Skip to main content
Erschienen in: Acta Neuropathologica 6/2012

Open Access 01.12.2012 | Review

Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited

verfasst von: Karl H. Plate, Alexander Scholz, Daniel J. Dumont

Erschienen in: Acta Neuropathologica | Ausgabe 6/2012

Abstract

The cellular and molecular mechanisms of tumor angiogenesis and its prospects for anti-angiogenic cancer therapy are major issues in almost all current concepts of both cancer biology and targeted cancer therapy. Currently, (1) sprouting angiogenesis, (2) vascular co-option, (3) vascular intussusception, (4) vasculogenic mimicry, (5) bone marrow-derived vasculogenesis, (6) cancer stem-like cell-derived vasculogenesis and (7) myeloid cell-driven angiogenesis are all considered to contribute to tumor angiogenesis. Many of these processes have been described in developmental angiogenesis; however, the relative contribution and relevance of these in human brain cancer remain unclear. Preclinical tumor models support a role for sprouting angiogenesis, vascular co-option and myeloid cell-derived angiogenesis in glioma vascularization, whereas a role for the other four mechanisms remains controversial and rather enigmatic. The anti-angiogenesis drug Avastin (Bevacizumab), which targets VEGF, has become one of the most popular cancer drugs in the world. Anti-angiogenic therapy may lead to vascular normalization and as such facilitate conventional cytotoxic chemotherapy. However, preclinical and clinical studies suggest that anti-VEGF therapy using bevacizumab may also lead to a pro-migratory phenotype in therapy resistant glioblastomas and thus actively promote tumor invasion and recurrent tumor growth. This review focusses on (1) mechanisms of tumor angiogenesis in human malignant glioma that are of particular relevance for targeted therapy and (2) controversial issues in tumor angiogenesis such as cancer stem-like cell-derived vasculogenesis and bone-marrow-derived vasculogenesis.

Introduction

In his landmark 1971 publication, Folkman [41] developed the idea that tumor growth is angiogenesis dependent and described for the first time the potential prospects of anti-angiogenic cancer therapy. From thereon, it took 13 years before fibroblast growth factor (FGF)-2, the first heparin-binding angiogenic growth factor, was identified and 18 years before vascular endothelial growth factor (VEGF), which by now has turned out to be the single most important angiogenesis factor in both health and disease, was described (for review see [38]). Napoleone Ferrara, one of the discoverers of VEGF, developed monoclonal antibodies to VEGF [70] that blocked tumor growth in vivo [71] and that paved the way for the development of bevacizumab, a monoclonal anti-VEGF antibody that is now used in a large number of clinical cancer trials, including glioblastoma, where anti-angiogenesis represents one treatment arm [104, 131]. It is now evident that the entire process of tumor-induced angiogenesis appears to be far more complex than initially envisioned [15]. Moreover, the idea that blockade of tumor angiogenesis is able to inhibit tumor growth in vivo has been confirmed in principal in both experimental and clinical settings; however, current evidence suggests that cancer cells are able to circumvent anti-angiogenic therapy and develop resistance to targeted mono-therapy [7, 129].

Cell biology of tumor angiogenesis

In a recent Cell snapshot Jain and Carmeliet listed six different principal cellular mechanisms under the heading of tumor angiogenesis. These include (1) classical sprouting angiogenesis, (2) vascular co-option, (3) vessel intussusception, (4) vasculogenic mimicry, (5) bone marrow derived vasculogenesis and (6) cancer stem-like cell derived vasculogenesis [62]. Another important mechanism of tumor angiogenesis is angiogenesis driven by blood derived infiltrating myeloid cells, a process that recently received considerable attention. Whether the above listed mechanisms of tumor angiogenesis (for overview see Fig. 1) are all operational in human glioblastoma is not entirely clear. Here, we briefly discuss the above mentioned angiogenesis mechanisms and review the existing evidence for their role in glioma biology.
Angiogenesis, the sprouting of capillaries from pre-existing vessels, has since long been considered the principal mechanism of brain vascularization, both during development and in brain cancer [80, 81, 111, 121]. A vascular sprout that is led by a filopodia-rich tip cell migrates toward an angiogenic stimulus (often VEGF) that is produced by tumor cells. Following the tip are a group of cells, entitled stalk cells, that divide, which promotes the elongation of the sprout (Fig. 2). Tip cells from two distinct sprouts may fuse to yield a larger sprout, a process that is thought to be modulated by the action of macrophages on these activated endothelial cells (Fig. 4). Thereafter, the newly formed vessel remodels to form a vascular lumen (that permits blood flow) and to attract mural cells that stabilize the vessel (reviewed in [16, 143]). Current evidence suggests that vascular sprouting represents a major mechanism of tumor angiogenesis. Analysis of cell division in glioblastoma, by MIB-1 staining, suggests that there are a considerable number of vascular endothelial cells undergo cell division suggesting that these cells may represent stalk cells. In addition to this, molecules typically expressed on endothelial tip cells such as VEGFR-2, Neuropilin-1, Angiopoietin-2 (Ang-2), Integrin-ß1 and others have been described to be present in glioma vessels in situ [11, 31, 114], implying that tip and stalk cell phenotypes co-exist in the glioblastoma vasculature.
Vascular co-option describes the infiltration of tumor cells into normal tissue and the adoption of the pre-existing vasculature [56]. When one takes this into consideration, vascular co-option may be viewed as part of the invasive phenotype that is intrinsic to all diffuse gliomas—rather than an active vascular process. Invoking the process of vascular co-option aligns well with the known migratory pattern of tumor cells along vessels which has been observed in numerous experimental glioma models in rodents. Vascular co-option may facilitate the infiltration of human gliomas. Of note, anti-VEGF treatment may drive glioma cells to utilize the co-option pathway thereby circumventing the impact of anti-VEGF treatment resulting in an increase in the number of migrating glioma cells, that may use pre-existing vessels as scaffolds for their migration pathways (see below).
Vessel intussusception describes the formation of a new vessel by vascular invagination, intra-luminar pillar formation and splitting. Vascular intussusception has initially been described in physiological vascular development [32] but more recently has been expanded to experimental tumors. It has been suggested that sprouting angiogenesis may switch to vascular intussusception to allow rapid development of new vessels [119]. The molecular mechanisms that drive vascular intussusception are currently poorly understood and whether intussusception occurs or plays a role in human glioma or tumor biology in general is currently unclear.
Vasculogenic mimicry is defined as a process where tumor cells replace endothelial cells and form a vessel with a lumen This phenomenon was first described (mainly based on PAS staining) in a subset of aggressive uveal melanomas [95] and later in other types of cancer but its overall occurrence and significance, if any, is highly controversial. Moreover, it is unclear whether vasculogenic mimicry represents an active process (e.g., cancer cells actively forming the vascular lumen) or whether vasculogenic mimicry is a consequence of vessel regression. Vessel regression is part of the vascular remodeling process that takes place in development but can also occur in tumors (Fig. 2a). During development, a normal and well-orchestrated realignment of the balance between growth/survival signals such as high levels of Ang-2 together with low levels of VEGF leads to vessel regression and endothelial cell apoptosis causing vessel pruning and vessel remodeling [50]. A similar situation (high levels of Ang-2 combined with low levels of VEGF) can occur in cancer after bevacizumab therapy and may therefore be responsible for intratumoral vessel regression. In addition to programmed vessel regression, cessation of blood flow due to development of shunts or the thrombosis of tumor vessels can lead to tumor vessel regression via down-regulation of the transcription factor kruppel-like factor2 (KLF2) and subsequent up-regulation of Ang-2 (Fig. 2b) [74, 145]. McDonald and colleagues have discussed the phenomenon of vasculogenic mimicry in detail and came to the conclusion that even in the prototype of cancer that shows vasculogenic mimicry, aggressive uveal melanomas, vascular channels are almost always lined by endothelial cells and not by cancer cells [98, 99], leading these authors generally doubt the significance of vasculogenic mimicry in tumor angiogenesis. Studies dealing with vasculogenic mimicry in human gliomas are sparse and mainly rely on PAS-staining to identify cancer cell-lined vascular channels. In one study, PAS-positive vascular channels devoid of CD34+ cells have been described in 2/45 human glioma specimens [155], and in 13 out of 101 samples in another study [89]. Recently, based on double-immunofluorescence staining, co-expression of GFAP (a marker for glioma cells) and VEGFR-2 (a marker for endothelial cells) has been reported in 7 out of 11 human GBM investigated and has been considered as evidence for vasculogenic mimicry [44], but this awaits confirmation. In summary, the functional and clinical relevance of vasculogenic mimicry appears questionable in light of the methodological problems encountered. Rather, we prefer to conclude from the data published that vasculogenic mimicry appears to be of little significance in glioblastoma angiogenesis, since most vessels appear to be lined by a proper endothelium on the luminal side.
Bone marrow-derived vasculogenesis describes the recruitment of circulating endothelial precursor cells to the tumor, their integration into the vessel wall and their terminal differentiation into an endothelial cell. From the onset of the original description of tumor vasculogenesis [2], this phenomenon has attracted a considerable amount of attention. This attention has stemmed from the fact that it has challenged the previous dogma that the postnatal vasculature (under both physiological and pathological conditions) can only proceed via angiogenesis but not vasculogenesis in order to adapt to altered physiological needs [121]. Mouse molecular studies that used chimeric mice with GFP-tagged bone marrow cells together with high-resolution confocal imaging and 3D-reconstruction revealed however that BM-derived endothelial cells in experimental gliomas represent only a small amount (less than 1 %) of all vascular endothelial cells [93]. However, it has been suggested that vasculogenesis is important for neovascularization after tumor irradiation [72]. Again, this issue has lead to a controversy since work from several laboratories suggests that bone marrow-derived cells do not incorporate into the vessel lumen to a significant extent but rather stay adjacent to the vessel, in a perivascular location [75]. These results suggest that bone marrow-derived cells support angiogenesis in a paracrine manner but presumably not by incorporation into the endothelial cell layer proper [76]. This imprecise use of the term “vasculogenesis” has facilitated the confusion in the literature about the process of tumor vasculogenesis [75]. Taken together, “real” tumor vasculogenesis appears to be a rare biological event that probably does not bear any clinical significance in human glioblastoma.
Cancer stem-like cell derived vasculogenesis: In 2010, two groups independently reported the transdifferentiation of GBM-derived stem-like cells into endothelial cells in vitro [120, 148]. The authors showed further that a proportion of vascular cells within human GBM contained genetic alterations that are typically encountered in glioblastoma cells (such as EGFR amplification) and are typically not seen in vascular endothelial cells. One group even reported that the majority of GBM vascular cells contained genetic alterations typically found in tumor cells [120]. The authors concluded that GBM-derived cancer stem-like cells can contribute to the vasculature by integrating into the vascular wall and transdifferentiate into endothelial cells, while retaining their genetic alterations. In contrast to this, recent work by Rodriguez [123] who examined GBM by detailed histological analysis demonstrated that in GBM (1) mutant vascular cells are extremely rare and (2) these cells are usually found in the perivascular space or vascular wall but were not lining the vessel lumen. Taken together, the existence of cancer stem-like cell-derived vasculogenesis in human glioblastoma is highly controversial and at best appears to be a very rare event.
The participation of bone marrow derived cells in driving tumor angiogenesis has achieved considerable attention recently (for reviews see [47, 66]). In particular, monocytes/macrophages can polarize into phenotypes that exert different functions in vivo. In contrast to M1 monocytes/macrophages, that are anti-tumorigenic, M2-polarized monocytes/macrophages are pro-angiogenic and thus drive tumor growth [135]. Current evidence suggests that monocytes/macrophages secrete a variety of pro-tumorigenic and pro-angiogenic growth factors (including VEGF). Experimental studies in glioma models suggest that blocking of monocyte/macrophage recruitment and/or function may block glioma growth (see below). As such, monocytes/macrophages represent an attractive target for anti-angiogenic tumor therapy although the relevance of bone marrow driven angiogenesis in human glioma still needs to be determined.

