Gill2016 (Grupo3)

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Biochimica et Biophysica Acta 1866 (2016) 87–105

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

journal homepage: www.elsevier.com/locate/bbacan

Review

Glycolysis inhibition as a cancer treatment and its role in an anti-tumour


immune response
Kheshwant S. Gill a,b, Philana Fernandes a, Tracey R. O'Donovan a, Sharon L. McKenna a, Kishore K. Doddakula b,
Derek G. Power a,c, Declan M. Soden a, Patrick F. Forde a,⁎
a
Cork Cancer Research Centre, Western Gateway Building, University College Cork, Cork, Ireland
b
Cardiothoracic Surgery Department, Cork University Hospital, Cork, Ireland
c
Department of Medical Oncology, Mercy University Hospital, Grenville Place, Cork, Ireland

a r t i c l e i n f o a b s t r a c t

Article history: Increased glycolysis is the main source of energy supply in cancer cells that use this metabolic pathway for ATP
Received 15 March 2016 generation. Altered energy metabolism is a biochemical fingerprint of cancer cells that represents one of the
Received in revised form 29 June 2016 “hallmarks of cancer”. The immune system can prevent tumour growth by eliminating cancer cells but this
Accepted 30 June 2016
editing process ultimately results in poorly immunogenic cells remaining allowing for unchallenged tumour
Available online xxxx
growth. In this review we look at the glycolysis pathway as a target for cancer treatments. We also examine
Keywords:
the interplay between the glycolysis modulation and the immune response as an anti-cancer therapy.
Glycolysis © 2016 Elsevier B.V. All rights reserved.
Glycolytic modulator
Immune-metabolic interaction
Electroporation

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
2. Glycolysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
2.1. Warburg hypothesis/effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
2.2. Cellular metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
2.3. Glycolysis in cancer cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
3. Rationale behind targeting glycolysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
3.1. Glycolytic targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
3.1.1. Hexokinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
3.1.2. Phosphofructokinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
3.1.3. Glucose transporters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
3.1.4. Pyruvate dehydrogenase kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
3.1.5. Lactate dehydrogenase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
3.1.6. Pyruvate kinase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
3.1.7. Fatty acid synthesis/fatty acid oxidation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
4. Relationship between cell glycolysis and immune cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
4.1. Immune cell metabolism and tumour microenvironment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92
4.1.1. Innate immune cell metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
4.1.2. Adaptive immune cell metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
5. Immune signaling and metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
6. Metabolic modulators and immune-glycolytic therapeutic implications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
6.1. Targeting tumour hypoxia-inducible factor-1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
6.2. Targeting uncoupling proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95

⁎ Corresponding author.
E-mail address: p.forde@ucc.ie (P.F. Forde).

http://dx.doi.org/10.1016/j.bbcan.2016.06.005
0304-419X/© 2016 Elsevier B.V. All rights reserved.
88 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

6.3. Targeting tumour glycolysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96


6.4. Targeting the mitochondria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
7. Delivery methods of glycolytic modulators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
7.1. Viral drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
7.2. Non-viral drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
7.2.1. Synthetic polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
7.2.2. Natural polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
7.3. Physical methods of drug-delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
7.3.1. Electroporation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
7.3.2. Other methods of physically facilitating drug-delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99

1. Introduction and (2) pathway that does not involve the mitochondria (glycolysis;
less energy-efficient and a shorter pathway). Oxidative phosphorylation
Intracellular chemical transformations sustain life and this is re- is a pathway involving mitochondrial respiration. Depending on intra-
ferred to as metabolism. Organisms grow and reproduce, respond to cellular requirements and available resources, cells undergo either gly-
their environment, and maintain their structure as a result of enzyme- colysis or oxidative phosphorylation to metabolise glucose. Glycolysis
catalyzed metabolism. Biochemical reactions in cells ensure daily oper- is inhibited (‘Pasteur effect’) by the presence of oxygen in most mam-
ations of a cell are accomplished. These reactions are either up- or malian cells. Oxygen allows the mitochondria to oxidise pyruvate to
downregulated depending on the cell's needs and functions. Monitoring carbon dioxide and water. The maintenance of energy production in
of the numerous cellular anabolic (breaking down of organic matter) various conditions of oxygen concentrations is met by the mammalian
and catabolic (metabolic component construction) pathways is crucial cells' metabolic adaptability.
to ensure they are balanced in a coordinated fashion at any given ‘Anaerobic glycolysis’ is whereby glucose is metabolised via glycoly-
time. Cells organise these reactions into different enzyme-catalyzed sis instead of mitochondrial oxidation in low levels of oxygen. This
pathways to achieve this goal. results in the conversion of pyruvate into lactate, which is then
Metabolic pathways are organised chemical reactions of metabolism exported. Interestingly, ‘aerobic glycolysis’ (glycolysis in the presence
where a chemical compound is converted into another one by a series of of oxygen) is also seen in cancer cells. This almost a century-old phe-
enzymatic activity. Enzymes are vital to metabolism. This is because nomenon in cancer cells is known as the ‘Warburg effect’, and was
they allow the organism to drive energy-consuming reactions that will hypothesised in 1924 by the German scientist Otto Heinrich Warburg.
not occur independently. This is achieved by coupling reactions with
spontaneous reactions which release energy. Enzymes speed up reac- 2.1. Warburg hypothesis/effect
tion rates as they act as catalysts besides allowing the metabolic path-
way regulation secondary to changes in the cell's environment or to Cancer cells mainly produce energy by an increased rate of glycolysis
other cellular signaling. (200 times more as compared to normal tissues of origin) followed by
Basic metabolic pathways and their components are similar across fermentation of lactate in the cytosol of the cell, even if oxygen is plen-
vast different species [1]. This is exemplified by a set of carboxylic acid tiful [5]. This is to fulfill their bioenergetic and biosynthetic demands to
(citric acid intermediates) which is present in all known organisms. support rapid proliferation. This observation in oncology is called the
The early appearance in evolutionary history and efficiency in retention ‘Warburg effect’. In normal cells however the rate of aerobic glycolysis
have likely led to the similarities in metabolism [2,3]. The process of is lower, and is followed by oxidative phosphorylation in the mitochon-
catabolism harvests energy by means of cellular respiration, and in dria, where pyruvate is oxidated [6–8]. It was postulated by Otto War-
anabolism, cellular components are constructed using energy e.g. burg that this metabolic change is the fundamental cause of cancer
nucleic acids and proteins. An example of a metabolic pathway which [9], a claim now known as the Warburg hypothesis. However, it was
is common in virtually all cells, both prokaryotic and eukaryotic, is then discovered that mutations in oncogenes and tumour suppressor
glycolysis. genes are responsible for malignant transformation. Instead of a cause,
the Warburg effect is considered to be more of a result of these genetic
2. Glycolysis mutations [10,11].

Glycolysis pathway of cellular respiration is a series of reactions that 2.2. Cellular metabolism
constitute the first phase of most carbohydrate catabolism. The word
glycolysis is derived from two Greek words and means the breakdown Normal cell metabolism derives about 70% of its energy from the
of something sweet. Glycolysis is an oxygen independent metabolic Krebs cycle and only 20% from glycolysis. Most of its energy production
pathway, meaning it does not use molecular oxygen for any of its is via oxidative phosphorylation. In contrast, cancer cells exhibit a defec-
reactions. It occurs in the cellular cytosol of most organisms. Embden– tive tricarboxylic acid (TCA) cycle and derive little or no energy from it.
Meyerhof–Parnas (EMP pathway) [4] is the most common type of glycol- Instead, they derive almost all their energy from glycolysis (fermenta-
ysis. Other types include the Entner-Doudoroff pathway and various tion phosphorylation) and in the absence of oxygen. Normal cells oper-
hetero- and homofermentative pathways. ate at normal metabolic levels and reproduce themselves at a regulated
The glycolytic pathway can be divided into two phases. Firstly, the phase by possessing hormones and enzymes behaving in balanced
preparatory phase (or investment phase) is where ATP is utilised and manner. Normal functioning cells orderly-divide to proliferate only
followed by production of ATP in the second Pay Off phase. In the cell, when in demand. Cancer cells on the other hand are overactive and re-
glucose catabolism occurs via two main pathways, (1) one which in- produce themselves in requiring more nutrients. These cells have over-
volves mitochondrial respiration (longer but energy-efficient pathway) active or underactive enzymes and hormones, and develop an aberrant
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 89

DNA or gene structure or acquire abnormal number of chromosomes. fashion, therefore the increased glucose uptake is not merely a biochem-
This continues to be created uncontrollably and ultimately these ical switch but also a metabolic transformation that is indispensible. On-
surpluses of cells form a mass. Tumour cells possess a high rate of cogenic activation (e.g. c-myc, Akt) up-regulates glycolytic activity in
glucose uptake and glycolytic metabolism. The majority of their en- cancer cells [24] via HIF-1α activation. Akt activates aerobic glycolysis
ergy is supplied by glycolysis, whilst maintaining increased rates of and more this makes cancer cells dependent on glycolysis to survive
lactic acid production. This is important for cancer cell survival in [25]. The anabolic and catabolic processes in cancer cells are coordinated
hypoxic environments. and regulated by networks of signaling pathways such as the mammali-
an Target of Rapamycin (mTOR). AMPK (energy sensing molecule) is
2.3. Glycolysis in cancer cells able to influence the activation mechanism of mTOR complex 1
(mTORC1). This in turn either delays or stops the energy consuming
Only about 10% of normal cells energy is produced by glycolysis. Mi- synthetic processes [26]. Activation of c-myc and HIF-1α genes regulates
tochondrial respiration contributes to the remaining 90%. A glucose the expression of glycolytic enzymes, which in turn are regulated by
molecule is metabolised into two molecules of pyruvate and NADH+ mTORC1 [26–28].
each, and gains 2 molecules of ATP. This is a markedly smaller amount It is evident that cancer cells benefit from aerobic glycolysis with the
of energy produced as compared to 38 molecules of ATP produced via PPP not only to promote tumour growth but also offering its therapy re-
mitochondrial respiration. Therefore, to obtain sufficient energy to sistance. Hence, tumour glycolysis provides the best possible targets for
grow and proliferate, cancer cells require an increased glucose uptake. therapeutic interventions. Fig. 1 is a pictorial representation of glycolytic
In hypoxic conditions, glucose via glycolysis is catabolised to lactate to pathway targets.
produce ATP. The mitochondriae are unable to produce ATP in oxidative
phosphorylation during hypoxia because oxygen is crucial for electron 3.1. Glycolytic targets
transport. Also, in hypoxic conditions, the monocarboxylate trans-
porters MCT4 (plasma membrane lactate transporter) are up regulated In addition to providing sufficient energy, the environment of cancer
via a HIF-1α mediated mechanism. This enables cells to dispose the cell glycolysis also promotes proliferation by providing its building
accumulation of lactic acid rapidly [12]. blocks, whilst supressing apoptosis at the same time. Cell death (ATP-in-
Given the small yield of energy via glycolysis, it is understood that dependent) will occur from rapid depletion of ATP as a result of direct
not only the metabolic switch to glycolysis from oxidative phosphoryla- glycolytic inhibition. This strategy will also likely damage normal
tion, but also mitochondrial dysfunction [13,14] is important for cancer healthy tissues. Non-malignant tissues e.g. smooth and skeletal muscle
cell growth. Cancer cells develop a high glucose uptake rate and glycol- and normal viscera will be affected as glycolytic inhibition is non-
ysis to produce energy. This increased rate of glycolysis in hypoxia leads selective. Translational potential of glycolytic inhibitors such as Hexoki-
to higher levels of lactate, which is sufficient for cancer cell survival. nase inhibitors and 3-Bromopyruvate are limited because of this reason.
Mitochondrial defects are commonly seen in cancer. The enzymes Their clinical trials have not been successfully completed. Pre-clinical
succinate dehydrogenase (SDH), isocitrate dehydrogenase (IDH) and data suggests the induction of necrosis in cancer and has also shown sig-
fumarate dehydrogenase (FDH) in the TCA cycle are affected by mito- nificant toxicities in animal models at even slightly-above therapeutic
chondrial DNA (mtDNA) mutations [15]. Besides that, the bioenergetics doses.
of cancer cells are affected by gene mutations in nuclear DNA (nDNA) of
the mitochondria [15]. Thus it is evident that a shift in cancer cell metab- 3.1.1. Hexokinase
olism can be caused by mitochondrial dysfunction. Having said that, Hexokinase (HK) is a tissue-specific isoenzyme which catalyzes the
there are several benefits of aerobic glycolysis that drive cancer cells phosphorylation of glucose to glucose-6-phosphate (G6P), after glucose
to favour it over mitochondrial oxidation [16]. The glycolytic rate in can- enters the cell via glucose transporters (GLUT). This ATP-dependent re-
cer cells are accelerated to about 100 fold thereby resulting in higher action is the first step in glycolysis, and is also rate limiting. G6P serves
ATP production [17,18], required for cellular maintenance and sufficient as the starting point for the sugar to enter the glycolic pathway or the
for cancer growth. Besides ATP production, the biosynthesis of macro- PPP, or for glycogen synthesis. HK has four isoforms, and they are desig-
molecules is essential for tumour cell proliferation [19]. Ribose-5- nated HK I, II, III, and IV (glucokinase found in hepatocytes) or A, B, C,
phosphate and NADPH (essential for nucleic acid and lipid synthesis) and D. Among these isoforms, HK-II plays a key role in the development
are produced via the Pentose Phosphate Pathway (PPP) promoted by of the Warburg phenotype exhibited by most types of cancer. HK-II iso-
the accumulation of these glycolytic intermediates. The antioxidant glu- form is found mainly in skeletal muscle and adipose tissues. Overex-
tathione (GSH; produced in presence of NADPH) maintains the redox pression of HK-II is also seen in some tumour tissues and associated
levels and counteracts chemotherapeutic agents thus protecting cancer with poor prognosis [29,30]. Phosphorylated HK-IIs are able to bind to
cells [20,21]. The transketolase (TKTL1) enzyme involved in the PPP has the voltage-dependent anion channels (VDACs) found on the outer mi-
been shown to improve cell survival in stressful times by affecting tochondrial membrane, besides its glucose phosphorylation activity.
chemosensitivity of cancer cells to chemotherapeutic drugs e.g. imatinib Negative feedback inhibition of G6P is prevented by this VDAC-HK in-
[22] and cetuximab [23]. In summary, aerobic glycolysis and the PPP teraction to ensure tumour cells entrap enough glucose. VDAC-HK inter-
provide multiple advantages to cancer cells and help tumour to progress action also confers HK direct access to ATP generated by mitochondriae.
besides resisting them from therapy. And this key signature of cancer Binding of BAX (protein apoptotic regulator which binds to VDAC in
metabolism (glycolysis) caters therapeutic intervention targets. normal cells) is interfered by VDAC-HKII interaction [31]. This VDAC-
HKII association also affects the pro-apoptotic functions of BAX in nor-
3. Rationale behind targeting glycolysis mal cells, where cytochrome C (located in the inter-membrane mito-
chondria space) escapes from the mitochondria thus triggering
Targeting the glycolytic pathway has the potential to be utilised as a apoptosis. This ultimately leads to tumour cellular apoptosis being
cancer treatment strategy. Tumour cells have a higher uptake of glucose prevented.
not only to meet the manifold requirements but also the high-energy HKs maintain the downhill concentration gradient that favours the
demand. Apart from providing energy, intermediates of the glycolytic facilitated transport of glucose into cells by catalyzing the first step in
pathway are also used for anabolic reactions such as glucose-6-phos- glycolysis. ‘Trapping’ of glucose and 2-deoxyhexoe glucose analogs
phate used to synthesise ribose-5-phosphate. This is then further used (e.g. 2-deoxyglucose, and 2-fluoro-2-deoxyglucose) within cells is due
in the synthesis of nucleic acids. Dihydroxyacetone is utilised in lipid to the addition of a charged phosphate group at the 6-position of hex-
synthesis. Tumour cells proliferate rapidly and in an uncontrollable ose. This is because charged hexose phosphates cannot easily cross the
90 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 91

