Elsevier

Seminars in Cancer Biology

Volume 43, April 2017, Pages 119-133
Seminars in Cancer Biology

Review
Therapeutic implications of tumor interstitial acidification

https://doi.org/10.1016/j.semcancer.2017.01.008Get rights and content

Abstract

Interstitial acidification is a hallmark of solid tumor tissues resulting from the combination of different factors, including cellular buffering systems, defective tissue perfusion and high rates of cellular metabolism. Besides contributing to tumor pathogenesis and promoting tumor progression, tumor acidosis constitutes an important intrinsic and extrinsic mechanism modulating therapy sensitivity and drug resistance. In fact, pharmacological properties of anticancer drugs can be affected not only by tissue structure and organization but also by the distribution of the interstitial tumor pH. The acidic tumor environment is believed to create a chemical barrier that limits the effects and activity of many anticancer drugs. In this review article we will discuss the general protumorigenic effects of acidosis, the role of tumor acidosis in the modulation of therapeutic efficacy and potential strategies to overcome pH-dependent therapy-resistance.

Introduction

Tumor tissues are characterized by an intricate organization that cannot be viewed simply as the outgrowth of cancer cells clones within a normal tissue. Instead, they consist of multiple cell types (both transformed and non-transformed) that cooperate through diverse mechanisms to maintain the microenvironment favorable for tumor cells survival and progression [1]. Unlike normal tissues with a structured capillary system that provides a uniform and structured blood flow to all cell layers, fast-dividing tumor cells often form chaotically organized structures. Cancer blood vessels are, in contrast to normal vessels, dilated, irregularly built (e.g., they may have dead ends) and leaky due to loose contacts of perivascular cells with endotheliocytes [2], [3]. This results, on the one hand, in extravasation of liquids and proteins that build interstitial pressure, and, on the other hand, in insufficient tissue supply with oxygen and nutrients. Thus, due to perfusion issues, tumor cell layers distantly located from the vessels receive inadequate supply of nutrients and have abnormal flow of the metabolites [4]. Taken together, tumors have a unique milieu that cannot be well-tolerated by healthy non-transformed cells. This environment is believed to exert selective pressure on the cells, thus driving propagation of the cells adapted to these harsh conditions.

Specific cancer microenvironment is a cause and a consequence of metabolic re-wiring of both tumor cells and cancer-associated non-transformed cells [5], [6]. While under physiological conditions all systems of an organism (e.g., breathing, excretion) are directed towards maintaining of a slightly alkaline, relatively constant pH, tumor cells shift their metabolism towards alternative pathways that contribute to the acidification of the interstitial space (the phenomenon referred to as tumor acidosis) [7], [8]. Low oxygen pressure within the tumor mass brings about metabolic switch of cancer cells that is characterized by elevated glycolytic activity and, hence, increased lactate production through monocarboxylate transporters (MCTs). Tumor-recruited normal cells were also shown to adapt to the microenvironment conditions through metabolic alterations, in their turn also contributing to the acidification of the extracellular environment. Common molecular hallmarks of tumor-associated metabolism connected to pH regulation are elevated activity and/or expression of proton pumps, carbonic anhydrases and ion channels responsible for the transport of metabolic acids to the extracellular space [9], [10].

It should be noted that local acidic pH often occurs also in a healthy organism. For example, the slightly acidic skin surface is believed to play a protective role against bacterial invasion. Low pH maintenance of the epidermis is achieved by the interplay between focal-adhesion kinases and the Na+/H+ exchanger type 1 (NHE1) [11], [12]. Besides, physiological inflammatory response is associated with local acidosis, presumably also due to its bacteriostatic effect. In this case, acidic conditions are achieved by protons secreted by macrophages [13], [14]. In a similar way, pH as low as 5.5 is induced in the skeleton via proton secreting osteoclasts during the process of bone resorption [15]. Under all the above-mentioned conditions, tissue acidosis is temporarily and spatially limited whereas tumors have constitutively low pH on their site. A tumor consists of cancer cells as well as recruited normal residing cells that are being exploited to maintain specific homeostasis within a tumor organ. Therefore, normally occurring mechanisms of pH decrease can be encountered within tumors (e.g., tumor-inflammation associated proton secretion by macrophages or osteoclast involvement). Importantly, although proton secretion does take place in tumors, tumor acidosis is achieved through the combination of proton extrusion and the secretion of acidic metabolites by tumor and tumor-associated cells [16].