Angiogenic signaling pathways in glioblastoma

Signaling pathways initiated from numerous growth factor receptors are known to play pivotal roles in developmental and tumor-mediated angiogenesis. In this review, we focus on three signaling pathways with a proven role in vascular biology and/or tumor angiogenesis that are currently being explored in clinical trials (Table 1) or represent major pharmaceutical targets that are under intense scrutiny. In addition to these angiogenic factors, further angiogenic signaling pathways are provided by axon guidance molecules that are known to play a role in both vascular and neuronal patterning (for recent review see [1, 37]. However, due to their prominent role in nervous system development and homeostasis it appears questionable whether these molecules will become targets for anti-angiogenic glioma therapy (see below and [115]).
Table 1
Current clinical trials targeting the VEGF/VEGFR or Ang/Tie2 signaling pathways in glioblastoma
Compound
Target
Combination
Phase
NCT number
Status
Company and/or trial center
Aflibercept
VEGF
Temozolomide Radiation
I
NCT00650923
Active
Sidney Kimmel Comprehensive Cancer Center
 
II
NCT00369590
Completed
Sidney Kimmel Comprehensive Cancer Center
AZD2171 (Cediranib Maleate, Recentin)
VEGF
Bevacizumab
I
NCT00458731
Active
M.D. Anderson Cancer Center
Bevacizumab
VEGF
Temozolomide
II
NCT00590681
Active
Genentech
Erlotinib
II
NCT00671970
Active
Genentech
Irinotecan Voronistat
I
NCT00762255
Active
Merck
Radiation Temozolomide
II
NCT01478321
Active
Northwestern University
Temozolomide
II
NCT01013285
Active
UCLA
 
II
NCT00271609
Active
National Cancer Institute
Temsirolimus
II
NCT00800917
Completed
Roch/Pfizer
Cetuximab Irinotecan
II
NCT00463073
Completed
 
Bevacizumab
VEGF
Irinotecan
II
NCT00463203
Completed
 
TPI287
I/II
NCT01582152
Recruiting
Archer Biosciences, Inc.
Voronistat
I/II
NCT01266031
Recruiting
Genentech
Temozolomide
II
NCT01149850
Recruiting
Genentech
Radiosurgery
I/II
NCT01086345
Recruiting
Case Comprehensive Cancer Center
Lomustine
II
NCT01067469
Recruiting
M.D. Anderson Cancer Center
Stereotactic Radiotherapy
I
NCT01392209
Recruiting
Genentech
Lenalidomide
I
NCT01183663
Recruiting
M.D. Anderson Cancer Center
Rindopepimut/GM-CSF
II
NCT01498328
Recruiting
Celldex Therapeutics
Irinotecan
II
NCT00393094
Terminated
National Cancer Institute
AEE788
VEGFR, EGFR/ErbB2
 
I/II
NCT00116376
Completed
Novartis
BIBF 1120
VEGFR, PDGFR, FGFR
 
II
NCT01251484
Active
Boehringer Ingelheim Pharmaceuticals University of Copenhagen
Pazopanib
VEGFR-1/2/3, PDGFR, c-Kit
Lapatinib
II
NCT00350727
Completed
GlaxoSmithKline
AMG 386 (Trebanabnib)
Ang-1/2
Bevacizumab
I/II
NCT01290263
Suspended
Amgen
 
I
NCT01538095
Recruiting
National Cancer Institute
Bevacizumab
II
NCT01609790
Recruiting
National Cancer Institute (NCI)
PF- 04856884 (CVX-060)
Ang-2
 
II
NCT01225510
Withdrawn
Pfizer
Ang Angiopoietin, VEGF vascular endothelial growth factor, VEGFR vascular endothelial growth factor receptor

VEGF signaling

VEGF is the principal angiogenesis factor in both embryonic development and tumor growth (for review see [38]). Deletion of either VEGF or VEGFR-1 or VEGFR-2 leads to embryonic lethality in the mouse due to severe defects in the developing vascular system [14, 42, 43, 133]. A role for VEGF in cerebral vascularization is illustrated by analysis of the conditional deletion of VEGF in the nervous system using a Nestin-cre/VEGF flox binary transgenic mouse to specifically delete VEGF in all Nestin-expressing cells. Mice carrying this deletion have a severe phenotype where the primitive perineural vascular plexus is able to develop, but vascular sprouts do not penetrate into the primitive avascular neuroectoderm. These mice have an avascular brain that causes microcephaly and massive periventricular apoptosis [116].
In GBM, VEGF is highly up-regulated, in particular in perinecrotic pseudopalisading cells [113, 134]. A major driving force of VEGF expression in GBM appears to be tumor hypoxia since VEGF and other hypoxia-inducible genes such as HIF-1 alpha and LDH are expressed in the same cell type [156]. Data derived from The Cancer Genome Atlas (TCGA) (http://​cancergenome.​nih.​gov) suggest that in GBM the entire VEGF/VEGF receptor system is predominantly orchestrated by the up-regulation of VEGF; however, the VEGF receptors-1, -2 and -3 are also up-regulated in the tumor vasculature. The vast majority of clinical trials that target tumor angiogenesis use drugs that target the VEGF signaling pathway, either by blocking VEGF or by interfering with VEGFR-2 signaling (see Table 1 and for review [100]). The most advanced VEGF-specific drug currently on the market is bevacizumab, a monoclonal antibody that blocks VEGF function. Bevacizumab is routinely used in combination with chemotherapy (e.g., irinotecan) for the treatment of recurrent glioblastoma [19, 142, 147]. Unfortunately, several recent clinical reports suggest that anti-VEGF treatment may in fact promote a pro-migratory cellular tumor phenotype in patients with recurrent glioblastoma (for example [24, 102]). Importantly, these clinical observations are supported by several pre-clinical studies that in principle show that anti-VEGF treatment in mice or rats promotes a pro-invasive phenotype and may even increase tumor metastasis [35, 68, 107]. A possible explanation for this phenomenon, which represents a major challenge and clinical drawback, might be the induction of hypoxia and up-regulation of hypoxia-inducible genes via the HIF-1 alpha pathway [117]. Indeed, it has been suggested that hypoxia-mediated migration of tumor cells is responsible for the development of “pseudopalisading” glioma cells surrounding necroses, a histological hallmark of glioblastoma [10]. As such, these cells may represent glioma cells that are hypoxic and “try to escape” from a low oxygen microenvironment [110, 114]. The molecular mechanisms underlying this event are only partly understood. The protooncogene c-met, for example, has been shown to be up-regulated by hypoxia and to trigger invasion [9, 36, 108]. Recently, it has been suggested that c-met is activated in GBM upon bevacizumab treatment in a VEGFR-2 and phosphotyrosine phosphatase (PTP1B)-dependent manner. In this scenario, bevacizumab-induced depletion of VEGF reduces PTP1B activity and promotes c-met phosphorylation. This mechanism may therefore account at least in part for the observed therapy-induced switch to a pro-migratory phenotype after bevacizumab treatment [20, 92]. However, this putative mechanism requires the co-expression of c-met together with VEGFR-2 on glioma cells in vivo. Whereas VEGFR-2 can be readily detected in glioma cell lines in vitro, most studies suggest that in both murine and human glioma specimens in vivo expression of VEGFR-2 is mainly confined to vascular endothelial cells [51, 52, 77, 87, 112114, 136], and potentially to a subset of cancer stem-like cells [49]. As such, it remains currently unclear whether the mechanism of bevacizumab-induced migration of glioma cells as proposed by Lu et al. is of relevance in human glioblastoma in vivo.

Tie2/Angiopoietin signaling

Pharmacological interference of the Tie2/Angiopoietin signaling pathway has received considerable attention recently (for review see [17, 58]). Several pre-clinical studies have shown that modulation of this pathway leads to alterations in vascular morphology and inhibition of tumor growth. Several types of angiopoietin inhibitors are now in phase I–III clinical trials (Table 1). The Tie2 receptor tyrosine kinase is expressed in endothelial cells and a subset of hematopoietic cells during development [34]. Similar to VEGF receptors, Tie2 is critical for normal vascular development [33]; however, in contrast to VEGF receptors that are down-regulated after embryonic angiogenesis has ceased, Tie2 appears to be constitutively expressed and phosphorylated in the adult vasculature [150]. This constitutive expression and activation suggests that signaling via Tie2 is important for the homeostasis of the mature vasculature (Fig. 3). Indeed, Tie2 signaling promotes stable vessels that are covered by pericytes. Tie2 is bound by three different ligands that engage the same binding site on the receptor. Angiopoietin-1 (Ang-1) and Angiopoietin-2 (Ang-2) are the first ligands to be discovered and are the best studied, whereas the function of Ang-4 is less understood. These ligands are thought to bind to Tie2 with roughly similar affinities and cause receptor activation [144]. In addition to Tie2, integrins serve as receptors for angiopoietins. A major observation has been that Ang-1 acts as a stimulating ligand for the Tie2 RTK that leads to receptor phosphorylation, whereas Ang-2 inhibits Tie2 phosphorylation, even in the presence of Ang-1 [64, 94]. Ang-1 can “seal” vessels in vivo and decrease vascular permeability [61]. Interestingly, the major regulator of the entire Tie2/Angiopoietin signaling pathway appears to be Ang-2. TCGA data for glioblastoma reveal that Ang-2 is up-regulated to a much higher extent than Ang-1, suggesting that Tie2 signaling is blocked in glioblastoma (http://​cancergenome.​nih.​gov). The consequences of blocking Tie2 signaling in vivo, as evidenced by several pre-clinical studies, appears to be a shift towards an immature vasculature, whereas Ang-1 appears to “normalize” the vasculature (see below). In addition to an impact on angiogenesis, Ang-2 was shown to mediate a pro-inflammatory phenotype. In Ang-2 null mice the inflammatory response to stimuli such as TNF-α was greatly diminished when compared to controls [39]. Vice versa, in mice engineered to express an inducible form of Ang-2 in the vasculature, myeloid cells increased significantly over time in almost all organs, even without any pathological stimulus [128]. These findings suggest that Ang-2 on its own is able to orchestrate an inflammatory response by specifically recruiting myeloid cells—and leaving lymphocytes behind (reviewed in [69]). Moreover, since Ang-2 is highly expressed in glioblastoma vessels [141], Ang-2 up-regulation may also account—at least in part—for the attraction of myeloid cells of the monocyte/macrophage lineage to human gliomas. As such, inhibiting Ang-2 may have at least two different effects on tumor growth: (1) a shift of balance towards mature vessels (e.g., vascular normalization) and subsequent decrease of intratumoral hypoxia (2) inhibition of monocyte/macrophage recruitment to tumors [57, 59, 86, 97].

Notch signaling pathway

The Notch signaling pathway plays an important role in organ development and recently has also received considerable attention in the vascular biology field recently (for review see [8, 78, 122]). Of the four Notch receptors, Notch1 and Notch4 are expressed on endothelial cells but Notch1 appears to be the most important for developmental angiogenesis. Similar to their receptors, Notch ligands are membrane-bound. Delta-like ligand4 (Dll4) appears to be the most important Notch ligand for stimulating angiogenesis, whereas another ligand, Jagged1, negatively regulates angiogenesis by competing with Dll4 [4, 78]. Notch in concert with VEGF signaling appears to be instrumental in the tip cell versus stalk cell fate decision that is essential for the initiation of sprouting angiogenesis (see Fig. 2). VEGF-A induces signaling in VEGFR-2 and -3, which are expressed in the tip cells leading to up-regulation of Dll4 and subsequent activation of Notch signaling in adjacent endothelial stalk cells. This signaling cascade places the Notch signaling pathway downstream of VEGFR-2 and R-3 signaling in endothelial cells. Once activated, Notch signaling can ultimately provide negative feedback on VEGFR signaling by up-regulating VEGFR-1 but inhibiting VEGFR-2 and R-3 expression in stalk cells, this signaling circuit provides a way for regulating tip versus stalk cell fate decisions [5]. The up-regulation of VEGFR-1, which acts as a decoy receptor, further contributes to the maintenance of the VEGF gradient. Blocking of Dll4–Notch interaction leads to hypervascular tumors that are nevertheless inhibited in their growth. This phenomenon has been entitled the “delta-paradox” and is probably based on the excessive production of non-functional vessels (e.g., tumor vessel abnormalization) [78]. Since Dll4 has been primarily detected in glioblastoma vasculature and not in glioma cells [65], targeting Dll4 by neutralizing antibodies is considered a potential anti-angiogenic tumor therapy. However, safety concerns have been arisen since it has been reported that in preclinical models, chronic Dll4 blockade abnormally activates endothelial cells and causes vascular neoplasms [152].