cell membrane. HKs, besides GLUT1, exert the main control (71%) of gly- expression pattern is often elevated in colon, breast, ovarian and thyroid
colytic flux in hepatocellular and cervical carcinoma cell lines [32]. cancer with a particular preference for PFK-2/FBPase [55].
Many anti-cancer strategies implicating HK-II as a target have been of The PFK inhibitor, chalcone-like 3-(3-pyridinyl)-1-(4-pyrinidyl)-2-
big interest recently. Publication of work on identifying HK-I–HK-IV propene-1-one (3PO), is a PFKFB3-specific inhibitor. 3-PO has been
inhibitors goes back to the mid-20th century e.g. dehydroascorbic found to decrease intracellular F2,6BP and glycolytic flux in transformed
acid, alloxan and other compounds were found to inhibit liver HK [33]. cells and to selectively affect ras-transformed cells and decrease
After phosphorylation by HK, 2-deoxyglucose (2DG), a structural an- tumourigenic growth in several adenocarcinomas in vivo [56]. In non-
alog of glucose, enters cells through GLUTs and selectively accumulates small cell lung carcinoma (NSCLC), 3-PO has been used in combination
in cancer cells. This is because they are not able to progress through the with ascorbic acid and was shown to have a synergistic activity. Apopto-
glycolytic pathway. Cellular energy metabolism and its survival are like- sis was induced with this combination strategy by an ROS-dependent
ly to be profoundly affected by inhibition of HK. Thus, HK is an attractive mechanism [57]. The effects of this compound are yet to be explored
target for anticancer agents. Lonidamide is an inhibitor of HK-II and has in clinical trials.
been shown to induce apoptosis and treat multidrug resistance in mul-
tiple cell lines [34–36]. Phase II clinical trials of Lonidamide for benign 3.1.3. Glucose transporters
prostatic hyperplasia treatment have been suspended due to its toxicity Glucose transporters (GLUTs) are a wide group of membrane
on the liver [36,37]. Combination of Lonidamide with Paclitaxel (che- proteins that facilitate the transport of glucose and other substrates
motherapeutic agent) is undergoing clinical trials for cancer treatment over a plasma membrane and entering into cells as nutrients. 12
[7,38]. GLUTS are encoded by human genome. A sodium-linked transporter
2-DG and 3-bromopyruvate (3-BrPA) are also well known HKII in- system (SGLT) and facilitative GLUTs [58] are the two different types
hibitors [39,40], which have shown to interrupt binding of HKII to the of membrane transporter proteins. These assist the transport of glucose
mitochondrion besides inducing cell death by depleting ATP and and other sugars through the lipid bilayer of the cell membrane down
inhibiting the cell cycle progression. 3-BrPA inhibition of glycolysis its concentration gradient. GLUT-1 is related to chemo- or radiotherapy
causes ATP depletion and this suppresses ATP-binding cassette (ABC) responses and rectal cancer stages [59]. Human cell lines of breast and
transporter activity and drug efflux leading to more drug retention. lung cancer (NSCLC) were shown to reduce in cellular proliferation by
This results in the ability of 3-BrPA in overcoming chemoresistance 50% and 75% respectively by inhibiting GLUT-1, and also mediated
and improving cancer therapeutics. apoptosis [60].
2-DG induces cancer cell death by depriving intracellular glucose Overexpression of GLUT-1 and/or GLUT-3 is associated with poor
levels. This results in induction of stress-related proteins [41,42], the prognosis of several types of human tumours [61,62]. GLUT-1 transport-
generation of free radicals [43], and inhibition of energy metabolism er is a potential target for anticancer therapy as inhibition of its expres-
[42–44]. Combination of 2-DG with ionizing radiation (IR) to treat ma- sion is associated with reduction in tumour growth [63]. Phloretin, a
lignant gliomas in recent clinical trials showed it to be well tolerated natural type of phenol found in the leaves of apples and pears, is a com-
with low toxicity levels and surrounding normal tissues being un- petitive GLUT inhibitor. It has been shown to inhibit tumour growth
harmed[45–47]. 2-DG was shown to activate pro-survival pathways in and/or trigger apoptosis in leukaemia, melanoma, liver, colon, and
cancer cells contrary to the wide believe as an anticancer agent [48]. breast cancers in vitro and in xenograft models. Another flavonoid,
Hypoxic cells have also demonstrated chemoresistance against 2-DG silybin (flavonolignan) was tested in men with prostate cancer [64]
[49]. This challenges the success of 2-DG as a single agent for and liver cancer (trials NCT00487721 and NCT01129570). It showed
antiglycolytic therapy. However, 2-DG showed good outcomes when this drug to have low tissue penetration and therefore needed high
combined with other treatments [50,51]. Clinical trials for 2DG as a gly- doses for effectiveness [64].
colytic inhibitor for prostate cancer (NCT00633087) and intracranial In contrast to flavonoids, fasentin, WZB117 and STF-31 are promi-
neoplasms (NCT00247403) have been suspended, mainly due to non- nent inhibitors of GLUT-1 that induce glucose deprivation, starvation
selectivity of the drug thus causing damage to non-cancerous cells too. and subsequent death in renal cell carcinoma cell lines and animal
models [65]. Overexpressed GLUTs have been explored as receptors to
carry nanoparticles loaded with drugs across the blood-brain barrier
3.1.2. Phosphofructokinase (BBB). Jiang et al. demonstrated that 2-DG-modified polymer nanopar-
Phosphofructokinase (PFK) is a glycolytic enzyme that catalyzes the ticles showed efficient GLUT-mediated transcytosis across BBB as well
irreversible transfer of a phosphate from ATP to fructose-6-phosphate. as endocytosis into glioma cells enabling targeted delivery of paclitaxel
PFK is considered to be a key regulatory enzyme for glycolysis because for brain glioma therapy [66]. These results show that the Warburg ef-
of the irreversible nature of this step in glycolysis, and its activity is ex- fect confers distinct characteristics to tumour cells that can be selective-
tremely sensitive to small changes in pH, with its activity dropping as ly targeted for anti-cancer therapy [65].
pH decreases [52,53]. PFKs activity is inhibited by allosteric regulation
by ATP itself when ATP levels are high in the cell. High intracellular 3.1.4. Pyruvate dehydrogenase kinase
levels of citrate also inhibit PFK activity. The enzyme ATP citrate lyase Mitochondrial activators increase mitochondrial influx and subse-
converts citrate into acetyl-CoA in the cytosol. PFK activity is inhibited quently increasing mitochondrial function. They have proven to be a
because the cell can get enough energy from the citric acid cycle and strategy towards inhibiting cancer growth. For example, dichloroacetate
thus not depend on glycolysis to supply carbons into the citric acid (DCA)–pyruvate dehydrogenase kinase (PDK) inhibitor acutely in-
cycle. The two types of PFKs are PFK-1 and PFK-2. Translation of four creases glucose oxidation (GO). Dromparis P et al. showed that the
mRNAs produces the minimum six isoenzymes of 6-phosphofructo-2- mitochondriae of cancer cells are not permanently damaged but rather
kinase/fructose-2.6-biphosphatase (PFK-2/FBPase). Concentration of suppressed, because GO occurs exclusively in the mitochondria. There
F2,6BP determines PFK-1 activity and thus promoting glycolysis is a solid reason for developing similar inhibiting-molecules as antican-
when levels are high. PFK-1 activity is stimulated by fructose-2,6- cer therapies. A study showed that DCA depolarised the hyperpolarised
bisphosphate (F-2,6-BP) and is often overexpressed in various primary membrane potential of non-small cell, Glioblastoma multiforme (GBM)
cancers, metastases and in transformed cells [54]. PFK-2/FBPase and breast cancer cell mitochondriae. This suggests the restoration of

Fig. 1. Schematic representation of the glycolytic pathway. Following uptake of glucose from the bloodstream into the tumour microenvironment, it is delivered intracellularly by GLUT
transporters. In glycolysis the glucose is processed by a sequence of reactions, which lead to the production of pyruvate (aerobic conditions) and lactate (anaerobic conditions). HIF-1
affects the expression of enzymes and transporters in the glycolysis pathway.
92 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

mitochondrial function [67]. By enhancing GO and lowering the produc- inhibiting PKM2. This results in the opposite to detrimental effects to tu-
tion of lactate, DCA decreases the glycolysis to GO ratio. Xenotransplant mour growth. Both normal and cancer cells need to generate macromol-
tumour size was shown to reduce in size when treated with DCA and it ecules such as proteins, lipids and nucleic acids. Cell proliferation will
also lessened the aggressiveness of breast cancer [68]. Non-cancerous not occur in the absence or low levels of these macromolecules even
cell mitochondriae were not affected with DCA treatment but cancer in the presence of ATPs. Shuttling of PEP from slowing down glycolysis
cells treated with DCA showed to have activated Kv-channels, increased will result in the production of macromolecules necessary for cellular
mROS and inhibited NFAT. Many other studies in colon [69], prostate proliferation [87–89]. However, there is still interest in inhibiting
[70], and endometrial [71] also showed regression in tumour size PKM2 as it is important to tumour metabolism. A combination of mod-
when treated with DCA. Mitaplatin (cisplatin + DCA) is a cancer selec- ulation of both, the transfer of an amine from glutamine during mito-
tive drug and treatment which induces mitochondrial-dependent apo- chondrial respiration and PK, has been suggested as a method for
ptosis. Furthermore, it also overcame the resistance towards cisplatin metabolic targeting of tumours. This is because glutamine metabolism
initially thus reducing proliferation of tumour. is important for tumour metabolism. Many steps in glutamine metabo-
lism have been shown to be regulated by Myc [90]. Novel inhibitors and
3.1.5. Lactate dehydrogenase nucleic acid therapies were designed to target PKM2 to achieve the de-
Lactate dehydrogenase (LDH) is a tetrameric enzyme with two sub- sired anticancer activity. Thallion Pharmaceuticals started a Phase II
types: LDH-A (skeletal muscle type or LDH-M) and LDH-B (heart type or clinical trials in 2007 with PKM2 inhibitor called TLN-232/CAP-232, ad-
LDH-H). In hypoxic cells, conversion of pyruvate into lactate (final step ministered to patients with refractory metastatic renal cell carcinoma
in the glycolytic pathway) is catalyzed by LDH-A whilst LDH-B catalyzes [91] and it was found to be well tolerated, but Phase III of this study
conversion of lactate into pyruvate. Lactate accumulation substantially was terminated due to legal reasons.
reduces intracellular pH and this is detrimental to the cell. Production PKM2 interacts with suppressor of cytokines signaling 3 (SOCS3)
of NAD+ from oxidation of the cofactor NADH is necessary from the re- making it highly immunomodulatory. Antigen-presenting ability of
duction step of pyruvate into lactate to continue glycolysis. Evidence DCs was disrupted with the PKM2-SOCS3 interaction [92]. T-cell upreg-
suggests that LDH-A, which is upregulated in invasive glycolytic can- ulation of glucose uptake and glycolysis during interaction with
cers, plays a critical role in cell proliferation. This allows the tumours antigen-presenting cells (APC) produces instant ATP to ensure a
to survive even in low oxygen levels. Elevated serum LDH levels are as- sustained immunogenic response to an antigen [93,94]. Furthermore,
sociated with poor prognosis in pancreatic [72], renal [73], nasopharyn- PKM2 has also been shown to interact with IgE receptor on the cells
geal [74] and lung cancers [75]. resulting in the inhibition of its activity [95].
LDH activity is not needed for pyruvate metabolism through the TCA
cycle, and therefore inhibitors of this enzyme should spare glucose me- 3.1.7. Fatty acid synthesis/fatty acid oxidation
tabolism of normal non-proliferating cells. Several studies have already In the cytoplasm, fatty acids are produced from acetyl-CoA, via
found that the inhibition of LDH-A in cancer cells could stimulate mito- malonyl-CoA, and catalyzed by fatty acid synthase (FAS). The synthesis
chondrial pyruvate metabolism (in a sense mimicking the effects of of cancer cell membrane requires a high amount of fatty acids and this
DCA), decrease mitochondrial membrane potentials and finally leading demand is fed from the Krebs cycle intermediate citrate [19] rather
to cancer cell death [76,77]. Oxidative stress is increased when intracel- than external sources. The metabolite citrate accumulates when en-
lular ATP depletion occurs as a result of treatment with FX11 (LDH in- zymes of the Krebs cycle are mutated. This has been shown to be as-
hibitor and antimalarial drug). Tumour cell death both in vitro and sociated with cancer. In pre-clinical breast cancer model, inhibition
in vivo has been shown with FX11 treatment [76]. FX11 inhibits pro- of fatty acid synthesis by the compound C75 also showed increased
gression of sizable human lymphoma and pancreatic cancer [76]. An- apoptotic levels and inhibition of tumour growth [96,97]. The en-
other LDH inhibitor, oxamate, sensitizes resistant cancer cells to zyme malonyl-CoA decarboxylase (MCD) catalyzes the conversion
chemotherapeutic agents. A synergistic inhibitory effect was shown in of malonyl-CoA into acetyl-CoA and carbon dioxide. Inhibition of
combinational treatment of paclitaxel and oxamate in paclitaxel- FAS results in increased levels of its substrate malonyl-CoA, which
resistant breast cancer cells, which had higher apoptotic levels [78]. allows FAS inhibitors to display their anti-tumour effects. Therefore,
Lactate suppresses the function of tumour-specific T cells and in- inhibition of MCD can be used to enhance the efficacy of FAS inhibi-
hibits tumour specific cytotoxic cell activity [79]. It also induces defect tors as anti-cancer agents [98]. Through the Randle cycle, fatty
in signal transduction [80]. Dendritic cells' (DC) Ag-presenting ability acid oxidation (FAO) inhibition indirectly promotes GO and thus
is altered by low pH levels [81]. Furthermore, high lactate levels also in- inhibiting growth of cancer.
hibit cytotoxic activity [82] and promote IL-23/IL-17 pro-inflammatory
pathway [83]. Inhibition of LDH has demonstrated promising effects in 4. Relationship between cell glycolysis and immune cells
preclinical investigations and its further progress depends on the out-
come of clinical trials. However, in order to translate into clinical prac- Like other cells, immune cells use up the nutrients through cellular
tice, further studies are warranted to demonstrate target specificity metabolism and thus modulating its growth, volume and ion integrity
and therapeutic efficacy of experimental agents. [99]. Immune cells utilise the ATPs produced to perform various
functional activities and its proliferation, in addition to housekeep-
3.1.6. Pyruvate kinase ing proliferation and sustenance [99,100]. Notable and rapid changes
Pyruvate kinase (PK) is a critical enzyme in the glycolysis pathway. in metabolism are required as most of these actions are thermody-
PK has two isoenzymes, encoded by the PKM gene: PKM1 and PKM2. namically taxing [99,100]. Besides that, immune cells must also be
PK converts phosphoenolpyruvate (PEP) to pyruvate at the final irre- able to increase ion signaling and the turnover of phospholipids,
versible rate-limiting glycolytic step, and produces one molecule of macromolecule manufacturing and ultimately facilitate changes in
ATP in the process. The fate of pyruvate depends on local oxygen con- the cytoskeleton [101]. Resting immune cells depend on imported
centration levels in normal cells. In the presence of oxygen, pyruvate en- glucose to meet their metabolic needs as they contain little amounts
ters the mitochondria where it is converted into acetyl-Co-A by of stored glycogen [93,102,103].
pyruvate dehydrogenase complex (PDC) or into alanine by transamina-
tion. However, in low oxygen levels pyruvate is converted into lactic 4.1. Immune cell metabolism and tumour microenvironment
acid by LDH [84].
The PKM2 is highly expressed in cancer cells [85,86] and the glyco- The diverse functions of immune cells give rise to the differences in
lytic flux regresses to limit the generation of ATP in cancer cells by metabolism between the immune and other cells in the body. To
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 93

generate ATP, alveolar cells depend on oxidative phosphorylation [104] New nucleotides and proteins are also synthesised during this time. Ef-
whereas those always in contact with oxygenated blood use mitochon- fector cells result from the division and differentiation of T cells. It is im-
drial respiration. APCs migrate and present the foreign antigens detect- portant to obtain sufficient amounts of nutrients and therefore driving
ed by travelling immune cells, to lymphocytes. There are epidermal DCs glycolysis [103] to maintain the quick change in cellular size and
[105] in the deep tissue layers of the skin (lower oxygen tension com- function.
pared to dermis layer [106]). To survive and function properly in hypox- T cells heavily depend on glycolysis, and their proliferation is halted
ic conditions, immune cells must be able to use different forms of even in the presence of glutamine abundance [128]. They generate ATP
metabolism [107]. Lymphocytes traverse the cell wall of the endotheli- and lactate via glycolysis by converting glucose to pyruvate [18]. This in
um into the targeted area upon activation within the secondary lym- turn converts NADH back to NAD+ thus retaining glycolysis. More lac-
phoid organs [108], resulting in a slightly hypoxic environment too tate means more NADH production [93], which are needed as macro-
and the loss of dependence on oxidative phosphorylation. Inflammatory molecular precursors by T cells. T cells have high energy requirement
sites are also hypoxic due to the blocking of blood vessels by innate and are not able to enhance mitochondrial respiration to meet their
phagocytes [109–111]. To promote more glycolytic pathways, immune needs. Anabolic metabolism is driven efficiently by glycolysis and this
cells need to localise and adapt to different oxygen concentration levels is important to maintain cellular function and longevity and especially
[112,113]. during times of pathogen clearance [103,129] e.g. seen in T cell activity.
Protecting the body from pathogens is important and both innate However, in situations of glucose shortage [130], some oxygen is still
and adaptive immune cell activation is vital for this. Metabolic choices consumed [131]. Upregulation of mitochondrial enzymatic activity is
within immune cells are not solely dictated by the tumour microenvi- driven by calcium flux from lymphocytic activation resulting in in-
ronment. Immune cells must be efficient in their nutrient metabolism. creased oxidative phosphorylation [132,133]. Unstable Teff cells under-
Here it would seem like immune cells should utilise the oxidative phos- go apoptosis in conditions of low IL-2 and low nutrient levels. Some
phorylation pathway which generates most energy, but however this is clone cells have higher mitochondrial concentration and therefore are
not the case. The immune cell specificity and reactivity-state dictates able to switch to oxidative phosphorylation. On the contrary, constantly
the amount of mitochondrial respiration used. To synthesise macromol- activated T cells rely on oxidative phosphorylation too in autoreactivity
ecules and proliferate, activated immune cells prefer using the much and in graft-versus host diseases [134,135].
faster glycolysis [18]. T regulator cells (Tregs) [136] and memory CD8+ T cell [137] gener-
ation relies on lipid oxidation. Memory is generated after antigen-
4.1.1. Innate immune cell metabolism clearance post T cell activation and apoptosis of T effector cells (Teff) oc-
Foreign body substances in the body are constantly monitored by curs secondary to shortage of growth factors and metabolic activity
naive APCs. Myeloid cells and granulocytes favour glycolysis at resting [138]. Metabolism of both memory T cells and naïve cells requires oxi-
state [114,115]. As mentioned above, APCs immediately upregulate dative phosphorylation. To ensure the survival of T cells, some impor-
co-stimulatory molecules and process and present antigens on their tant changes happen during an immune response. Memory cells have
cell surface once an antigen is phagocytosed, requiring a greater level a higher affinity towards TCRs. This ensures survival in scarce-nutrient
of ATP. However, APCs continue to depend on glycolysis [114,115]. conditions and low homeostatic engagements [139]. CD8 T cells have
When stimulated by toll-like receptors (TLRs), DCs undergo metabolic extra respiratory capacities (necessary for ATP production in stressful
changes and increase their glycolytic rate [116]. Classically activated times), and they will be memory cells [140]. This also ensures cellular
macrophages (M1; proinflammatory) are regulated by glycolysis where- survival [141,142]. Noxa gene blocking generates CD4 T cell memory
as alternatively activated macrophages (M2; anti-inflammatory), rely [143,144].
more mitochondrial respiration [117,118]. Granulocytes e.g. neutrophils
also favour glycolysis [114,119]. As the first mediators at foreign body 5. Immune signaling and metabolism
sites, neutrophils' rapid degranulation results to cell death [120]. Neutro-
phils need to act quickly but it is not necessary to depend on greater ATP Almost every immune cell type such as T cells, B cells, NK cells, baso-
production to survive. This makes more sense for granulocytes and APCs phils, DCs, neutrophils, myeloid-derived suppressor cells (MCDCs) and
(fast turnover rates) to use the glycolytic pathway to generate the high NKT cells is well recognised in cancer progression [145–147]. Cells com-
demands of ATP. Lymphocytes switch their metabolic need to glycolysis municate with their environment through a variety of cell-surface re-
when activated, whilst rely heavily on oxidative phosphorylation in qui- ceptors that recognise and bind molecules present in the extracellular
escence. Memory is generated by lymphocytes (in oxidative phosphory- environment. Immune cells are activated when ligand binds with cell
lation state) after clearance of an antigen. This shows that they fluctuate membrane receptors and a reaction known as signal transduction is
their metabolic pathways [121]. triggered when this binding happens. In response to cellular signaling,
immune cells secrete cytokines (often produced in cascades) that bind
4.1.2. Adaptive immune cell metabolism to specific membrane receptors. These membrane receptors then signal
Plentiful supply of nutrients and T-cell receptor (TCR) stimulation at the cell via second messengers, often tyrosine kinases, to change the
resting state is necessary for T cells to function adequately. To generate activity of the cells. Interleukins are manufactured by one leukocyte to
ATP reserves (via catabolic metabolism), naïve lymphocytes are sub- act on other leukocytes as signaling ligands. They are the biggest class
jected to oxidative phosphorylation [103]. Following stimulation by an of cytokines.
antigen, quiescent immune cells quickly mobilise to the source [122]. Immune cells possess clear metabolic characteristics that influence
Quiescence is not only preserved by the high-ATP turnover of cell their immunological functions. For example, when DCs and T cells are
cycle proteins [123,124] but also by the upregulation of kinase inhib- activated, the metabolism of glucose and amino acids alters [148].
itors. Quiescent cells, via autophagy, are able to extract nutrients Also, energy, iron, and lipid metabolisms distinct metabolic characteris-
from degraded proteins [99,125,126]. Shortage of TCR interaction tics are related to macrophage polarisation [146]. The engagement of
downregulates the transport of glucose and ATP. The mitochondrial the Major Histocompatibility Complex-peptide on APC cells is required
potential will also be reduced [127]. BAX induction followed by for the stimulation of T cells via the T-Cell Receptor (TCR). Also, down-
apoptosis results from the lack of glucose uptake and the naïve T stream signaling and effector function depend on co-stimulation during
cell population can be controlled by this stringency. T cell activation. However, T cell anergy and deletion results from a lack
Upon antigen stimulation, lymphocytes (B and T cells) need to be- of co-stimulation [149,150]. The archetypal co-stimulatory molecule for
come rapidly activated, like the innate cells. Lymphocytes considerably T cell activation is the CD28 and it promotes T cell differentiation via its
enlarge their size within the first 24 h post-mitogen stimulation [121]. ability to bind CD80/CD86 on APCs [151,152]. The ability of CD28 to
94 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

