica S, Vacca A, Nico B, Crivellato E, Roccaro AM, Dammacco F. Tryptase-positive mast cells correlate positively with bone marrow angiogenesis in B-cell chronic lymphocytic leukemia. Leukemia. 2003;17:1428–1430. doi: 10.1038/sj.leu.2402970. [DOI] [PubMed] [Google Scholar]
  • 71.Molica S, Montillo M, Ribatti D, Mirabelli R, Tedeschi A, Ricci F, Veronese S, Vacca A, Morra E. Intense reversal of bone marrow angiogenesis after sequential fludarabine-induction and alemtuzumab-consolidation therapy in advanced chronic lymphocytic leukemia. Haematologica. 2007;92:1367–1374. [PubMed] [Google Scholar]
  • 72.Ribatti D. The discovery of endothelial progenitor cells. An historical review. Leuk Res. 2007;31:439–444. doi: 10.1016/j.leukres.2006.10.014. [DOI] [PubMed] [Google Scholar]
  • 73.Igreja C, Courinha M, Cachaco AS, Pereira T, Cabecadas J, da Silva MG, Dias S. Characterization and clinical relevance of circulating and biopsyderived endothelial progenitor cells in lymphoma patients. Haematologica. 2007;92:469–477. doi: 10.3324/haematol.10723. [DOI] [PubMed] [Google Scholar]
  • 74.Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, Chadburn A, Heissig B, Marks W, Witte L, et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med. 2001;7:1194–1201. doi: 10.1038/nm1101-1194. [DOI] [PubMed] [Google Scholar]
  • 75.Monestiroli S, Mancuso P, Burlini A, Pruneri G, Dell'Agnola C, Gobbi A, Martinelli G, Bertolini F. Kinetics and viability of circulating endothelial cells as surrogate angiogenesis marker in an animal model of human lymphoma. Cancer Res. 2001;61:4341–4344. [PubMed] [Google Scholar]
  • 76.Mancuso P, Burlini A, Pruneri G, Goldhirsch A, Martinelli G, Bertolini F. Resting and activated endothelial cells are increased in the peripheral blood of cancer patients. Blood. 2001;97:3658–3661. doi: 10.1182/blood.v97.11.3658. [DOI] [PubMed] [Google Scholar]
  • 77.Seto M. Genomic profiles in B cell lymphoma. Int J Hematol. 2010;92:238–245. doi: 10.1007/s12185-010-0662-1. [DOI] [PubMed] [Google Scholar]
  • 78.Streubel B, Chott A, Huber D, Exner M, Jäger U, Wagner O, Schwarzinger I. Lymphoma-specific genetic aberrations in microvascular endothelial cells in B-cell lymphomas. N Engl J Med. 2004;351:250–259. doi: 10.1056/NEJMoa033153. [DOI] [PubMed] [Google Scholar]
  • 79.Huang X, Bai X, Cao Y, Wu J, Huang M, Tang D, Tao S, Zhu T, Liu Y, Yang Y, et al. Lymphoma endothelium preferentially expresses Tim-3 and facilitates the progression of lymphoma by mediating immune evasion. J Exp Med. 2010;207:505–520. doi: 10.1084/jem.20090397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Folkman J. Antiangiogenesis in cancer therapy—endostatin and its mechanisms of action. Exp Cell Res. 2006;312:594–607. doi: 10.1016/j.yexcr.2005.11.015. [DOI] [PubMed] [Google Scholar]
  • 81.Standker L, Schrader M, Kanse SM, Jurgens M, Forssmann WG, Preissner KT. Isolation and characterization of the circulating form of human endostatin. FEBS Lett. 1997;420:129–133. doi: 10.1016/s0014-5793(97)01503-2. [DOI] [PubMed] [Google Scholar]
  • 82.Bono P, Teerenhovi L, Joensuu H. Elevated serum endostatin is associated with poor outcome in patients with non-Hodgkin lymphoma. Cancer. 2003;97:2762–2775. doi: 10.1002/cncr.11399. [DOI] [PubMed] [Google Scholar]
