2,631
Views
35
CrossRef citations to date
0
Altmetric
Review

Vessel pruning or healing: endothelial metabolism as a novel target?

, , &
Pages 239-247 | Received 11 Oct 2016, Accepted 11 Jan 2017, Published online: 27 Jan 2017

ABSTRACT

Introduction: Antiangiogenic drugs were originally designed to starve tumors by cutting off their vascular supply. Unfortunately, when these agents are used as monotherapy or in combination with chemotherapy, they provide only modest survival benefits in the order of weeks to months in most cancer patients. Strategies normalizing the disorganized tumor vasculature offer the potential to increase tumor perfusion and oxygenation, and to improve the efficacy of radio-, chemo- and immunotherapy, while reducing metastasis.

Areas covered: This review discusses tumor vascular normalization (TVN) as an alternative strategy for anti-angiogenic cancer treatment. We summarize (pre)-clinical strategies that have been developed to normalize tumor vessels as well as their potential to enhance standard therapy. Notably, we describe how targeting endothelial cell metabolism offers new possibilities for antiangiogenic therapy through evoking TVN.

Expert opinion: Several drugs targeting VEGF signaling are now clinically used for antiangiogenic cancer treatment. However, excessive blood vessel pruning impedes perfusion and causes tumor hypoxia, known to promote cancer cell dissemination and impair radio-, chemo- and immunotherapy. Normalized vessels lessen tumor hypoxia, impair cancer cell intravasation and enhance anticancer treatment. New data indicate that targeting endothelial cell metabolism is an alternative strategy of antiangiogenic cancer treatment via promotion of TVN.

1. Introduction

Angiogenesis is the process of new blood vessel formation [Citation1]. It occurs throughout life in both health and disease and relies on migration, proliferation, and differentiation of endothelial cells (ECs), which line the inside wall of blood vessels. In established vessels, ECs have a cobblestone-like appearance and are often referred to as phalanx cells [Citation2]. However, in the presence of pro-angiogenic signals such as growth factors and hypoxia, ECs can rapidly switch to an angiogenic state and become motile and invasive [Citation3,Citation4]. Upon detection of pro-angiogenic stimuli, such as VEGF (vascular endothelial growth factor), ECs lose their adherence junctions, and matrix metalloproteases degrade the basement membrane, thereby creating a scaffold for EC migration [Citation3,Citation4]. In response to such stimuli, a vessel sprout emerges in which individual ECs adopt distinct and functionally specialized phenotypes [Citation3].

Vessel sprouting is initiated by the differentiation of ECs into specialized tip and stalk cells. Tip cells extend long filopodia and guide the new sprout toward an angiogenic stimulus but proliferate rarely, whereas stalk cells have fewer filopodia, proliferate to elongate the branch and form a vascular lumen [Citation3,Citation4]. When the new branch is formed and perfused, ECs regain their quiescent phalanx cell phenotype. Recruitment of pericytes and vascular smooth muscle cells, together referred to as mural cells, provides stability, maintains vessel integrity and regulates perfusion [Citation3,Citation4]. Furthermore, extracellular matrix proteins, laid down by both phalanx cells and pericytes, establish a basement membrane at the basal side of the endothelium [Citation3Citation5].

Blood vessels not only deliver oxygen and nutrients to the body’s tissues but also support diseases such as cancer [Citation6]. Rapidly growing tumors are in continuous demand for oxygen and nutrients. Therefore, they have an excessive production of angiogenic stimuli, which creates an imbalance in pro- versus antiangiogenic signaling. This results in an abnormal, leaky, and hypoperfused vascular network, characterized by hypoxia, acidosis, and high interstitial fluid pressure. This hostile tumor microenvironment stimulates the production of pro-angiogenic factors even more and fuels an endless self-reinforcing loop of nonproductive angiogenesis [Citation7,Citation8]. Paradoxically, however, this nonproductive angiogenesis leads to the formation of less functional vessels. This results in hypoxic sites within the tumor, deprived of nutrients and growth factors, creating a hostile microenvironment, from where cancer cells attempt to escape, thereby favoring cancer cell invasion and dissemination, further aided by the leaky endothelium, through which cancer cells can escape [Citation7,Citation8]. Moreover, the abnormal tumor vasculature impairs perfusion and thus drug delivery and distribution. Together, these features stimulate overall malignancy [Citation7,Citation9,Citation10].

Therapeutic approaches for inhibition of angiogenesis have been developed to treat cancer and have led to the approval of several antiangiogenic drugs. So far, innumerable patients have benefited from these therapies but limited efficacy and resistance pose clinical challenges [Citation8,Citation11Citation13]. A novel paradigm is to heal the abnormal tumor vasculature in a process called tumor vessel normalization [Citation10]. Tumor vascular normalization (TVN) strategies reduce metastasis and improve the response to conventional anticancer therapies [Citation10,Citation14].

2. Role of abnormal vasculature in tumor progression and therapeutic strategies

2.1. Tumor vessels are highly abnormal

Tumor blood vessels are structurally and functionally highly abnormal. They are tortuous, leaky, irregular and form a chaotic network. Moreover, they are heterogeneous in size and shape, ranging from capillaries to big, thin-walled vessels [Citation7,Citation9,Citation10] (). ECs lining tumor vessels have an irregular shape and are disorganized. They establish weak junctions, which results in a loose association, promoting EC trans-migration from their resident site. In certain regions, ECs are stacked upon each other and extend multiple protrusions within the lumen, thus obstructing the blood flow, while at other sites, ECs move away from their position, leaving gaps behind. In addition, mural cells show abnormal structural features. They have an abnormal shape and are often loosely associated with ECs or absent. Finally, the basement membrane of tumor vessels is also abnormal. In some tumors, it is unusually tick, while very thin, discontinuous, or absent in others [Citation7,Citation9,Citation10].

Figure 1. Tumor vessels are structurally and functionally abnormal.

