Elsevier

Pharmacological Research

Volume 150, December 2019, 104511
Pharmacological Research

Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy

https://doi.org/10.1016/j.phrs.2019.104511Get rights and content

Highlights

  • Cancer is the second major cause of mortality worldwide despite current chemotherapy.

  • Targeting aerobic glycolysis has become a research focus for developing anticancer drugs.

  • These antiglycolytic agents might also sensitize tumor cells to other cytotoxic therapies.

  • Although preclinical studies are promising, antiglycolytic therapy has not yet been translated into clinical practice.

  • This review presents the current state of glycolytic cancer therapy discussing the challenges and opportunitie.

Abstract

Most solid tumor cells adapt to their heterogeneous microenvironment by depending largely on aerobic glycolysis for energy production, a phenomenon called the Warburg effect, which is a hallmark of cancer. The altered energy metabolism not only provides cancer cell with ATP for cellular energy, but also generate essential metabolic intermediates that play a pivotal role in the biosynthesis of macromolecules, to support cell proliferation, invasiveness, and chemoresistance. The cellular metabolic reprogramming in cancer is regulated by several oncogenic proteins and tumor suppressors such as hypoxia-inducible factor (HIF-1), Myc, p53, and PI3K/Akt/mTOR pathway. A better understanding of the mechanisms involved in the regulation of aerobic glycolysis can help in developing glycolytic inhibitors as anticancer agents. These metabolic antiglycolytic agents could be more effective if used in drug combinations to combat cancer. Several preclinical and early clinical studies have shown the effectiveness of targeting the glycolytic pathway as a therapeutic approach to suppress cancer progression. This review aimed to present the most recent data on the emerging drug candidate targeting enzymes and intermediates involved in glucose metabolism to provide therapeutic opportunities and challenges for antiglycolytic cancer therapy.

Introduction

Cancer incidence is rapidly growing with an expected increase of 70 % over the next two decades. Cancer is ranked as a leading cause of death, accounting for 13 % of all mortality worldwide (GLOBOCAN 2018 database) [1]. Metabolism of cancer cells is different from that of normal cells, which allows them to sustain a high rate of proliferation and resist signals of apoptosis [2]. Normally, cells utilize multiple metabolic pathways to produce energy depending on the availability of metabolites and biosynthetic requirements for cellular function. Cells typically use glycolysis to convert glucose into pyruvate in the cytosol. The pyruvate is further metabolized in the mitochondria by oxidative phosphorylation (OxPhos) through the tricarboxylic acid (TCA) cycle and electron transport chain (ETC), to produce the energy-storing adenosine triphosphate (ATP). Under hypoxic conditions, cells can utilize anaerobic glycolysis, and converts pyruvate into lactate, producing much less amount of ATP, but at a faster rate [3]. Mitochondrial oxidation of one glucose molecule yields 36 molecules of ATP, while its metabolism to lactate by glycolysis produces only 2 ATP molecules. Under aerobic conditions, cells can also utilize fatty-acid oxidation (called beta-oxidation) or glutamine oxidation, if these metabolites are available [3].

Tumor cells, unlike normal cells, depend largely on glycolysis for producing energy even in the presence of adequate levels of oxygen, a process termed aerobic glycolysis [4]. Thus, inhibition of glycolytic pathways has the potential to provide an effective approach to cancer research aiming to develop new targeted anticancer agents. This approach has been proven effective in suppressing tumor progression, and several of these glycolytic inhibitors are currently under investigation in preclinical and clinical studies with promising results [[5], [6], [7]]. This review will present the most recent data on the emerging candidate agents targeting glycolytic enzymes and intermediates to be useful in cancer therapy.

Section snippets

Tumor glucose metabolism and Warburg effect

Cancer progression involves an inappropriate proliferation of cells, which have enhanced abilities for energy production to resist metabolic stresses [8]. Tumor cell metabolism is reprogrammed in favor of aerobic glycolysis despite the presence of plentiful oxygen [9]. This observation was first reported many decades ago by the German scientist Otto Warburg and is thus referred to as the “Warburg effect” [10]. This metabolic alteration to a high glycolysis rate has been observed in a variety of

Dual metabolic phenotype and hybrid state

The glycolytic phenotype is known to be expressed by many cancers [21], but the dependence of cancer cells on such phenotype remains unclear. It has been demonstrated cancer cell death can be induced by reversing the glycolytic state to OxPhos [22]. The glycolytic cancer cells can exhibit a non-glycolytic phenotype under acidic conditions by intracellular lactic acid accumulation. Lactic acidosis is a common consequence of the Warburg effect in most solid tumors [23]. Tumor cells cultured with

Reverse Warburg effect or metabolic coupling

Another type of tumor metabolism has been recently observed in certain types of cancers called the “reverse Warburg effect” or “metabolic coupling” which have high mitochondrial respiration and low glycolysis rate [40]. In this type, the tumor cells and adjacent stromal fibroblasts form a two-compartment model of cancer metabolism, where aerobic glycolysis occurs in fibroblasts, and the generated metabolites are transferred to malignant cells, to fuel the TCA cycle and maintain ATP generation [