  • 83.Bertolini F, Fusetti L, Mancuso P, Gobbi A, Corsini C, Ferrucci PF, Martinelli G, Pruneri G. Endostatin, an antiangiogenic drug, induces tumor stabilization after chemotherapy or anti-CD20 therapy in a NOD/SCID mouse model of human high-grade non-Hodgkin lymphoma. Blood. 2000;96:282–287. [PubMed] [Google Scholar]
  • 84.Capillo M, Mancuso P, Gobbi A, Monestiroli S, Pruneri G, Dell'Agnola C, Martinelli G, Shultz L, Bertolini F. Continuous infusion of endostatin inhibits differentiation, mobilization, and clonogenic potential of endothelial cell progenitors. Clin Cancer Res. 2003;9:377–382. [PubMed] [Google Scholar]
  • 85.Ribatti D, Vacca A. Therapeutic renaissance of thalidomide in the treatment of haematological malignancies. Leukemia. 2005;19:1523–1531. doi: 10.1038/sj.leu.2403852. [DOI] [PubMed] [Google Scholar]
  • 86.Smith SM, Grinblatt D, Johnson JL, Niedzwiecki D, Rizzieri D, Bartlett NL, Cheson BD. Cancer and Leukemia Group B Thalidomide has limited single-agent activity in relapsed or refractory indolent non-Hodgkin lymphomas: a phase II trial of the Cancer and Leukemia Group B. Br J Haematol. 2008;140:313–319. doi: 10.1111/j.1365-2141.2007.06937.x. [DOI] [PubMed] [Google Scholar]
  • 87.Pro B, Younes A, Albitar M, Dang NH, Samaniego F, Romaguera J, McLaughlin P, Hagemeister FB, Rodriguez MA, Clemons M, et al. Thalidomide for patients with recurrent lymphoma. Cancer. 2004;100:1186–1189. doi: 10.1002/cncr.20070. [DOI] [PubMed] [Google Scholar]
  • 88.Chanan-Khan A, Miller KC, Takeshita K, Koryzna A, Donohue K, Bernstein ZP, Mohr A, Klippenstein D, Wallace P, Zeldis JB, et al. Results of a phase 1 clinical trial of thalidomide in combination with fludarabine as initial therapy for patients with treatment-requiring chronic lymphocytic leukemia (CLL) Blood. 2005;106:3348–3352. doi: 10.1182/blood-2005-02-0669. [DOI] [PubMed] [Google Scholar]
  • 89.Strupp C, Aivado M, Germing U, Gattermann N, Haas R. Angioimmunoblastic lymphadenopathy (AILD) may respond to thalidomide treatment: two case reports. Leuk Lymphoma. 2002;43:133–137. doi: 10.1080/10428190210190. [DOI] [PubMed] [Google Scholar]
  • 90.Dogan A, Ngu LSP, Ng SH, Cervi PL. Pathology and clinical features of angioimmunoblastic T-cell lymphoma after successful treatment with thalidomide. Leukemia. 2005;19:873–875. doi: 10.1038/sj.leu.2403710. [DOI] [PubMed] [Google Scholar]
  • 91.Wiernik PH, Lossos IS, Tuscano JM, Justice G, Vose JM, Cole CE, Lam W, McBride K, Wride K, Pietronigro D, et al. Lenalidomide monotherapy in relapsed or refractory aggressive non-Hodgkin's lymphoma. J Clin Oncol. 2008;26:1–6. doi: 10.1200/JCO.2007.15.3429. [DOI] [PubMed] [Google Scholar]
  • 92.Habermann TM, Lossos IS, Justice G, Vose JM, Wiernik PH, McBride K, Wride K, Ervin-Haynes A, Takeshita K, Pietronigro D, et al. Lenalidomide oral monotherapy produces a high response rate in patients with relapsed or refractory mantle cell lymphoma. Br J Haematol. 2009;145:344–349. doi: 10.1111/j.1365-2141.2009.07626.x. [DOI] [PubMed] [Google Scholar]
  • 93.Witzig TE, Wiernik PH, Moore T, Reeder C, Cole C, Justice G, Kaplan H, Voralia M, Pietronigro D, Takeshita K, et al. Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin's lymphoma. J Clin Oncol. 2009;27:5404–5409. doi: 10.1200/JCO.2008.21.1169. [DOI] [PubMed] [Google Scholar]