Endothelial cells lining tumor vessels demonstrate aberrations in shape, they are hyperproliferative and hypermigrative and are often separated by wide and irregular inter-endothelial junctions. In addition, tumor vessels are covered by fewer pericytes, which are often detached from endothelial cells. These structural abnormalities lead to hypoperfusion and hypoxia, which stimulate cancer cells to escape and metastasize in distant organs. Moreover, the accompanying functional aberrations limit delivery and distribution of chemotherapeutics to and into the tumors. BM: basement membrane. Adapted from [Citation9].

Figure 1. Tumor vessels are structurally and functionally abnormal.Endothelial cells lining tumor vessels demonstrate aberrations in shape, they are hyperproliferative and hypermigrative and are often separated by wide and irregular inter-endothelial junctions. In addition, tumor vessels are covered by fewer pericytes, which are often detached from endothelial cells. These structural abnormalities lead to hypoperfusion and hypoxia, which stimulate cancer cells to escape and metastasize in distant organs. Moreover, the accompanying functional aberrations limit delivery and distribution of chemotherapeutics to and into the tumors. BM: basement membrane. Adapted from [Citation9].

Abnormalities in tumor vessel morphology impair perfusion. Blood flow often changes direction, flowing rapidly in certain vessels and being stagnant in others. In addition, the structural abnormalities lead to increased vessel permeability. Hence, intravascular fluids and plasma proteins can easily extravasate and increase interstitial fluid pressure, further impeding blood flow. Moreover, these leaky vessels facilitate intravasation of cancer cells and metastasis [Citation8Citation10]. The combination of poor perfusion and increased interstitial fluid pressure creates hypoxic and acidic sites. In such conditions, invasive cancer cells are selected. Overall, the aforementioned tumor vessel abnormalities increase metastasis and impair the delivery and efficacy of systemically administered drugs, therefore reducing the efficacy of anticancer therapies [Citation7Citation10].

2.2. Vascular normalization as anticancer strategy: an emerging paradigm

Anti-VEGF therapies were originally designed to restore the balance between pro- and antiangiogenic molecules and the vascular architecture of tumors by pruning immature vessels [Citation15Citation17]. Early preclinical studies were promising and demonstrated a significant tumor growth delay and reduced metastasis [Citation16,Citation18]. Unfortunately, the effects of such anti-VEGF agents in cancer patients during clinical trials have not fulfilled the expected hopes as the survival improvement by antiangiogenic therapy is rather modest [Citation19]. Monotherapy with anti-VEGF monoclonal antibody often failed to attain substantial response rates or survival benefits [Citation20Citation22]. These data suggest that anti-VEGF therapy alone cannot efficiently induce sustained tumor shrinkage or complete tumor eradication in most cancer patients.

However, anti-VEGF therapy in combination with systemic chemotherapy has often shown improvements in progression-free survival when compared with chemotherapy [Citation21,Citation23Citation28]. This has been interpreted to imply that anti-VEGF therapy may enhance the efficacy of systemic chemotherapy. These findings are counterintuitive since anti-VEGF therapies were designed to promote vascular pruning; yet, the efficacy of chemotherapy relies on adequate tumor blood supply to ensure drug delivery. Thus, tumor vessel pruning by anti-VEGF treatment should theoretically decrease rather than enhance the chemotherapy efficacy [Citation10]. The hypothesis of vascular normalization may resolve this paradox [Citation29]. This hypothesis posits that rather than destroying vessels, antiangiogenic therapy might restore the normal structure and function of tumor vessels by ‘healing’ abnormal disorganized and dysfunctional tumor vessels. These changes would improve tumor oxygenation, thereby reducing metastasis and improving therapy responses, in part through more efficient drug delivery and lessened tumor hypoxia (which can improve the efficacy of several chemo- or radiotherapeutics, given that they rely on the conversion of oxygen to its radicals, and of immunotherapy [see below]). Thus, anticancer therapies given upon normalization might achieve greater efficacy [Citation29].

Initial evidence for vascular normalization stems from preclinical studies with anti-VEGF, which show that blockade of VEGF signaling remodels the abnormal tumor vasculature into a more normal vasculature [Citation14]. Of relevance, the kinetics of vascular normalization determined the overall outcome of combined antiangiogenic and conventional therapy. Preclinical and clinical tumor studies with anti-VEGF agents identified a ‘normalization window,’ typically occurring within a few days after therapy onset followed by a closure coinciding with the loss of normalization features [Citation10]. The transient nature of the normalization of tumor vessels may relate either to excessively high or continued dosing of antiangiogenic therapy or to the development of resistance by activation of alternative pro-angiogenic pathways [Citation10]. High doses of antiangiogenic drugs prune the immature tumor vessels, causing a rapid reduction in blood perfusion and consequent increase in hypoxia, which is a strong stimulus for cancer cells to invade, intravasate, and metastasize [Citation8]. On the contrary, properly timed low dose of anti-VEGF therapy promotes the formation of less tortuous vessels, normal basement membrane, and greater vessel coverage by pericytes. Indeed, low doses of anti-VEGFR2 antibody normalized the breast cancer vasculature and improved tissue perfusion, reprogramming the tumor microenvironment from immunosuppression toward enhanced cancer vaccine therapy [Citation30].

Other mechanisms might contribute to the vascular normalization effect of anti-VEGF agents. For instance, upon anti-VEGFR2 inhibition, mural cells upregulate angiopoietin-1 (ANG-1), promoting vessel maturation, stability, and integrity [Citation31]. The ANG/TIE-2 system represents another target for antiangiogenic therapies to induce tumor vessel normalization [Citation1,Citation8,Citation10]. In contrast to ANG-1, ANG-2 is primarily synthesized by ECs and antagonizes the effects of ANG-1. ANG-2 destabilizes blood vessels and promotes vascular permeability. Hence, preclinical reports demonstrated that blockade of ANG-2 induces vessel normalization by recruitment of pericytes and tightening of endothelial junctions [Citation32,Citation33]. Moreover, combined targeting of VEGF and ANG-2 showed a greater degree of vessel normalization [Citation34].