The concept of vascular normalization

The concept of “vascular normalization” has been introduced by Jain [60]. This concept states that anti-angiogenic (e.g., Bevacizumab) treatment merely affects the immature vasculature and leaves the mature vessels unaltered. Indeed, it has been shown that VEGF withdrawal leads to selective ablation of immature blood vessels [6]. As such, a “normalized” vasculature results as a consequence of anti-VEGF treatment, leading to increased perfusion of the tumor and subsequent increase of oxygenation. Vascular normalization is thought to interrupt the vicious circle that is driven by hypoxia and that leads to up-regulation of VEGF, resulting in the growth of immature—partly unperfused—vessels and a subsequent increase in tumor hypoxia [23]. Some researchers believe that vessel normalization followed by cytotoxic chemotherapy and/or radiotherapy should be the ultimate goal of any anti-angiogenic therapy [62]. Indeed, in an experimental glioma model, intratumoral vascular normalization restored blood–brain barrier properties by suppressing edema formation [118] and a clinical study showed increased blood perfusion in GBM after anti-angiogenic therapy [138]. In order to achieve vascular normalization, several potential targets beside anti-VEGF therapy have been identified that include (1) Tie2/Angiopoietin signaling (see above), (2) prolylhydroxylases, which are upstream of the HIF-signaling pathway [23], (3) blockade of the VEGF receptor-1 ligand placenta growth factor (PlGF) [40], (4) blocking of TGF-ß signaling [31] and (5) increase of vascular wnt/ß-catenin signaling [118].

The role of myeloid cells in tumor angiogenesis

Current studies suggest that infiltrating myeloid cells contribute significantly to tumor angiogenesis, presumably by secreting pro-angiogenic factors including VEGF, stromal-derived factor-1 (SDF-1) and others (Fig. 4, for review see [47, 101]). Grunewald et al. [48] have shown that myeloid cells are (1) recruited to tumors, (2) “educated” by the tumor microenvironment and (3) support tumor growth by secretion of SDF-1. In another study, chimeric mice were generated by transplanting bone marrow from VEGFR-1 signaling deficient mice into lethally irradiated wild-type mice. When Gl261 glioma cells were transplanted intracranially into these mice, tumor growth was significantly inhibited when compared to mice harboring a normal bone marrow. Subsequent studies showed that VEGFR-1 signaling deficient bone marrow chimeras displayed a defect in monocyte migration leading to significant lower amount of myeloid cells in the tumor tissue [67]. In a preclinical study, myeloid cell infiltration has been shown to be associated with tumor progression after anti-angiogenic therapy [109]. In a clinical study, a decrease in the number of VEGFR-1 + monocytes in the blood correlated with improved response after bevacizumab treatment [25]. Most of the myeloid cells that support tumor growth through the stimulation of angiogenesis appear to be monocytes/macrophages with M2 polarity (TAMs, tumor-associated macrophages) [135]. In contrast, bone marrow-derived non-hematopoietic mesenchymal stem cells may in fact suppress glioma growth through the inhibition of angiogenesis [54]. Interestingly, one distinct subtype of monocytes with particular importance for tumor angiogenesis and immunosuppression seems to be Tie2-expressing monocytes (TEMs) [28]. TEMs represent a subset of circulating blood monocytes that are recruited from the circulation into tumors and locally support tumor growth in a paracrine manner [88]. Although the number of TEMs within a tumor appears relatively small (e.g., <10 % of infiltrating monocytes express Tie2) [21], loss of function studies using suicide gene approaches have shown that the specific depletion of TEMs is able to block tumor angiogenesis and tumor growth [29]. The number of infiltrating macrophages has been shown to be of prognostic relevance in breast cancer [85], rectal cancer [132], classic Hodgkin’s lymphoma [139], adenocarcinoma of the lung [157] and hepatocellular carcinoma [27, 96]. Therefore, both TEMs and TAMs clearly have evolved as novel targets for anti-angiogenic cancer therapy [26, 66].

Future prospects for anti-angiogenic therapy

The first specific anti-angiogenic drug that received FDA approval in 2004 was bevacizumab, a neutralizing monoclonal antibody to VEGF. Bevacizumab is widely used in different types of cancer and significantly inhibits tumor growth in many cancer types (see Table 1 for current anti-angiogenesis trials in glioblastoma). However, drugs that interfere with the function of the vascular system may also cause—sometimes severe—undesired effects [3, 84]. VEGFR-2 is not only expressed on endothelial cells but also on hemangioblasts and angioblasts [121] and in the nervous system (see below). Documented undesired effects of bevacizumab therapy include hypertension, induction of bleeding disorders and deep venous thrombosis [100, 103]. Chronic inhibition of the Notch pathway by Dll4 blockade using inhibitory antibodies may even lead to the induction of vascular neoplasms [152]. In addition, many vascular growth factors have been shown to be involved in the development and homeostasis of the nervous system by directly interacting with neurons (for review see [37, 115, 124, 125]). VEGF, for example, acts as a survival factor for neurons in vitro and in vivo and promotes neurogenesis, most likely through interaction with VEGFR-2 expressed on neurons [13, 22, 63, 127, 149, 153]. Partial deletion of VEGF in mice leads to loss of motoneurons and subsequently to a phenotype that highly resembles amyotrophic lateral sclerosis (ALS) in humans [106]. Indeed, polymorphisms in the vegfa gene are associated with a higher risk to develop ALS in humans [83] and delivery of recombinant VEGF has been used successfully to protect motoneurons in a rat model of ALS [140]. Currently, intracerebroventricular delivered VEGF is in clinical phase I/II trial for patients with ALS. Further, in line with a direct effect of VEGF on the nervous system, anti-VEGF receptor treatment has been reported to lead a painful sensory neuropathy [146].
In addition to the above-mentioned adverse effects of anti-VEGF treatment, pre-clinical and clinical reports suggest that cancer cells may develop resistance to anti-angiogenic therapy by different mechanisms that include (1) switch to a pro-migratory phenotype, (2) up-regulation of other pro-angiogenic molecules [151] and (3) the increased recruitment of myeloid cells that support tumor growth (reviewed in [7, 66, 129]). As such, novel therapies are currently under development in both academia and pharmaceutical companies that include (1) novel VEGF inhibitors such as the VEGF trap [46, 55], (2) drugs that target the Tie2/Angiopoietin signaling pathway (reviewed in [17, 58]), (3) double inhibition of VEGF and angiopoietins [12, 18, 73], (4) double inhibition of VEGF and c-met [130, 154], (5) inhibition of PlGF [30, 40] and (6) drugs that block myeloid cell recruitment and/or their polarization [91], among others. However, it has been suggested that intratumoral vessel are heterogenous and that, for example, VEGF-dependent and -independent vessels may co-exist within a given tumor [137]. These observations speak in favor of a multi-modal therapy that may combine different types of anti-angiogenic therapy or anti-angiogenic therapy together with cytotoxic therapy. The integrin inhibitor cilengitide, for example, targets both tumor cells and the vasculature [105], and is now in phase III clinical trial for glioblastoma patients [79, 126]. In our personal view, targeting the Tie2 signaling pathway by blocking Ang-2 or stimulating Ang-1 (or both simultaneously) may be particularly promising since three mechanisms of tumor angiogenesis may be targeted similarly: (1) sprouting angiogenesis and (2) myeloid cell infiltration may be blocked, whereas (3) vascular normalization will be enhanced [17, 58]. Further, reported side effects of Ang-2 blocking appear to be sparse compared to other anti-angiogenic therapies employed [53, 82]. However, this has to be proven further in the clinical trials that are currently under way. It may also be necessary to stratify patients so that those that have a shift in the Ang-1/Ang-2 balance or other angiogenic factors are identified and the appropriate therapy initiated. For example, the monitoring of expression patterns of angiogenesis factors and their receptor in the tumor biopsy or the evaluation of biomarkers in the serum or cerebrospinal fluid of patients may provide useful indicators of a tumor’s susceptibility to a particular targeted therapy [100]. Promising biomarkers that may indicate therapy responsiveness in anti-angiogenic therapy include serum Ang-2 levels and the number of circulating VEGFR-1 + monocytes [25, 45, 90]. It will be exciting to watch whether any of the newly developed therapies will be able to significantly affect survival of brain cancer patients.