Warburg effect and tumor acidosis

The microenvironment of solid tumors is markedly heterogenous, so tumor cells tend to increase their uptake of glucose to maintain energy production [13]. Increased glycolysis and decreased mitochondrial oxidation lead to increased formation of lactic acid, increased glutaminolysis, increased beta-oxidation of fatty acids and activation of the pentose phosphate pathway [45]. To maintain the intracellular pH (pHi), tumor cells promptly export the excess intracellular acid load to the

Tumor glycolysis and its clinical relevance to cancer progression

The glycolysis process takes place in the cytoplasm by converting glucose into pyruvate through nine reaction steps, involving several glycolytic enzymes. First, glucose is transported into tumor cells at a high rate by glucose transporters (GLUTs), GLUT1 and sodium-glucose linked transporter 1 (SGLT1) which are overexpressed in most cancers [50]. Glucose is phosphorylated into glucose-6-phosphate by hexokinase (HK), a rate-limiting step that provides direct feedback inhibition to preserve

Non-glycolytic functions of glycolytic enzymes and metabolic intermediates

Many glycolytic enzymes have also important roles in several non-glycolytic processes involved in cellular functions that support cancer cell survival and growth [84]. For instance, the mitochondrial membrane-bound HK2 can antagonize the proapoptotic pathway in cancer cells [85]. Also, HK2 acting as a nuclear enzyme is involved in transcriptional regulation of some nuclear proteins [84]. Similarly, GAPDH has a critical role in maintaining the cellular redox balance by catalyzing the production

Molecular regulation of tumor glycolysis

It is increasingly evident that coordinated networks of signaling pathways regulate reprogramming of cancer cells to balance their metabolic state, supporting tumor growth and stress resistance. Several studies have demonstrated the affection of cancer cell metabolism by many regulators including protooncogenes (e.g. Myc), transcription factors (e.g. HIF-1), signaling pathways (e.g. PI3K/Akt/mTOR), and tumor suppressors (e.g. p53) [97]. The c-Myc is a transcription factor, encoded by Myc

Targeting tumor metabolism and Glycolytic inhibitors

Recent cancer research focuses on selective inhibition of metabolic pathways to deprive cancer cells of essential metabolic needs and interfere with tumor growth. The improved understanding of aerobic glycolysis as a hallmark of cancer and underlying mechanisms may pave the way for the development of targeted metabolic agents for antiglycolytic cancer therapy [114]. There are several approaches to disrupt energy production and prevent glucose utilization by cancer cells. Indeed,

Conclusion and future perspective for metabolic cancer therapy

Although aerobic glycolysis is a known signature of cancer cells, targeting this pathway for therapy has not yet been successfully translated into clinical practice. Recently, this hallmark of cancer metabolism has become a focus of cancer research and drug discovery, aiming for the introduction of effective metabolic agents as a promising strategy to combat cancer. These candidate drugs might also sensitize tumor cells to other more effective cytotoxic therapies. Many emerging drugs have been

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Declaration of Competing Interest

The authors declare that there is no conflict of interests.

Acknowledgments

We thank Dr. Abdul-Salam Noor Waly, Dean Faculty of Medicine, Umm Al-Qura University, Makkah, for providing access to the Saudi Digital Library.

References (282)

  • L. Zhang et al.

    Hypoxia induces epithelial-mesenchymal transition via activation of SNAI1 by hypoxia-inducible factor -1α in hepatocellular carcinoma

    BMC Cancer

    (2013)
  • K. Sun et al.

    Oxidized ATM-mediated glycolysis enhancement in breast cancer-associated fibroblasts contributes to tumor invasion through lactate as metabolic coupling

    E. Bio. Medicine

    (2019)
  • S. Taylor et al.

    Microenvironment acidity as a major determinant of tumor chemoresistance: proton pump inhibitors (PPIs) as a novel therapeutic approach

    Drug Resist. Updat. Rev. Comment. Antimicrob. Anticancer Chemother.

    (2015)
  • S.P. Mathupala et al.

    Hexokinase-2 bound to mitochondria: cancer’s stygian link to the “Warburg Effect” and a pivotal target for effective therapy

    Semin. Cancer Biol.

    (2009)
  • W. Luo et al.

    Emerging roles of PKM2 in cell metabolism and cancer progression

    Trends Endocrinol. Metab.

    (2012)
  • R.J. Deberardinis et al.

    Brick by brick: metabolism and tumor cell growth

    Curr. Opin. Genet. Dev.

    (2008)
  • D.S. Backos et al.

    The role of glutathione in brain tumor drug resistance

    Biochem. Pharmacol.

    (2012)
  • C.W. Lu et al.

    Induction of pyruvate dehydrogenase kinase-3 by hypoxia-inducible factor-1 promotes metabolic switch and drug resistance, J. Biol

    Chem.

    (2008)
  • O. Feron

    Pyruvate into lactate and back: from the Warburg effect to symbiotic energy fuel exchange in cancer cells

    Radiother. Oncol.