2.3. Emerging successful tumor vessel normalization strategies

In addition to targeting the VEGF/VEGFR and ANG/TIE-2 axes, a number of strategies that alter other molecules in stromal and cancer cells can induce vascular normalization. The oxygen sensor prolyl hydroxylase domain protein 2 (PHD2) plays an important role in mediating abnormalities in tumor vessels. In the tumor microenvironment, lack of oxygen reduces the enzymatic activity of PHD2 on one hand, and possibly as a feedback, also upregulates the expression levels of this enzyme. Nonetheless, hypoxia results in the activation of hypoxia-inducible transcription factors, key mediators of transcriptional response to hypoxia [Citation35]. In turn, VEGF is upregulated, thereby promoting abnormal angiogenesis [Citation36]. Studies in genetically modified mice have shown that global (stromal) or endothelial-specific PHD2 haplodeficiency does not impair physiological angiogenesis but induces sustained normalization of tumor vessels [Citation2,Citation37]. There are currently no pharmacological agents available to inhibit PHD2 specifically in ECs. In addition, global PHD2 haplodeficiency also induces tumor vessel normalization in part by decreasing the activation and contraction of cancer-associated fibroblasts (CAFs) [Citation38]. Mechanistically, PHD2 haplodeficient cancer cells inhibit CAF-induced matrix deposition that cancer cells use as a migration scaffold for dissemination, thereby reducing metastasis [Citation38]. This suggests that administration of a pharmacological blocker inhibiting PHD2 in both stromal and cancer cell compartments might offer therapeutic benefit by reducing metastatic disease.

Specific targeting of the transmembrane glycoprotein L1 on tumor vasculature also promotes tumor vessel normalization, resulting in reduced tumor growth and metastasis [Citation39]. The regulator of G-protein signaling 5 (RGS5) is a marker of mural cells that has been identified as a key mediator of the abnormal tumor vasculature [Citation40]. Tumor vessels of RGS5-deficient mice were characterized by reduced vessel permeability and leakage, increased structural homogeneity, improved oxygenation, and coverage by more mature pericytes. This enhanced the influx of immune effector cells into the tumor and prolonged the survival of the tumor-bearing mice [Citation40], offering alternative therapeutic opportunities for immunotherapy and anticancer therapy [Citation40]. The modulation of tumor-associated macrophages (TAMs) may be another way to control tumor vessel abnormalities. For instance, histidine-rich glycoprotein suppressed the expression of placental growth factor and induced the polarization of TAMs, promoting TVN and enhancement of immunity [Citation41].

The anti-malaric drug chloroquine also yielded promising results in cancer patients [Citation42]. While its anticancer cell activity relies on the blockade of cancer cell autophagy at a high dose, a low dose of chloroquine targets ECs via an autophagy-independent increase in Notch signaling, which promotes tumor EC quiescence and thereby tumor vessel normalization [Citation43,Citation44]. A number of FDA-approved drugs also target molecules that indirectly contribute to the tumor vessel abnormalities, namely by targeting cancer cells that subsequently affect tumor vessels. Examples include the human epidermal growth factor receptor 2, the phosphoinositide-3-kinase/AKT serine/threonine kinase 1/mammalian target of rapamycin (mTOR) axis, Ras, and the EGF receptor (reviewed in Ref. [Citation10]).

Taken together, the aforementioned studies suggest that targeting stromal, cancer, and other cells in the tumor milieu may improve the efficacy of tumor vessel normalization. The challenge for the future is to explore whether these approaches also promote sustained tumor vessel normalization in patients in clinical settings, as observed in preclinical studies.

2.4. Vessel normalization as strategy to improve cancer immunotherapy

In addition to the examples mentioned above, a growing body of evidence suggests that tumor vessel normalization may also enhance the efficacy of immunotherapy [Citation30,Citation40,Citation41,Citation45]. During tumor progression, cancer cells co-opt immune checkpoint pathways to promote immune evasion, particularly by cytotoxic T cells [Citation46]. Different strategies to interfere with ligand–receptor interactions involved in immune checkpoint pathways have been developed and have entered clinical practice [Citation47]. However, several hurdles need to be overcome to further improve such treatments.

Indeed, the efficacy of anticancer immunotherapy by blocking immune checkpoints is hampered by hypoxia and poor infiltration of T cells inside the tumor, as a result of the poor perfusion in the disorganized tumor vessels [Citation48]. Abnormal tumor vessels also limit the adhesion and extravasation of leukocytes and impair their infiltration inside the tumor core [Citation7,Citation9]. Hypoxia increases the immunosuppressive nature of the stromal tumor environment, by impairing T-cell effector functions (T-cell receptor signaling, proliferation, and cytokine production by T cells) [Citation48,Citation49]. In contrast, hyperoxia increases cytotoxic T-cell performance, which correlates with better clinical responses to blockade of the immune checkpoint molecule programed death 1 (PD-1) [Citation50]. In addition, nutrient deprivation (such as glucose) impedes T-cell proliferation and activation into CD8+ effector cells [Citation51].

Hence, tumor vessel normalization increases tumor perfusion, and thereby oxygen and nutrient supply, and thus can be expected to improve the overall anticancer immunotherapy response. This hypothesis is supported by findings that anti-VEGF therapy promotes antitumor immunity, in part by improving vessel function and reducing hypoxia [Citation30]. Restoring vessel integrity improves tumor perfusion and decreases interstitial fluid pressure, processes that would be expected to improve the influx of immune cells into the tumor [Citation40,Citation52]. Moreover, the fewer gaps between ECs in the normalized vasculature might create a more continuous vascular surface to support leucocyte rolling and diapedesis [Citation53]. However, the exact mechanism by which tumor vessel normalization strategies increase the influx of immune cells into the tumor, while reducing cancer cell escape, remains to be defined. The increased antitumor immunity responses observed upon anti-VEGF therapy might be also related to the inhibition of the immunosuppressive functions exerted by VEGF on effector T cells [Citation54]. Thus, blocking VEGF signaling enhances effector T-cell function by increasing their activation and delivery to the tumor (via tumor vessel normalization) on one hand and by inhibiting the VEGF-induced upregulation of inhibitory immune checkpoints, on the other hand [Citation30,Citation55].