  • 94.Czuczman MS, Vose JM, Witzig TE, Zinzani PL, Buckstein R, Polikoff J, Li J, Pietronigro D, Ervin-Haynes A, Reeder CB. The differential effect of lenalidomide monotherapy in patients with relapsed or refractory transformed non-Hodgkin lymphoma of distinct histological origin. Br J Haematol. 2011;154:477–481. doi: 10.1111/j.1365-2141.2011.08781.x. [DOI] [PubMed] [Google Scholar]
  • 95.Chanan-Khan A, Miller KC, Musial L, Lawrence D, Padmanabhan S, Takeshita K, Porter CW, Goodrich DW, Bernstein ZP, Wallace P, et al. Clinical efficacy of lenalidomide in patients with relapsed or refractory chronic lymphocytic leukemia: results of a phase II study. J Clin Oncol. 2006;24:5343–5349. doi: 10.1200/JCO.2005.05.0401. [DOI] [PubMed] [Google Scholar]
  • 96.Ferrajoli A, Lee BN, Schlette EJ, O'Brien SM, Gao H, Wen S, Wierda WG, Estrov Z, Faderl S, Cohen EN, et al. Lenalidomide induces complete and partial remissions in patients with relapsed and refractory chronic lymphocytic leukemia. Blood. 2008;111:5291–5297. doi: 10.1182/blood-2007-12-130120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kaufmann H, Raderer M, Wohrer S, Püspök A, Bankier A, Zielinski C, Chott A, Drach J. Antitumor activity of rituximab plus thalidomide in patients with relapsed/refractory mantle cell lymphoma. Blood. 2004;104:2269–2271. doi: 10.1182/blood-2004-03-1091. [DOI] [PubMed] [Google Scholar]
  • 98.Adams J. The proteasome: a suitable antineoplastic target. Nat Rev Cancer. 2004;4:349–360. doi: 10.1038/nrc1361. [DOI] [PubMed] [Google Scholar]
  • 99.Goy A, Younes A, McLaughlin P, Pro B, Romaguera JE, Hagemeister F, Fayad L, Dang NH, Samaniego F, Wang M, et al. Phase II study of proteasome inhibitor bortezomib in relapsed or refractory B-cell non-Hodgkin's lymphoma. J Clin Oncol. 2005;23:667–675. doi: 10.1200/JCO.2005.03.108. [DOI] [PubMed] [Google Scholar]
  • 100.Fisher RI, Bernstein SH, Kahl BS, Djulbegovic B, Robertson MJ, de Vos S, Epner E, Krishnan A, Leonard JP, Lonial S, et al. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2006;24:4867–4874. doi: 10.1200/JCO.2006.07.9665. [DOI] [PubMed] [Google Scholar]
  • 101.Belch A, Kouroukis CT, Crump M, Sehn L, Gascovne RD, Klasa R, Powers J, Wright J, Eisenhauer EA. A phase II study of bortezomib in mantle cell lymphoma: the National Cancer Institute of Canada Clinical Trials Group trial IND.150. Ann Oncol. 2007;18:116–121. doi: 10.1093/annonc/mdl316. [DOI] [PubMed] [Google Scholar]
  • 102.de Vos S, Goy A, Dakhil SR, Saleh MN, McLaughlin P, Belt R, Flowers CR, Knapp M, Hart L, Patel-Donnelly D, et al. Multicenter randomized phase II study of weekly or twice-weekly bortezomib plus rituximab in patients with relapsed or refractory follicular or marginal-zone B-cell lymphoma. J Clin Oncol. 2009;27:5023–5030. doi: 10.1200/JCO.2008.17.7980. [DOI] [PubMed] [Google Scholar]
  • 103.O'Connor OA, Portlock C, Moskowitz C, Hamlin P, Straus D, Gerecitano J, Gonen M, Dumitrescu O, Sarasohn D, Butos J, et al. Time to treatment response in patients with follicular lymphoma treated with bortezomib is longer compared with other histologic subtypes. Clin Cancer Res. 2010;16:719–726. doi: 10.1158/1078-0432.CCR-08-2647. [DOI] [PubMed] [Google Scholar]