Acknowledgments

We are grateful to Prof. Michel Mittelbronn for his help in obtaining and interpreting data derived from the The Cancer Genome Atlas (TCGA). The work of the corresponding author is supported by (1) Deutsche Forschungsgemeinschaft (DFG): (a) Collaborative Research Center (SFB/TR23) grants B7, C1 and C4, (b) Excellence Cluster Cardiopulmonary-Systems (ECCPS), and (2) Helmholtz Society/BMBF (a) Deutsches Konsortium für Translationale Krebsforschung (DKTK), (b) Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK). D.J.D. is the Canadian Research Chair for Angiogenic and Lymphangiogenic Signaling; Funding was provided by the Heart and Stroke Foundation of Canada (NA5425).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Literatur
1.
Zurück zum Zitat Adams RH, Eichmann A (2010) Axon guidance molecules in vascular patterning. Cold Spring Harb Perspect Biol 2(5):a001875PubMedCrossRef Adams RH, Eichmann A (2010) Axon guidance molecules in vascular patterning. Cold Spring Harb Perspect Biol 2(5):a001875PubMedCrossRef
2.
Zurück zum Zitat Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM (1997) Isolation of putative progenitor endothelial cells for angiogenesis. Science 275(5302):964–967PubMedCrossRef Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM (1997) Isolation of putative progenitor endothelial cells for angiogenesis. Science 275(5302):964–967PubMedCrossRef
3.
Zurück zum Zitat Belcik JT, Qi Y, Kaufmann BA et al (2012) Cardiovascular and systemic microvascular effects of anti-vascular endothelial growth factor therapy for cancer. J Am Coll Cardiol 60(7):618–625PubMedCrossRef Belcik JT, Qi Y, Kaufmann BA et al (2012) Cardiovascular and systemic microvascular effects of anti-vascular endothelial growth factor therapy for cancer. J Am Coll Cardiol 60(7):618–625PubMedCrossRef
4.
Zurück zum Zitat Benedito R, Roca C, Sörensen I, Adams S, Gossler A, Fruttiger M, Adams RH (2009) The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell 137(6):1124–1135PubMedCrossRef Benedito R, Roca C, Sörensen I, Adams S, Gossler A, Fruttiger M, Adams RH (2009) The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell 137(6):1124–1135PubMedCrossRef
5.
Zurück zum Zitat Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O, Duarte A, Pytowski B, Adams RH (2012) Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF–VEGFR2 signalling. Nature 484(7392):110–114PubMedCrossRef Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O, Duarte A, Pytowski B, Adams RH (2012) Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF–VEGFR2 signalling. Nature 484(7392):110–114PubMedCrossRef
6.
Zurück zum Zitat Benjamin LE, Hemo I, Keshet E (1998) A plasticity window for blood vessel remodelling is defined by pericyte coverage of the preformed endothelial network and is regulated by PDGF-B and VEGF. Development 125(9):1591–1598PubMed Benjamin LE, Hemo I, Keshet E (1998) A plasticity window for blood vessel remodelling is defined by pericyte coverage of the preformed endothelial network and is regulated by PDGF-B and VEGF. Development 125(9):1591–1598PubMed
7.
Zurück zum Zitat Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603PubMedCrossRef Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603PubMedCrossRef
8.
Zurück zum Zitat Blanco R, Gerhardt H (2012) VEGF and Notch in tip and stalk cell selection. Cold Spring Harb Perspect Med October 1–20 (published online) Blanco R, Gerhardt H (2012) VEGF and Notch in tip and stalk cell selection. Cold Spring Harb Perspect Med October 1–20 (published online)
9.
Zurück zum Zitat Bottaro DP, Liotta LA (2003) Cancer: out of air is not out of action. Nature 423(6940):593–595PubMedCrossRef Bottaro DP, Liotta LA (2003) Cancer: out of air is not out of action. Nature 423(6940):593–595PubMedCrossRef
10.
Zurück zum Zitat Brat DJ, Castellano-Sanchez AA, Hunter SB, Pecot M, Cohen C, Hammond EH, Devi SN, Kaur B, Van Meir EG (2004) Pseudopalisades in glioblastoma are hypoxic, express extracellular matrix proteases, and are formed by an actively migrating cell population. Cancer Res 64(3):920–927PubMedCrossRef Brat DJ, Castellano-Sanchez AA, Hunter SB, Pecot M, Cohen C, Hammond EH, Devi SN, Kaur B, Van Meir EG (2004) Pseudopalisades in glioblastoma are hypoxic, express extracellular matrix proteases, and are formed by an actively migrating cell population. Cancer Res 64(3):920–927PubMedCrossRef
11.
Zurück zum Zitat Broholm H, Laursen H (2004) Vascular endothelial growth factor (VEGF) receptor neuropilin-1’s distribution in astrocytic tumors. APMIS 112(4–5):257–263PubMedCrossRef Broholm H, Laursen H (2004) Vascular endothelial growth factor (VEGF) receptor neuropilin-1’s distribution in astrocytic tumors. APMIS 112(4–5):257–263PubMedCrossRef
12.
Zurück zum Zitat Brown JL, Cao ZA, Pinzon-Ortiz M et al (2010) A human monoclonal anti-ANG2 antibody leads to broad antitumor activity in combination with VEGF inhibitors and chemotherapy agents in preclinical models. Mol Cancer Ther 9(1):145–156PubMedCrossRef Brown JL, Cao ZA, Pinzon-Ortiz M et al (2010) A human monoclonal anti-ANG2 antibody leads to broad antitumor activity in combination with VEGF inhibitors and chemotherapy agents in preclinical models. Mol Cancer Ther 9(1):145–156PubMedCrossRef
13.
Zurück zum Zitat Cao L, Jiao X, Zuzga DS, Liu Y, Fong DM, Young D, During MJ (2004) VEGF links hippocampal activity with neurogenesis, learning and memory. Nat Genet 36(8):827–835PubMedCrossRef Cao L, Jiao X, Zuzga DS, Liu Y, Fong DM, Young D, During MJ (2004) VEGF links hippocampal activity with neurogenesis, learning and memory. Nat Genet 36(8):827–835PubMedCrossRef
14.
Zurück zum Zitat Carmeliet P, Ferreira V, Breier G et al (1996) Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 380(6573):435–439PubMedCrossRef Carmeliet P, Ferreira V, Breier G et al (1996) Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature 380(6573):435–439PubMedCrossRef
15.
Zurück zum Zitat Carmeliet P, Jain RK (2011) Molecular mechanisms and clinical applications of angiogenesis. Nature 473(7347):298–307PubMedCrossRef Carmeliet P, Jain RK (2011) Molecular mechanisms and clinical applications of angiogenesis. Nature 473(7347):298–307PubMedCrossRef
16.
Zurück zum Zitat Carmeliet P, De Smet F, Loges S, Mazzone M (2009) Branching morphogenesis and antiangiogenesis candidates: tip cells lead the way. Nat Rev Clin Oncol 6(6):315–326PubMedCrossRef Carmeliet P, De Smet F, Loges S, Mazzone M (2009) Branching morphogenesis and antiangiogenesis candidates: tip cells lead the way. Nat Rev Clin Oncol 6(6):315–326PubMedCrossRef
17.
Zurück zum Zitat Cascone T, Heymach JV (2012) Targeting the Angiopoietin/Tie2 pathway: cutting tumor vessels with a double-edged sword? J Clin Oncol 30(4):441–444PubMedCrossRef Cascone T, Heymach JV (2012) Targeting the Angiopoietin/Tie2 pathway: cutting tumor vessels with a double-edged sword? J Clin Oncol 30(4):441–444PubMedCrossRef
18.
Zurück zum Zitat Chae SS, Kamoun WS, Farrar CT, Kirkpatrick ND, Niemeyer E, de Graaf AMA, Sorensen AG, Munn LL, Jain RK, Fukumura D (2010) Angiopoietin-2 interferes with anti-VEGFR2-induced vessel normalization and survival benefit in mice bearing gliomas. Clin Cancer Res 16(14):3618–3627PubMedCrossRef Chae SS, Kamoun WS, Farrar CT, Kirkpatrick ND, Niemeyer E, de Graaf AMA, Sorensen AG, Munn LL, Jain RK, Fukumura D (2010) Angiopoietin-2 interferes with anti-VEGFR2-induced vessel normalization and survival benefit in mice bearing gliomas. Clin Cancer Res 16(14):3618–3627PubMedCrossRef
19.
Zurück zum Zitat Chamberlain MC (2011) Bevacizumab for the treatment of recurrent glioblastoma. Clin Med Insights Oncol 5:117–129PubMedCrossRef Chamberlain MC (2011) Bevacizumab for the treatment of recurrent glioblastoma. Clin Med Insights Oncol 5:117–129PubMedCrossRef
20.
Zurück zum Zitat Claesson-Welsh L (2012) Receptor talk and tumor cell walk in glioblastoma. Cancer Cell 22(1):1–2PubMedCrossRef Claesson-Welsh L (2012) Receptor talk and tumor cell walk in glioblastoma. Cancer Cell 22(1):1–2PubMedCrossRef
21.
Zurück zum Zitat Coffelt SB, Tal AO, Scholz A et al (2010) Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions. Cancer Res 70(13):5270–5280PubMedCrossRef Coffelt SB, Tal AO, Scholz A et al (2010) Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions. Cancer Res 70(13):5270–5280PubMedCrossRef
22.
Zurück zum Zitat Darland DC, Cain JT, Berosik MA, Saint-Geniez M, Odens PW, Schaubhut GJ, Frisch S, Stemmer-Rachamimov A, Darland T, D’Amore PA (2011) Vascular endothelial growth factor (VEGF) isoform regulation of early forebrain development. Dev Biol 358(1):9–22PubMedCrossRef Darland DC, Cain JT, Berosik MA, Saint-Geniez M, Odens PW, Schaubhut GJ, Frisch S, Stemmer-Rachamimov A, Darland T, D’Amore PA (2011) Vascular endothelial growth factor (VEGF) isoform regulation of early forebrain development. Dev Biol 358(1):9–22PubMedCrossRef
23.
Zurück zum Zitat De Bock K, Cauwenberghs S, Carmeliet P (2010) Vessel abnormalization: another hallmark of cancer? Molecular mechanisms and therapeutic implications. Curr Opin Genet Dev 21(1):1–7 De Bock K, Cauwenberghs S, Carmeliet P (2010) Vessel abnormalization: another hallmark of cancer? Molecular mechanisms and therapeutic implications. Curr Opin Genet Dev 21(1):1–7
24.
Zurück zum Zitat de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, Conrad CA (2010) Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol 12(3):233–242PubMedCrossRef de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, Conrad CA (2010) Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol 12(3):233–242PubMedCrossRef
25.
Zurück zum Zitat de Groot JF, Piao Y, Tran H et al (2011) Myeloid biomarkers associated with glioblastoma response to anti-VEGF therapy with aflibercept. Clin Cancer Res 17(14):4872–4881PubMedCrossRef de Groot JF, Piao Y, Tran H et al (2011) Myeloid biomarkers associated with glioblastoma response to anti-VEGF therapy with aflibercept. Clin Cancer Res 17(14):4872–4881PubMedCrossRef
26.
Zurück zum Zitat De Palma M, Lewis CE (2011) Cancer: macrophages limit chemotherapy. Nature 472(7343):303–304PubMedCrossRef De Palma M, Lewis CE (2011) Cancer: macrophages limit chemotherapy. Nature 472(7343):303–304PubMedCrossRef
27.
Zurück zum Zitat De Palma M, Coukos G, Semela D (2012) TIE2-expressing monocytes: A novel cellular biomarker for hepatocellular carcinoma? Hepatology. doi:10.1002/hep.26025 De Palma M, Coukos G, Semela D (2012) TIE2-expressing monocytes: A novel cellular biomarker for hepatocellular carcinoma? Hepatology. doi:10.​1002/​hep.​26025
28.
Zurück zum Zitat De Palma M, Murdoch C, Venneri MA, Naldini L, Lewis CE (2007) Tie2-expressing monocytes: regulation of tumor angiogenesis and therapeutic implications. Trends Immunol 28(12):519–524PubMedCrossRef De Palma M, Murdoch C, Venneri MA, Naldini L, Lewis CE (2007) Tie2-expressing monocytes: regulation of tumor angiogenesis and therapeutic implications. Trends Immunol 28(12):519–524PubMedCrossRef
29.
Zurück zum Zitat De Palma M, Venneri MA, Galli R, Sergi LS, Politi LS, Sampaolesi M, Naldini L (2005) Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 8(3):211–226PubMedCrossRef De Palma M, Venneri MA, Galli R, Sergi LS, Politi LS, Sampaolesi M, Naldini L (2005) Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 8(3):211–226PubMedCrossRef