2.5. Vessel normalization: a clinical perspective

Unfortunately, the high expectations of antiangiogenic therapies could not be fulfilled, as the high therapy efficacy observed in preclinical models could not be fully reproduced in clinical trials [Citation56]. Therapy effects in cancer patients are mostly short lived due to intrinsic refractoriness or development of acquired resistance upon antiangiogenic therapy. Several modes of resistance have been identified preclinically, but they are less well characterized in the clinical setting [Citation57,Citation58].

Limitations to better clinically characterize the mode of action of antiangiogenic therapies are related to multiple reasons. Amongst those, one reason may relate to the fact that most antiangiogenic drugs are used for the treatment of advanced stage disease (mostly metastatic), where tissue sampling is only possible from accessible tumors (for instance, rectal cancer via rectoscopy); and therefore, translational investigations are often limited to blood sampling or imaging studies [Citation19]. Nevertheless, in a subset of patients, tumor vessel normalization has been observed upon administration of antiangiogenic therapy [Citation59] ().

Table 1. Clinical studies investigating vascular normalization in humans.

The first studies investigating the influence of the VEGF neutralizing antibody bevacizumab on tumor vessel normalization were performed in rectal cancer patients. Bevacizumab monotherapy decreased the interstitial fluid pressure, while tumor blood vessels were covered with more pericytes as compared to pre-therapy investigations [Citation67]. Long-term follow-up of the subsequent phase II study showed high local tumor control rate and promising disease-free and overall survival upon neoadjuvant bevacizumab treatment in combination with standard chemoradiotherapy [Citation60], suggesting that vessel normalization features might induce a better therapeutic outcome. In support of this hypothesis, treatment with cediranib (a VEGFR tyrosine kinase inhibitor) resulted in increased tumor perfusion and prolonged survival in a subgroup of patients with recurrent glioblastoma multiforme [Citation61]. Accordingly, improved tumor vascularization after bevacizumab combination therapy with carboplatin and nab-paclitaxel was associated with longer survival in patients with advanced non-small cell lung cancer [Citation62]. Overall, the abovementioned studies may imply that antiangiogenic therapy regimens that promote tumor vessel normalization offer benefit to cancer patients. Clearly, the number of clinical studies showing beneficial outcome of tumor vessel normalization is still limited, and additional studies are needed to corroborate these initial clinical findings. In addition, it is of utmost importance to identify predictive biomarkers for vascular normalization and therapy stratification in future clinical trials [Citation19].

A recent clinical trial, including a small number of patients with metastatic renal cell carcinoma treated with bevacizumab in combination with an anti-PD-ligand 1 antibody showed that the combination treatment increases the intra-tumoral infiltration of CD8+ T cells, thus enhancing the anticancer immune-specific responses [Citation68]. The ongoing early phase I/II clinical trials will further reveal whether the anti-VEGF therapies in combination with checkpoint inhibitors increase the anticancer effects [Citation69].

Mechanistically, it has been hypothesized (and supported by preclinical evidence) that upon tumor vessel normalization, the improved vessel functionality leads to enhanced tumor delivery of chemotherapeutics [Citation70]. However, some clinical trials investigating the effect of antiangiogenic drugs on tumor delivery showed that antiangiogenic drugs reduced (rather than increased) the delivery of chemotherapy or biological drugs [Citation71,Citation66]. A reduction in tumor perfusion and vessel permeability has been proposed to explain the decreased tumor uptake of chemotherapeutics upon bevacizumab therapy [Citation72]. Further, drug penetration in tumors was enhanced only when the chemotherapeutic agent was administered within a defined interval after anti-VEGF therapy [Citation73,Citation74]. Together, while tumor vessel normalization is an attractive strategy to impede metastasis and improve chemo-, radio-, and immunotherapy in preclinical models, and initial signs have been recognized in clinical settings, other studies also highlight the importance of testing different treatment regimen schedules and dosing of bevacizumab in order to examine their effect on perfusion and permeability and to identify the normalization window in which the antiangiogenic agent exerts beneficial effects. Nonetheless, it will remain challenging to clinically translate the preclinical efficacy of VEGF-signaling blockers to induce long-lasting tumor vessel normalization required for optimal therapeutic benefit. Hence, we will need additional agents with a completely different mechanism, which are capable of inducing persistent tumor vessel normalization (as for instance documented in mice for chloroquine, PHD2 gene haplodeficiency, etc. – see above).

2.6. Tumor vessel normalization by targeting EC metabolism

An entirely different and new antiangiogenic approach to promote tumor vessel normalization is to target EC metabolism. This strategy is based on the postulate that EC metabolism is the engine onto which pro-angiogenic signals like VEGF and others converge and that ‘cooling down the overheated metabolism’ of ECs can paralyze angiogenic ECs and reduce pathological angiogenesis, regardless of how many angiogenic signals are still present upon neutralization of VEGF [Citation3,Citation75,Citation76]. Since ECs are highly glycolytic [Citation77], targeting glycolysis might provide an alternative new therapeutic opportunity for reducing pathological angiogenesis. Indeed, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) is a key regulator of glycolysis in ECs. Inhibition of PFKFB3, either by genetic loss in ECs or by global pharmacological blockade with the small molecule 3PO (3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one), reduces vessel sprouting by inhibiting EC proliferation and migration, not only in physiological but also in pathological conditions of inflammation and tissue injury [Citation77,Citation78]. Notably, however, PFKFB3 silencing or blockade reduces EC glycolysis only partially by no more than 35%, but still sufficient to normalize the hyper-glycolysis of sprouting ECs to maintenance levels found in quiescent ECs [Citation77,Citation78]. Thus, even a modest decrease in glycolysis was sufficient to impair vessel sprouting by promoting quiescence [Citation78]. In addition, the effect of 3PO in vivo was transient because of its short half-life (30 min) and rapid clearance. As a result of the partial, transient reduction (not elimination) of glycolysis, and the fact that ECs are more glycolysis addicted than other cell types, the effect of 3PO was well tolerated [Citation78,Citation79].