trigger a build-up of glycolytic intermediates and thus enhancing glu- To link metabolism and immunity, adipocytes release adipokines,
cose metabolism [93], stimulating glycolysis and increasing the expres- functioning similarly to macrophages [94,189,190]. These adipokines
sion of glucose transporters [94] is similar to insulin-receptor signaling. include IL-1, IL-6, IFNγ, TNFα, and MCP1. Lymphocytes and monocytes
In the hypoxic tumour microenvironment, the glycolytic shift is re- are recruited into the adipose tissue by adipokines, which in turn pro-
quired by cancer cells to survive. This glycolytic shift is triggered by mul- motes both pro- and anti-inflammatory functions. Macrophage entry
tiple mechanisms and is discussed elsewhere [153,154]. The effects of into the adipose tissue is induced by adipocyte hypertrophy [191].
CD28 are nullified by CTLA-4 and this reduces glycolysis and making This hypertrophy leads to MCP-1 production and a core with low levels
cells dormant [155]. The PI3K-Akt-mTOR pathway is crucial for bridging of oxygen and is secondary to overnutrition. These infiltrating macro-
the gap between metabolism and T cell activation. CD28, IL-2 and TCR phages are modulated by lymphocytes which are associated with
engagement leads to PI3K-dependent Akt activation. This then increases adipose tissue [192]. An anti-inflammatory macrophage phenotype in
activities of enzymes of glycolysis secondary to the surplus of glucose seen in lean mice with higher levels of Treg cells [193]. Also, recruitment
transporters on the plasma membrane of cells [103,156,157]. Myc (reg- of more proinflammatory macrophages is associated with proinflam-
ulator gene that codes for a transcription factor) induces glucose trans- matory Teff cells seen in the fat of obese mice [194]. This contributes to
porters and glycolytic enzymes, and it is activated by PI3K, MAPK and insulin resistance [195,196].
NF-κB [158,159]. In resting T cells, AKT together with STAT5 influence
glucose uptake into cells [160]. Lymphocytes only express the GLUT1 6. Metabolic modulators and immune-glycolytic
glucose transporter unlike other cell types. GLUT1 will be internalized therapeutic implications
in the absence of sufficient cytokine and/or TCR stimulation. This leads
to lower glucose uptake across the plasma membrane secondary to The immunology of tumour microenvironment is very complex and
down regulation of surface expression, and eventually decreasing the makes the development of modulators of metabolism challenging. The
viability of the cell. CD28-mediated Akt signaling is vital for expression main reasons of limited success of cancer immunotherapy include
and trafficking of GLUT1 to the surface of the cell that in turn increases poor infiltration of effector cells into solid tumours and poor host re-
glucose uptake [93,161]. Following B cell receptor (BCR) engagement, B sponses towards tumour antigens. Furthermore, the intrinsically immu-
cells also increase GLUT1 expression [102]. PI3K regulates glycolysis and nosuppressive nature of the tumour microenvironment adds to this
GLUT1 and is involved critically in immunoglobulin synthesis and B cell poor success rate. The effectivity of cancer immunotherapy depends
proliferation [162]. Even though critical for lymphocyte activation, on the balance of immune responses from tumour protection towards
GLUT1 overexpression can result in hyperactive lymphocytes and its rejection [197–199]. The microenvironment of tumours is therefore
other pathologies [161]. Therefore, a balance must exist. also to be considered in order to achieve better antitumour responses
Another important regulator of metabolism in immune cells is in future. Spontaneous regression has been witnessed in metastatic
mTOR [163]. They couple nutrient sensing to metabolic outcomes like melanoma treated with adoptive cell therapy (ACT) using autologous
glycolysis, fatty-acid and protein synthesis. mTOR enhances the expres- tumour infiltrating lymphocytes (TIL) [200–202]. (Table 1.)
sion of HIF1α [164,165] and thus mediating the glycolytic process. In all
mammalian cells, and not only DCs and T cells, mTOR is a downstream 6.1. Targeting tumour hypoxia-inducible factor-1
target of AKT signaling [166]. Protein synthesis [167] and mRNA trans-
lation are induced by the ability of mTOR to sense nutrient availability Hypoxia inducible factors (HIF) are comprised of an oxygen labile α
[168,169]. T cell proliferation will be blocked [170] without proper subunit (HIF-α) and a β-subunit, expressed throughout various cell
mTOR signaling and anabolic storage processes will be decreased [171, types irrespective of oxygen levels [203]. HIF-1α (one of three isoforms)
172]. T cell anergy [173,174] results from inhibition of the PI3K-Akt- expression in tissues is universal and was the first to be discovered [204]
mTOR pathway. T cells that are deficient of mTOR differentiate into reg- whereas HIF-2αs are expressed in the vasculature endothelium. Tu-
ulatory T cells (Treg) and not effector T cells (Teff) [175]. The role of PI3K- mour microenvironment is hypoxic (≤2% oxygen) and HIF-1 modulates
Akt in glycolysis is reported for immune cells like macrophages, DCs and metabolism in these conditions. HIF-1α levels are not only regulated by
T cells is reported elsewhere [164,166,176]. T cell migration results from oxygen levels but also by cytokines IL-1β and TNF-α [205]. Further-
links between mTOR and chemokine-dependent signaling [177–179]. more, DNA binding of HIF-1 stimulated by p44/42 MAP kinase can
This link also results in cancer metastasis [180,181]. To initiate an effec- also increase HIF-1 levels [206]. It is an important signaling molecule
tive adaptive immune response, it is important to have proper metabol- for proliferation and growth of tumours. Aerobic glycolysis is promoted
ic signaling. This can also reveal targets for therapeutic intervention. in increased pyruvate levels [207]. HIF-1α binds to GLUT1 [208] and gly-
Oxidative phosphorylation (lower activity rate) is still necessary colytic enzymes to facilitate this metabolic switch. For example, cancer
for functionality although immune cells mainly use glycolysis to cell dependence on oxidative phosphorylation is reduced due to low
metabolise. Cytokine binding and cytokine receptor expression levels of acetyl-CoA (converted from pyruvate) because of increased
have also been connected with metabolism. TNF binding to specific PDK activity; secondary to HIF-1α mediated hypoxia responsive ele-
cellular receptors [182] and IL-2R expression on lymphocytes are ments [209]. This leads to lactate build-up in cells. This switch in metab-
reduced [183] if the electron transport chain in mitochondrial respi- olism preserves cellular viability in hypoxic times. Increased levels of
ration is blocked. The importance of proinflammatory cytokines in HIF-1 activation promote tumour growth, angiogenesis and metastasis
metabolic signaling is shown in a study where TNFα-deficient by reducing cytochrome c levels causing caspase-9 and -3 inactivation
mice were protected from insulin resistance secondary to obesity that prevents cell death [210] suggesting poorer prognosis, whilst low
[184,185]. Cytokines of the innate immune system e.g. IL-1, IL-6, IL-3 activity levels of HIF are associated with decreased tumour metabolism,
and IL-7 also play a role in cellular metabolism. Each cytokine binds to growth and vascularity [211].
a specific receptor on the cell surface, and is involved in the coordina- Hypoxic macrophages and DCs also switch their metabolism to gly-
tion of T cell function with metabolic needs. IL-1 can prevent fatty colysis by HIF-1α and mTOR. Activated lymphocytes undergo similar
acid synthesis [186], whereas IL-6 can both increase the levels of lipid metabolic changes to hypoxic cells. This questions the role of HIF-1s in
and glucose metabolism [187]. A metabolic shift to glycolysis from oxi- tweaking T cell activation. Immune cells need to survive and function
dative phosphorylation is caused by IL-3 [188]. Besides that, IL-3 is also properly in hypoxic conditions too, and this highlights the importance
important in supporting GLUT1 on lymphocyte surfaces [157] and to aid of HIF-1 activation [107]. In activated T cells, GLUT1 expressions are in-
in the growth of lymphoid and myeloid cells. Lastly, IL-7 must be pres- creased secondary to HIF-1 induced gene expressions that improve via-
ent to sustain uptake of glucose by activated AKT in T cells (both resting bility and survival of immune cells during aerobic glycolysis [212,213].
and activated states) [128]. Furthermore, T cell differentiation is also regulated by HIF-1. However,
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 95

Table 1
Current glycolytic and immune modulator clinical trials.

Modulator Target Effect Tumour type targeted Stage Study number

Vemurafenib plus BRAF BRAF/MEK inhibitor Metastatic melanoma Phase 2 NCT02414750


Cobimetinib
Response to neoadjuvant Determine the correlation between histopathological response Breast cancer NCT01440413
CT based on AT IR in (pCR) and induction of tumour immunity in response to
localized breast cancer neoadjuvant chemotherapy
2-deoxy-2-(18)fluoro-D-- Immunotherapy for FDG PET measure the response to immune checkpoint Melanoma, renal cell Phase 0 NCT01666353
glucose (FDG) melanoma blockade therapy carcinoma, non-small
lung cancer
Abraxane with or without Gene expression profile Humanised monoclonal antibody combined with Abraxane to Breast cancer Phase 2 NCT01307891
Tigatuzumab and the serve as targeting agent for triple negative breast cancer
proliferation/apoptotic patients
markers
K562-GM allogene Cancer testis antigen Patients with allogenic tumour express high levels of multiple Sarcoma, melanoma, Phase 1 NCT01313429
(CTA) CTA in combination with depletion with T regulatory cells germ cell tumour
TGFB2-Antisense-GMCSF TGGB2 Expression of TGFB2/GMCSF following transcription by Advance and Phase 1 NCT00684294
electroporation and irradiation of autologous cancer tissue non-curable solid
tumour
EMD 273066 EpCAM EMD 273066 is an experimental biological drug that may Ovarian, prostate, Phase 1 NCT00132522
increase the immune response to certain cancers colorectal or
non-small cell lung
cancers
MVA-brachyury-TRICOM Brachyury protein Vaccine it teaches immune cell to target which protein is Lung, breast, prostate, Phase 1 NCT02179515
present in some tumour cells, and it can help tumour cells ovarian
spread to other parts of the body.
Sipuleucel-T PA2024 Anti-androgen treatment started before or after sipuleucel-T Prostate cancer Phase 2 NCT01431391
leads to superior augmentation of immune response to
sipuleucel-T
2-Deoxy-D-glucose Hexokinase Inhibits the metabolism of glucose in the cells of the body Lung, breast, prostate Phase 1/2 NCT00633087
AP23573 mTORC1 Inhibits mTORC1 Solid tumours Phase 2 NCT00110188
Ipilimumab and Anti-CTLA-4 monoclonal Ipilimumab augments the effects of chemotherapy in animal Malignant melanoma Phase 2 NCT01654692
Fotemustrin antibody models relies on the ability of the cytotoxic agent to induce
apoptosis of tumour cells

Tregs are powered via lipid oxidation [136]. Innate cell functions e.g. ATP miR100 (mTOR signaling inhibitor) has been identified in clear cell
generation, glycolytic enzymes and GLUT1 expressions, are modulated ovarian cancer [224]. Delivery of these promising HIF-1 inhibitors
by HIF-1 levels, as seen in macrophages and neutrophils [214]. When should be encapsulated with nanoparticles to ensure targeted delivery
HIF-1 levels are low, innate cell functions deteriorate and thus resulting and avoiding nonspecific toxicity, as demonstrated by successful
in decreased abilities in T cell activation and its pathogen killing capabil- antitumour siRNA delivery to hypoxic tumour sites [225].
ities [115,215]. This is coupled by weakened granulocyte responses and
APC phagocytosis in hypoxic conditions [216,217] leading to poor host 6.2. Targeting uncoupling proteins
defenses. However by affecting the innate and adaptive immune cells,
both HIF-1 and chronic inflammation can initiate tumour advancement Mitochondrial ATPs are produced by coupling the electron transport
and autoimmunity. Glycolysis upregulated by HIF-1α has been linked chain (ETC) and ADP-ATP phosphorylation. However, this process is not
with tumour-associated-macrophages (TAMs), which are macrophages efficient in the presence of uncoupling proteins (UCP), which are anion
containing perinecrotic and hypoxic tumour tissues that aid in tumour transporters of the mitochondrial inner membrane [226]. Besides that,
angiogenesis and growth. UCPs also reduce the production of mitochondrial-free radicals [227]
Both HIF-1 and HIF-2 levels are raised in malignancy e.g. in bladder, for example by decreasing pyruvate entry into the TCA cycle [228,
colon, ovarian, pancreatic, prostate, hepatocellular, and glial cancers. 229], and are also involved in thermogenesis e.g. UCP1 involvement in
HIF-1α has been widely studied as a glycolytic target for cancer therapy, thermogenesis activity of brown tissue [230]. Three main uncoupling
both at pre-clinical and clinical stages [218], given its extensive involve- proteins which are UCP1, UCP2 and UCP3 have different uncoupling ac-
ment in regulating glycolysis. Cytotoxic agents are developed with aim tivities [226,231] and play different roles in mitochondrial physiology
to block HIF-1α expression, transcription and translation and also to in- [232]. UCP2 knockout results in resistance to infections [233,234] due
terrupt binding of targets. Digoxin for example inhibits HIF-1 gene ex- to NF-κB activation and increased levels of reactive oxygen species
pression and tumour proliferation [219], besides blocking RORγt- (ROS) in immune cells, making UCP2 an immunometabolic target.
dependent TH17 differentiation [220]. HIF-1 represses Treg differentia- High levels of mitochondrial UPC2 are found in activated T cells owing
tion and therefore inhibiting HIF may elevate suppressive T cells. This to its need for high proliferation via glycolysis. Low ROS levels are also
phenomenon may be advantageous in protecting against self-antigen necessary for expressing genes and signaling activity [235]. Uncoupling
recognition. Active CD8 T cells may still be able to kill tumours if HIF-1 the ETC when high levels of ROS are generated resets homeostasis,
is blocked. Other approaches of HIF-1 blockade include induction of particularly in the innate immune and T cell interplay. UPC2 is pro-
HIF-1α degradation by HSP90 inhibitor and 1-β-2 methoxyestradiol, glycolytic and is shown by its ability to stabilise the glyceraldehyde 3-
HRE-binding HIF-1 inhibition by doxorubicin and daunorubicin, and phosphate dehydrogenase (GAPDH) in cancer cell cytoplasm [236].
also bortezomib which inhibits HIF-1 transcription [221,222]. More re- UCP2 supports the glycolytic flux by its antioxidant effects of GAPDH ox-
cent genetic approaches targeting HIF-1α inhibition include siRNA ther- idation and nuclear translocation inhibition. With this, they prevent
apy EZN-2968, tested in hepatic metastasis. However, issue of toxicity cancer cells from stimulating cell death mechanisms. UCP2 decreases
arose in a number of patients with this approach [223]. Micro-RNA glucose-stimulated insulin release [237]. This enables them to control
(miRNA)-mediated therapy is also being tested in glycolytic inhibition. the autoreactive T cell responses. Increased levels of UCP2 are found in
96 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