31.
Zurück zum Zitat Dieterich LC, Mellberg S, Langenkamp E et al (2012) Transcriptional profiling of human glioblastoma vessels indicates a key role of VEGF-A and TGFβ2 in vascular abnormalization. J Pathol. doi:10.1002/path.4072 Dieterich LC, Mellberg S, Langenkamp E et al (2012) Transcriptional profiling of human glioblastoma vessels indicates a key role of VEGF-A and TGFβ2 in vascular abnormalization. J Pathol. doi:10.​1002/​path.​4072
32.
Zurück zum Zitat Djonov V, Schmid M, Tschanz SA, Burri PH (2000) Intussusceptive angiogenesis: its role in embryonic vascular network formation. Circ Res 86(3):286–292PubMedCrossRef Djonov V, Schmid M, Tschanz SA, Burri PH (2000) Intussusceptive angiogenesis: its role in embryonic vascular network formation. Circ Res 86(3):286–292PubMedCrossRef
33.
Zurück zum Zitat Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein M, Auerbach A, Breitman ML (1994) Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo. Genes Dev 8(16):1897–1909PubMedCrossRef Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein M, Auerbach A, Breitman ML (1994) Dominant-negative and targeted null mutations in the endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of the embryo. Genes Dev 8(16):1897–1909PubMedCrossRef
34.
Zurück zum Zitat Dumont DJ, Yamaguchi TP, Conlon RA, Rossant J, Breitman ML (1992) tek, a novel tyrosine kinase gene located on mouse chromosome 4, is expressed in endothelial cells and their presumptive precursors. Oncogene 7(8):1471–1480PubMed Dumont DJ, Yamaguchi TP, Conlon RA, Rossant J, Breitman ML (1992) tek, a novel tyrosine kinase gene located on mouse chromosome 4, is expressed in endothelial cells and their presumptive precursors. Oncogene 7(8):1471–1480PubMed
35.
Zurück zum Zitat Ebos JML, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS (2009) Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15(3):232–239PubMedCrossRef Ebos JML, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS (2009) Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15(3):232–239PubMedCrossRef
36.
Zurück zum Zitat Eckerich C, Zapf S, Fillbrandt R, Loges S, Westphal M, Lamszus K (2007) Hypoxia can induce c-Met expression in glioma cells and enhance SF/HGF-induced cell migration. Int J Cancer 121(2):276–283PubMedCrossRef Eckerich C, Zapf S, Fillbrandt R, Loges S, Westphal M, Lamszus K (2007) Hypoxia can induce c-Met expression in glioma cells and enhance SF/HGF-induced cell migration. Int J Cancer 121(2):276–283PubMedCrossRef
38.
Zurück zum Zitat Ferrara N (2002) VEGF and the quest for tumour angiogenesis factors. Nat Rev Cancer 2(10):795–803PubMedCrossRef Ferrara N (2002) VEGF and the quest for tumour angiogenesis factors. Nat Rev Cancer 2(10):795–803PubMedCrossRef
39.
Zurück zum Zitat Fiedler U, Reiss Y, Scharpfenecker M et al (2006) Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med 12(2):235–239PubMedCrossRef Fiedler U, Reiss Y, Scharpfenecker M et al (2006) Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med 12(2):235–239PubMedCrossRef
40.
Zurück zum Zitat Fischer C, Jonckx B, Mazzone M et al (2007) Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell 131(3):463–475PubMedCrossRef Fischer C, Jonckx B, Mazzone M et al (2007) Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell 131(3):463–475PubMedCrossRef
41.
Zurück zum Zitat Folkman J (1971) Tumor angiogenesis: therapeutic implications. N Engl J Med 285(21):1182–1186PubMedCrossRef Folkman J (1971) Tumor angiogenesis: therapeutic implications. N Engl J Med 285(21):1182–1186PubMedCrossRef
42.
Zurück zum Zitat Fong GH, Rossant J, Gertsenstein M, Breitman ML (1995) Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 376(6535):66–70PubMedCrossRef Fong GH, Rossant J, Gertsenstein M, Breitman ML (1995) Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 376(6535):66–70PubMedCrossRef
43.
Zurück zum Zitat Fong GH, Zhang L, Bryce DM, Peng J (1999) Increased hemangioblast commitment, not vascular disorganization, is the primary defect in flt-1 knock-out mice. Development 126(13):3015–3025PubMed Fong GH, Zhang L, Bryce DM, Peng J (1999) Increased hemangioblast commitment, not vascular disorganization, is the primary defect in flt-1 knock-out mice. Development 126(13):3015–3025PubMed
44.
Zurück zum Zitat Francescone R, Scully S, Bentley B, Yan W, Taylor SL, Oh D, Moral L, Shao R (2012) Glioblastoma-derived tumor cells induce vasculogenic mimicry through Flk-1 protein activation. J Biol Chem 287(29):24821–24831PubMedCrossRef Francescone R, Scully S, Bentley B, Yan W, Taylor SL, Oh D, Moral L, Shao R (2012) Glioblastoma-derived tumor cells induce vasculogenic mimicry through Flk-1 protein activation. J Biol Chem 287(29):24821–24831PubMedCrossRef
45.
Zurück zum Zitat Goede V, Coutelle O, Neuneier J et al (2010) Identification of serum Angiopoietin-2 as a biomarker for clinical outcome of colorectal cancer patients treated with bevacizumab-containing therapy. Br J Cancer 103(9):1407–1414PubMedCrossRef Goede V, Coutelle O, Neuneier J et al (2010) Identification of serum Angiopoietin-2 as a biomarker for clinical outcome of colorectal cancer patients treated with bevacizumab-containing therapy. Br J Cancer 103(9):1407–1414PubMedCrossRef
46.
Zurück zum Zitat Gomez-Manzano C, Holash J, Fueyo J, Xu J, Conrad CA, Aldape KD, de Groot JF, Bekele BN, Yung WKA (2008) VEGF Trap induces antiglioma effect at different stages of disease. Neuro Oncol 10(6):940–945PubMedCrossRef Gomez-Manzano C, Holash J, Fueyo J, Xu J, Conrad CA, Aldape KD, de Groot JF, Bekele BN, Yung WKA (2008) VEGF Trap induces antiglioma effect at different stages of disease. Neuro Oncol 10(6):940–945PubMedCrossRef
47.
Zurück zum Zitat Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell 140(6):883–899PubMedCrossRef Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and cancer. Cell 140(6):883–899PubMedCrossRef
48.
Zurück zum Zitat Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Yung S, Chimenti S, Landsman L, Abramovitch R, Keshet E (2006) VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell 124(1):175–189PubMedCrossRef Grunewald M, Avraham I, Dor Y, Bachar-Lustig E, Itin A, Yung S, Chimenti S, Landsman L, Abramovitch R, Keshet E (2006) VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell 124(1):175–189PubMedCrossRef
49.
Zurück zum Zitat Hamerlik P, Lathia JD, Rasmussen R et al (2012) Autocrine VEGF–VEGFR2–Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J Exp Med 209(3):507–520PubMedCrossRef Hamerlik P, Lathia JD, Rasmussen R et al (2012) Autocrine VEGF–VEGFR2–Neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J Exp Med 209(3):507–520PubMedCrossRef
50.
51.
Zurück zum Zitat Hatva E, Kaipainen A, Mentula P, Jääskeläinen J, Paetau A, Haltia M, Alitalo K (1995) Expression of endothelial cell-specific receptor tyrosine kinases and growth factors in human brain tumors. Am J Pathol 146(2):368–378PubMed Hatva E, Kaipainen A, Mentula P, Jääskeläinen J, Paetau A, Haltia M, Alitalo K (1995) Expression of endothelial cell-specific receptor tyrosine kinases and growth factors in human brain tumors. Am J Pathol 146(2):368–378PubMed
52.
Zurück zum Zitat He T, Smith N, Saunders D et al (2011) Molecular MRI assessment of vascular endothelial growth factor receptor-2 in rat C6 gliomas. J Cell Mol Med 15(4):837–849PubMedCrossRef He T, Smith N, Saunders D et al (2011) Molecular MRI assessment of vascular endothelial growth factor receptor-2 in rat C6 gliomas. J Cell Mol Med 15(4):837–849PubMedCrossRef
53.
Zurück zum Zitat Herbst RS, Hong D, Chap L et al (2009) Safety, pharmacokinetics, and antitumor activity of AMG 386, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors. J Clin Oncol 27(21):3557–3565PubMedCrossRef Herbst RS, Hong D, Chap L et al (2009) Safety, pharmacokinetics, and antitumor activity of AMG 386, a selective angiopoietin inhibitor, in adult patients with advanced solid tumors. J Clin Oncol 27(21):3557–3565PubMedCrossRef
54.
Zurück zum Zitat Ho IA, Toh HC, Ng WH, Teo YL, Guo CM, Hui KM, Lam PY (2012) Human bone marrow-derived mesenchymal stem cells suppress human glioma growth through inhibition of angiogenesis. Stem Cells. doi:10.1002/stem.1247 Ho IA, Toh HC, Ng WH, Teo YL, Guo CM, Hui KM, Lam PY (2012) Human bone marrow-derived mesenchymal stem cells suppress human glioma growth through inhibition of angiogenesis. Stem Cells. doi:10.​1002/​stem.​1247
55.
Zurück zum Zitat Holash J (2002) VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA 99(17):11393–11398PubMedCrossRef Holash J (2002) VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA 99(17):11393–11398PubMedCrossRef
56.
Zurück zum Zitat Holash J, Wiegand SJ, Yancopoulos GD (1999) New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietins and VEGF. Oncogene 18(38):5356–5362PubMedCrossRef Holash J, Wiegand SJ, Yancopoulos GD (1999) New model of tumor angiogenesis: dynamic balance between vessel regression and growth mediated by angiopoietins and VEGF. Oncogene 18(38):5356–5362PubMedCrossRef
57.
Zurück zum Zitat Holopainen T, Saharinen P, D’Amico G et al (2012) Effects of Angiopoietin-2-blocking antibody on endothelial cell–cell junctions and lung metastasis. J Natl Cancer Inst 104(6):461–475PubMedCrossRef Holopainen T, Saharinen P, D’Amico G et al (2012) Effects of Angiopoietin-2-blocking antibody on endothelial cell–cell junctions and lung metastasis. J Natl Cancer Inst 104(6):461–475PubMedCrossRef
58.
Zurück zum Zitat Huang H, Bhat A, Woodnutt G, Lappe R (2010) Targeting the ANGPT–TIE2 pathway in malignancy. Nat Rev Cancer 10(8):575–585PubMedCrossRef Huang H, Bhat A, Woodnutt G, Lappe R (2010) Targeting the ANGPT–TIE2 pathway in malignancy. Nat Rev Cancer 10(8):575–585PubMedCrossRef
59.
Zurück zum Zitat Huang H, Lai J-Y, Do J et al (2011) Specifically targeting Angiopoietin-2 inhibits angiogenesis, Tie2-expressing monocyte infiltration, and tumor growth. Clin Cancer Res 17(5):1001–1011PubMedCrossRef Huang H, Lai J-Y, Do J et al (2011) Specifically targeting Angiopoietin-2 inhibits angiogenesis, Tie2-expressing monocyte infiltration, and tumor growth. Clin Cancer Res 17(5):1001–1011PubMedCrossRef
60.
Zurück zum Zitat Jain RK (2005) Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706):58–62PubMedCrossRef Jain RK (2005) Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706):58–62PubMedCrossRef
62.
63.
Zurück zum Zitat Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA (2002) Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci USA 99(18):11946–11950PubMedCrossRef Jin K, Zhu Y, Sun Y, Mao XO, Xie L, Greenberg DA (2002) Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc Natl Acad Sci USA 99(18):11946–11950PubMedCrossRef
64.
Zurück zum Zitat Jones N, Iljin K, Dumont DJ, Alitalo K (2001) Tie receptors: new modulators of angiogenic and lymphangiogenic responses. Nat Rev Mol Cell Biol 2(4):257–267PubMedCrossRef Jones N, Iljin K, Dumont DJ, Alitalo K (2001) Tie receptors: new modulators of angiogenic and lymphangiogenic responses. Nat Rev Mol Cell Biol 2(4):257–267PubMedCrossRef
65.