This type of anti-glycolytic approach differs substantially from previous anti-glycolytic anticancer therapies, which were not always successful [Citation80,Citation81], mainly because they attempted to eliminate glycolysis completely and permanently, which causes adverse effects. Indeed, the non-metabolizable glucose analog 2-deoxy-d-glucose (2DG), which reduces glycolysis by 80%, causes ATP depletion and EC death [Citation78]. Since high 2DG doses are needed to compete with the high levels of glucose in the blood, its effects are toxic.

Deletion of both PFKFB3 alleles in ECs decreases angiogenesis and perfusion in tumors [Citation82]. A more recent study showed however that endothelial haplodeficiency of PFKFB3 does not inhibit tumor growth but reduces metastasis and improves the delivery and response to chemotherapy, by normalizing tumor vessels [Citation83]. Treatment with a low dose of 3PO, which reduces glycolysis by only 15% in ECs, induces similar effects [Citation83]. Since ECs’ lining tumor blood vessels have a much higher glycolytic rate than healthy ECs, they are more sensitive to PFKFB3 blockade [Citation83]. This can explain why even the deletion of one allele of PFKFB3 or the use of a low dose of 3PO already induces tumor vessel normalization [Citation83]. Unlike traditional antiangiogenic agents [Citation84Citation86], PFKFB3 haplodeficiency or blockade does not reduce tumor vessel density or total vascular area. Instead, PFKFB3 inhibition enlarges the vessel lumen and stabilizes tumor vessels by increasing vessel maturation through pericyte recruitment. These morphological changes improve tumor perfusion and thereby lower tumor hypoxia [Citation83]. Overall, PFKFB3 blockade reduces metastasis and increases chemotherapy delivery and efficacy ().

Figure 2. Targeting endothelial cell metabolism induces tumor vessel normalization.

  • a. Schematic representation of the glycolytic pathway converting glucose into pyruvate. PFKFB3 is a key regulator of glycolysis by producing fructose-2,6-bisphosphate (F2,6P2), the most potent allosteric activator of phosphofructokinase-1 (PFK-1). G6P, glucose-6-phosphate; F6P, fructose-6-phosphate; F2,6P2, fructose-2,6-bisphosphate; PFK, phospho-fructokinase; 3PG, 3-phospho glyceraldehyde; TCA, tricarboxylic acid cycle; ATP, cellular adenosine 5ʹ-trisphosphate.

  • b. Upon inhibition of PFKFB3 in hyperglycolytic tumor endothelial cells, tumor vessels show smoother endothelial surface, reduced intercellular gaps, more prominent basement membrane and increased pericyte coverage. All these changes improve tumor vessel perfusion and thereby lower hypoxia, contributing to reduced invasion, intravasation and metastasis. Adapted from [Citation8].

Figure 2. Targeting endothelial cell metabolism induces tumor vessel normalization. a. Schematic representation of the glycolytic pathway converting glucose into pyruvate. PFKFB3 is a key regulator of glycolysis by producing fructose-2,6-bisphosphate (F2,6P2), the most potent allosteric activator of phosphofructokinase-1 (PFK-1). G6P, glucose-6-phosphate; F6P, fructose-6-phosphate; F2,6P2, fructose-2,6-bisphosphate; PFK, phospho-fructokinase; 3PG, 3-phospho glyceraldehyde; TCA, tricarboxylic acid cycle; ATP, cellular adenosine 5ʹ-trisphosphate.b. Upon inhibition of PFKFB3 in hyperglycolytic tumor endothelial cells, tumor vessels show smoother endothelial surface, reduced intercellular gaps, more prominent basement membrane and increased pericyte coverage. All these changes improve tumor vessel perfusion and thereby lower hypoxia, contributing to reduced invasion, intravasation and metastasis. Adapted from [Citation8].

3. Conclusions

Since the concept of tumor vessel normalization for anticancer therapy was proposed in 2001, promising progress has been made in preclinical studies and clinical trials. By restoring the balance between pro- and antiangiogenic factors in the tumor microenvironment, antiangiogenic therapy using adequate antiangiogenic agents and delivery regimens is able to ‘heal’ the perturbed tumor vessels and to restore a more normal tumor vasculature, capable of more efficiently delivery of cytotoxic drugs and other therapies such as immunotherapies to tumors. While preclinical studies have amply demonstrated the therapeutic benefit of tumor vessel normalization strategies, translation to the clinic is now required to provide proof of evidence for similar benefit in cancer patients. However, this requires clinical development of new alternative strategies, capable of inducing more long-lasting and more efficient tumor vessel normalization. Targeting endothelial metabolism by lowering glycolysis in tumor ECs is emerging as a novel anticancer therapeutic approach, capable of inducing tumor vessel normalization, and hence reducing metastasis while improving chemo-, radio-, and immunotherapy.

4. Expert opinion

More than a decade of clinical experience reveals that we have yet to realize the full potential of antiangiogenic therapy. Most antiangiogenic agents received market authorization after providing proof of efficacy in a phase III trial that PFS or overall survival is prolonged compared to the most efficacious available therapy. However, these studies did not sufficiently investigate the fundamental mechanisms of how antiangiogenic drugs in combination with chemotherapy produce clinical benefit, and the exact time window in which the antiangiogenic agents exert the greatest benefit remains largely undefined.

Cancer patients receive disrupting scheduling of anti-VEGF regimens, which are often interrupted because of toxicity, resistance, or high costs. Discontinuation of antiangiogenic therapy might cause a rebound effect, leading to disease progression and metastasis (as observed at least in preclinical studies [Citation11,Citation12,Citation85,Citation87]). One possible alternative, which has been poorly explored to date, might be to administer low (i.e. lower than maximally tolerated) doses of antiangiogenic drugs with the objective to promote tumor vessel normalization. In this case, the increased anticancer activity would not result from vessel pruning upon administration of high doses of antiangiogenic drugs, but rather from the restoration of a more functional tumor microenvironment that not only facilitates the delivery of chemotherapy and immune effector cells to the tumor but also impairs metastasis. Since targeting VEGF signaling often normalizes tumor vessels transiently and may ultimately provoke vessel regression, the development of alternative antiangiogenic strategies with a fundamentally distinct mechanism is mandated.

The recent data that the glycolytic regulator PFKFB3 controls vessel sprouting and its blockade promotes tumor vessel normalization [Citation83] raise the question if strategies targeting EC metabolism could increase the response of cancer patients to current anticancer treatment and represent a complementary or alternative antiangiogenic approach. A clinical trial using a small chemical PFKFB3 blocker has been initiated to target, in the first instance, cancer cells by using maximally tolerated doses [Citation88], based on preclinical studies designed to inhibit cancer cell proliferation [Citation89]. Since ECs are very sensitive to even small changes in glycolysis levels and, in fact, more responsive to PFKFB3 blockade than various cancer cell lines in vitro [Citation83], a clinical trial with another design using lower doses of the PFKFB3 blocker will be required to test whether PFKFB3 blockade can impair metastasis, while improving standard care therapy by promoting tumor vessel normalization. It will remain to be determined whether a high, or rather a low, dose of the PFKFB3 blocker is capable of inducing tumor vessel normalization, and its associated therapeutic benefits of reduced metastasis and improved response to chemotherapy.

Article highlights

  • Anti-VEGF therapy promotes vascular pruning

  • Excessive vascular pruning increases the risk of metastasis and impairs drug delivery and efficacy

  • Tumor vessel normalization represents an alternative strategy for antiangiogenic cancer treatment

  • Targeting endothelial cell glycolysis normalizes tumor vessels

  • PFKFB3 blockade reduces metastasis and promotes chemotherapy efficacy

This box summarizes key points contained in the article.

Declaration of interest

P. Carmeliet declares to be named as inventor on patent applications, claiming subject matter related to the results described in this paper. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Additional information

Funding

A. R. Cantelmo and J. Kalucka are supported by the Research Foundation Flanders (FWO). A. Pircher is supported by the Austrian Science Fund FWF (J3730-B26). The work of P. Carmeliet is supported by FWO-Vlaanderen, the Foundation against Cancer, the Belgian Science Policy (IUAP P7/03), long-term structural funding Methusalem funding by the Flemish Government, AXA Research Fund, an ERC Advanced Research Grant (EU-ERC269073).

References

  • Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011 May 19;473(7347):298–307.
  • Mazzone M, Dettori D, Leite de Oliveira R, et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell. 2009 Mar 6;136(5):839–851.
  • de Bock K, Georgiadou M, Carmeliet P. Role of endothelial cell metabolism in vessel sprouting. Cell Metab. 2013 Nov 5;18(5):634–647.
  • Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011 Sep 16;146(6):873–887.
  • Kalluri R. Basement membranes: structure, assembly and role in tumour angiogenesis. Nat Rev Cancer. 2003 Jun;3(6):422–433.
  • Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000 Sep 14;407(6801):249–257.
  • de Bock K, Cauwenberghs S, Carmeliet P. Vessel abnormalization: another hallmark of cancer? Molecular mechanisms and therapeutic implications. Curr Opin Genet Dev. 2011 Feb;21(1):73–79.
  • de Bock K, Mazzone M, Carmeliet P. Antiangiogenic therapy, hypoxia, and metastasis: risky liaisons, or not? Nat Rev Clin Oncol. 2011 Jul;8(7):393–404.
  • Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov. 2011 Jun;10(6):417–427.
  • Goel S, Duda DG, Xu L, et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev. 2011 Jul;91(3):1071–1121.
  • Ebos JM, Lee CR, Cruz-Munoz W, et al. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell. 2009 Mar 3; 15(3):232–239.
  • Paez-Ribes M, Allen E, Hudock J, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009 Mar 3;15(3):220–231.
  • Welti J, Loges S, Dimmeler S, et al. Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J Clin Invest. 2013 Aug;123(8):3190–3200.
  • Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014 Nov 10;26(5):605–622.
  • Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971 Nov 18;285(21):1182–1186.
  • Kim KJ, Li B, Winer J, et al. Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature. 1993 Apr 29;362(6423):841–844.
  • Yuan F, Chen Y, Dellian M, et al. Time-dependent vascular regression and permeability changes in established human tumor xenografts induced by an anti-vascular endothelial growth factor/vascular permeability factor antibody. Proc Natl Acad Sci U S A. 1996 Dec 10;93(25):14765–14770.
  • Warren RS, Yuan H, Matli MR, et al. Regulation by vascular endothelial growth factor of human colon cancer tumorigenesis in a mouse model of experimental liver metastasis. J Clin Investig. 1995;95(4):1789–1797.
  • Jayson GC, Kerbel R, Ellis LM, et al. Antiangiogenic therapy in oncology: current status and future directions. Lancet. 2016 Jul 30;388(10043):518–529.
  • Cobleigh MA, Langmuir VK, Sledge GW, et al. A phase I/II dose-escalation trial of bevacizumab in previously treated metastatic breast cancer. Semin Oncol. 2003 Oct;30(5 Suppl 16):117–124.
  • Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol. 2007 Apr 20;25(12):1539–1544.
  • Jain RK, Duda DG, Clark JW, et al. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol. 2006 Jan;3(1):24–40.
  • Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004 Jun 3;350(23):2335–2342.
  • Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007 Dec 27;357(26):2666–2676.
  • Reck M, Von Pawel J, Zatloukal P, et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAiL. J Clin Oncol. 2009 Mar 10;27(8):1227–1234.
  • Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006 Dec 14;355(24):2542–2550.
  • Tebbutt NC, Wilson K, Gebski VJ, et al. Capecitabine, bevacizumab, and mitomycin in first-line treatment of metastatic colorectal cancer: results of the Australasian Gastrointestinal Trials Group randomized phase III MAX study. J Clin Oncol. 2010 Jul 1;28(19):3191–3198.
  • Saltz LB, Clarke S, Diaz-Rubio E, et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol. 2008 Apr 20;26(12):2013–2019.
  • Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med. 2001 Sep;7(9):987–989.
  • Huang Y, Yuan J, Righi E, et al. Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc Natl Acad Sci U S A. 2012 Oct 23;109(43):17561–17566.
  • Winkler F, Kozin SV, Tong RT, et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell. 2004 Dec;6(6):553–563.
  • Falcon BL, Hashizume H, Koumoutsakos P, et al. Contrasting actions of selective inhibitors of angiopoietin-1 and angiopoietin-2 on the normalization of tumor blood vessels. Am J Pathol. 2009 Nov;175(5):2159–2170.
  • Nasarre P, Thomas M, Kruse K, et al. Host-derived angiopoietin-2 affects early stages of tumor development and vessel maturation but is dispensable for later stages of tumor growth. Cancer Res. 2009 Feb 15;69(4):1324–1333.
  • Koh YJ, Kim HZ, Hwang SI, et al. Double antiangiogenic protein, DAAP, targeting VEGF-A and angiopoietins in tumor angiogenesis, metastasis, and vascular leakage. Cancer Cell. 2010 Aug 9;18(2):171–184.
  • Kaelin WG Jr., Ratcliffe PJ. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell. 2008 May 23;30(4):393–402.
  • Simons M, Gordon E, Claesson-Welsh L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat Rev Mol Cell Biol. 2016 Oct;17(10):611-625.
  • de Bock K, De Smet F, Leite de Oliveira R, et al. Endothelial oxygen sensors regulate tumor vessel abnormalization by instructing phalanx endothelial cells. J Mol Med. 2009;87(6):561–569.
  • Kuchnio A, Moens S, Bruning U, et al. The cancer cell oxygen sensor PHD2 promotes metastasis via activation of cancer-associated fibroblasts. Cell Rep. 2015 Aug 11;12(6):992–1005.
  • Magrini E, Villa A, Angiolini F, et al. Endothelial deficiency of L1 reduces tumor angiogenesis and promotes vessel normalization. J Clin Invest. 2014 Oct;124(10):4335–4350.
  • Hamzah J, Jugold M, Kiessling F, et al. Vascular normalization in Rgs5-deficient tumours promotes immune destruction. Nature. 2008 May 15;453(7193):410–414.
  • Rolny C, Mazzone M, Tugues S, et al. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PIGF. Cancer Cell. 2011 Jan 18;19(1):31–44.
  • Maes H, Rubio N, Garg AD, et al. Autophagy: shaping the tumor microenvironment and therapeutic response. Trends Mol Med. 2013 Jul;19(7):428–446.
  • Maes H, Kuchnio A, Carmeliet P, et al. Chloroquine anticancer activity is mediated by autophagy-independent effects on the tumor vasculature. Mol Cell Oncol. 2016 Jan;3:e970097.
  • Maes H, Kuchnio A, Peric A, et al. Tumor vessel normalization by chloroquine independent of autophagy. Cancer Cell. 2014 Aug 11;26(2):190–206.
  • Huang Y, Goel S, Duda DG, et al. Vascular normalization as an emerging strategy to enhance cancer immunotherapy. Cancer Res. 2013 May 15;73(10):2943–2948.
  • Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012 Apr;12(4):252–264.
  • Wu YL, Liang J, Zhang W, et al. Immunotherapies: the blockade of inhibitory signals. Int J Biol Sci. 2012;8(10):1420–1430.
  • Bellone M, Calcinotto A. Ways to enhance lymphocyte trafficking into tumors and fitness of tumor infiltrating lymphocytes. Front Oncol. 2013;3:231.
  • Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008 Oct 6;27(45):5904–5912.
  • Hatfield SM, Kjaergaard J, Lukashev D, et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med. 2015 Mar 4;7(277):277ra30.
  • Cham CM, Gajewski TF. Glucose availability regulates IFN-gamma production and p70S6 kinase activation in CD8+ effector T cells. J Immunol. 2005 Apr 15;174(8):4670–4677.
  • Baronzio G, Schwartz L, Kiselevsky M, et al. Tumor interstitial fluid as modulator of cancer inflammation, thrombosis, immunity and angiogenesis. Anticancer Res. 2012 Feb;32(2):405–414.
  • Peske JD, Thompson ED, Gemta L, et al. Effector lymphocyte-induced lymph node-like vasculature enables naive T-cell entry into tumours and enhanced anti-tumour immunity. Nat Commun. 2015 May;6(6):7114.
  • Voron T, Colussi O, Marcheteau E, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med. 2015 Feb 9;212(2):139–148.
  • Terme M, Pernot S, Marcheteau E, et al. VEGFA-VEGFR pathway blockade inhibits tumor-induced regulatory T-cell proliferation in colorectal cancer. Cancer Res. 2013 Jan 15;73(2):539–549.
  • Bottsford-Miller JN, Coleman RL, Sood AK. Resistance and escape from antiangiogenesis therapy: clinical implications and future strategies. J Clin Oncol. 2012 Nov 10;30(32):4026–4034.
  • Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008 Aug;8(8):592–603.
  • Ribatti D. Tumor refractoriness to anti-VEGF therapy. Oncotarget. 2016 Jul 19;7(29):46668-46677.
  • Jain RK. Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J Clin Oncol. 2013 Jun 10;31(17):2205–2218.
  • Willett CG, Duda DG, di Tomaso E, et al. Efficacy, safety, and biomarkers of neoadjuvant bevacizumab, radiation therapy, and fluorouracil in rectal cancer: a multidisciplinary phase II study. J Clin Oncol. 2009 Jun 20;27(18):3020–3026.
  • Sorensen AG, Emblem KE, Polaskova P, et al. Increased survival of glioblastoma patients who respond to antiangiogenic therapy with elevated blood perfusion. Cancer Res. 2012 Jan 15;72(2):402–407.
  • Heist RS, Duda DG, Sahani DV, et al. Improved tumor vascularization after anti-VEGF therapy with carboplatin and nab-paclitaxel associates with survival in lung cancer. Proc Natl Acad Sci U S A. 2015 Feb 3;112(5):1547–1552.
  • van der Veldt AA, Lubberink M, Bahce I, et al. Rapid decrease in delivery of chemotherapy to tumors after anti-VEGF therapy: implications for scheduling of anti-angiogenic drugs. Cancer Cell. 2012 Jan 17;21(1):82–91.
  • Tolaney SM, Boucher Y, Duda DG, et al. Role of vascular density and normalization in response to neoadjuvant bevacizumab and chemotherapy in breast cancer patients. Proc Natl Acad Sci U S A. 2015 Nov 17;112(46):14325–14330.
  • Wong AL, Sundar R, Wang TT, et al. Phase Ib/II randomized, open-label study of doxorubicin and cyclophosphamide with or without low-dose, short-course sunitinib in the pre-operative treatment of breast cancer. Oncotarget. 2016 Sep 27;7(39):64089-64099.
  • Zissen MH, Kunz P, Subbarayan M, et al. 18F-5-fluorouracil dynamic positron emission tomography/computed tomography shows decreased tracer activity after bevacizumab in colorectal metastases. Nucl Med Commun. 2011 May;32(5):343–347.
  • Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med. 2004 Feb;10(2):145–147.
  • Wallin JJ, Bendell JC, Funke R, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624.
  • Ott PA, Hodi FS, Buchbinder EI. Inhibition of immune checkpoints and vascular endothelial growth factor as combination therapy for metastatic melanoma: an overview of rationale, preclinical evidence, and initial clinical data. Front Oncol. 2015;5:202.
  • Arjaans M, Schroder CP, Oosting SF, et al. VEGF pathway targeting agents, vessel normalization and tumor drug uptake: from bench to bedside. Oncotarget. 2016 Apr 19;7(16):21247–21258.
  • Oosting SF, Brouwers AH, van Es SC, et al. 89Zr-bevacizumab PET visualizes heterogeneous tracer accumulation in tumor lesions of renal cell carcinoma patients and differential effects of antiangiogenic treatment. J Nucl Med. 2015 Jan;56(1):63–69.
  • Cesca M, Morosi L, Berndt A, et al. Bevacizumab-induced inhibition of angiogenesis promotes a more homogeneous intratumoral distribution of paclitaxel, improving the antitumor response. Mol Cancer Ther. 2016 Jan;15(1):125–135.
  • Dickson PV, Hamner JB, Sims TL, et al. Bevacizumab-induced transient remodeling of the vasculature in neuroblastoma xenografts results in improved delivery and efficacy of systemically administered chemotherapy. Clin Cancer Res. 2007 Jul 1;13(13):3942–3950.
  • Liu Y, Suzuki M, Masunaga S, et al. Effect of bevacizumab treatment on p-boronophenylalanine distribution in murine tumor. J Radiat Res. 2013 Mar 1;54(2):260–267.
  • Eelen G, De Zeeuw P, Simons M, et al. Endothelial cell metabolism in normal and diseased vasculature. Circ Res. 2015 Mar 27;116(7):1231–1244.
  • Treps L, Conradi LC, Harjes U, et al. Manipulating angiogenesis by targeting endothelial metabolism: hitting the engine rather than the drivers-a new perspective? Pharmacol Rev. 2016 Jul;68(3):872–887.
  • de Bock K, Georgiadou M, Schoors S, et al. Role of PFKFB3-driven glycolysis in vessel sprouting. Cell. 2013 Aug 1;154(3):651–663.
  • Schoors S, De Bock K, Cantelmo AR, et al. Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis. Cell Metab. 2014 Jan 7;19(1):37–48.
  • Schoors S, Cantelmo AR, Georgiadou M, et al. Incomplete and transitory decrease of glycolysis: a new paradigm for anti-angiogenic therapy? Cell Cycle. 2014;13(1):16–22.
  • Granchi C, Minutolo F. Anticancer agents that counteract tumor glycolysis. ChemMedChem. 2012 Aug;7(8):1318–1350.
  • Raez LE, Papadopoulos K, Ricart AD, et al. A phase I dose-escalation trial of 2-deoxy-D-glucose alone or combined with docetaxel in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2013 Feb;71(2):523–530.
  • Xu Y, An X, Guo X, et al. Endothelial PFKFB3 plays a critical role in angiogenesis. Arterioscler Thromb Vasc Biol. 2014 Jun;34(6):1231–1239.
  • Cantelmo AR, Conradi LC, Brajic A, et al. Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy. Cancer Cell. 2016 Dec 12;30(6):968–985.
  • Crawford Y, Ferrara N. VEGF inhibition: insights from preclinical and clinical studies. Cell Tissue Res. 2009 Jan;335(1):261–269.
  • Ebos JM, Kerbel RS. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat Rev Clin Oncol. 2011 Apr;8(4):210–221.
  • Saharinen P, Eklund L, Pulkki K, et al. VEGF and angiopoietin signaling in tumor angiogenesis and metastasis. Trends Mol Med. 2011 Jul;17(7):347–362.
  • Yang Y, Zhang Y, Iwamoto H, et al. Discontinuation of anti-VEGF cancer therapy promotes metastasis through a liver revascularization mechanism. Nat Commun. 2016;7:12680.
  • Redman RPP, Kurman M, Tabolsky G, et al. A phase I, dose-escalation, multi-center study of PFK-158 in patients with advanced solid malignancies explores a first-in-man inhbibitor of glycolysis. J Clin Oncol. 2015;vol 33 no 15(suppl TPS2606). Abstract ASCO Gastrointestinal Cancers Symposium, May 2015.
  • Clem BF, O’Neal J, Klarer AC, et al. Clinical development of cancer therapeutics that target metabolism. QJM. 2016 Jun;109(6):367–372.