leukaemia, kidney, colorectal, lung, breast, prostate, ovarian, bladder, downregulates cytokine receptors and thus reduces T cell signaling
oesophageal and testicular cancers [238]. [264], ameliorates Experimental Autoimmune Encephalomyelitis
Uncouplers have not been utilised in the treatment of autoreactive T (EAE) and enhances Treg cells [265]. Other studies showed the antioxi-
cells. Rottlerin (a chemical mitochondrial uncoupler) has been shown to dants' anti-inflammatory ability by restricting NF-κB activation [266–
decrease apoptosis in autoimmune-diseased alveolar macrophages 268] and glycolytic reduction in lymphoma cells through LDH activity
[239]. A fatty-liver (non-alcoholic) diseased model showed to have re- inhibition, by α-tocopherol (an antioxidant found in Vitamin E) [269].
duced insulin resistance when treated with 2,4-dinitrophenol [202]. Metalloporphyrins are a group of catalytic antioxidants that can
This drug also causes adhesion build-up particularly seen in post- completely remove toxic ROS. They show no toxicity and are able to
peritoneal surgery [240]. Uncouplers are cell-specific making them a modulate the immune function. It is an alternative metabolic therapy
potential therapy target for immune diseases (high oxidative stress). that may be beneficial in reducing inflammation and potentiating can-
Uncoupling inhibitors on the other hand can be used to treat standard cer regression. They have been shown to reduce incidence of type 1 di-
chemotherapy-resistant cancers [241]. abetes [258,270],activate NF-κB [268] and reduce CD8 Teff cell functions
[271]. Metalloporphyrins can reduce hypoxia, block HIF-1α activity and
6.3. Targeting tumour glycolysis suppress growth of tumour [272]. Early studies suggest administration
of metalloporphyrins decreases aerobic glycolysis, which in turn de-
Immune cells are highly dependent on glucose metabolism. Anti- creased T cell differentiation resulting in quiescence.
glycolytics limit immunity e.g. Rapamycin inhibits glucose metabolism
by blocking the mTOR downstream of PI3K-Akt and ultimately lowering 7. Delivery methods of glycolytic modulators
proliferation rate of cells due to reduced T and B cell activity. It can also
lead to hyperglycaemia because of this blockade. Rapamycin can affect T Current cancer treatment involves surgery, chemotherapy, radio-
cell differentiation and memory at the immune cell level. Furthermore, therapy or a combination of these. With the aid of cancer imaging tech-
besides causing a glycolytic switch to oxidative phosphorylation whilst nologies, the removal becomes more tumour-selective. Tumours may
increasing CD8 T cell memory response quality, rapamycin can also im- recur because cancer cells are not completely eliminated from the
itate dietary restriction and improve longevity [242] in memory T cells. body. Disease recurrence is a major challenge in cancer treatment. Nu-
Rapamycin can initiate Treg development in CD4 T cells[243,244] and merous anticancer reagents have been developed to suppress the
also higher infection rates[245,246] are seen with rapamycin treatment growth of residual or inoperable tumours. The identification of glycolyt-
because of its immunosuppressive capabilities. ic inhibitors for cancer therapy has led to the specific destruction of neo-
3-Bromopyruvate (3BP) (HK inhibitor) is an anti-glycolytic also plastic cells in several types of cancer. However, many types of cancer
used in cancer treatment. Cell death occurs with 3BP treatment as intra- still require effective anticancer drugs. To enhance anticancer activity
cellular ATP levels deplete [247]. 3BP is selective to cancer cells and whilst minimizing adverse effects and toxicity to healthy surrounding
therefore it is less toxic to healthy surrounding tissues [248,249], and tissues, the cancer-selective delivery of therapeutic molecules is de-
treatment with it can increase tumour immunity with higher infiltra- sired. Viral or non-viral route is the main category of drug delivery
tion of killer cells whilst diminishing the restraining tumour microenvi- methods. Each type of vector has their advantages and limitations.
ronment. Localized treatment especially intra-tumoural approach Innovations in micro/nanoparticle designs, new drugs e.g. post-
induces anti-tumour CD8 memory T cells. T cells may be harmed via transcriptional gene silencing nucleotides, and advanced new bio-
systemic treatment with 3BP because of the switch of metabolism to ox- materials result in new drug delivery methods.
idative phosphorylation from glycolysis.
7.1. Viral drug delivery system
6.4. Targeting the mitochondria
Drug delivery refers to various approaches and formulations, differ-
The mitochondrial ETC produces ROS and redox-damaged byproducts ent technologies incorporated into developing vectors and different sys-
that result in cellular death and therefore ETC mutations can decrease the tems for transporting a pharmaceutical composite. In doing so, the
amount of ROS and eventually prolong life [250,251]. NF-κB deactivation desired therapeutic effect has to be safely achieved. Viral delivery sys-
(reducing cytokine-induced adhesion of endothelial cells [252,253]) and tems, virosomes, consist of viral envelope components of inactivated
defects in phagocytes are caused by inhibition of various parts of the viral particles. They act as a vehicle for therapeutic molecules.
mitochondrial ETC [254]. Metformin is widely used in clinical practice Virosomes are composed of a phospholipid bilayer. Glycoproteins on
as an anti-hyperglycaemic. Its mechanism of action includes increasing the viral surface protrude from these vesicle surfaces [273–275].
skeletal muscle glucose uptake and reducing gloconeogenesis in the Virosomes do not contain a lot of preservative and their preparation
liver in order to diminish circulating insulin levels. UCP2 levels are in- usually does not involve complicated techniques [276]. Virosomes are
creased with the use of metformin [255]. In conditions of low oxygen, able to transport macromolecules e.g. drugs, nucleic acids, and proteins
metformin is able to block the mTOR function by AMP-activated protein to erythrocytes, hepatocytes, immune cells and glioma effectively [277–
kinase (AMPK) activation which itself increases glucose uptake. All 279]. In conventional drug delivery systems, the therapeutic molecules
these eventually lead to inhibition of cell proliferation [256,257]. Also, get degraded prior to reaching the target cells. However, virosomes are
metformin is able to inhibit cancer cell oxygen consumption by hindering able to deliver these drug molecules to target cells by virosome-
complex I of the ETC. Targeted tumour therapy with metformin causes T lysosome membrane fusion without being affected by the host defense
cells to kill cancerous cells and reduce tumour growth. Metformin mechanism. Many recombinant virus based delivery systems have
can regulate the immune system and the increased levels of memory emerged lately and these have been used for gene therapy and for vac-
CD8 T cells prove this post-administration of the drug on ova-specific T cine purposes [280]. By combining fusion peptide from influenza virus
cells, alongside tumour regression. haemagglutinin (HA), gene transfer efficiency of receptor-mediated
Another approach of metabolic control for immune regulation is the gene delivery systems has been enhanced. HVJ liposomes have been
usage of antioxidants across an array of pathologies from autoimmunity, widely used in animal models for cancer treatment. To prevent melano-
neurodegeneration and cancer [258–262]. Antioxidants are known to ma growth, melanoma-associated antigen gene or RNA has been
reduce ROS levels e.g. hydrogen peroxide (H2O2) is metabolised by the injected into spleen or skeletal muscle [281]. This has successfully in-
antioxidant glutathione during aerobic respiration [263]. There are voked immunity to prevent melanoma growth. Newer viral vaccine sys-
various ways in which antioxidants improve the immune system. tems offer advantages over conventional vaccines; however, they have
For example, epigallocatechin-3-gallate (antioxidant in green tea) their own limitations and side effects [282,283]. These viral vaccines
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 97

include adenovirus types 2 and 5, pox-, lenti-, retro-, and alphaviruses, siRNA existed in tumour tissue for more than one week when the
adeno-associated virus and lastly the herpes simplex virus (HSV). siRNA/atelocollagen complex was intravenously injected into tumour-
They can delivery antigens to specific subsets of immune cells effective- bearing mice. Intravenously administered SiRNA-atelocollagen complex
ly such as the APCs which can potentially act as adjuvants. Viral vectors into mice with bone metastasis from human prostate cancer [298]
can be combined with other vaccines and administered to result in en- revealed marked suppression of metastatic tumours in both types of
hanced immune responses. Both RNA and DNA viruses have been used siRNA in an in vivo imaging analysis.
as vaccines to express the antigenic/therapeutic protein in vivo and to
stimulate potent specific humoral and cellular immune responses [284]. 7.2.2. Natural polymers
Other polymer-based delivery systems include cationised gelatins.
These gelatins are prepared by the introduction of amine residues to
7.2. Non-viral drug delivery system
the carboxyl groups of gelatin via chemical means. Crosslinking
cationised gelatin with glutaraldehyde result in cationised gelatin
A promising technology of the delivery of variety of compounds to
hydrogels. These hydrogels have different in vivo degradabilities as
target sites is liposomes and liposomal-based delivery systems. Lipo-
the carrier of DNA. The plasmid DNA incorporated gelatin prolongs
somes are a non-viral delivery system and are used as a vehicle for ad-
the duration of gene expression. The antitumour plasmid DNA of NK4s
ministration of macromolecules into cells. In liposomes, water-soluble
(protein made up of the NH2-terminal hairpin and the four-kringled do-
molecules incorporate into a lipid bilayer-vesicle. They have been
mains of hepatocyte growth factor) biological activity is promoted by
utilised to target tumour tissues by systemic administration. To improve
the controlled release technology [299]. NK4 is an HGF antagonist that
the transfection efficiency and to reduce the cytotoxicity of the lipoplex,
inhibits the ability of HGF to promote tumour metastasis and angiogen-
numerous cationic lipids have been synthesised [285–288]. Lipoplex
esis. The mice with which received subcutaneous injection (controlled
with HLA-B7 and b-2 microglobulin genes were found to induce
release approach) of hydrogel microspheres had significant prolonged
antitumour immunity in HLA-B7-negative melanoma patients. Also,
survival as compared to mice that received NK4 plasmid DNA in
Allovectin-7 (a liposomal drug) has been clinically tested to treat meta-
solution.
static melanoma [287]. Glioblastoma patients in Japan were treated
Chitosan, prepared from chitin by deacetylation, is a positively
with the IFN-b gene. These were delivered by cationic liposomes
charged linear polysaccharide consisting of one to four linked N-
which were composed of N-(a-trimethy-lammonioacetyl)-didodecyl-
acetyl- D -glucosamine and D -glucosamine subunits, and interacts
D-glutamate chloride (TMAG) [289] dilauroyl phosphatidylcholine,
strongly with negatively charged DNA [300]. Chitin is a component
and dioleoyl phosphatidylethanolamine. Doxil which is a liposomal
of fungi and exoskeleton of crustaceans' cell walls. Chitosan is a non-
preparation composed of the high phase-transition temperature phos-
toxic polymer and can be digested by enzymes in the blood and
pholipid hydrogenated soy phosphatidylcholine (HSPC) and cholester-
human intestines. These enzymes include chitinases or lysozymes. In-
ol, is currently in use for the treatment of recurrent breast cancer [290,
terest to this polymer as a drug delivery system in pharmaceutical re-
291].
search was based on the above-mentioned special characteristics of
chitosan. Galactosylated, trimethylated and N-dodecylated chitosan
7.2.1. Synthetic polymers [285] are examples of chitosan derivatives. Chitosan is also used for
Currently, synthetic polymers are the most actively researched areas the intravenous delivery of siRNA for cancer therapy [301]. Another
of biomedicine. They have easily adjustable characteristics. Polymer- form of DNA delivery into cultured cells is polyethylenimine (PEI).
based nanoparticle delivery systems e.g. polymeric nanoparticles, The proton sponge effect causes the escape of DNA from the endosome
polymeric micelles, and dendrimers are newly innovative candidates to the cytoplasm, enhanced by PEI [302]. Stearic acid modified PEI-
to not only diagnose but also to monitor disease status and treat cancer based nanoparticles (more effective combination than PEI/siRNA com-
diseases. This is mainly due to their special features of enhanced perme- plex) were developed to deliver siRNA against signal transducer and
ability, biocompatibility and biodegradability, improved bioavailability activator of transcription 3 (STAT3) to suppress mouse melanoma
and transport efficiency, low toxicity depot effect, and lastly targeted in vivo [303].
delivery [292]. Less toxic synthetic polymers like poly-D,L-lactide-co-
glycolide (PLGA) and polylactic acid (PLA) also provide biological com- 7.3. Physical methods of drug-delivery
patibility [293]. ‘Smart’ polymers undergo conformational changes in
response to changes in the environment such as the temperature and Drug deliveries mediated by different physical methods have grown
surrounding pH [294]. They are able to employ in different fields and significantly over the past decade and a half. Systems of drug delivery
in response to specific physiological triggers they are able to release aim to deliver the therapeutic amounts of drug to targeted sites in the
the contained drugs. Polymeric micelles are block-copolymers, which body, and to promptly achieve and maintain the desired drug concen-
consist of two distinct linear polymers, are conjugated to therapeutic trations [304]. Physically facilitating drug-delivery (PFDD) systems can
molecules by an electrostatic interaction, hydrophobic interaction or be classified mainly as photo-, ultrasound-, magnetic-, and electrical-
metal complex formation. Doxorubicin was encapsulated into PEG-b- based modulation systems. This vast literature on PFDD is impossible
polyaspartic acid copolymer. PEG-b-polyaspartamide with a 1,2- to be covered in this review. Physical methods to push the DNA into
diaminoethane side chain is used as a block-copolymer for DNA encap- cells have been developed to enhance gene delivery based on naked
sulation. This micelle for DNA encapsulation enhanced the penetration DNA injection, firstly reported for skeletal muscle [305] and subse-
into tumour tissue and enabled gene expression in the hypoxic state quently reported for other tissues such as the thyroid, cardiac muscle,
[295]. The amount of doxorubicin in tumours by pH-sensitive micelles skin and liver [306].
was approximately fourfold higher than that by pH-insensitive micelles,
and more efficient tumour regression was achieved by pH-sensitive 7.3.1. Electroporation
micelles than by conventional micelles [296]. Electroporation (EP) is a technique where electrical fields are
Atelocollagen is a bovine type I collagen that is treated with pepsin applied to cells to increase the permeability of the cell membrane. The
and solubilised by protease. However, its physical properties are virtual- cell membrane represents a physical barrier to the intracellular transfer
ly identical to those of natural collagens, which are not solubilised. of macromolecules, peptides, hydrophilic drugs, or DNA to be intro-
Natural substances do not possess the superior characteristics found in duced into the cell [307]. The phospholipid bilayer of the plasma
atelocollagen. The atelocollagen (gel biomaterial in clinical use) has a membrane restricts the transmembrane movement of hydrophilic and
positive charge and can be a carrier of DNA and siRNA [297]. The polar molecules, other than by specific channel or receptor-mediated
98 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

assistance. EP is the strategy that involves direct application of an elec- late 1990s. It enhances capillary endothelial and parenchymal cell per-
trical field to target tissues or tumours to allow passage of various mol- meability. It began with investigations in intravascular injection of plas-
ecules including those that destabilise the cell membrane, by surpassing mid DNA solution for gene delivery in whole animals [329–331]. This
the cell membrane barrier. Both in vitro and in vivo cellular membranes delivery method is now a common method for DNA and RNA delivery
become transiently permeable through short high-voltage electric field in rodents and proven to be a useful method for delivery for a wide
pulse application. These voltages are optimised to balance its reversible range of applications [332]. For example, the hydrodynamic tail vein in-
and irreversible effects [308,309]. Cellular membrane composition jection of plasmid DNA has been used to assess the involvement of spe-
[310], its size and shape [311,312], and cellular suspension density cific genes in the development or regression of pathophysiological
[313] are factors that influence the extent of cellular permeabilisation conditions. Gene transfer efficiency is dependent on the elasticity of
after EP. EP with high electric currents causes tissue damage but having the target organ as its delivery to the cells of the organ is achieved by
said that, malignant tissues are more affected by electrochemotherapy pressure e.g. a soluble form of the fetal liver kinase-1 gene, a receptor
(ECT) as compared to normal tissues [314]. EP at a lower voltage with for vascular endothelial growth factor (VEGF), delivered by hydrody-
similar transfection efficiency has been developed to solve the problem namic injection suppressed VEGF-driven angiogenesis and inhibited
of irreversible EP [315]. There are two types of electrodes, the plate type, the growth of renal cell carcinoma and lung carcinoma in mice [333]. In-
which provides more stable results, and the needle type that is more travascular hydrodynamic limb vein delivery of the human gp100
useful for transfection in various tissues. Newer electrode designs are melanoma-associated antigen gene to skeletal muscle was used for mel-
being developed and used in clinical trials for internal tumours e.g. anoma vaccination [334].
liver metastasis from colorectal cancer [316]. In addition to the use of
drugs, EP can also be used to deliver a wide range of potentially thera- 8. Conclusion
peutic agents including proteins, dyes, tracers, antibodies, oligonucleo-
tides, RNA and DNA [317]. EP is used in animal models of cancer Increased aerobic glycolysis is commonly exhibited in cancer cells. In
treatments for the intramuscular delivery of IL-12 and IL-27 genes and order to generate ATP, malignant cells adapt to metabolic changes mak-
also for the regional tumour delivery of various therapeutic molecules, ing them addicted to and dependent on glycolysis. Cancer cell survival is
including microRNA against mutant K-ras [318]. at an advantage given the alterations in energy metabolism with in-
In clinical practice, reversible EP e.g. ECT, is an anti-cancer strat- creased glycolytic enzymatic activity and other molecules promoting
egy used with combination of chemotherapy drugs (intravenous or longevity. Increased glycolysis results in an acidic tumour microenvi-
intratumoural bleomycin administration followed by topical EP for ronment making it conducive for a selection of cancer cells with high
recurrent cutaneous breast cancer) [319]. Irreversible EP is another survival capacity.
anti-cancer strategy where cellular membranes do not reseal when The immune condition of tumour microenvironments is an impor-
higher electric fields are applied and ultimately results in cell death. tant factor in tumour prevention, development, and progression. Signif-
This approach however does not include combination with other icant evidences have been provided in several studies regarding the
drugs. The study of melanoma treatment is one of the more relevant correlation of disease outcome with tumour microenvironment status.
applications of EP. Human dendritic cells were transfected with tumour The future of cancer immunotherapies depends on recognizing the con-
mRNA using EP and then injected into melanoma patients in a Phase I/II cept of switching the immune response to a tumour-destructive profile,
study [320]. A Phase I trial of the direct injection of the IL-12 gene by EP instead of a tumour-promoting one. Manipulating immunological pa-
was performed with melanoma patients, and the safety of the method rameters in the tumour microenvironment may be adequate in tipping
was shown [321]. Gene therapy (IL-12) has been proven as safe, effec- the balance of host response towards effective immunity.
tive, reproducible and titratable by means of EP-assisted delivery The complex metabolic switch, immune-glycolytic interactions and
(gene electrotransfer) since the initial human trials [321]. tumour microenvironment present a major challenge in cancer treat-
ment. Over the years, many potential drugs have been developed,
7.3.2. Other methods of physically facilitating drug-delivery systems some of which are currently in preclinical and clinical stages. However,
Since the 1950s, the use of ultrasound has been known to improve several such promising drug candidates have been discontinued during
targeted drug delivery in human tissues [322]. Ultrasound is also avail- various stages of development due to poor bioavailability, unfavourable
able for the delivery of therapeutic molecules to tissue [323]. This trans- pharmacokinetic profile and associated nonspecific toxicity. Cancer im-
fection method depends on cavitation activity, one of the biological munotherapy has been revived with the successes of immune check-
actions of ultrasound. Contrast reagents such as Optison and Levovist, point blockade. However, a better understanding of immune response
which consist of gas-filled lipid or albumin-coated particles, are mixed mechanisms and checkpoint blockade resistance needs to be attained
with DNA. Ultrasound energies rupture these bubbles and subsequently in order to reach the goals of achieving substantial antitumour re-
enhance the membrane permeability. With these activities, DNA is in- sponses. The development of combination therapies (e.g. combination
corporated into target cells and most of the membranes are recovered therapy with ipilimumab and electrochemotherapy) [335] which in-
in 60 s. Increment in the mechanical index, frequency and exposure crease therapeutic benefits and reduce its side effects are necessary for
time increase the transfection efficiency. It is essential to determine the group of patients who do not benefit from a single checkpoint
the optimum conditions for each case as these parameters are also blockade.
closely related to cell damage. Tissue damage caused by sonoporation The burgeoning field of nanodelivery system has shown tremendous
seems to be less than the damage caused by EP. However, the delivery promise in circumventing many of the drawbacks associated with con-
efficacy is comparable to that of EP [324]. Sonoporation is also used ventional drug delivery approaches. This has opened avenues to revisit
for enhancing the liposomal delivery of the IFN-b gene to tumours application of several discontinued chemotherapeutic agents for the
[325] and regional intratumoural delivery of toxic compounds, such as treatment of different medical conditions. Recent literature survey
bleomycin and the cytolethal toxin gene, by sonoporation was success- clearly indicates that the advantages of nanocarriers have also led to nu-
ful in mice [326]. Advantages of ultrasound-based delivery system (high merous reports on delivery of drugs targeted against glycolytic path-
frequency sonoporation) include good-recorded safety usage in clinical way. In solid tumours, drug penetration is a critical aspect to consider
trials, minimally invasive, heterogeneous skin permeability and its abil- when looking at therapeutic strategies. With the advent of novel tu-
ity to deliver small molecules and transfer genes of interest locally [327, mour-penetrating nanoparticles, these nanodelivery systems open the
328]. possibility of delivering therapeutic agents into tumour parenchyma.
The application of controlled hydrodynamic pressure in capillaries is Apart from using small molecule and biologics individually in targeting
known as hydrodynamic delivery, and this method was incepted in the aerobic glycolysis, combination therapy with small molecules and
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 99