  • 104.Sehn LH, MacDonald D, Rubin S, Cantin G, Rubinger M, Lemieux B, Basi S, Imrie K, Gascoyne RD, Sussman J, et al. Bortezomib added to R-CVP is safe and effective for previously untreated advanced-stage follicular lymphoma: a phase II study by the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2011;29:3396–3401. doi: 10.1200/JCO.2010.33.6594. [DOI] [PubMed] [Google Scholar]
  • 105.Stopeck AT, Unger JM, Rimsza LM, Bellamy WT, Iannone M, Persky DO, Leblanc M, Fisher RI, Miller TP. A phase II trial of single agent bevacizumab in patients with relapsed, aggressive non-Hodgkin lymphoma: Southwest Oncology Group study S0108. Leuk Lymphoma. 2009;50:728–735. doi: 10.1080/10428190902856808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Farohani M, Treweeke AT, Toh CH, Till KJ, Harris RJ, Cawley JC, Zuzel M, Chen H. Autocrine VEGF mediates the antiapoptotic effect of CD154 on CLL cells. Leukemia. 2005;19:524–530. doi: 10.1038/sj.leu.2403631. [DOI] [PubMed] [Google Scholar]
  • 107.Till KJ, Spiller DG, Harris RJ, Chen H, Zuzel M, Cawley JC. CLL, but not normal, B cells are dependent on autocrine VEGF and α4β1 integrin for chemokine-induced motility on and through endothelium. Blood. 2005;105:4813–4819. doi: 10.1182/blood-2004-10-4054. [DOI] [PubMed] [Google Scholar]
  • 108.Rasheed W, Bishton M, Johnstone RW, Prince HM. Histone deacetylase inhibitors in lymphoma and solid malignancies. Expert Rev Anticancer Ther. 2008;8:413–432. doi: 10.1586/14737140.8.3.413. [DOI] [PubMed] [Google Scholar]
  • 109.Richon VM, O'Brien JP. Histone deacetylase inhibitors: a new class of potential therapeutic agents for cancer treatment. Clin Cancer Res. 2002;8:662–664. [PubMed] [Google Scholar]
  • 110.Medina V, Edmonds B, Young GP, James R, Appleton S, Zalewski PD. Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway. Cancer Res. 1997;57:3697–3707. [PubMed] [Google Scholar]
  • 111.Deaoanne CF, Bonjeean K, Servotte S, Devy L, Colige A, Clausse N, Blacher S, Verdin E, Foidart JM, Nusgens BV, et al. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene. 2002;21:427–436. doi: 10.1038/sj.onc.1205108. [DOI] [PubMed] [Google Scholar]
  • 112.Duvic M, Talpur R, Ni X, Zhang C, Hazarika P, Kelly C, Chiao JH, Reilly JF, Ricker JL, Richon VM, et al. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL) Blood. 2007;109:31–39. doi: 10.1182/blood-2006-06-025999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Crump M, Coiffier B, Jacobsen ED, Sun L, Ricker JL, Xie H, Frankel SR, Randolph SS, Cheson BD. Phase II trial of oral vorinostat (suberoylanilide hydroxamic acid) in relapsed diffuse large-B-cell lymphoma. Ann Oncol. 2008;19:964–969. doi: 10.1093/annonc/mdn031. [DOI] [PubMed] [Google Scholar]
  • 114.Ellis L, Pan Y, Smyth GK, George DJ, McCormack C, Williams-Truax R, Mita M, Beck J, Burris H, Ryan G, et al. Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res. 2008;14:4500–4510. doi: 10.1158/1078-0432.CCR-07-4262. [DOI] [PubMed] [Google Scholar]
  • 115.Heider U, Kaiser M, Sterz J, Zavrski I, Jakob C, Fleissner C, Eucker J, Possinger K, Sezer O. Histone deacetylase inhibitors reduce VEGF production and induce growth suppression and apoptosis in human mantle cell lymphoma. Eur J Haematol. 2006;76:42–50. doi: 10.1111/j.1600-0609.2005.00546.x. [DOI] [PubMed] [Google Scholar]

Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES