Elsevier

Biochemical Pharmacology

Volume 89, Issue 2, 15 May 2014, Pages 162-170
Biochemical Pharmacology

Commentary
Metabolic effects of antiangiogenic drugs in tumors: Therapeutic implications

https://doi.org/10.1016/j.bcp.2014.02.018Get rights and content

Abstract

Antiangiogenic therapy has become a mainstay of cancer therapeutics, but clinical responses are generally short-term owing to the development of secondary resistance. Tumor starvation by antiangiogenic drugs is largely attributed to increased hypoxia and impaired nutrients supply, suggesting that angiogenesis inhibition causes remarkable metabolic perturbations in the tumor microenvironment. We review here recent acquisitions concerning metabolic effects of angiogenesis blockade in tumors and discuss the possibility that some metabolic features of tumor cells – i.e. their dependency from glucose as primary energy substrate – might affect tumor responses to anti-vascular endothelial growth factor treatment. Moreover, we discuss the hypothesis that anti-angiogenic therapy might foster metabolic evolution of tumors. The therapeutic implications of this hypothesis will be discussed further here.

Introduction

Angiogenesis is a tightly regulated biological process through which new blood vessels are generated from pre-existing ones. Following seminal observations by Folkman and other investigators in the 1970s [1], angiogenesis became a vivid area in cancer research and stimulated the search for conceptually new treatments. The identification of vascular endothelial growth factor (VEGF) as a key driver of the angiogenic process [2] has converted the VEGF/VEGF receptor (VEGFR) signaling pathway into an attractive therapeutic target. VEGF blockers belong to two broad categories including (I) VEGF ligand-blocking drugs and (II) drugs blocking VEGFR signaling. Not surprisingly, given the redundancy of factors involved in the regulation of the angiogenic process, also other angiogenic factors and signaling pathways – such as fibroblast growth factors (FGFs) and angiopoietins – have been considered as potential therapeutic targets [3], [4]. After a decade of clinical research, we are currently at a critical point in the development of antiangiogenic drugs for cancer, as their therapeutic activity in patients has been substantially lower than expected based on preclinical findings. The development of assays to interrogate the dependence of individual tumors from any of these key angiogenic pathways will certainly be fundamental to personalize and possibly improve efficacy of antiangiogenic therapy.

Since the field of angiogenesis inhibitors is very large and the topic has been extensively reviewed elsewhere [5], [6], [7], we will briefly overview only a fraction of the antiangiogenic agents available, focusing on those in late stage clinical trials or approved by the regulatory agencies, being aware that more antiangiogenic drugs will enter the clinical arena in the coming years. A summary of the main characteristics and pharmacologic targets of these drugs is presented in Table 1, whereas their structure is shown in Fig. 1. Next, we will review literature linking antiangiogenic therapy to tumor metabolism, explain why we consider this an interesting area of investigation and outline possible therapeutic implications of these studies.

Section snippets

Bevacizumab

A recombinant humanized monoclonal anti-VEGF antibody which binds to all active isoforms of VEGF-A and neutralizes their activity by preventing binding of VEGF to its receptors. Based on the positive outcome of several phase III clinical trials, in the United States (U.S.) bevacizumab was approved for four types of cancer including metastatic renal cell carcinoma (mRCC), metastatic colorectal cancer (mCRC), non-small cell lung cancer (NSCLC) and recurrent glioblastoma (rGBM) [8]. In Europe,

Vascular responses to antiangiogenic therapies

The rationale of antiangiogenic therapy is grounded on the concept of starving the tumor of oxygen and nutrients, thereby limiting the capacity of tumors to grow. Since most of these drugs, and bevacizumab in particular, generally lack direct cytotoxic or cytostatic effects on tumor cells, their therapeutic activity has been attributed to effects on the tumor microenvironment. The best characterized effect of many of these drugs is induction of hypoxia, which has been observed in many

Metabolic effects of antiangiogenic therapy: Looking beyond hypoxia

As anticipated above, the best recognized metabolic effect of antiangiogenic therapy is hypoxia, which leads to stabilization of HIF-1, a transcriptional factor which coordinates adaptation of cells to hypoxic conditions by modulation of gene sets involved in the regulation of cell proliferation, apoptosis, angiogenesis and metabolism [40]. Once activated, HIF-1 induces the transcription of genes encoding glucose transporters and most glycolytic enzymes. In addition, HIF-1 activates the

Metabolic evolution of tumors treated with anti-VEGF therapy

One of the intriguing questions related to the metabolic changes caused by antiangiogenic therapy is whether they are transient or stable (Fig. 3). At least some of these metabolic changes are indeed HIF-1α-driven and therefore could reflect adaptation to the hypoxic microenvironment rather than stable metabolic changes in tumor cells. Increased expression of glycolysis-associated markers following bevacizumab has been observed in previous studies [55], but it was considered to depend on

Antiangiogenic therapy may sensitize tumors to glycolytic metabolism-targeting drugs

Conceivably, alteration of glucose metabolism in tumors treated with antiangiogenic drugs may render them more likely to respond to drugs targeting the glycolytic metabolism. In this respect, several “druggable” targets have been identified along the glycolytic pathway [66], but there is still a paucity of investigations on the potential advantage of combining antiangiogenic therapies with agents counteracting glycolysis. One problem could be efficient delivery of the drugs to tumors, given the

Conclusions

Understanding metabolic perturbations associated with administration of antiangiogenic drugs is a novel area of investigation, which will greatly profit of advanced technologies such as hyperpolarized MRI and other innovative imaging techniques, which enable serial evaluation of metabolic parameters. Therapeutic targeting of dis-regulated metabolic pathways, such as glycolysis, is potentially a very attractive strategy because it takes advantage of increased dependence of tumor cells on

Review criteria

Data for this Review were obtained by searching the PubMed database before 1 December 2013 using search terms including but not restricted to “antiangiogenic therapy”, “VEGF inhibitor”, “VEGF blockade”, “anti-VEGF therapy”, “bevacizumab”, “sunitinib”, “mass spectrometry”, “magnetic resonance spectroscopy”, “PET”, “metabolism”, “glycolysis”. Reference lists from identified articles were also searched for additional articles. Only full-text articles published in English were considered.

Conflict of interest statement

The Authors declare no conflict of interest.

References (70)

  • J. Folkman

    Tumor angiogenesis: therapeutic implications

    N Engl J Med

    (1971)
  • D.W. Leung et al.

    Vascular endothelial growth factor is a secreted angiogenic mitogen

    Science

    (1989)
  • H. Huang et al.

    Targeting the ANGPT-TIE2 pathway in malignancy

    Nat Rev Cancer

    (2010)
  • C. Lieu et al.

    Beyond VEGF: inhibition of the fibroblast growth factor pathway and antiangiogenesis

    Clin Cancer Res

    (2011)
  • P. Carmeliet et al.

    Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases

    Nat Rev Drug Discovery

    (2011)
  • A. Grothey et al.

    Targeting angiogenesis: progress with anti-VEGF treatment with large molecules

    Nat Rev Clin Oncol

    (2009)
  • G.C. Jayson et al.

    Antiangiogenic therapy—evolving view based on clinical trial results

    Nat Rev Clin Oncol

    (2012)
  • L.R. Feliz et al.

    Anti-vascular endothelial growth factor therapy in the era of personalized medicine

    Cancer Chemother Pharmacol

    (2013)
  • I.A. Boere et al.

    It takes two to tango: combinations of conventional cytotoxics with compounds targeting the vascular endothelial growth factor-vascular endothelial growth factor receptor pathway in patients with solid malignancies

    Cancer Sci

    (2010)
  • R.J. Motzer et al.

    Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma

    J Clin Oncol

    (2009)
  • G.M. Blumenthal et al.

    FDA approval summary: sunitinib for the treatment of progressive well-differentiated locally advanced or metastatic pancreatic neuroendocrine tumors

    Oncologist

    (2012)
  • J.M. Ebos et al.

    Antiangiogenic therapy: impact on invasion, disease progression, and metastasis

    Nat Rev Clin Oncol

    (2011)
  • B. Escudier et al.

    Sorafenib in advanced clear-cell renal-cell carcinoma

    N Engl J Med

    (2007)
  • C.N. Sternberg et al.

    Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial

    J Clin Oncol

    (2010)
  • H.J. Schmoll et al.

    Cediranib with mFOLFOX6 versus bevacizumab with mFOLFOX6 as first-line treatment for patients with advanced colorectal cancer: a double-blind, randomized phase III study [HORIZON III]

    J Clin Oncol

    (2012)
  • F.A. Raja et al.

    Initial toxicity assessment of ICON6: a randomised trial of cediranib plus chemotherapy in platinum-sensitive relapsed ovarian cancer

    Br J Cancer

    (2011)
  • S. Tejpar et al.

    Overcoming resistance to antiangiogenic therapies

    Oncologist

    (2012)
  • N.G. Chau et al.

    Vandetanib for the treatment of medullary thyroid cancer

    Clin Cancer Res

    (2013)
  • H. Huang et al.

    Specifically targeting angiopoietin-2 inhibits angiogenesis, Tie2-expressing monocyte infiltration, and tumor growth

    Clin Cancer Res

    (2011)
  • N. Turner et al.

    Fibroblast growth factor signalling: from development to cancer

    Nat Rev Cancer

    (2010)
  • J.M. Llovet et al.

    Brivanib in patients with advanced hepatocellular carcinoma who were intolerant to sorafenib or for whom sorafenib failed: results from the randomized phase III BRISK-PS study

    J Clin Oncol

    (2013)
  • G. Bergers et al.

    Modes of resistance to anti-angiogenic therapy

    Nat Rev Cancer

    (2008)
  • J. Pouyssegur et al.

    Hypoxia signalling in cancer and approaches to enforce tumour regression

    Nature

    (2006)
  • K. De Bock et al.

    Antiangiogenic therapy, hypoxia, and metastasis: risky liaisons, or not

    Nat Rev Clin Oncol

    (2011)
  • A.C. Yopp et al.

    Antiangiogenic therapy for primary liver cancer: correlation of changes in dynamic contrast-enhanced magnetic resonance imaging with tissue hypoxia markers and clinical response

    Ann Surg Oncol

    (2011)
  • Cited by (20)

    • New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects

      2022, Drug Resistance Updates
      Citation Excerpt :

      Angiogenesis and metabolism are entwined processes that support tumor growth and progression (Goncalves et al., 2021; Icard et al., 2018; Kopecka et al., 2020; Narayanan et al., 2020; Pi et al., 2022). AAD treatment-induced metabolic reprogramming, also called metabolic symbiosis or metabolic adaptation, has emerged as a novel mechanism of antiangiogenic therapy resistance in tumors, which is associated with hypoxia and glycolytic process that are induced by antiangiogenic therapy (Jiménez-Valerio and Casanovas, 2017; Quintieri et al., 2014). Studies have revealed that metabolic symbiosis is characterized by the differential expression of GLUT1, GLUT3, MCT1, and MCT4, and aberrant mTOR signaling in the TME (Allen et al., 2016; Jimenez-Valerio et al., 2016; Kuang et al., 2017; Pisarsky et al., 2016).

    • Tumor Microenvironment Regulation of Tumor Expansion

      2021, Tumor Microenvironment Regulation of Tumor Expansion
    • Three-dimensional in vitro models of angiogenesis

      2020, Biomaterials for 3D Tumor Modeling
    • Antiangiogenic compounds: well-established drugs versus emerging natural molecules

      2018, Cancer Letters
      Citation Excerpt :

      Lenalodomide belongs to the same type of inhibitors as thalidomine, being also used against multiple myeloma by inhibiting VEGF expression (Table 1) [114]. Despite the various molecules involved in the angiogenic process, most of these drugs have greater affinity for acting on tyrosine kinases, particularly the VEGF and respective receptor [104,105]. For example, bevacizumab is a monoclonal antibody that specifically recognize and binds to VEGF, making this growth factor unable to activate the receptor [108].

    • Linking metabolic reprogramming to therapy resistance in cancer

      2017, Biochimica et Biophysica Acta - Reviews on Cancer
      Citation Excerpt :

      Notably, however, some studies involving experimental tumors treated with antiangiogenic drugs, such as bevacizumab, provide evidence of the existence of MCT4+ tumor regions uncoupled from necrotic and bona fide hypoxic areas [36]. This observation, along with the metabolic characterization of ex vivo tumor cells obtained from tumors treated with anti-VEGF therapy [38], raises the hypothesis that in parallel with hypoxia-driven metabolic plasticity, antiangiogenic therapy could elicit in vivo selection of tumor cells with stable metabolic changes, e.g. enhanced glycolysis [43]. This hypothesis underscores the possible existence of intra-tumor metabolic heterogeneity not only imposed by local pathophysiological conditions, such as hypoxia or glucose deprivation, or adaptation to a specific environment [44], but rather intrinsic to tumor cells.

    View all citing articles on Scopus
    1

    M.S was a recipient of the Erasmus Mundus Fellowship (Al-Fihri Program).

    View full text