    (2009)
  • J.W. Kim et al.

    Multifaceted roles of glycolytic enzymes

    Trends Biochem. Sci.

    (2005)
  • N. Majewski et al.

    Hexokinase-mitochondria interaction mediated by Akt is required to inhibit apoptosis in the presence or absence of Bax and Bak

    Mol. Cell

    (2004)
  • W. Yang et al.

    W.K. Alfred Yung, Z. Lu, PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis

    Cell

    (2012)
  • N.A. Demarse et al.

    Direct binding of glyceraldehyde 3-phosphate dehydrogenase to telomeric DNA protects telomeres against chemotherapy-induced rapid degradation

    J. Mol. Biol.

    (2009)
  • N. Harada et al.

    Glyceraldehyde-3-phosphate dehydrogenase enhances transcriptional activity of androgen receptor in prostate cancer cells

    J. Biol. Chem.

    (2007)
  • O. Popanda et al.

    Modulation of DNA polymerases alpha, delta and epsilon by lactate dehydrogenase and 3-phosphoglycerate kinase

    Biochim. Biophys. Acta

    (1998)
  • R. Diaz-Ruiz et al.

    Mitochondrial oxidative phosphorylation is regulated by fructose 1,6-bisphosphate. A possible role in Crabtree effect induction?

    J. Biol. Chem.

    (2008)
  • F. Bray et al.

    Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

    CA-Cancer J. Clin.

    (2018)
  • H. Liu et al.

    Hypersensitization of tumor cells to glycolytic inhibitors

    Biochemistry

    (2001)
  • H. Pelicano et al.

    Glycolysis inhibition for anticancer treatment

    Oncogene

    (2006)
  • S. Ganapathy-Kanniappan et al.

    Tumor glycolysis as a target for cancer therapy: progress and prospects

    Mol. Cancer

    (2013)
  • L.J. Savic et al.

    Targeting glucose metabolism in cancer: new class of agents for loco-regional and systemic therapy of liver cancer and beyond? Hepat

    Oncol.

    (2016)
  • X.S. Chen et al.

    Anticancer strategies based on the metabolic profile of tumor cells: therapeutic targeting of the Warburg effect

    Acta Pharmacol. Sin.

    (2016)
  • N.S. Akins et al.

    Inhibition of glycolysis and glutaminolysis: an emerging drug discovery approach to combat Cancer

    Curr. Top. Med. Chem.

    (2018)
  • M.G. Vander Heiden et al.

    Understanding the Warburg effect: the metabolic requirements of cell proliferation

    Science

    (2009)
  • O. Warburg

    On the origin of cancer cells

    Science

    (1956)
  • O. Warburg et al.

    Über Den Stoffwechsel Der Carcinomzelle

    Biochem. Zeitschr.

    (1924)
  • H.R. Christofk et al.

    The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth

    Nature

    (2008)
  • M. Weiler-Sagie et al.

    18F-FDG avidity in lymphoma readdressed: a study of 766 patients

    J. Nucl. Med.

    (2010)
  • S.Y. Lunt

    M.G. Vander Heiden, Aerobic glycolysis: meeting the metabolic requirements of cell proliferation

    Annu. Rev. Cell Dev. Biol.

    (2011)
  • R.A. Gatenby et al.

    Why do cancers have high aerobic glycolysis?

    Nat. Rev. Cancer

    (2004)
  • H. Wu et al.

    Central role of lactic acidosis in cancer cell resistance to glucose deprivation-induced cell death

    J. Pathol.

    (2012)
  • W. Lu et al.

    Novel role of NOX in supporting aerobic glycolysis in cancer cells with mitochondrial dysfunction and as a potential target for cancer therapy

    PLoS Biol.

    (2012)
  • C.L. Lu et al.

    Tumor cells switch to mitochondrial oxidative phosphorylation under radiation via mTOR-Mediated hexokinase II Inhibition-A warburg-reversing effect

    PLoS One

    (2015)
  • J. Xie et al.

    Beyond Warburg effect–dual metabolic nature of cancer cells

    Sci. Rep.

    (2014)
  • C.V. Dang

    Rethinking the Warburg effect with Myc micromanaging glutamine metabolism

    Cancer Res.

    (2010)
  • M.C. Caino et al.

    PI3K therapy reprograms mitochondrial trafficking to fuel tumor cell invasion

    Proc. Natl. Acad. Sci.

    (2015)
  • C.J. De Saedeleer et al.

    Lactate activates HIF-1 in oxidative but not in Warburg-phenotype human tumor cells

    PLoS One

    (2012)
  • D. Jia et al.

    Elucidating cancer metabolic plasticity by coupling gene regulation with metabolic pathways

    Proc. Natl. Acad. Sci. U.S.A.

    (2019)
  • V.S. LeBleu et al.

    PGC-1α mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis

    Nat. Cell Biol.

    (2014)
  • P.L. Pedersen

    Mitochondria in relation to cancer metastasis: introduction to a minireview series

    J. Bioenergy Biomembr.

    (2012)
  • Cited by (303)

    View all citing articles on Scopus
    View full text