Tissue samples from patients with head and neck cancer as well as musculoskeletal cancers were shown to be acidic [17], [18], [19]. Moreover, tumor acidosis is regarded as one of the contributing factors to therapy resistance. Partially, this is due to the mutational profile of stress-adapted cells, and, partially, due to the pH-induced modifications that occur to a drug in the extracellular environment. Interestingly, measuring the acidity of a tumor by non-invasive methods proved to predict antitumor activity and the efficiency of a therapy more accurately than the routinely used glucose-uptake imaging with lower tumor acidity correlating with better drug response [20]. Taken together, tumor acidosis is viewed as an important hallmark of cancer that contributes to cancer development, progression and therapy response [21], [22], [23]. In this review article we will try to update the current evidences about the pathogenic role of tumor acidosis and we will specifically discuss how tumor acidosis may negatively affect the efficacy of different therapeutic strategies. Finally, we will identify potential strategies to overcome pH-mediated therapy resistance.

Section snippets

Pathogenic role of tumor interstitial acidification

Extracellular acidification has evolved from a phenotypic feature of cancer cells to an important hallmark of cancer [24], [25], [26]. Metabolic peculiarities of tumor cells have been attracting the attention of cancer researchers for long time (since the “Warburg effect” was described, implying that cancer cells rely on glycolysis even in the presence of oxygen). However, it has become increasingly clear that tumor cell metabolism is more complex than that and involves the use of different

Acid/base balance and drug resistance

pH homeostasis is fundamental for the regulation of many biochemical processes and the reversed pHi/pHe gradient observed in cancer cells modulates sensitivity to growth factors, cell cycle and apoptosis. Also, the pH gradient across cellular membranes is a crucial factor determining the passive diffusion of small molecules. The therapeutic implications of tumor acidosis represent the main objective of this review and in the next sections we will discuss such issues, keeping in mind both the

Strategies to target cancer cells in an acidic environment

To overcome pH-mediated therapy resistance that is related to the acidification of tumor microenvironment, different strategies have been proposed, either by using molecules that are more active in an acidic environment or by the modulation of tumor interstitial pH in order to increase drug efficacy. In this last case, the counteraction of tumor acidification and the restoring of a normal extracellular pH in the tumor environment have three major goals: to improve the efficacy of weakly basic

Conclusive remarks

Tumor acidosis has been known as a feature of solid tumors for many decades and the identification of metabolic alterations as important hallmarks of cancer has contributed to acknowledge tumor acidosis a self-standing role as a driver of malignancy. New pathobiological effects of acidosis are being discovered and will contribute to a better knowledge of the molecular mechanisms driving tumor development and progression. These studies, together with appropriate consideration of the charge

Conflict of interest

No conflict of interests.

Funding source

This study is partially funded by grants from Karolinska Institute.

Acknowledgements

This study is partially funded by grants from the Karolinska Institute (to ADM) and from the Italian Association for Cancer Research (#15608 to NB and #14191 to SA).

References (223)

  • A.S. Tan et al.

    Mitochondrial genome acquisition restores respiratory function and tumorigenic potential of cancer cells without mitochondrial DNA

    Cell Metab.

    (2015)
  • G. Qing et al.

    ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation

    Cancer Cell.

    (2012)
  • C. Ryder et al.

    Acidosis promotes Bcl-2 family-mediated evasion of apoptosis: involvement of acid-sensing G protein-coupled receptor Gpr65 signaling to Mek/Erk

    J. Biol. Chem.

    (2012)
  • S.J. Reshkin et al.

    Phosphoinositide 3-kinase is involved in the tumor-specific activation of human breast cancer cell Na(+)/H(+) exchange, motility, and invasion induced by serum deprivation

    J. Biol. Chem.

    (2000)
  • R. Stern et al.

    Lactate stimulates fibroblast expression of hyaluronan and CD44: the Warburg effect revisited

    Exp. Cell Res.

    (2002)
  • L. Xu et al.

    Acidic extracellular pH induces vascular endothelial growth factor (VEGF) in human glioblastoma cells via ERK1/2 MAPK signaling pathway: mechanism of low pH-induced VEGF

    J. Biol. Chem.

    (2002)
  • T. Morita et al.

    Clastogenicity of low pH to various cultured mammalian cells

    Mutat. Res.

    (1992)
  • J.A. Kellum

    Metabolic acidosis in patients with sepsis: epiphenomenon or part of the pathophysiology

    Crit. Care Resusc.

    (2004)
  • S.R. Walmsley et al.