nucleic acids is another potential approach that can be explored. Combi- [25] R.L. Elstrom, D.E. Bauer, M. Buzzai, R. Karnauskas, M.H. Harris, D.R. Plas, H. Zhuang,
R.M. Cinalli, A. Alavi, C.M. Rudin, C.B. Thompson, Akt stimulates aerobic glycolysis
nation delivery of therapeutics using nanoparticle-based delivery in cancer cells, Cancer Res. 64 (2004) 3892–3899.
systems has been successfully reported to achieve synergistic effects [26] A. Csibi, J. Blenis, Appetite for destruction: the inhibition of glycolysis as a therapy
of therapeutic agents especially in multidrug-resistant cancer. for tuberous sclerosis complex-related tumors, BMC Biol. 9 (2011) 69.
[27] K. Duvel, J.L. Yecies, S. Menon, P. Raman, A.I. Lipovsky, A.L. Souza, E. Triantafellow,
Q. Ma, R. Gorski, S. Cleaver, M.G. Vander Heiden, J.P. MacKeigan, P.M. Finan, C.B.
Conflict of interest Clish, L.O. Murphy, B.D. Manning, Activation of a metabolic gene regulatory net-
work downstream of mTOR complex 1, Mol. Cell 39 (2010) 171–183.
[28] X. Jiang, H. Kenerson, L. Aicher, R. Miyaoka, J. Eary, J. Bissler, R.S. Yeung, The tuber-
There is no conflict of interest. ous sclerosis complex regulates trafficking of glucose transporters and glucose up-
take, Am. J. Pathol. 172 (2008) 1748–1756.
[29] D. Palmieri, D. Fitzgerald, S.M. Shreeve, E. Hua, J.L. Bronder, R.J. Weil, S. Davis, A.M.
Acknowledgements
Stark, M.J. Merino, R. Kurek, H.M. Mehdorn, G. Davis, S.M. Steinberg, P.S. Meltzer, K.
Aldape, P.S. Steeg, Analyses of resected human brain metastases of breast cancer
This work was funded by the Breakthrough Cancer Research, reveal the association between up-regulation of hexokinase 2 and poor prognosis,
Glenlee, Western Road, Cork, Ireland. Mol. Cancer Res. 7 (2009) 1438–1445.
[30] S.Y. Peng, P.L. Lai, H.W. Pan, L.P. Hsiao, H.C. Hsu, Aberrant expression of the glyco-
lytic enzymes aldolase B and type II hexokinase in hepatocellular carcinoma are
References predictive markers for advanced stage, early recurrence and poor prognosis,
Oncol. Rep. 19 (2008) 1045–1053.
[1] N.R. Pace, The universal nature of biochemistry, Proc. Natl. Acad. Sci. U. S. A. 98 [31] J.G. Pastorino, N. Shulga, J.B. Hoek, Mitochondrial binding of hexokinase II in-
(2001) 805–808. hibits Bax-induced cytochrome c release and apoptosis, J. Biol. Chem. 277
[2] O. Ebenhoh, R. Heinrich, Evolutionary optimization of metabolic pathways. Theo- (2002) 7610–7618.
retical reconstruction of the stoichiometry of ATP and NADH producing systems, [32] S. Rodriguez-Enriquez, A. Marin-Hernandez, J.C. Gallardo-Perez, R. Moreno-
Bull. Math. Biol. 63 (2001) 21–55. Sanchez, Kinetics of transport and phosphorylation of glucose in cancer cells, J.
[3] E. Melendez-Hevia, T.G. Waddell, M. Cascante, The puzzle of the Krebs citric acid Cell. Physiol. 221 (2009) 552–559.
cycle: assembling the pieces of chemically feasible reactions, and opportunism in [33] S.K. Bhattacharya, Inhibition of liver hexokinase by dehydroascorbic acid and allox-
the design of metabolic pathways during evolution, J. Mol. Evol. 43 (1996) an, Nature 184 (Suppl. 21) (1959) 1638–1640.
293–303. [34] D. Del Bufalo, A. Biroccio, S. Soddu, N. Laudonio, C. D'Angelo, A. Sacchi, G. Zupi,
[4] M.A. Keller, A.V. Turchyn, M. Ralser, Non-enzymatic glycolysis and pentose phos- Lonidamine induces apoptosis in drug-resistant cells independently of the p53
phate pathway-like reactions in a plausible Archean ocean, Mol. Syst. Biol. 10 gene, J. Clin. Invest. 98 (1996) 1165–1173.
(2014) 725. [35] Y.C. Li, K.P. Fung, T.T. Kwok, C.Y. Lee, Y.K. Suen, S.K. Kong, Mitochondrial targeting
[5] K.O. Alfarouk, D. Verduzco, C. Rauch, A.K. Muddathir, H.H. Adil, G.O. Elhassan, M.E. drug lonidamine triggered apoptosis in doxorubicin-resistant HepG2 cells, Life Sci.
Ibrahim, J. David Polo Orozco, R.A. Cardone, S.J. Reshkin, S. Harguindey, Glycolysis, 71 (2002) 2729–2740.
tumor metabolism, cancer growth and dissemination. A new pH-based [36] L. Ravagnan, I. Marzo, P. Costantini, S.A. Susin, N. Zamzami, P.X. Petit, F. Hirsch, M.
etiopathogenic perspective and therapeutic approach to an old cancer question, Goulbern, M.F. Poupon, L. Miccoli, Z. Xie, J.C. Reed, G. Kroemer, Lonidamine triggers
Oncoscience 1 (2014) 777–802. apoptosis via a direct, Bcl-2-inhibited effect on the mitochondrial permeability
[6] K.O. Alfarouk, A.K. Muddathir, M.E. Shayoub, Tumor acidity as evolutionary spite, transition pore, Oncogene 18 (1999) 2537–2546.
Cancer 3 (2011) 408–414. [37] M.K. Brawer, Lonidamine: basic science and rationale for treatment of prostatic
[7] R.A. Gatenby, R.J. Gillies, Why do cancers have high aerobic glycolysis? Nat. Rev. proliferative disorders, Rev. Urol. 7 (Suppl. 7) (2005) S21–S26.
Cancer 4 (2004) 891–899. [38] H. Pelicano, D.S. Martin, R.H. Xu, P. Huang, Glycolysis inhibition for anticancer
[8] J.W. Kim, C.V. Dang, Cancer's molecular sweet tooth and the Warburg effect, Cancer treatment, Oncogene 25 (2006) 4633–4646.
Res. 66 (2006) 8927–8930. [39] R.L. Aft, F.W. Zhang, D. Gius, Evaluation of 2-deoxy-D-glucose as a chemotherapeu-
[9] O. Warburg, On the origin of cancer cells, Science (New York, N.Y.) 123 (1956) tic agent: mechanism of cell death, Br. J. Cancer 87 (2002) 805–812.
309–314. [40] P. Dell'Antone, Targets of 3-bromopyruvate, a new, energy depleting, anticancer
[10] J.S. Bertram, The molecular biology of cancer, Mol. Asp. Med. 21 (2000) 167–223. agent, Med. Chem. (Shariqah (United Arab Emirates)) 5 (2009) 491–496.
[11] D. Grander, How do mutated oncogenes and tumor suppressor genes cause can- [41] X. Liu, A.K. Gupta, P.M. Corry, Y.J. Lee, Hypoglycemia-induced c-Jun phosphoryla-
cer? Med. Oncol. (Northwood, Lond., Engl.) 15 (1998) 20–26. tion is mediated by c-Jun N-terminal kinase 1 and Lyn kinase in drug-
[12] M.S. Ullah, A.J. Davies, A.P. Halestrap, The plasma membrane lactate transporter resistant human breast carcinoma MCF-7/ADR cells, J. Biol. Chem. 272
MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent (1997) 11690–11693.
mechanism, J. Biol. Chem. 281 (2006) 9030–9037. [42] D.R. Spitz, J.E. Sim, L.A. Ridnour, S.S. Galoforo, Y.J. Lee, Glucose deprivation-induced
[13] Y. Hu, W. Lu, G. Chen, P. Wang, Z. Chen, Y. Zhou, M. Ogasawara, D. Trachootham, L. oxidative stress in human tumor cells. A fundamental defect in metabolism? Ann.
Feng, H. Pelicano, P.J. Chiao, M.J. Keating, G. Garcia-Manero, P. Huang, K-rasG12V N. Y. Acad. Sci. 899 (2000) 349–362.
transformation leads to mitochondrial dysfunction and a metabolic switch from [43] K. Nomura, H. Imai, T. Koumura, M. Arai, Y. Nakagawa, Mitochondrial phospholipid
oxidative phosphorylation to glycolysis, Cell Res. 22 (2012) 399–412. hydroperoxide glutathione peroxidase suppresses apoptosis mediated by a mito-
[14] W. Lu, Y. Hu, G. Chen, Z. Chen, H. Zhang, F. Wang, L. Feng, H. Pelicano, H. Wang, M.J. chondrial death pathway, J. Biol. Chem. 274 (1999) 29294–29302.
Keating, J. Liu, W. McKeehan, H. Wang, Y. Luo, P. Huang, Novel role of NOX in [44] O. Kaplan, G. Navon, R.C. Lyon, P.J. Faustino, E.J. Straka, J.S. Cohen, Effects of 2-
supporting aerobic glycolysis in cancer cells with mitochondrial dysfunction and deoxyglucose on drug-sensitive and drug-resistant human breast cancer cells: tox-
as a potential target for cancer therapy, PLoS Biol. 10 (2012), e1001326. icity and magnetic resonance spectroscopy studies of metabolism, Cancer Res. 50
[15] D.C. Wallace, Mitochondria and cancer, Nat. Rev. Cancer 12 (2012) 685–698. (1990) 544–551.
[16] A.C. de Souza, G.Z. Justo, D.R. de Araujo, A.D. Cavagis, Defining the molecular basis [45] B.K. Mohanti, G.K. Rath, N. Anantha, V. Kannan, B.S. Das, B.A. Chandramouli, A.K.
of tumor metabolism: a continuing challenge since Warburg's discovery, Cell. Banerjee, S. Das, A. Jena, R. Ravichandran, U.P. Sahi, R. Kumar, N. Kapoor, V.K.
Physiol. Biochem. 28 (2011) 771–792. Kalia, B.S. Dwarakanath, V. Jain, Improving cancer radiotherapy with 2-deoxy-D-
[17] J.W. Locasale, L.C. Cantley, Altered metabolism in cancer, BMC Biol. 8 (2010) 88. glucose: phase I/II clinical trials on human cerebral gliomas, Int. J. Radiat. Oncol.
[18] T. Pfeiffer, S. Schuster, S. Bonhoeffer, Cooperation and competition in the evolution Biol. Phys. 35 (1996) 103–111.
of ATP-producing pathways, Science (New York, N.Y.) 292 (2001) 504–507. [46] D. Singh, A.K. Banerji, B.S. Dwarakanath, R.P. Tripathi, J.P. Gupta, T.L. Mathew, T.
[19] R.J. Deberardinis, N. Sayed, D. Ditsworth, C.B. Thompson, Brick by brick: metabo- Ravindranath, V. Jain, Optimizing cancer radiotherapy with 2-deoxy-D-glucose
lism and tumor cell growth, Curr. Opin. Genet. Dev. 18 (2008) 54–61. dose escalation studies in patients with glioblastoma multiforme, Strahlenther.
[20] D.S. Backos, C.C. Franklin, P. Reigan, The role of glutathione in brain tumor drug re- Onkol. 181 (2005) 507–514 (… [et al]).
sistance, Biochem. Pharmacol. 83 (2012) 1005–1012. [47] N.K. Venkataramanaa, P.K. Venkatesh, B.S. Dwarakanath, S. Vani, Protective ef-
[21] N. Traverso, R. Ricciarelli, M. Nitti, B. Marengo, A.L. Furfaro, M.A. Pronzato, U.M. fect on normal brain tissue during a combinational therapy of 2-deoxy- D -
Marinari, C. Domenicotti, Role of glutathione in cancer progression and glucose and hypofractionated irradiation in malignant gliomas, Asian J.
chemoresistance, Oxidative Med. Cell. Longev. 2013 (2013) 972913. Neurosurg. 8 (2013) 9–14.
[22] F. Zhao, A. Mancuso, T.V. Bui, X. Tong, J.J. Gruber, C.R. Swider, P.V. Sanchez, J.J. Lum, [48] D. Zhong, L. Xiong, T. Liu, X. Liu, X. Liu, J. Chen, S.Y. Sun, F.R. Khuri, Y. Zong, Q. Zhou,
N. Sayed, J.V. Melo, A.E. Perl, M. Carroll, S.W. Tuttle, C.B. Thompson, Imatinib resis- W. Zhou, The glycolytic inhibitor 2-deoxyglucose activates multiple prosurvival
tance associated with BCR-ABL upregulation is dependent on HIF-1alpha-induced pathways through IGF1R, J. Biol. Chem. 284 (2009) 23225–23233.
metabolic reprograming, Oncogene 29 (2010) 2962–2972. [49] J.C. Maher, M. Wangpaichitr, N. Savaraj, M. Kurtoglu, T.J. Lampidis, Hypoxia-
[23] F. Monteleone, R. Rosa, M. Vitale, C. D'Ambrosio, M. Succoio, L. Formisano, L. Nappi, inducible factor-1 confers resistance to the glycolytic inhibitor 2-deoxy-D-glucose,
M.F. Romano, A. Scaloni, G. Tortora, R. Bianco, N. Zambrano, Increased anaerobic Mol. Cancer Ther. 6 (2007) 732–741.
metabolism is a distinctive signature in a colorectal cancer cellular model of resis- [50] B. Dwarakanath, V. Jain, Targeting glucose metabolism with 2-deoxy-D-glucose for
tance to antiepidermal growth factor receptor antibody, Proteomics 13 (2013) improving cancer therapy, Future Oncol. (Lond. Engl.) 5 (2009) 581–585.
866–877. [51] G. Maschek, N. Savaraj, W. Priebe, P. Braunschweiger, K. Hamilton, G.F. Tidmarsh,
[24] S. Rimpi, J.A. Nilsson, Metabolic enzymes regulated by the Myc oncogene are pos- L.R. De Young, T.J. Lampidis, 2-Deoxy-D-glucose increases the efficacy of
sible targets for chemotherapy or chemoprevention, Biochem. Soc. Trans. 35 adriamycin and paclitaxel in human osteosarcoma and non-small cell lung cancers
(2007) 305–310. in vivo, Cancer Res. 64 (2004) 31–34.
100 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

[52] M. Erecinska, J. Deas, I.A. Silver, The effect of pH on glycolysis and phosphofructo- induction of oxidative stress mediated mitochondrial pathway apoptosis, Breast
kinase activity in cultured cells and synaptosomes, J. Neurochem. 65 (1995) Cancer Res. Treat. 131 (2012) 791–800.
2765–2772. [78] M. Zhou, Y. Zhao, Y. Ding, H. Liu, Z. Liu, O. Fodstad, A.I. Riker, S. Kamarajugadda, J.
[53] B. Trivedi, W.H. Danforth, Effect of pH on the kinetics of frog muscle phosphofruc- Lu, L.B. Owen, S.P. Ledoux, M. Tan, Warburg effect in chemosensitivity: targeting
tokinase, J. Biol. Chem. 241 (1966) 4110–4112. lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol, Mol.
[54] S. Vora, J.P. Halper, D.M. Knowles, Alterations in the activity and isozymic profile of Cancer 9 (2010) 33.
human phosphofructokinase during malignant transformation in vivo and in vitro: [79] K. Fischer, P. Hoffmann, S. Voelkl, N. Meidenbauer, J. Ammer, M. Edinger, E.
transformation- and progression-linked discriminants of malignancy, Cancer Res. Gottfried, S. Schwarz, G. Rothe, S. Hoves, K. Renner, B. Timischl, A. Mackensen, L.
45 (1985) 2993–3001. Kunz-Schughart, R. Andreesen, S.W. Krause, M. Kreutz, Inhibitory effect of tumor
[55] T. Atsumi, J. Chesney, C. Metz, L. Leng, S. Donnelly, Z. Makita, R. Mitchell, R. Bucala, cell-derived lactic acid on human T cells, Blood 109 (2007) 3812–3819.
High expression of inducible 6-phosphofructo-2-kinase/fructose-2,6- [80] A.N. Mendler, B. Hu, P.U. Prinz, M. Kreutz, E. Gottfried, E. Noessner, Tumor lactic ac-
bisphosphatase (iPFK-2; PFKFB3) in human cancers, Cancer Res. 62 (2002) idosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation, Int. J.
5881–5887. Cancer 131 (2012) 633–640.
[56] B. Clem, S. Telang, A. Clem, A. Yalcin, J. Meier, A. Simmons, M.A. Rasku, S. [81] E. Gottfried, L.A. Kunz-Schughart, S. Ebner, W. Mueller-Klieser, S. Hoves, R.
Arumugam, W.L. Dean, J. Eaton, A. Lane, J.O. Trent, J. Chesney, Small- Andreesen, A. Mackensen, M. Kreutz, Tumor-derived lactic acid modulates den-
molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glyco- dritic cell activation and antigen expression, Blood 107 (2006) 2013–2021.
lytic flux and tumor growth, Mol. Cancer Ther. 7 (2008) 110–120. [82] B. Fischer, B. Muller, K.G. Fischer, N. Baur, W. Kreutz, Acidic pH inhibits non-
[57] S.B. Vuyyuri, J. Rinkinen, E. Worden, H. Shim, S. Lee, K.R. Davis, Ascorbic acid and a MHC-restricted killer cell functions, Clin. Immunol. (Orlando, Fla.) 96 (2000)
cytostatic inhibitor of glycolysis synergistically induce apoptosis in non-small cell 252–263.
lung cancer cells, PLoS One 8 (2013), e67081. [83] H. Shime, M. Yabu, T. Akazawa, K. Kodama, M. Matsumoto, T. Seya, N. Inoue,
[58] M.B. Calvo, A. Figueroa, E.G. Pulido, R.G. Campelo, L.A. Aparicio, Potential role of Tumor-secreted lactic acid promotes IL-23/IL-17 proinflammatory pathway, J.
sugar transporters in cancer and their relationship with anticancer therapy, Int. J. Immunol. (Baltim. Md.: 1950) 180 (2008) 7175–7183.
Endocrinol. (2010). [84] S. Yeluri, B. Madhok, K.R. Prasad, P. Quirke, D.G. Jayne, Cancer's craving for sugar:
[59] S. Brophy, K.M. Sheehan, D.A. McNamara, J. Deasy, D.J. Bouchier-Hayes, E.W. Kay, an opportunity for clinical exploitation, J. Cancer Res. Clin. Oncol. 135 (2009)
GLUT-1 expression and response to chemoradiotherapy in rectal cancer, Int. J. Can- 867–877.
cer 125 (2009) 2778–2782. [85] H.R. Christofk, M.G. Vander Heiden, M.H. Harris, A. Ramanathan, R.E. Gerszten, R.
[60] S. Rastogi, S. Banerjee, S. Chellappan, G.R. Simon, Glut-1 antibodies induce growth Wei, M.D. Fleming, S.L. Schreiber, L.C. Cantley, The M2 splice isoform of pyruvate
arrest and apoptosis in human cancer cell lines, Cancer Lett. 257 (2007) 244–251. kinase is important for cancer metabolism and tumour growth, Nature 452
[61] A. Krzeslak, K. Wojcik-Krowiranda, E. Forma, P. Jozwiak, H. Romanowicz, A. (2008) 230–233.
Bienkiewicz, M. Brys, Expression of GLUT1 and GLUT3 glucose transporters in en- [86] S. Mazurek, C.B. Boschek, F. Hugo, E. Eigenbrodt, Pyruvate kinase type M2 and its
dometrial and breast cancers, Pathol. Oncol. Res. 18 (2012) 721–728. role in tumor growth and spreading, Semin. Cancer Biol. 15 (2005) 300–308.
[62] M.L. Macheda, S. Rogers, J.D. Best, Molecular and cellular regulation of glucose [87] M. Fang, Z. Shen, S. Huang, L. Zhao, S. Chen, T.W. Mak, X. Wang, The ER UDPase
transporter (GLUT) proteins in cancer, J. Cell. Physiol. 202 (2005) 654–662. ENTPD5 promotes protein N-glycosylation, the Warburg effect, and proliferation
[63] Y. Noguchi, A. Saito, Y. Miyagi, S. Yamanaka, D. Marat, C. Doi, T. Yoshikawa, A. in the PTEN pathway, Cell 143 (2010) 711–724.
Tsuburaya, T. Ito, S. Satoh, Suppression of facilitative glucose transporter 1 mRNA [88] S. Marshall, V. Bacote, R.R. Traxinger, Discovery of a metabolic pathway mediating
can suppress tumor growth, Cancer Lett. 154 (2000) 175–182. glucose-induced desensitization of the glucose transport system. Role of
[64] T.W. Flaig, M. Glode, D. Gustafson, A. van Bokhoven, Y. Tao, S. Wilson, L.J. Su, Y. Li, G. hexosamine biosynthesis in the induction of insulin resistance, J. Biol. Chem. 266
Harrison, R. Agarwal, E.D. Crawford, M.S. Lucia, M. Pollak, A study of high-dose oral (1991) 4706–4712.
silybin-phytosome followed by prostatectomy in patients with localized prostate [89] M.G. Vander Heiden, L.C. Cantley, C.B. Thompson, Understanding the Warburg ef-
cancer, Prostate 70 (2010) 848–855. fect: the metabolic requirements of cell proliferation, Science (New York, N.Y.)
[65] D.A. Chan, P.D. Sutphin, P. Nguyen, S. Turcotte, E.W. Lai, A. Banh, G.E. Reynolds, J.T. 324 (2009) 1029–1033.
Chi, J. Wu, D.E. Solow-Cordero, M. Bonnet, J.U. Flanagan, D.M. Bouley, E.E. Graves, [90] J.B. Wang, J.W. Erickson, R. Fuji, S. Ramachandran, P. Gao, R. Dinavahi, K.F. Wilson,
W.A. Denny, M.P. Hay, A.J. Giaccia, Targeting GLUT1 and the Warburg effect in A.L. Ambrosio, S.M. Dias, C.V. Dang, R.A. Cerione, Targeting mitochondrial gluta-
renal cell carcinoma by chemical synthetic lethality, Sci. Transl. Med. 3 (2011), minase activity inhibits oncogenic transformation, Cancer Cell 18 (2010) 207–219.
94ra70. [91] P.E. Porporato, S. Dhup, R.K. Dadhich, T. Copetti, P. Sonveaux, Anticancer targets in
[66] X. Jiang, H. Xin, Q. Ren, J. Gu, L. Zhu, F. Du, C. Feng, Y. Xie, X. Sha, X. Fang, Nanopar- the glycolytic metabolism of tumors: a comprehensive review, Front. Pharmacol. 2
ticles of 2-deoxy-D-glucose functionalized poly(ethylene glycol)-co- (2011) 49.
poly(trimethylene carbonate) for dual-targeted drug delivery in glioma treatment, [92] Z. Zhang, Q. Liu, Y. Che, X. Yuan, L. Dai, B. Zeng, G. Jiao, Y. Zhang, X. Wu, Y. Yu, Y.
Biomaterials 35 (2014) 518–529. Zhang, R. Yang, Antigen presentation by dendritic cells in tumors is disrupted by
[67] S. Bonnet, S.L. Archer, J. Allalunis-Turner, A. Haromy, C. Beaulieu, R. Thompson, C.T. altered metabolism that involves pyruvate kinase M2 and its interaction with
Lee, G.D. Lopaschuk, L. Puttagunta, S. Bonnet, G. Harry, K. Hashimoto, C.J. Porter, SOCS3, Cancer Res. 70 (2010) 89–98.
M.A. Andrade, B. Thebaud, E.D. Michelakis, A mitochondria-K+ channel axis is sup- [93] K.A. Frauwirth, J.L. Riley, M.H. Harris, R.V. Parry, J.C. Rathmell, D.R. Plas, R.L. Elstrom,
pressed in cancer and its normalization promotes apoptosis and inhibits cancer C.H. June, C.B. Thompson, The CD28 signaling pathway regulates glucose metabo-
growth, Cancer Cell 11 (2007) 37–51. lism, Immunity 16 (2002) 769–777.
[68] R.C. Sun, M. Fadia, J.E. Dahlstrom, C.R. Parish, P.G. Board, A.C. Blackburn, Reversal of [94] G. Matarese, A. La Cava, The intricate interface between immune system and me-
the glycolytic phenotype by dichloroacetate inhibits metastatic breast cancer cell tabolism, Trends Immunol. 25 (2004) 193–200.
growth in vitro and in vivo, Breast Cancer Res. Treat. 120 (2010) 253–260. [95] M.H. Oak, H. Cheong, K.M. Kim, Activation of Fc epsilon RI inhibits the pyruvate ki-
[69] B.M. Madhok, S. Yeluri, S.L. Perry, T.A. Hughes, D.G. Jayne, Dichloroacetate induces nase through direct interaction with the gamma-chain, Int. Arch. Allergy Immunol.
apoptosis and cell-cycle arrest in colorectal cancer cells, Br. J. Cancer 102 (2010) 119 (1999) 95–100.
1746–1752. [96] G. Hatzivassiliou, F. Zhao, D.E. Bauer, C. Andreadis, A.N. Shaw, D. Dhanak, S.R.
[70] W. Cao, S. Yacoub, K.T. Shiverick, K. Namiki, Y. Sakai, S. Porvasnik, C. Urbanek, C.J. Hingorani, D.A. Tuveson, C.B. Thompson, ATP citrate lyase inhibition can suppress
Rosser, Dichloroacetate (DCA) sensitizes both wild-type and over expressing Bcl- tumor cell growth, Cancer Cell 8 (2005) 311–321.
2 prostate cancer cells in vitro to radiation, Prostate 68 (2008) 1223–1231. [97] E.S. Pizer, J. Thupari, W.F. Han, M.L. Pinn, F.J. Chrest, G.L. Frehywot, C.A. Townsend,
[71] J.Y. Wong, G.S. Huggins, M. Debidda, N.C. Munshi, I. De Vivo, Dichloroacetate in- F.P. Kuhajda, Malonyl-coenzyme-A is a potential mediator of cytotoxicity induced
duces apoptosis in endometrial cancer cells, Gynecol. Oncol. 109 (2008) 394–402. by fatty-acid synthase inhibition in human breast cancer cells and xenografts, Can-
[72] F. Tas, F. Aykan, S. Alici, E. Kaytan, A. Aydiner, E. Topuz, Prognostic factors in pan- cer Res. 60 (2000) 213–218.
creatic carcinoma: serum LDH levels predict survival in metastatic disease, Am. J. [98] J.F. Cheng, M. Chen, B. Liu, Z. Hou, T. Arrhenius, A.M. Nadzan, Design and synthesis
Clin. Oncol. 24 (2001) 547–550. of heterocyclic malonyl-CoA decarboxylase inhibitors, Bioorg. Med. Chem. Lett. 16
[73] A.J. Armstrong, D.J. George, S. Halabi, Serum lactate dehydrogenase predicts for (2006) 695–700.
overall survival benefit in patients with metastatic renal cell carcinoma treated [99] S. Krauss, M.D. Brand, F. Buttgereit, Signaling takes a breath—new quantitative per-
with inhibition of mammalian target of rapamycin, J. Clin. Oncol. Off. J. Am. Soc. spectives on bioenergetics and signal transduction, Immunity 15 (2001) 497–502.
Clin. Oncol. 30 (2012) 3402–3407. [100] F. Buttgereit, G.R. Burmester, M.D. Brand, Bioenergetics of immune functions: fun-
[74] Y. Jin, X. Ye, L. Shao, B.C. Lin, C.X. He, B.B. Zhang, Y.P. Zhang, Serum lactic dehydro- damental and therapeutic aspects, Immunol. Today 21 (2000) 192–199.
genase strongly predicts survival in metastatic nasopharyngeal carcinoma treated [101] G.R. Crabtree, N.A. Clipstone, Signal transmission between the plasma membrane
with palliative chemotherapy, Eur. J. Cancer (Oxf. Engl.: 1990) 49 (2013) and nucleus of T lymphocytes, Annu. Rev. Biochem. 63 (1994) 1045–1083.
1619–1626. [102] C.A. Doughty, B.F. Bleiman, D.J. Wagner, F.J. Dufort, J.M. Mataraza, M.F. Roberts, T.C.
[75] M.I. Koukourakis, A. Giatromanolaki, E. Sivridis, G. Bougioukas, V. Didilis, K.C. Chiles, Antigen receptor-mediated changes in glucose metabolism in B lympho-
Gatter, A.L. Harris, Lactate dehydrogenase-5 (LDH-5) overexpression in non- cytes: role of phosphatidylinositol 3-kinase signaling in the glycolytic control of
small-cell lung cancer tissues is linked to tumour hypoxia, angiogenic factor pro- growth, Blood 107 (2006) 4458–4465.
duction and poor prognosis, Br. J. Cancer 89 (2003) 877–885. [103] C.J. Fox, P.S. Hammerman, C.B. Thompson, Fuel feeds function: energy metabolism
[76] A. Le, C.R. Cooper, A.M. Gouw, R. Dinavahi, A. Maitra, L.M. Deck, R.E. Royer, D.L. and the T-cell response, Nat. Rev. Immunol. 5 (2005) 844–852.
Vander Jagt, G.L. Semenza, C.V. Dang, Inhibition of lactate dehydrogenase A induces [104] A. Ouiddir, C. Planes, I. Fernandes, A. VanHesse, C. Clerici, Hypoxia upregulates ac-
oxidative stress and inhibits tumor progression, Proc. Natl. Acad. Sci. U. S. A. 107 tivity and expression of the glucose transporter GLUT1 in alveolar epithelial cells,
(2010) 2037–2042. Am. J. Respir. Cell Mol. Biol. 21 (1999) 710–718.
[77] Z.Y. Wang, T.Y. Loo, J.G. Shen, N. Wang, D.M. Wang, D.P. Yang, S.L. Mo, X.Y. Guan, [105] G. Schuler, R.M. Steinman, Murine epidermal Langerhans cells mature into potent
J.P. Chen, LDH-A silencing suppresses breast cancer tumorigenicity through immunostimulatory dendritic cells in vitro, J. Exp. Med. 161 (1985) 526–546.
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 101

[106] G.D. Winter, Oxygen and epidermal wound healing, Adv. Exp. Med. Biol. 94 (1977) [137] K. Araki, A.P. Turner, V.O. Shaffer, S. Gangappa, S.A. Keller, M.F. Bachmann, C.P.
673–678. Larsen, R. Ahmed, mTOR regulates memory CD8 T-cell differentiation, Nature
[107] M. Sitkovsky, D. Lukashev, Regulation of immune cells by local-tissue oxygen ten- 460 (2009) 108–112.
sion: HIF1 alpha and adenosine receptors, Nat. Rev. Immunol. 5 (2005) 712–721. [138] M. Hockel, P. Vaupel, Tumor hypoxia: definitions and current clinical, biologic, and
[108] U.H. von Andrian, C.R. Mackay, T-cell function and migration. Two sides of the molecular aspects, J. Natl. Cancer Inst. 93 (2001) 266–276.
same coin, N. Engl. J. Med. 343 (2000) 1020–1034. [139] J.C. Rathmell, M.G. Vander Heiden, M.H. Harris, K.A. Frauwirth, C.B. Thompson, In
[109] F. Arnold, D. West, S. Kumar, Wound healing: the effect of macrophage and tumour the absence of extrinsic signals, nutrient utilization by lymphocytes is insufficient
derived angiogenesis factors on skin graft vascularization, Br. J. Exp. Pathol. 68 to maintain either cell size or viability, Mol. Cell 6 (2000) 683–692.
(1987) 569–574. [140] G.J. van der Windt, B. Everts, C.H. Chang, J.D. Curtis, T.C. Freitas, E. Amiel, E.J. Pearce,
[110] G. Helmlinger, F. Yuan, M. Dellian, R.K. Jain, Interstitial pH and pO2 gradients in E.L. Pearce, Mitochondrial respiratory capacity is a critical regulator of CD8+ T cell
solid tumors in vivo: high-resolution measurements reveal a lack of correlation, memory development, Immunity 36 (2012) 68–78.
Nat. Med. 3 (1997) 177–182. [141] S.W. Choi, A.A. Gerencser, D.G. Nicholls, Bioenergetic analysis of isolated
[111] H.P. Simmen, H. Battaglia, P. Giovanoli, J. Blaser, Analysis of pH, pO2 and pCO2 in cerebrocortical nerve terminals on a microgram scale: spare respiratory capacity
drainage fluid allows for rapid detection of infectious complications during the and stochastic mitochondrial failure, J. Neurochem. 109 (2009) 1179–1191.
follow-up period after abdominal surgery, Infection 22 (1994) 386–389. [142] D.G. Nicholls, Spare respiratory capacity, oxidative stress and excitotoxicity,
[112] D. Masopust, V. Vezys, A.L. Marzo, L. Lefrancois, Preferential localization of effector Biochem. Soc. Trans. 37 (2009) 1385–1388.
memory cells in nonlymphoid tissue, Science (New York, N.Y.) 291 (2001) [143] N.L. Alves, I.A. Derks, E. Berk, R. Spijker, R.A. van Lier, E. Eldering, The Noxa/Mcl-1
2413–2417. axis regulates susceptibility to apoptosis under glucose limitation in dividing T
[113] E. Roman, E. Miller, A. Harmsen, J. Wiley, U.H. Von Andrian, G. Huston, S.L. Swain, cells, Immunity 24 (2006) 703–716.
CD4 effector T cell subsets in the response to influenza: heterogeneity, migration, [144] M. Yamashita, M. Kuwahara, A. Suzuki, K. Hirahara, R. Shinnaksu, H. Hosokawa, A.
and function, J. Exp. Med. 196 (2002) 957–968. Hasegawa, S. Motohashi, A. Iwama, T. Nakayama, Bmi1 regulates memory CD4 T
[114] N. Borregaard, T. Herlin, Energy metabolism of human neutrophils during phago- cell survival via repression of the Noxa gene, J. Exp. Med. 205 (2008) 1109–1120.
cytosis, J. Clin. Invest. 70 (1982) 550–557. [145] G. Bindea, B. Mlecnik, M. Tosolini, A. Kirilovsky, M. Waldner, Anna C. Obenauf, H.
[115] T. Cramer, Y. Yamanishi, B.E. Clausen, I. Forster, R. Pawlinski, N. Mackman, V.H. Angell, T. Fredriksen, L. Lafontaine, A. Berger, P. Bruneval, Wolf H. Fridman, C. Beck-
Haase, R. Jaenisch, M. Corr, V. Nizet, G.S. Firestein, H.P. Gerber, N. Ferrara, R.S. er, F. Pagès, Michael R. Speicher, Z. Trajanoski, J. Galon, Spatiotemporal dynamics of
Johnson, HIF-1alpha is essential for myeloid cell-mediated inflammation, Cell intratumoral immune cells reveal the immune landscape in human cancer, Immu-
112 (2003) 645–657. nity, 39 782-795.
[116] C.M. Krawczyk, T. Holowka, J. Sun, J. Blagih, E. Amiel, R.J. DeBerardinis, J.R. Cross, E. [146] S.K. Biswas, A. Mantovani, Macrophage plasticity and interaction with lymphocyte
Jung, C.B. Thompson, R.G. Jones, E.J. Pearce, Toll-like receptor-induced changes in subsets: cancer as a paradigm, Nat. Immunol. 11 (2010) 889–896.
glycolytic metabolism regulate dendritic cell activation, Blood 115 (2010) [147] D. Hanahan, Lisa M. Coussens, Accessories to the crime: functions of cells recruited
4742–4749. to the tumor microenvironment, Cancer Cell, 21 309-322.
[117] A. Lacy-Hulbert, K.J. Moore, Designer macrophages: oxidative metabolism fuels in- [148] Erika L. Pearce, Edward J. Pearce, Metabolic pathways in immune cell activation
flammation repair, Cell Metab. 4 (2006) 7–8. and quiescence, Immunity, 38 633-643.
[118] D. Vats, L. Mukundan, J.I. Odegaard, L. Zhang, K.L. Smith, C.R. Morel, R.A. Wagner, [149] F.A. Harding, J.G. McArthur, J.A. Gross, D.H. Raulet, J.P. Allison, CD28-mediated sig-
D.R. Greaves, P.J. Murray, A. Chawla, Oxidative metabolism and PGC-1beta attenu- nalling co-stimulates murine T cells and prevents induction of anergy in T-cell
ate macrophage-mediated inflammation, Cell Metab. 4 (2006) 13–24. clones, Nature 356 (1992) 607–609.
[119] D.A. Healy, R.W. Watson, P. Newsholme, Glucose, but not glutamine, protects [150] A.D. Wells, New insights into the molecular basis of T cell anergy: anergy factors,
against spontaneous and anti-Fas antibody-induced apoptosis in human neutro- avoidance sensors, and epigenetic imprinting, J. of Immunol. (Baltim. Md.: 1950)
phils, Clin. Sci. (Lond. Engl.: 1979) 103 (2002) 179–189. 182 (2009) 7331–7341.
[120] J.S. Savill, A.H. Wyllie, J.E. Henson, M.J. Walport, P.M. Henson, C. Haslett, Macro- [151] V.K. Kuchroo, M.P. Das, J.A. Brown, A.M. Ranger, S.S. Zamvil, R.A. Sobel, H.L. Weiner,
phage phagocytosis of aging neutrophils in inflammation. Programmed cell N. Nabavi, L.H. Glimcher, B7-1 and B7-2 costimulatory molecules activate differen-
death in the neutrophil leads to its recognition by macrophages, J. Clin. Invest. 83 tially the Th1/Th2 developmental pathways: application to autoimmune disease
(1989) 865–875. therapy, Cell 80 (1995) 707–718.
[121] E.L. Pearce, Metabolism in T cell activation and differentiation, Curr. Opin. [152] A.I. Sperling, J.A. Bluestone, The complexities of T-cell co-stimulation: CD28 and
Immunol. 22 (2010) 314–320. beyond, Immunol. Rev. 153 (1996) 155–182.
[122] D. Schmid, G.R. Burmester, R. Tripmacher, A. Kuhnke, F. Buttgereit, Bioenergetics of [153] R.A. Cairns, I.S. Harris, T.W. Mak, Regulation of cancer cell metabolism, Nat. Rev.
human peripheral blood mononuclear cell metabolism in quiescent, activated, and Cancer 11 (2011) 85–95.
glucocorticoid-treated states, Biosci. Rep. 20 (2000) 289–302. [154] Patrick S. Ward, Craig B. Thompson, Metabolic reprogramming: a cancer hallmark
[123] S.C. Land, P.W. Hochachka, Protein turnover during metabolic arrest in turtle hepa- even Warburg did not anticipate, Cancer Cell, 21 297-308.
tocytes: role and energy dependence of proteolysis, Am. J. Phys. 266 (1994) [155] J.C. Rathmell, R.L. Elstrom, R.M. Cinalli, C.B. Thompson, Activated Akt promotes in-
C1028–C1036. creased resting T cell size, CD28-independent T cell growth, and development of
[124] M.F. Princiotta, D. Finzi, S.B. Qian, J. Gibbs, S. Schuchmann, F. Buttgereit, J.R. autoimmunity and lymphoma, Eur. J. Immunol. 33 (2003) 2223–2232.
Bennink, J.W. Yewdell, Quantitating protein synthesis, degradation, and endoge- [156] K.A. Frauwirth, C.B. Thompson, Regulation of T lymphocyte metabolism, J.
nous antigen processing, Immunity 18 (2003) 343–354. Immunol. 172 (2004) 4661–4665.
[125] J.J. Lum, R.J. DeBerardinis, C.B. Thompson, Autophagy in metazoans: cell survival in [157] H.L. Wieman, J.A. Wofford, J.C. Rathmell, Cytokine stimulation promotes glucose
the land of plenty, Nat. Rev. Mol. Cell Biol. 6 (2005) 439–448. uptake via phosphatidylinositol-3 kinase/Akt regulation of Glut1 activity and traf-
[126] Z. Yang, D.J. Klionsky, Eaten alive: a history of macroautophagy, Nat. Cell Biol. 12 ficking, Mol. Biol. Cell 18 (2007) 1437–1446.
(2010) 814–822. [158] R. Grumont, P. Lock, M. Mollinari, F.M. Shannon, A. Moore, S. Gerondakis, The
[127] J.C. Rathmell, C.J. Fox, D.R. Plas, P.S. Hammerman, R.M. Cinalli, C.B. Thompson, Akt- mitogen-induced increase in T cell size involves PKC and NFAT activation of Rel/
directed glucose metabolism can prevent Bax conformation change and promote NF-kappaB-dependent c-myc expression, Immunity 21 (2004) 19–30.
growth factor-independent survival, Mol. Cell. Biol. 23 (2003) 7315–7328. [159] R. Wang, C.P. Dillon, L.Z. Shi, S. Milasta, R. Carter, D. Finkelstein, L.L. McCormick, P.
[128] N.J. Maciver, S.R. Jacobs, H.L. Wieman, J.A. Wofford, J.L. Coloff, J.C. Rathmell, Glucose Fitzgerald, H. Chi, J. Munger, D.R. Green, The transcription factor Myc controls meta-
metabolism in lymphocytes is a regulated process with significant effects on im- bolic reprogramming upon T lymphocyte activation, Immunity 35 (2011) 871–882.
mune cell function and survival, J. Leukoc. Biol. 84 (2008) 949–957. [160] J.A. Wofford, H.L. Wieman, S.R. Jacobs, Y. Zhao, J.C. Rathmell, IL-7 promotes Glut1
[129] H. Lu, R.A. Forbes, A. Verma, Hypoxia-inducible factor 1 activation by aerobic gly- trafficking and glucose uptake via STAT5-mediated activation of Akt to support
colysis implicates the Warburg effect in carcinogenesis, J. Biol. Chem. 277 (2002) T-cell survival, Blood 111 (2008) 2101–2111.
23111–23115. [161] S.R. Jacobs, C.E. Herman, N.J. Maciver, J.A. Wofford, H.L. Wieman, J.J. Hammen, J.C.
[130] F. Buttgereit, M.D. Brand, M. Muller, ConA induced changes in energy metabolism Rathmell, Glucose uptake is limiting in T cell activation and requires CD28-
of rat thymocytes, Biosci. Rep. 12 (1992) 381–386. mediated Akt-dependent and independent pathways, J. Immunol. 180 (2008)
[131] F.M. Marelli-Berg, H. Fu, C. Mauro, Molecular mechanisms of metabolic 4476–4486.
reprogramming in proliferating cells: implications for T-cell-mediated immunity, [162] D.A. Fruman, S.B. Snapper, C.M. Yballe, L. Davidson, J.Y. Yu, F.W. Alt, L.C. Cantley,
Immunology 136 (2012) 363–369. Impaired B cell development and proliferation in absence of phosphoinositide 3-
[132] F. Buttgereit, M.D. Brand, A hierarchy of ATP-consuming processes in mammalian kinase p85alpha, Science (New York, N.Y.) 283 (1999) 393–397.
cells, Biochem. J. 312 (Pt 1) (1995) 163–167. [163] G.M. Delgoffe, J.D. Powell, mTOR: taking cues from the immune microenviron-
[133] M.R. Duchen, Contributions of mitochondria to animal physiology: from homeo- ment, Immunology 127 (2009) 459–465.
static sensor to calcium signalling and cell death, J. Physiol. 516 (Pt 1) (1999) 1–17. [164] S.-C. Cheng, J. Quintin, R.A. Cramer, K.M. Shepardson, S. Saeed, V. Kumar, E.J.
[134] E. Gatza, D.R. Wahl, A.W. Opipari, T.B. Sundberg, P. Reddy, C. Liu, G.D. Glick, J.L. Ferrara, Giamarellos-Bourboulis, J.H.A. Martens, N.A. Rao, A. Aghajanirefah, G.R. Manjeri,
Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis Y. Li, D.C. Ifrim, R.J.W. Arts, B.M.J.W. van der Veer, P.M.T. Deen, C. Logie, L.A.
and arrests graft-versus-host disease, Sci. Transl. Med. 3 (2011) (67ra68). O'Neill, P. Willems, F.L. van de Veerdonk, J.W.M. van der Meer, A. Ng, L.A.B.
[135] D.R. Wahl, B. Petersen, R. Warner, B.C. Richardson, G.D. Glick, A.W. Opipari, Charac- Joosten, C. Wijmenga, H.G. Stunnenberg, R.J. Xavier, M.G. Netea, mTOR- and HIF-
terization of the metabolic phenotype of chronically activated lymphocytes, Lupus 1α-mediated aerobic glycolysis as metabolic basis for trained immunity, Science
19 (2010) 1492–1501. (New York, N.Y.) 345 (2014).
[136] R.D. Michalek, V.A. Gerriets, S.R. Jacobs, A.N. Macintyre, N.J. MacIver, E.F. Mason, [165] J.J. Howell, B.D. Manning, mTOR couples cellular nutrient sensing to organismal
S.A. Sullivan, A.G. Nichols, J.C. Rathmell, Cutting edge: distinct glycolytic and lipid metabolic homeostasis, Trends Endocrinol. Metab., 22 94-102.
oxidative metabolic programs are essential for effector and regulatory CD4+ T [166] C.M. Krawczyk, T. Holowka, J. Sun, J. Blagih, E. Amiel, R.J. DeBerardinis, J.R. Cross, E.
cell subsets, J. Immunol. (Baltim. Md.: 1950) 186 (2011) 3299–3303. Jung, C.B. Thompson, R.G. Jones, E.J. Pearce, Toll-like receptor-induced changes in
102 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

glycolytic metabolism regulate dendritic cell activation, Blood 115 (2010) [195] V.Z. Rocha, E.J. Folco, G. Sukhova, K. Shimizu, I. Gotsman, A.H. Vernon, P. Libby, In-
4742–4749. terferon-gamma, a Th1 cytokine, regulates fat inflammation: a role for adaptive
[167] G.J. Brunn, C.C. Hudson, A. Sekulic, J.M. Williams, H. Hosoi, P.J. Houghton, J.C. immunity in obesity, Circ. Res. 103 (2008) 467–476.
Lawrence Jr., R.T. Abraham, Phosphorylation of the translational repressor PHAS- [196] S. Winer, Y. Chan, G. Paltser, D. Truong, H. Tsui, J. Bahrami, R. Dorfman, Y. Wang, J.
I by the mammalian target of rapamycin, Science (New York, N.Y.) 277 (1997) Zielenski, F. Mastronardi, Y. Maezawa, D.J. Drucker, E. Engleman, D. Winer, H.M.
99–101. Dosch, Normalization of obesity-associated insulin resistance through immuno-
[168] S.G. Dann, G. Thomas, The amino acid sensitive TOR pathway from yeast to mam- therapy, Nat. Med. 15 (2009) 921–929.
mals, FEBS Lett. 580 (2006) 2821–2829. [197] T.F. Gajewski, Cancer immunotherapy, Mol. Oncol. 6 (2012) 242–250.
[169] J.J. Howell, B.D. Manning, mTOR couples cellular nutrient sensing to organismal [198] A. Mantovani, P. Romero, A.K. Palucka, F.M. Marincola, Tumour immunity: effector
metabolic homeostasis, Trends Endocrinol. Metab. 22 (2011) 94–102. response to tumour and role of the microenvironment, Lancet (Lond. Engl.) 371
[170] J.E. Kay, L. Kromwel, S.E. Doe, M. Denyer, Inhibition of T and B lymphocyte prolif- (2008) 771–783.
eration by rapamycin, Immunology 72 (1991) 544–549. [199] S.A. Quezada, K.S. Peggs, T.R. Simpson, J.P. Allison, Shifting the equilibrium in cancer
[171] N.F. Brown, M. Stefanovic-Racic, I.J. Sipula, G. Perdomo, The mammalian target of immunoediting: from tumor tolerance to eradication, Immunol. Rev. 241 (2011)
rapamycin regulates lipid metabolism in primary cultures of rat hepatocytes, 104–118.
Metab. Clin. Exp. 56 (2007) 1500–1507. [200] B. Bodey, Spontaneous regression of neoplasms: new possibilities for immunother-
[172] I.J. Sipula, N.F. Brown, G. Perdomo, Rapamycin-mediated inhibition of mammalian apy, Expert. Opin. Biol. Ther. 2 (2002) 459–476.
target of rapamycin in skeletal muscle cells reduces glucose utilization and in- [201] L.V. Kalialis, K.T. Drzewiecki, H. Klyver, Spontaneous regression of metastases from
creases fatty acid oxidation, Metab. Clin. Exp. 55 (2006) 1637–1644. melanoma: review of the literature, Melanoma Res. 19 (2009) 275–282.
[173] J.D. Powell, C.G. Lerner, R.H. Schwartz, Inhibition of cell cycle progression by [202] F. Saleh, W. Renno, I. Klepacek, G. Ibrahim, H. Dashti, S. Asfar, A. Behbehani,
rapamycin induces T cell clonal anergy even in the presence of costimulation, J. H. Al-Sayer, A. Dashti, Direct evidence on the immune-mediated
Immunol. (Baltim. Md.: 1950) 162 (1999) 2775–2784. spontaneous regression of human cancer: an incentive for pharmaceutical
[174] Y. Zheng, S.L. Collins, M.A. Lutz, A.N. Allen, T.P. Kole, P.E. Zarek, J.D. Powell, A role for companies to develop a novel anti-cancer vaccine, Curr. Pharm. Des. 11
mammalian target of rapamycin in regulating T cell activation versus anergy, J. (2005) 3531–3543.
Immunol. (Baltim. Md.: 1950) 178 (2007) 2163–2170. [203] P. Jaakkola, D.R. Mole, Y.M. Tian, M.I. Wilson, J. Gielbert, S.J. Gaskell, A. von
[175] G.M. Delgoffe, T.P. Kole, Y. Zheng, P.E. Zarek, K.L. Matthews, B. Xiao, P.F. Worley, S.C. Kriegsheim, H.F. Hebestreit, M. Mukherji, C.J. Schofield, P.H. Maxwell, C.W. Pugh,
Kozma, J.D. Powell, The mTOR kinase differentially regulates effector and regulato- P.J. Ratcliffe, Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation com-
ry T cell lineage commitment, Immunity 30 (2009) 832–844. plex by O2-regulated prolyl hydroxylation, Science (New York, N.Y.) 292 (2001)
[176] P.J. Siska, J.C. Rathmell, T cell metabolic fitness in antitumor immunity, Trends 468–472.
Immunol., 36 257-264. [204] J.M. Heddleston, Z. Li, J.D. Lathia, S. Bao, A.B. Hjelmeland, J.N. Rich, Hypoxia induc-
[177] D. Finlay, D. Cantrell, Phosphoinositide 3-kinase and the mammalian target of ible factors in cancer stem cells, Br. J. Cancer 102 (2010) 789–795.
rapamycin pathways control T cell migration, Ann. N. Y. Acad. Sci. 1183 (2010) [205] T. Hellwig-Burgel, K. Rutkowski, E. Metzen, J. Fandrey, W. Jelkmann, Interleukin-
149–157. 1beta and tumor necrosis factor-alpha stimulate DNA binding of hypoxia-
[178] R. Munk, P. Ghosh, M.C. Ghosh, T. Saito, M. Xu, A. Carter, F. Indig, D.D. Taub, D.L. inducible factor-1, Blood 94 (1999) 1561–1567.
Longo, Involvement of mTOR in CXCL12 mediated T cell signaling and migration, [206] D.E. Richard, E. Berra, E. Gothie, D. Roux, J. Pouyssegur, p42/p44 mitogen-activated
PLoS One 6 (2011), e24667. protein kinases phosphorylate hypoxia-inducible factor 1alpha (HIF-1alpha) and
[179] T.T. Murooka, R. Rahbar, L.C. Platanias, E.N. Fish, CCL5-mediated T-cell chemotaxis enhance the transcriptional activity of HIF-1, J. Biol. Chem. 274 (1999)
involves the initiation of mRNA translation through mTOR/4E-BP1, Blood 111 32631–32637.
(2008) 4892–4901. [207] R.A. Gatenby, R.J. Gillies, Glycolysis in cancer: a potential target for therapy, Int. J.
[180] I. Hashimoto, K. Koizumi, M. Tatematsu, T. Minami, S. Cho, N. Takeno, A. Biochem. Cell Biol. 39 (2007) 1358–1366.
Nakashima, H. Sakurai, S. Saito, K. Tsukada, I. Saiki, Blocking on the CXCR4/mTOR [208] C. Chen, N. Pore, A. Behrooz, F. Ismail-Beigi, A. Maity, Regulation of glut1 mRNA by
signalling pathway induces the anti-metastatic properties and autophagic cell hypoxia-inducible factor-1. Interaction between H-ras and hypoxia, J. Biol. Chem.
death in peritoneal disseminated gastric cancer cells, Eur. J. cancer (Oxf. Engl. : 276 (2001) 9519–9525.
1990) 44 (2008) 1022–1029. [209] J.W. Kim, I. Tchernyshyov, G.L. Semenza, C.V. Dang, HIF-1-mediated expression of
[181] J. Wang, Y. Lu, J. Wang, A.E. Koch, J. Zhang, R.S. Taichman, CXCR6 induces prostate pyruvate dehydrogenase kinase: a metabolic switch required for cellular adapta-
cancer progression by the AKT/mammalian target of rapamycin signaling pathway, tion to hypoxia, Cell Metab. 3 (2006) 177–185.
Cancer Res. 68 (2008) 10367–10376. [210] E. Sasabe, Y. Tatemoto, D. Li, T. Yamamoto, T. Osaki, Mechanism of HIF-1alpha-de-
[182] J.A. Sanchez-Alcazar, I. Hernandez, M.P. De la Torre, I. Garcia, E. Santiago, M.T. pendent suppression of hypoxia-induced apoptosis in squamous cell carcinoma
Munoz-Yague, J.A. Solis-Herruzo, Down-regulation of tumor necrosis factor recep- cells, Cancer Sci. 96 (2005) 394–402.
tors by blockade of mitochondrial respiration, J. Biol. Chem. 270 (1995) [211] G. Powis, L. Kirkpatrick, Hypoxia inducible factor-1alpha as a cancer drug target,
23944–23950. Mol. Cancer Ther. 3 (2004) 647–654.
[183] H. Karlsson, L. Nassberger, In vitro metabolic inhibition of the human lymphocyte: in- [212] D.E. Richard, E. Berra, J. Pouyssegur, Nonhypoxic pathway mediates the induction
fluence on the expression of interleukin-2 receptors, Immunol. Cell Biol. 70 (Pt 5) of hypoxia-inducible factor 1alpha in vascular smooth muscle cells, J. Biol. Chem.
(1992) 309–313. 275 (2000) 26765–26771.
[184] C.S. Choi, J.J. Fillmore, J.K. Kim, Z.X. Liu, S. Kim, E.F. Collier, A. Kulkarni, A. Distefano, [213] H. Zhong, K. Chiles, D. Feldser, E. Laughner, C. Hanrahan, M.M. Georgescu, J.W. Si-
Y.J. Hwang, M. Kahn, Y. Chen, C. Yu, I.K. Moore, R.M. Reznick, T. Higashimori, G.I. mons, G.L. Semenza, Modulation of hypoxia-inducible factor 1alpha expression
Shulman, Overexpression of uncoupling protein 3 in skeletal muscle protects by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP
against fat-induced insulin resistance, J. Clin. Invest. 117 (2007) 1995–2003. pathway in human prostate cancer cells: implications for tumor angiogenesis
[185] J.P. Warne, Tumour necrosis factor alpha: a key regulator of adipose tissue mass, J. and therapeutics, Cancer Res. 60 (2000) 1541–1545.
Endocrinol. 177 (2003) 351–355. [214] N. Dehne, B. Brune, HIF-1 in the inflammatory microenvironment, Exp. Cell Res.
[186] T. Matsuki, R. Horai, K. Sudo, Y. Iwakura, IL-1 plays an important role in lipid me- 315 (2009) 1791–1797.
tabolism by regulating insulin levels under physiological conditions, J. Exp. Med. [215] J. Jantsch, D. Chakravortty, N. Turza, A.T. Prechtel, B. Buchholz, R.G. Gerlach, M.
198 (2003) 877–888. Volke, J. Glasner, C. Warnecke, M.S. Wiesener, K.U. Eckardt, A. Steinkasserer, M.
[187] V. Wallenius, K. Wallenius, B. Ahren, M. Rudling, H. Carlsten, S.L. Dickson, C. Hensel, C. Willam, Hypoxia and hypoxia-inducible factor-1 alpha modulate
Ohlsson, J.O. Jansson, Interleukin-6-deficient mice develop mature-onset obesity, lipopolysaccharide-induced dendritic cell activation and function, Journal of im-
Nat. Med. 8 (2002) 75–79. munology (Baltim. Md. : 1950) 180 (2008) 4697–4705.
[188] D.E. Bauer, M.H. Harris, D.R. Plas, J.J. Lum, P.S. Hammerman, J.C. Rathmell, J.L. Riley, [216] E. Panther, S. Corinti, M. Idzko, Y. Herouy, M. Napp, A. la Sala, G. Girolomoni, J.
C.B. Thompson, Cytokine stimulation of aerobic glycolysis in hematopoietic cells Norgauer, Adenosine affects expression of membrane molecules, cytokine and
exceeds proliferative demand, FASEB J. 18 (2004) 1303–1305. chemokine release, and the T-cell stimulatory capacity of human dendritic cells,
[189] B. Cousin, O. Munoz, M. Andre, A.M. Fontanilles, C. Dani, J.L. Cousin, P. Laharrague, Blood 101 (2003) 3985–3990.
L. Casteilla, L. Penicaud, A role for preadipocytes as macrophage-like cells, FASEB J. [217] M. Thiel, A. Chouker, A. Ohta, E. Jackson, C. Caldwell, P. Smith, D. Lukashev, I.
13 (1999) 305–312. Bittmann, M.V. Sitkovsky, Oxygenation inhibits the physiological tissue-
[190] K.E. Wellen, G.S. Hotamisligil, Obesity-induced inflammatory changes in adipose protecting mechanism and thereby exacerbates acute inflammatory lung injury,
tissue, J. Clin. Invest. 112 (2003) 1785–1788. PLoS Biol. 3 (2005), e174.
[191] K. Takahashi, S. Mizuarai, H. Araki, S. Mashiko, A. Ishihara, A. Kanatani, H. Itadani, H. [218] G.L. Semenza, HIF-1 inhibitors for cancer therapy: from gene expression to drug
Kotani, Adiposity elevates plasma MCP-1 levels leading to the increased CD11b- discovery, Curr. Pharm. Des. 15 (2009) 3839–3843.
positive monocytes in mice, J. Biol. Chem. 278 (2003) 46654–46660. [219] H. Zhang, D.Z. Qian, Y.S. Tan, K. Lee, P. Gao, Y.R. Ren, S. Rey, H. Hammers, D. Chang,
[192] N. Ouchi, J.L. Parker, J.J. Lugus, K. Walsh, Adipokines in inflammation and metabolic R. Pili, C.V. Dang, J.O. Liu, G.L. Semenza, Digoxin and other cardiac glycosides inhibit
disease, Nat. Rev. Immunol. 11 (2011) 85–97. HIF-1alpha synthesis and block tumor growth, Proc. Natl. Acad. Sci. U. S. A. 105
[193] M. Feuerer, L. Herrero, D. Cipolletta, A. Naaz, J. Wong, A. Nayer, J. Lee, A.B. Goldfine, (2008) 19579–19586.
C. Benoist, S. Shoelson, D. Mathis, Lean, but not obese, fat is enriched for a unique [220] J.R. Huh, M.W. Leung, P. Huang, D.A. Ryan, M.R. Krout, R.R. Malapaka, J. Chow,
population of regulatory T cells that affect metabolic parameters, Nat. Med. 15 N. Manel, M. Ciofani, S.V. Kim, A. Cuesta, F.R. Santori, J.J. Lafaille, H.E. Xu, D.Y.
(2009) 930–939. Gin, F. Rastinejad, D.R. Littman, Digoxin and its derivatives suppress TH17 cell
[194] S. Nishimura, I. Manabe, M. Nagasaki, K. Eto, H. Yamashita, M. Ohsugi, M. Otsu, K. differentiation by antagonizing RORgammat activity, Nature 472 (2011)
Hara, K. Ueki, S. Sugiura, K. Yoshimura, T. Kadowaki, R. Nagai, CD8+ effector T 486–490.
cells contribute to macrophage recruitment and adipose tissue inflammation in [221] B. Onnis, A. Rapisarda, G. Melillo, Development of HIF-1 inhibitors for cancer ther-
obesity, Nat. Med. 15 (2009) 914–920. apy, J. Cell. Mol. Med. 13 (2009) 2780–2786.
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 103