Zurück zum Zitat Jubb AM, Browning L, Campo L, Turley H, Steers G, Thurston G, Harris AL, Ansorge O (2012) Expression of vascular Notch ligands Delta-like 4 and Jagged-1 in glioblastoma. Histopathology 60(5):740–747PubMedCrossRef Jubb AM, Browning L, Campo L, Turley H, Steers G, Thurston G, Harris AL, Ansorge O (2012) Expression of vascular Notch ligands Delta-like 4 and Jagged-1 in glioblastoma. Histopathology 60(5):740–747PubMedCrossRef
67.
Zurück zum Zitat Kerber M, Reiss Y, Wickersheim A et al (2008) Flt-1 signaling in macrophages promotes glioma growth in vivo. Cancer Res 68(18):7342–7351PubMedCrossRef Kerber M, Reiss Y, Wickersheim A et al (2008) Flt-1 signaling in macrophages promotes glioma growth in vivo. Cancer Res 68(18):7342–7351PubMedCrossRef
68.
Zurück zum Zitat Keunen O, Johansson M, Oudin A et al (2011) Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma. Proc Natl Acad Sci USA 108(9):3749–3754PubMedCrossRef Keunen O, Johansson M, Oudin A et al (2011) Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma. Proc Natl Acad Sci USA 108(9):3749–3754PubMedCrossRef
69.
70.
Zurück zum Zitat Kim KJ, Li B, Houck K, Winer J, Ferrara N (1992) The vascular endothelial growth factor proteins: identification of biologically relevant regions by neutralizing monoclonal antibodies. Growth Factors 7(1):53–64PubMedCrossRef Kim KJ, Li B, Houck K, Winer J, Ferrara N (1992) The vascular endothelial growth factor proteins: identification of biologically relevant regions by neutralizing monoclonal antibodies. Growth Factors 7(1):53–64PubMedCrossRef
71.
Zurück zum Zitat Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, Ferrara N (1993) Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362(6423):841–844PubMedCrossRef Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS, Ferrara N (1993) Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362(6423):841–844PubMedCrossRef
72.
Zurück zum Zitat Kioi M, Vogel H, Schultz G, Hoffman RM, Harsh GR, Brown JM (2010) Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest 120(3):694–705PubMedCrossRef Kioi M, Vogel H, Schultz G, Hoffman RM, Harsh GR, Brown JM (2010) Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest 120(3):694–705PubMedCrossRef
73.
Zurück zum Zitat Koh YJ, Kim HZ, Hwang S-I et al (2010) Double antiangiogenic protein, DAAP, targeting VEGF-A and angiopoietins in tumor angiogenesis, metastasis, and vascular leakage. Cancer Cell 18(2):171–184PubMedCrossRef Koh YJ, Kim HZ, Hwang S-I et al (2010) Double antiangiogenic protein, DAAP, targeting VEGF-A and angiopoietins in tumor angiogenesis, metastasis, and vascular leakage. Cancer Cell 18(2):171–184PubMedCrossRef
74.
Zurück zum Zitat Korn C, Augustin HG (2012) Born to die: blood vessel regression research coming of age. Circulation 125(25):3063–3065PubMedCrossRef Korn C, Augustin HG (2012) Born to die: blood vessel regression research coming of age. Circulation 125(25):3063–3065PubMedCrossRef
75.
Zurück zum Zitat Kozin SV, Duda DG, Munn LL, Jain RK (2011) Is vasculogenesis crucial for the regrowth of irradiated tumours? Nat Rev Cancer 11(7):532PubMedCrossRef Kozin SV, Duda DG, Munn LL, Jain RK (2011) Is vasculogenesis crucial for the regrowth of irradiated tumours? Nat Rev Cancer 11(7):532PubMedCrossRef
76.
Zurück zum Zitat Kozin SV, Kamoun WS, Huang Y, Dawson MR, Jain RK, Duda DG (2010) Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res 70(14):5679–5685PubMedCrossRef Kozin SV, Kamoun WS, Huang Y, Dawson MR, Jain RK, Duda DG (2010) Recruitment of myeloid but not endothelial precursor cells facilitates tumor regrowth after local irradiation. Cancer Res 70(14):5679–5685PubMedCrossRef
77.
Zurück zum Zitat Kuczynski EA, Patten SG, Coomber BL (2011) VEGFR2 expression and TGF-β signaling in initial and recurrent high-grade human glioma. Oncology 81(2):126–134PubMedCrossRef Kuczynski EA, Patten SG, Coomber BL (2011) VEGFR2 expression and TGF-β signaling in initial and recurrent high-grade human glioma. Oncology 81(2):126–134PubMedCrossRef
78.
Zurück zum Zitat Kuhnert F, Kirshner JR, Thurston G (2011) Dll4-Notch signaling as a therapeutic target in tumor angiogenesis. Vascular Cell 3(1):20PubMedCrossRef Kuhnert F, Kirshner JR, Thurston G (2011) Dll4-Notch signaling as a therapeutic target in tumor angiogenesis. Vascular Cell 3(1):20PubMedCrossRef
79.
Zurück zum Zitat Kurozumi K, Ichikawa T, Onishi M, Fujii K, Date I (2012) Cilengitide treatment for malignant glioma: current status and future direction. Neurol Med Chir (Tokyo) 52(8):539–547CrossRef Kurozumi K, Ichikawa T, Onishi M, Fujii K, Date I (2012) Cilengitide treatment for malignant glioma: current status and future direction. Neurol Med Chir (Tokyo) 52(8):539–547CrossRef
80.
Zurück zum Zitat Kurz H, Korn J, Eggli PS, Huang R, Christ B (2001) Embryonic central nervous system angiogenesis does not involve blood-borne endothelial progenitors. J Comp Neurol 436(3):263–274PubMedCrossRef Kurz H, Korn J, Eggli PS, Huang R, Christ B (2001) Embryonic central nervous system angiogenesis does not involve blood-borne endothelial progenitors. J Comp Neurol 436(3):263–274PubMedCrossRef
81.
Zurück zum Zitat Kurz H, Korn J, Christ B (2004) Morphogenesis of embryonic CNS vessels. Cancer Treat Res 117:33–50PubMedCrossRef Kurz H, Korn J, Christ B (2004) Morphogenesis of embryonic CNS vessels. Cancer Treat Res 117:33–50PubMedCrossRef
82.
Zurück zum Zitat Kümpers P, Gueler F, David S et al (2011) The synthetic Tie2 agonist peptide vasculotide protects against vascular leakage and reduces mortality in murine abdominal sepsis. Crit Care 15(5):R261PubMedCrossRef Kümpers P, Gueler F, David S et al (2011) The synthetic Tie2 agonist peptide vasculotide protects against vascular leakage and reduces mortality in murine abdominal sepsis. Crit Care 15(5):R261PubMedCrossRef
83.
Zurück zum Zitat Lambrechts D, Storkebaum E, Morimoto M et al (2003) VEGF is a modifier of amyotrophic lateral sclerosis in mice and humans and protects motoneurons against ischemic death. Nat Genet 34(4):383–394PubMedCrossRef Lambrechts D, Storkebaum E, Morimoto M et al (2003) VEGF is a modifier of amyotrophic lateral sclerosis in mice and humans and protects motoneurons against ischemic death. Nat Genet 34(4):383–394PubMedCrossRef
84.
Zurück zum Zitat Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, Desai S, Ferrara N, Nagy A, Roos KP, Iruela-Arispe ML (2007) Autocrine VEGF signaling is required for vascular homeostasis. Cell 130(4):691–703PubMedCrossRef Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, Desai S, Ferrara N, Nagy A, Roos KP, Iruela-Arispe ML (2007) Autocrine VEGF signaling is required for vascular homeostasis. Cell 130(4):691–703PubMedCrossRef
85.
Zurück zum Zitat Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL (1996) Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res 56(20):4625–4629PubMed Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J, Harris AL (1996) Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res 56(20):4625–4629PubMed
86.
Zurück zum Zitat Leow CC, Coffman K, Inigo I et al (2012) MEDI3617, a human anti-angiopoietin 2 monoclonal antibody, inhibits angiogenesis and tumor growth in human tumor xenograft models. Int J Oncol 40(5):1321–1330PubMed Leow CC, Coffman K, Inigo I et al (2012) MEDI3617, a human anti-angiopoietin 2 monoclonal antibody, inhibits angiogenesis and tumor growth in human tumor xenograft models. Int J Oncol 40(5):1321–1330PubMed
87.
Zurück zum Zitat Leung SY, Chan AS, Wong MP, Yuen ST, Cheung N, Chung LP (1997) Expression of vascular endothelial growth factor and its receptors in pilocytic astrocytoma. Am J Surg Pathol 21(8):941–950PubMedCrossRef Leung SY, Chan AS, Wong MP, Yuen ST, Cheung N, Chung LP (1997) Expression of vascular endothelial growth factor and its receptors in pilocytic astrocytoma. Am J Surg Pathol 21(8):941–950PubMedCrossRef
88.
Zurück zum Zitat Lewis CE, De Palma M, Naldini L (2007) Tie2-expressing monocytes and tumor angiogenesis: regulation by hypoxia and Angiopoietin-2. Cancer Res 67(18):8429–8432PubMedCrossRef Lewis CE, De Palma M, Naldini L (2007) Tie2-expressing monocytes and tumor angiogenesis: regulation by hypoxia and Angiopoietin-2. Cancer Res 67(18):8429–8432PubMedCrossRef
89.
Zurück zum Zitat Liu X-M, Zhang Q-P, Mu Y-G, Zhang X-H, Sai K, Pang JC-S, Ng H-K, Chen Z-P (2011) Clinical significance of vasculogenic mimicry in human gliomas. J Neurooncol 105(2):173–179PubMedCrossRef Liu X-M, Zhang Q-P, Mu Y-G, Zhang X-H, Sai K, Pang JC-S, Ng H-K, Chen Z-P (2011) Clinical significance of vasculogenic mimicry in human gliomas. J Neurooncol 105(2):173–179PubMedCrossRef
90.
Zurück zum Zitat Llovet JM, Peña CEA, Lathia CD, Shan M, Meinhardt G, Bruix J, SHARP Investigators Study Group (2012) Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res 18(8):2290–2300PubMedCrossRef Llovet JM, Peña CEA, Lathia CD, Shan M, Meinhardt G, Bruix J, SHARP Investigators Study Group (2012) Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res 18(8):2290–2300PubMedCrossRef
91.
Zurück zum Zitat Loges S, Schmidt T, Carmeliet P (2009) “Antimyeloangiogenic” therapy for cancer by inhibiting PlGF. Clin Cancer Res 15(11):3648–3653PubMedCrossRef Loges S, Schmidt T, Carmeliet P (2009) “Antimyeloangiogenic” therapy for cancer by inhibiting PlGF. Clin Cancer Res 15(11):3648–3653PubMedCrossRef
92.
Zurück zum Zitat Lu KV, Chang JP, Parachoniak CA et al (2012) VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell 22(1):21–35PubMedCrossRef Lu KV, Chang JP, Parachoniak CA et al (2012) VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell 22(1):21–35PubMedCrossRef
93.
Zurück zum Zitat Machein MR, Renninger S, de Lima-Hahn E, Plate KH (2003) Minor contribution of bone marrow-derived endothelial progenitors to the vascularization of murine gliomas. Brain Pathol 13(4):582–597PubMedCrossRef Machein MR, Renninger S, de Lima-Hahn E, Plate KH (2003) Minor contribution of bone marrow-derived endothelial progenitors to the vascularization of murine gliomas. Brain Pathol 13(4):582–597PubMedCrossRef
94.
Zurück zum Zitat Maisonpierre PC, Suri C, Jones PF et al (1997) Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 277(5322):55–60PubMedCrossRef Maisonpierre PC, Suri C, Jones PF et al (1997) Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 277(5322):55–60PubMedCrossRef
95.
Zurück zum Zitat Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, Trent JM, Meltzer PS, Hendrix MJ (1999) Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155(3):739–752PubMedCrossRef Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, Trent JM, Meltzer PS, Hendrix MJ (1999) Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 155(3):739–752PubMedCrossRef
96.
Zurück zum Zitat Matsubara T, Kanto T, Kuroda S et al (2012) TIE2-expressing monocytes as a diagnostic marker for hepatocellular carcinoma correlated with angiogenesis. Hepatology. doi:10.1002/hep.25965 Matsubara T, Kanto T, Kuroda S et al (2012) TIE2-expressing monocytes as a diagnostic marker for hepatocellular carcinoma correlated with angiogenesis. Hepatology. doi:10.​1002/​hep.​25965
97.
Zurück zum Zitat Mazzieri R, Pucci F, Moi D et al (2011) Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell 19(4):431–433CrossRef Mazzieri R, Pucci F, Moi D et al (2011) Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell 19(4):431–433CrossRef
98.
Zurück zum Zitat McDonald DM, Foss AJ (2000) Endothelial cells of tumor vessels: abnormal but not absent. Cancer Metastasis Rev 19(1–2):109–120PubMedCrossRef McDonald DM, Foss AJ (2000) Endothelial cells of tumor vessels: abnormal but not absent. Cancer Metastasis Rev 19(1–2):109–120PubMedCrossRef
99.
Zurück zum Zitat McDonald DM, Munn L, Jain RK (2000) Vasculogenic mimicry: how convincing, how novel, and how significant? Am J Pathol 156(2):383–388PubMedCrossRef McDonald DM, Munn L, Jain RK (2000) Vasculogenic mimicry: how convincing, how novel, and how significant? Am J Pathol 156(2):383–388PubMedCrossRef
101.
Zurück zum Zitat Murdoch C, Muthana M, Coffelt SB, Lewis CE (2008) The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 8(8):618–631PubMedCrossRef Murdoch C, Muthana M, Coffelt SB, Lewis CE (2008) The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 8(8):618–631PubMedCrossRef
102.
Zurück zum Zitat Narayana A, Gruber D, Kunnakkat S, Golfinos JG, Parker E, Raza S, Zagzag D, Eagan P, Gruber ML (2012) A clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastoma. J Neurosurg 116(2):341–345PubMedCrossRef Narayana A, Gruber D, Kunnakkat S, Golfinos JG, Parker E, Raza S, Zagzag D, Eagan P, Gruber ML (2012) A clinical trial of bevacizumab, temozolomide, and radiation for newly diagnosed glioblastoma. J Neurosurg 116(2):341–345PubMedCrossRef
103.
Zurück zum Zitat Norden AD, Bartolomeo J, Tanaka S et al (2012) Safety of concurrent bevacizumab therapy and anticoagulation in glioma patients. J Neurooncol 106(1):121–125PubMedCrossRef Norden AD, Bartolomeo J, Tanaka S et al (2012) Safety of concurrent bevacizumab therapy and anticoagulation in glioma patients. J Neurooncol 106(1):121–125PubMedCrossRef
104.
Zurück zum Zitat Norden AD, Drappatz J, Wen PY (2009) Antiangiogenic therapies for high-grade glioma. Nat Rev Neurol 5(11):610–620PubMedCrossRef Norden AD, Drappatz J, Wen PY (2009) Antiangiogenic therapies for high-grade glioma. Nat Rev Neurol 5(11):610–620PubMedCrossRef
105.
Zurück zum Zitat Onishi M, Ichikawa T, Kurozumi K, Fujii K, Yoshida K, Inoue S, Michiue H, Chiocca EA, Kaur B, Date I (2012) Bimodal anti-glioma mechanisms of cilengitide demonstrated by novel invasive glioma models. Neuropathology. doi:10.1111 Onishi M, Ichikawa T, Kurozumi K, Fujii K, Yoshida K, Inoue S, Michiue H, Chiocca EA, Kaur B, Date I (2012) Bimodal anti-glioma mechanisms of cilengitide demonstrated by novel invasive glioma models. Neuropathology. doi:10.​1111
106.
Zurück zum Zitat Oosthuyse B, Moons L, Storkebaum E et al (2001) Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat Genet 28(2):131–138PubMedCrossRef Oosthuyse B, Moons L, Storkebaum E et al (2001) Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration. Nat Genet 28(2):131–138PubMedCrossRef
107.
Zurück zum Zitat Pàez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, Inoue M, Bergers G, Hanahan D, Casanovas O (2009) Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15(3):220–231PubMedCrossRef Pàez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Viñals F, Inoue M, Bergers G, Hanahan D, Casanovas O (2009) Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15(3):220–231PubMedCrossRef
108.
Zurück zum Zitat Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM (2003) Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 3(4):347–361PubMedCrossRef Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM (2003) Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell 3(4):347–361PubMedCrossRef
109.
Zurück zum Zitat Piao Y, Liang J, Holmes L, Zurita AJ, Henry V, Heymach JV, de Groot JF (2012) Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro Oncol. doi:10.1093/neuonc/nos158 PubMed Piao Y, Liang J, Holmes L, Zurita AJ, Henry V, Heymach JV, de Groot JF (2012) Glioblastoma resistance to anti-VEGF therapy is associated with myeloid cell infiltration, stem cell accumulation, and a mesenchymal phenotype. Neuro Oncol. doi:10.​1093/​neuonc/​nos158 PubMed
110.
Zurück zum Zitat Plate KH (1999) Mechanisms of angiogenesis in the brain. J Neuropathol Exp Neurol 58(4):313–320PubMedCrossRef Plate KH (1999) Mechanisms of angiogenesis in the brain. J Neuropathol Exp Neurol 58(4):313–320PubMedCrossRef
111.
Zurück zum Zitat Plate KH, Breier G, Risau W (1994) Molecular mechanisms of developmental and tumor angiogenesis. Brain Pathol 4(3):207–218PubMedCrossRef Plate KH, Breier G, Risau W (1994) Molecular mechanisms of developmental and tumor angiogenesis. Brain Pathol 4(3):207–218PubMedCrossRef
112.
Zurück zum Zitat Plate KH, Breier G, Millauer B, Ullrich A, Risau W (1993) Up-regulation of vascular endothelial growth factor and its cognate receptors in a rat glioma model of tumor angiogenesis. Cancer Res 53(23):5822–5827PubMed Plate KH, Breier G, Millauer B, Ullrich A, Risau W (1993) Up-regulation of vascular endothelial growth factor and its cognate receptors in a rat glioma model of tumor angiogenesis. Cancer Res 53(23):5822–5827PubMed
113.
Zurück zum Zitat Plate KH, Breier G, Weich HA, Risau W (1992) Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 359(6398):845–848PubMedCrossRef Plate KH, Breier G, Weich HA, Risau W (1992) Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 359(6398):845–848PubMedCrossRef
114.
Zurück zum Zitat Plate KH, Breier G, Weich HA, Mennel HD, Risau W (1994) Vascular endothelial growth factor and glioma angiogenesis: coordinate induction of VEGF receptors, distribution of VEGF protein and possible in vivo regulatory mechanisms. Int J Cancer 59(4):520–529PubMedCrossRef Plate KH, Breier G, Weich HA, Mennel HD, Risau W (1994) Vascular endothelial growth factor and glioma angiogenesis: coordinate induction of VEGF receptors, distribution of VEGF protein and possible in vivo regulatory mechanisms. Int J Cancer 59(4):520–529PubMedCrossRef
115.
Zurück zum Zitat Raab S, Plate KH (2007) Different networks, common growth factors: shared growth factors and receptors of the vascular and the nervous system. Acta Neuropathol 113(6):607–626PubMedCrossRef Raab S, Plate KH (2007) Different networks, common growth factors: shared growth factors and receptors of the vascular and the nervous system. Acta Neuropathol 113(6):607–626PubMedCrossRef
116.
Zurück zum Zitat Raab S, Beck H, Gaumann A, Yüce A, Gerber H-P, Plate K, Hammes H-P, Ferrara N, Breier G (2004) Impaired brain angiogenesis and neuronal apoptosis induced by conditional homozygous inactivation of vascular endothelial growth factor. Thromb Haemost 91(3):595–605PubMed Raab S, Beck H, Gaumann A, Yüce A, Gerber H-P, Plate K, Hammes H-P, Ferrara N, Breier G (2004) Impaired brain angiogenesis and neuronal apoptosis induced by conditional homozygous inactivation of vascular endothelial growth factor. Thromb Haemost 91(3):595–605PubMed
117.
Zurück zum Zitat Rapisarda A, Melillo G (2012) Overcoming disappointing results with antiangiogenic therapy by targeting hypoxia. Nat Rev Clin Oncol 9(7):378–390PubMedCrossRef Rapisarda A, Melillo G (2012) Overcoming disappointing results with antiangiogenic therapy by targeting hypoxia. Nat Rev Clin Oncol 9(7):378–390PubMedCrossRef
118.
Zurück zum Zitat Reis M, Czupalla CJ, Ziegler N et al (2012) Endothelial Wnt/ß-catenin signaling inhibits glioma angiogenesis and normalizes tumor blood vessels by inducing PDGF-B expression. J Exp Med 209(9):1611–1627PubMedCrossRef Reis M, Czupalla CJ, Ziegler N et al (2012) Endothelial Wnt/ß-catenin signaling inhibits glioma angiogenesis and normalizes tumor blood vessels by inducing PDGF-B expression. J Exp Med 209(9):1611–1627PubMedCrossRef
119.
Zurück zum Zitat Ribatti D, Djonov V (2012) Intussusceptive microvascular growth in tumors. Cancer Lett 316(2):126–131PubMedCrossRef Ribatti D, Djonov V (2012) Intussusceptive microvascular growth in tumors. Cancer Lett 316(2):126–131PubMedCrossRef
120.
Zurück zum Zitat Ricci-Vitiani L, Pallini R, Biffoni M et al (2010) Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468(7325):824–828PubMedCrossRef Ricci-Vitiani L, Pallini R, Biffoni M et al (2010) Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 468(7325):824–828PubMedCrossRef
122.
Zurück zum Zitat Roca C, Adams RH (2007) Regulation of vascular morphogenesis by Notch signaling. Genes Dev 21(20):2511–2524PubMedCrossRef Roca C, Adams RH (2007) Regulation of vascular morphogenesis by Notch signaling. Genes Dev 21(20):2511–2524PubMedCrossRef
123.
Zurück zum Zitat Rodriguez FJ, Orr BA, Ligon KL, Eberhart CG (2012) Neoplastic cells are a rare component in human glioblastoma microvasculature. Oncotarget 3(1):98–106PubMed Rodriguez FJ, Orr BA, Ligon KL, Eberhart CG (2012) Neoplastic cells are a rare component in human glioblastoma microvasculature. Oncotarget 3(1):98–106PubMed
124.
Zurück zum Zitat Rosenstein JM, Krum JM, Ruhrberg C (2010) VEGF in the nervous system. Organogenesis 6(2):107–114PubMedCrossRef Rosenstein JM, Krum JM, Ruhrberg C (2010) VEGF in the nervous system. Organogenesis 6(2):107–114PubMedCrossRef
125.
Zurück zum Zitat Ruiz de Almodovar C, Lambrechts D, Mazzone M, Carmeliet P (2009) Role and therapeutic potential of VEGF in the nervous system. Physiol Rev 89(2):607–648PubMedCrossRef Ruiz de Almodovar C, Lambrechts D, Mazzone M, Carmeliet P (2009) Role and therapeutic potential of VEGF in the nervous system. Physiol Rev 89(2):607–648PubMedCrossRef
126.
Zurück zum Zitat Scaringi C, Minniti G, Caporello P, Enrici RM (2012) Integrin inhibitor cilengitide for the treatment of glioblastoma: a brief overview of current clinical results. Anticancer Res 32(10):4213–4223PubMed Scaringi C, Minniti G, Caporello P, Enrici RM (2012) Integrin inhibitor cilengitide for the treatment of glioblastoma: a brief overview of current clinical results. Anticancer Res 32(10):4213–4223PubMed
127.
Zurück zum Zitat Schanzer A, Wachs F-P, Wilhelm D, Acker T, Cooper-Kuhn C, Beck H, Winkler J, Aigner L, Plate KH, Kuhn HG (2004) Direct stimulation of adult neural stem cells in vitro and neurogenesis in vivo by vascular endothelial growth factor. Brain Pathol 14(3):237–248PubMedCrossRef Schanzer A, Wachs F-P, Wilhelm D, Acker T, Cooper-Kuhn C, Beck H, Winkler J, Aigner L, Plate KH, Kuhn HG (2004) Direct stimulation of adult neural stem cells in vitro and neurogenesis in vivo by vascular endothelial growth factor. Brain Pathol 14(3):237–248PubMedCrossRef
128.
Zurück zum Zitat Scholz A, Lang V, Henschler R et al (2011) Angiopoietin-2 promotes myeloid cell infiltration in a β2-integrin-dependent manner. Blood 118(18):5050–5059PubMedCrossRef Scholz A, Lang V, Henschler R et al (2011) Angiopoietin-2 promotes myeloid cell infiltration in a β2-integrin-dependent manner. Blood 118(18):5050–5059PubMedCrossRef
129.
Zurück zum Zitat Sennino B, McDonald DM (2012) Controlling escape from angiogenesis inhibitors. Nat Rev Cancer 12(10):699–709PubMedCrossRef Sennino B, McDonald DM (2012) Controlling escape from angiogenesis inhibitors. Nat Rev Cancer 12(10):699–709PubMedCrossRef
130.
Zurück zum Zitat Sennino B, Ishiguro-Oonuma T, Wei Y et al (2012) Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov 2(3):270–287PubMedCrossRef Sennino B, Ishiguro-Oonuma T, Wei Y et al (2012) Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov 2(3):270–287PubMedCrossRef
131.
Zurück zum Zitat Seystahl K, Weller M (2012) Is there a world beyond bevacizumab in targeting angiogenesis in glioblastoma? Expert Opin Investig Drugs 21(5):605–617PubMedCrossRef Seystahl K, Weller M (2012) Is there a world beyond bevacizumab in targeting angiogenesis in glioblastoma? Expert Opin Investig Drugs 21(5):605–617PubMedCrossRef
132.
Zurück zum Zitat Shabo I, Olsson H, Sun X-F, Svanvik J (2009) Expression of the macrophage antigen CD163 in rectal cancer cells is associated with early local recurrence and reduced survival time. Int J Cancer 125(8):1826–1831PubMedCrossRef Shabo I, Olsson H, Sun X-F, Svanvik J (2009) Expression of the macrophage antigen CD163 in rectal cancer cells is associated with early local recurrence and reduced survival time. Int J Cancer 125(8):1826–1831PubMedCrossRef
133.
Zurück zum Zitat Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC (1995) Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376(6535):62–66PubMedCrossRef Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC (1995) Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376(6535):62–66PubMedCrossRef
134.
Zurück zum Zitat Shweiki D, Itin A, Soffer D, Keshet E (1992) Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359(6398):843–845PubMedCrossRef Shweiki D, Itin A, Soffer D, Keshet E (1992) Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature 359(6398):843–845PubMedCrossRef
135.
Zurück zum Zitat Sica A, Schioppa T, Mantovani A, Allavena P (2006) Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer 42(6):717–727PubMedCrossRef Sica A, Schioppa T, Mantovani A, Allavena P (2006) Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur J Cancer 42(6):717–727PubMedCrossRef
136.
Zurück zum Zitat Sikkema AH, de Bont ESJM, Molema G, Dimberg A, Zwiers PJ, Diks SH, Hoving EW, Kamps WA, Peppelenbosch MP, den Dunnen WFA (2011) Vascular endothelial growth factor receptor 2 (VEGFR-2) signalling activity in paediatric pilocytic astrocytoma is restricted to tumour endothelial cells. Neuropathol Appl Neurobiol 37(5):538–548PubMedCrossRef Sikkema AH, de Bont ESJM, Molema G, Dimberg A, Zwiers PJ, Diks SH, Hoving EW, Kamps WA, Peppelenbosch MP, den Dunnen WFA (2011) Vascular endothelial growth factor receptor 2 (VEGFR-2) signalling activity in paediatric pilocytic astrocytoma is restricted to tumour endothelial cells. Neuropathol Appl Neurobiol 37(5):538–548PubMedCrossRef
137.
Zurück zum Zitat Sitohy B, Nagy JA, Dvorak HF (2012) Anti-VEGF/VEGFR therapy for cancer: reassessing the target. Cancer Res 72(8):1909–1914PubMedCrossRef Sitohy B, Nagy JA, Dvorak HF (2012) Anti-VEGF/VEGFR therapy for cancer: reassessing the target. Cancer Res 72(8):1909–1914PubMedCrossRef
138.
Zurück zum Zitat Sorensen AG, Emblem KE, Polaskova P et al (2012) Increased survival of glioblastoma patients who respond to antiangiogenic therapy with elevated blood perfusion. Cancer Res 72(2):402–407PubMedCrossRef Sorensen AG, Emblem KE, Polaskova P et al (2012) Increased survival of glioblastoma patients who respond to antiangiogenic therapy with elevated blood perfusion. Cancer Res 72(2):402–407PubMedCrossRef
139.
Zurück zum Zitat Steidl C, Lee T, Shah SP et al (2010) Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med 362(10):875–885PubMedCrossRef Steidl C, Lee T, Shah SP et al (2010) Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma. N Engl J Med 362(10):875–885PubMedCrossRef
140.
Zurück zum Zitat Storkebaum E, Lambrechts D, Dewerchin M et al (2005) Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat Neurosci 8(1):85–92PubMedCrossRef Storkebaum E, Lambrechts D, Dewerchin M et al (2005) Treatment of motoneuron degeneration by intracerebroventricular delivery of VEGF in a rat model of ALS. Nat Neurosci 8(1):85–92PubMedCrossRef
141.
Zurück zum Zitat Stratmann A, Risau W, Plate KH (1998) Cell type-specific expression of Angiopoietin-1 and Angiopoietin-2 suggests a role in glioblastoma angiogenesis. Am J Pathol 153(5):1459–1466PubMedCrossRef Stratmann A, Risau W, Plate KH (1998) Cell type-specific expression of Angiopoietin-1 and Angiopoietin-2 suggests a role in glioblastoma angiogenesis. Am J Pathol 153(5):1459–1466PubMedCrossRef
142.
Zurück zum Zitat Thompson EM, Frenkel EP, Neuwelt EA (2011) The paradoxical effect of bevacizumab in the therapy of malignant gliomas. Neurology 76(1):87–93PubMedCrossRef Thompson EM, Frenkel EP, Neuwelt EA (2011) The paradoxical effect of bevacizumab in the therapy of malignant gliomas. Neurology 76(1):87–93PubMedCrossRef
143.
Zurück zum Zitat Tung JJ, Tattersall IW, Kitajewski J (2012) Tips, stalks, tubes: Notch-mediated cell fate determination and mechanisms of tubulogenesis during angiogenesis. Cold Spring Harb Perspect Med 2(2):a006601PubMed Tung JJ, Tattersall IW, Kitajewski J (2012) Tips, stalks, tubes: Notch-mediated cell fate determination and mechanisms of tubulogenesis during angiogenesis. Cold Spring Harb Perspect Med 2(2):a006601PubMed
144.
Zurück zum Zitat Valenzuela DM, Griffiths JA, Rojas J et al (1999) Angiopoietins 3 and 4: diverging gene counterparts in mice and humans. Proc Natl Acad Sci USA 96(5):1904–1909PubMedCrossRef Valenzuela DM, Griffiths JA, Rojas J et al (1999) Angiopoietins 3 and 4: diverging gene counterparts in mice and humans. Proc Natl Acad Sci USA 96(5):1904–1909PubMedCrossRef
145.
Zurück zum Zitat van Agtmaal EL, Bierings R, Dragt BS, Leyen TA, Fernandez-Borja M, Horrevoets AJG, Voorberg J (2012) The shear stress-induced transcription factor KLF2 affects dynamics and Angiopoietin-2 content of Weibel-Palade bodies. PLoS One 7(6):e38399PubMedCrossRef van Agtmaal EL, Bierings R, Dragt BS, Leyen TA, Fernandez-Borja M, Horrevoets AJG, Voorberg J (2012) The shear stress-induced transcription factor KLF2 affects dynamics and Angiopoietin-2 content of Weibel-Palade bodies. PLoS One 7(6):e38399PubMedCrossRef
146.
Zurück zum Zitat Verheyen A, Peeraer E, Nuydens R et al (2012) Systemic anti-vascular endothelial growth factor therapies induce a painful sensory neuropathy. Brain 135(Pt 9):2629–2641PubMedCrossRef Verheyen A, Peeraer E, Nuydens R et al (2012) Systemic anti-vascular endothelial growth factor therapies induce a painful sensory neuropathy. Brain 135(Pt 9):2629–2641PubMedCrossRef
147.
Zurück zum Zitat Vredenburgh JJ, Desjardins A, Herndon JE et al (2007) Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 25(30):4722–4729PubMedCrossRef Vredenburgh JJ, Desjardins A, Herndon JE et al (2007) Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol 25(30):4722–4729PubMedCrossRef
148.
Zurück zum Zitat Wang R, Chadalavada K, Wilshire J, Kowalik U, Hovinga KE, Geber A, Fligelman B, Leversha M, Brennan C, Tabar V (2010) Glioblastoma stem-like cells give rise to tumour endothelium. Nature 468(7325):829–833PubMedCrossRef Wang R, Chadalavada K, Wilshire J, Kowalik U, Hovinga KE, Geber A, Fligelman B, Leversha M, Brennan C, Tabar V (2010) Glioblastoma stem-like cells give rise to tumour endothelium. Nature 468(7325):829–833PubMedCrossRef
149.
Zurück zum Zitat Wick A, Wick W, Waltenberger J, Weller M, Dichgans J, Schulz JB (2002) Neuroprotection by hypoxic preconditioning requires sequential activation of vascular endothelial growth factor receptor and Akt. J Neurosci 22(15):6401–6407PubMed Wick A, Wick W, Waltenberger J, Weller M, Dichgans J, Schulz JB (2002) Neuroprotection by hypoxic preconditioning requires sequential activation of vascular endothelial growth factor receptor and Akt. J Neurosci 22(15):6401–6407PubMed
150.
Zurück zum Zitat Wong AL, Haroon ZA, Werner S, Dewhirst MW, Greenberg CS, Peters KG (1997) Tie2 expression and phosphorylation in angiogenic and quiescent adult tissues. Circ Res 81(4):567–574PubMedCrossRef Wong AL, Haroon ZA, Werner S, Dewhirst MW, Greenberg CS, Peters KG (1997) Tie2 expression and phosphorylation in angiogenic and quiescent adult tissues. Circ Res 81(4):567–574PubMedCrossRef
151.
Zurück zum Zitat Xu L, Duda DG, di Tomaso E et al (2009) Direct evidence that bevacizumab, an anti-VEGF antibody, up-regulates SDF1alpha, CXCR4, CXCL6, and neuropilin 1 in tumors from patients with rectal cancer. Cancer Res 69(20):7905–7910PubMedCrossRef Xu L, Duda DG, di Tomaso E et al (2009) Direct evidence that bevacizumab, an anti-VEGF antibody, up-regulates SDF1alpha, CXCR4, CXCL6, and neuropilin 1 in tumors from patients with rectal cancer. Cancer Res 69(20):7905–7910PubMedCrossRef
152.
Zurück zum Zitat Yan M, Callahan CA, Beyer JC, Allamneni KP, Zhang G, Ridgway JB, Niessen K, Plowman GD (2010) Chronic DLL4 blockade induces vascular neoplasms. Nature 463(7282):E6–E7PubMedCrossRef Yan M, Callahan CA, Beyer JC, Allamneni KP, Zhang G, Ridgway JB, Niessen K, Plowman GD (2010) Chronic DLL4 blockade induces vascular neoplasms. Nature 463(7282):E6–E7PubMedCrossRef
153.
Zurück zum Zitat Yang X, Cepko CL (1996) Flk-1, a receptor for vascular endothelial growth factor (VEGF), is expressed by retinal progenitor cells. J Neurosci 16(19):6089–6099PubMed Yang X, Cepko CL (1996) Flk-1, a receptor for vascular endothelial growth factor (VEGF), is expressed by retinal progenitor cells. J Neurosci 16(19):6089–6099PubMed
154.
Zurück zum Zitat You W-K, Sennino B, Williamson CW, Falcón B, Hashizume H, Yao L-C, Aftab DT, McDonald DM (2011) VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer. Cancer Res 71(14):4758–4768PubMedCrossRef You W-K, Sennino B, Williamson CW, Falcón B, Hashizume H, Yao L-C, Aftab DT, McDonald DM (2011) VEGF and c-Met blockade amplify angiogenesis inhibition in pancreatic islet cancer. Cancer Res 71(14):4758–4768PubMedCrossRef
155.
Zurück zum Zitat Yue W-Y, Chen Z-P (2005) Does vasculogenic mimicry exist in astrocytoma? J Histochem Cytochem 53(8):997–1002PubMedCrossRef Yue W-Y, Chen Z-P (2005) Does vasculogenic mimicry exist in astrocytoma? J Histochem Cytochem 53(8):997–1002PubMedCrossRef
156.
Zurück zum Zitat Zagzag D, Zhong H, Scalzitti JM, Laughner E, Simons JW, Semenza GL (2000) Expression of hypoxia-inducible factor 1alpha in brain tumors: association with angiogenesis, invasion, and progression. Cancer 88(11):2606–2618PubMedCrossRef Zagzag D, Zhong H, Scalzitti JM, Laughner E, Simons JW, Semenza GL (2000) Expression of hypoxia-inducible factor 1alpha in brain tumors: association with angiogenesis, invasion, and progression. Cancer 88(11):2606–2618PubMedCrossRef
157.
Zurück zum Zitat Zhang BC, Gao J, Wang J, Rao ZG, Wang BC, Gao JF (2011) Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma. Med Oncol 28(4):1447–1452PubMedCrossRef Zhang BC, Gao J, Wang J, Rao ZG, Wang BC, Gao JF (2011) Tumor-associated macrophages infiltration is associated with peritumoral lymphangiogenesis and poor prognosis in lung adenocarcinoma. Med Oncol 28(4):1447–1452PubMedCrossRef
Metadaten
Titel
Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited
verfasst von
Karl H. Plate
Alexander Scholz
Daniel J. Dumont
Publikationsdatum
01.12.2012
Verlag
Springer-Verlag
Erschienen in
Acta Neuropathologica / Ausgabe 6/2012
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-012-1066-5

Weitere Artikel der Ausgabe 6/2012

Acta Neuropathologica 6/2012 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neu im Fachgebiet Neurologie

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.