    Neutrophil energetics and oxygen sensing

    Blood

    (2014)
  • B. Everts et al.

    Commitment to glycolysis sustains survival of NO-producing inflammatory dendritic cells

    Blood

    (2012)
  • K. Fischer et al.

    Inhibitory effect of tumor cell-derived lactic acid on human T cells

    Blood

    (2007)
  • K. Peter et al.

    Lactic acid delays the inflammatory response of human monocytes

    Biochem. Biophys. Res. Commun.

    (2015)
  • M. Nakanishi et al.

    Acidic microenvironments induce lymphangiogenesis and IL-8 production via TRPV1 activation in human lymphatic endothelial cells

    Exp. Cell Res.

    (2016)
  • V. Plaks et al.

    The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells

    Cell Stem Cell

    (2015)
  • D. Huang et al.

    Anti-angiogenesis or pro-angiogenesis for cancer treatment: focus on drug distribution

    Int. J. Clin. Exp. Med.

    (2015)
  • M.R. Horsman et al.

    Pathophysiological basis for the formation of the tumor microenvironment

    Front. Oncol.

    (2016)
  • T. Matsubara et al.

    Additive influence of extracellular pH, oxygen tension, and pressure on invasiveness and survival of human osteosarcoma cells

    Front. Oncol.

    (2013)
  • L.E. Gerweck et al.

    Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer

    Cancer Res.

    (1996)
  • W.F. Boron

    Regulation of intracellular pH

    Adv. Physiol. Educ.

    (2004)
  • A. McIntyre et al.

    The role of pH regulation in cancer progression

    Recent Results Cancer Res.

    (2016)
  • P. Swietach et al.

    The chemistry, physiology and pathology of pH in cancer

    Philos. Trans. R Soc. Lond. B: Biol. Sci.

    (2014)
  • G. Weiss et al.

    Macrophage defense mechanisms against intracellular bacteria

    Immunol. Rev.

    (2015)
  • R.S. Flannagan et al.

    Antimicrobial mechanisms of macrophages and the immune evasion strategies of staphylococcus aureus

    Pathogens

    (2015)
  • G. Lamonte et al.

    Acidosis induces reprogramming of cellular metabolism to mitigate oxidative stress

    Cancer Metab.

    (2013)
  • M.N. Loja et al.

    Optical molecular imaging detects changes in extracellular pH with the development of head and neck cancer

    Int. J. Cancer

    (2013)
  • Z. Luo et al.

    Widefield optical imaging of changes in uptake of glucose and tissue extracellular pH in head and neck cancer

    Cancer Prevent. Res.

    (2014)
  • T. Matsubara et al.

    Acridine orange used for photodynamic therapy accumulates in malignant musculoskeletal tumors depending on pH gradient

    Anticancer Res.

    (2006)
  • D.L. Longo et al.

    In vivo imaging of tumor metabolism and acidosis by combining PET and MRI-CEST pH imaging

    Cancer Res.

    (2016)
  • S. Fais et al.

    Microenvironmental acidosis in carcinogenesis and metastases: new strategies in prevention and therapy

    Cancer Metastasis Rev.

    (2014)
  • R.J. Gillies et al.

    Metabolism and its sequelae in cancer evolution and therapy

    Cancer J.

    (2015)
  • A. Schulze et al.

    How cancer metabolism is tuned for proliferation and vulnerable to disruption

    Nature

    (2012)
  • R.J. Gillies et al.

    31P-MRS measurements of extracellular pH of tumors using 3-aminopropylphosphonate

    Am. J. Physiol.

    (1994)
  • B.T. Mortensen et al.

    Changing bone marrow micro-environment during development of acute myeloid leukaemia in rats

    Br. J. Haematol.

    (1998)
  • P. Vaupel et al.

    Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review

    Cancer Res.

    (1989)
  • R. Morais et al.

    Tumor-forming ability in athymic nude mice of human cell lines devoid of mitochondrial DNA

    Cancer Res.

    (1994)
  • P.H. Maxwell et al.

    Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth

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

    (1997)
  • T.G. Graeber et al.

    Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours

    Nature

    (1996)
  • M.C. Lloyd et al.

    Darwinian dynamics of intratumoral heterogeneity: not solely random mutations but also variable environmental selection forces

    Cancer Res.

    (2016)
  • H. Wu et al.

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

    J. Pathol.

    (2012)
  • V.H. Villar et al.

    Glutaminolysis and autophagy in cancer

    Autophagy

    (2015)
  • Cited by (0)

    View full text