[222] S. Welsh, R. Williams, L. Kirkpatrick, G. Paine-Murrieta, G. Powis, Antitumor activity hepatitis C virus: an analysis of the Scientific Registry of Transplant Recipients Da-
and pharmacodynamic properties of PX-478, an inhibitor of hypoxia-inducible tabase, Liver Transpl. 18 (2012) 1029–1036.
factor-1alpha, Mol. Cancer Ther. 3 (2004) 233–244. [247] S. Ganapathy-Kanniappan, M. Vali, R. Kunjithapatham, M. Buijs, L.H. Syed, P.P. Rao,
[223] W. Jeong, A. Rapisarda, S.R. Park, R.J. Kinders, A. Chen, G. Melillo, B. Turkbey, S. Ota, B.K. Kwak, R. Loffroy, J.F. Geschwind, 3-Bromopyruvate: a new targeted
S.M. Steinberg, P. Choyke, J.H. Doroshow, S. Kummar, Pilot trial of EZN-2968, antiglycolytic agent and a promise for cancer therapy, Curr. Pharm. Biotechnol.
an antisense oligonucleotide inhibitor of hypoxia-inducible factor-1 alpha 11 (2010) 510–517.
(HIF-1alpha), in patients with refractory solid tumors, Cancer Chemother. [248] M. Buijs, J.A. Vossen, J.F. Geschwind, T. Ishimori, J.M. Engles, O. Acha-Ngwodo, R.L.
Pharmacol. 73 (2014) 343–348. Wahl, M. Vali, Specificity of the anti-glycolytic activity of 3-bromopyruvate con-
[224] A.K. Nagaraja, C.J. Creighton, Z. Yu, H. Zhu, P.H. Gunaratne, J.G. Reid, E. Olokpa, H. firmed by FDG uptake in a rat model of breast cancer, Investig. New Drugs 27
Itamochi, N.T. Ueno, S.M. Hawkins, M.L. Anderson, M.M. Matzuk, A link between (2009) 120–123.
mir-100 and FRAP1/mTOR in clear cell ovarian cancer, Mol.Endocrinol. (Baltim. [249] Y.H. Ko, B.L. Smith, Y. Wang, M.G. Pomper, D.A. Rini, M.S. Torbenson, J. Hullihen, P.L.
Md.) 24 (2010) 447–463. Pedersen, Advanced cancers: eradication in all cases using 3-bromopyruvate ther-
[225] X.Q. Liu, M.H. Xiong, X.T. Shu, R.Z. Tang, J. Wang, Therapeutic delivery of siRNA si- apy to deplete ATP, Biochem. Biophys. Res. Commun. 324 (2004) 269–275.
lencing HIF-1 alpha with micellar nanoparticles inhibits hypoxic tumor growth, [250] J. Feng, F. Bussiere, S. Hekimi, Mitochondrial electron transport is a key determi-
Mol. Pharm. 9 (2012) 2863–2874. nant of life span in Caenorhabditis elegans, Dev. Cell 1 (2001) 633–644.
[226] J. Hughes, F. Criscuolo, Evolutionary history of the UCP gene family: gene duplica- [251] S. Rea, T.E. Johnson, A metabolic model for life span determination in
tion and selection, BMC Evol. Biol. 8 (2008) 306. Caenorhabditis elegans, Dev. Cell 5 (2003) 197–203.
[227] V.P. Skulachev, Uncoupling: new approaches to an old problem of bioenergetics, [252] Y. Hattori, K. Suzuki, S. Hattori, K. Kasai, Metformin inhibits cytokine-induced nu-
Biochim. Biophys. Acta 1363 (1998) 100–124. clear factor kappaB activation via AMP-activated protein kinase activation in vas-
[228] F. Bouillaud, UCP2, not a physiologically relevant uncoupler but a glucose sparing cular endothelial cells, Hypertension 47 (2006) 1183–1188.
switch impacting ROS production and glucose sensing, Biochim. Biophys. Acta [253] S.A. Kim, H.C. Choi, Metformin inhibits inflammatory response via AMPK-PTEN
1787 (2009) 377–383. pathway in vascular smooth muscle cells, Biochem. Biophys. Res. Commun. 425
[229] Y. Emre, T. Nubel, Uncoupling protein UCP2: when mitochondrial activity meets (2012) 866–872.
immunity, FEBS Lett. 584 (2010) 1437–1442. [254] C. Josse, S. Legrand-Poels, B. Piret, F. Sluse, J. Piette, Impairment of the mitochondri-
[230] M. Klingenberg, S.G. Huang, Structure and function of the uncoupling protein from al electron chain transport prevents NF-kappa B activation by hydrogen peroxide,
brown adipose tissue, Biochim. Biophys. Acta 1415 (1999) 271–296. Free Radic. Biol. Med. 25 (1998) 104–112.
[231] E. Couplan, M. del Mar Gonzalez-Barroso, M.C. Alves-Guerra, D. Ricquier, M. [255] A. Anedda, E. Rial, M.M. Gonzalez-Barroso, Metformin induces oxidative stress in
Goubern, F. Bouillaud, No evidence for a basal, retinoic, or superoxide-induced white adipocytes and raises uncoupling protein 2 levels, J. Endocrinol. 199
uncoupling activity of the uncoupling protein 2 present in spleen or lung mito- (2008) 33–40.
chondria, J. Biol. Chem. 277 (2002) 26268–26275. [256] R.J. Shaw, N. Bardeesy, B.D. Manning, L. Lopez, M. Kosmatka, R.A. DePinho, L.C.
[232] J. Mozo, G. Ferry, A. Studeny, C. Pecqueur, M. Rodriguez, J.A. Boutin, F. Cantley, The LKB1 tumor suppressor negatively regulates mTOR signaling, Cancer
Bouillaud, Expression of UCP3 in CHO cells does not cause uncoupling, but con- Cell 6 (2004) 91–99.
trols mitochondrial activity in the presence of glucose, Biochem. J. 393 (2006) [257] M. Zakikhani, R. Dowling, I.G. Fantus, N. Sonenberg, M. Pollak, Metformin is an AMP
431–439. kinase-dependent growth inhibitor for breast cancer cells, Cancer Res. 66 (2006)
[233] D. Arsenijevic, H. Onuma, C. Pecqueur, S. Raimbault, B.S. Manning, B. Miroux, E. 10269–10273.
Couplan, M.C. Alves-Guerra, M. Goubern, R. Surwit, F. Bouillaud, D. Richard, S. [258] M.M. Delmastro, J.D. Piganelli, Oxidative stress and redox modulation potential in
Collins, D. Ricquier, Disruption of the uncoupling protein-2 gene in mice reveals type 1 diabetes, Clin. Dev. Immunol. 2011 (2011) 593863.
a role in immunity and reactive oxygen species production, Nat. Genet. 26 [259] M. Esrefoglu, Oxidative stress and benefits of antioxidant agents in acute and
(2000) 435–439. chronic hepatitis, Hepat. Mon. 12 (2012) 160–167.
[234] Y. Bai, H. Onuma, X. Bai, A.V. Medvedev, M. Misukonis, J.B. Weinberg, W. Cao, J. [260] M.M. Essa, R.K. Vijayan, G. Castellano-Gonzalez, M.A. Memon, N. Braidy, G.J.
Robidoux, L.M. Floering, K.W. Daniel, S. Collins, Persistent nuclear factor-kappa B Guillemin, Neuroprotective effect of natural products against Alzheimer's disease,
activation in Ucp2−/− mice leads to enhanced nitric oxide and inflammatory cy- Neurochem. Res. 37 (2012) 1829–1842.
tokine production, J. Biol. Chem. 280 (2005) 19062–19069. [261] V. Fuchs-Tarlovsky, Role of antioxidants in cancer therapy, Nutrition (Burbank, Los
[235] A. Rupprecht, A.U. Brauer, A. Smorodchenko, J. Goyn, K.E. Hilse, I.G. Shabalina, C. Angel. County, Calif.) 29 (2013) 15–21.
Infante-Duarte, E.E. Pohl, Quantification of uncoupling protein 2 reveals its main [262] H. Zhu, Y.R. Li, Oxidative stress and redox signaling mechanisms of inflammatory
expression in immune cells and selective up-regulation during T-cell proliferation, bowel disease: updated experimental and clinical evidence, Exp. Biol. Med. (May-
PLoS One 7 (2012), e41406. wood, N.J.) 237 (2012) 474–480.
[236] I. Dando, C. Fiorini, E.D. Pozza, C. Padroni, C. Costanzo, M. Palmieri, M. Donadelli, [263] J.C. Fernandez-Checa, C. Garcia-Ruiz, A. Colell, A. Morales, M. Mari, M. Miranda, E.
UCP2 inhibition triggers ROS-dependent nuclear translocation of GAPDH and au- Ardite, Oxidative stress: role of mitochondria and protection by glutathione,
tophagic cell death in pancreatic adenocarcinoma cells, Biochim. Biophys. Acta BioFactors (Oxf. Engl.) 8 (1998) 7–11.
1833 (2013) 672–679. [264] J. Wang, M. Pae, S.N. Meydani, D. Wu, Epigallocatechin-3-gallate inhibits expres-
[237] C.Y. Zhang, G. Baffy, P. Perret, S. Krauss, O. Peroni, D. Grujic, T. Hagen, A.J. Vidal- sion of receptors for T cell regulatory cytokines and their downstream signaling
Puig, O. Boss, Y.B. Kim, X.X. Zheng, M.B. Wheeler, G.I. Shulman, C.B. Chan, B.B. in mouse CD4+ T cells, J. Nutr. 142 (2012) 566–571.
Lowell, Uncoupling protein-2 negatively regulates insulin secretion and is a [265] J. Wang, Z. Ren, Y. Xu, S. Xiao, S.N. Meydani, D. Wu, Epigallocatechin-3-gallate ame-
major link between obesity, beta cell dysfunction, and type 2 diabetes, Cell 105 liorates experimental autoimmune encephalomyelitis by altering balance among
(2001) 745–755. CD4+ T-cell subsets, Am. J. Pathol. 180 (2012) 221–234.
[238] V. Ayyasamy, K.M. Owens, M.M. Desouki, P. Liang, A. Bakin, K. Thangaraj, D.J. [266] R. Checker, S.K. Sandur, D. Sharma, R.S. Patwardhan, S. Jayakumar, V. Kohli, G. Sethi,
Buchsbaum, A.F. LoBuglio, K.K. Singh, Cellular model of Warburg effect identifies B.B. Aggarwal, K.B. Sainis, Potent anti-inflammatory activity of ursolic acid, a
tumor promoting function of UCP2 in breast cancer and its suppression by genipin, triterpenoid antioxidant, is mediated through suppression of NF-kappaB, AP-1
PLoS One 6 (2011), e24792. and NF-AT, PLoS One 7 (2012), e31318.
[239] J.M. Brown, C.M. Schwanke, M.A. Pershouse, J.C. Pfau, A. Holian, Effects of rottlerin [267] C.H. Guo, P.J. Liu, K.P. Lin, P.C. Chen, Nutritional supplement therapy improves
on silica-exacerbated systemic autoimmune disease in New Zealand mixed mice, oxidative stress, immune response, pulmonary function, and quality of life in
Am. J. Phys. Lung Cell. Mol. Phys. 289 (2005) L990–L998. allergic asthma patients: an open-label pilot study, Altern. Med. Rev. 17
[240] V.I. Shavell, N.M. Fletcher, Z.L. Jiang, G.M. Saed, M.P. Diamond, Uncoupling oxida- (2012) 42–56.
tive phosphorylation with 2,4-dinitrophenol promotes development of the adhe- [268] H.M. Tse, M.J. Milton, J.D. Piganelli, Mechanistic analysis of the immunomodulatory
sion phenotype, Fertil. Steril. 97 (2012) 729–733. effects of a catalytic antioxidant on antigen-presenting cells: implication for their
[241] E. Dalla Pozza, C. Fiorini, I. Dando, M. Menegazzi, A. Sgarbossa, C. Costanzo, M. use in targeting oxidation-reduction reactions in innate immunity, Free Radic.
Palmieri, M. Donadelli, Role of mitochondrial uncoupling protein 2 in cancer cell Biol. Med. 36 (2004) 233–247.
resistance to gemcitabine, Biochim. Biophys. Acta 1823 (2012) 1856–1863. [269] R. Sharma, M. Vinayak, Antioxidant alpha-tocopherol checks lymphoma promo-
[242] D.E. Harrison, R. Strong, Z.D. Sharp, J.F. Nelson, C.M. Astle, K. Flurkey, N.L. Nadon, J.E. tion via regulation of expression of protein kinase C-alpha and c-Myc genes and
Wilkinson, K. Frenkel, C.S. Carter, M. Pahor, M.A. Javors, E. Fernandez, R.A. Miller, glycolytic metabolism, Leuk. Lymphoma 53 (2012) 1203–1210.
Rapamycin fed late in life extends lifespan in genetically heterogeneous mice, Na- [270] M.M. Delmastro, A.J. Styche, M.M. Trucco, C.J. Workman, D.A.A. Vignali, J.D.
ture 460 (2009) 392–395. Piganelli, Modulation of redox balance leaves murine diabetogenic TH1 T cells
[243] M. Battaglia, A. Stabilini, B. Migliavacca, J. Horejs-Hoeck, T. Kaupper, M.G. “LAG-3-ing” behind, Diabetes 61 (2012) 1760–1768.
Roncarolo, Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ [271] M.M. Sklavos, H.M. Tse, J.D. Piganelli, Redox modulation inhibits CD8 T cell effector
regulatory T cells of both healthy subjects and type 1 diabetic patients, J. Immunol. function, Free Radic. Biol. Med. 45 (2008) 1477–1486.
(Baltim. Md.: 1950) 177 (2006) 8338–8347. [272] Z.N. Rabbani, I. Spasojevic, X. Zhang, B.J. Moeller, S. Haberle, J. Vasquez-Vivar, M.W.
[244] H. Kopf, G.M. de la Rosa, O.M. Howard, X. Chen, Rapamycin inhibits differentiation Dewhirst, Z. Vujaskovic, I. Batinic-Haberle, Antiangiogenic action of redox-
of Th17 cells and promotes generation of FoxP3+ T regulatory cells, Int. modulating Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnTE-2-
Immunopharmacol. 7 (2007) 1819–1824. PyP(5+), via suppression of oxidative stress in a mouse model of breast tumor,
[245] J.A. Fielhaber, S.F. Carroll, A.B. Dydensborg, M. Shourian, A. Triantafillopoulos, S. Free Radic. Biol. Med. 47 (2009) 992–1004.
Harel, S.N. Hussain, M. Bouchard, S.T. Qureshi, A.S. Kristof, Inhibition of mammalian [273] D. Koppers-Lalic, M.M. Hogenboom, J.M. Middeldorp, D.M. Pegtel, Virus-modified
target of rapamycin augments lipopolysaccharide-induced lung injury and apopto- exosomes for targeted RNA delivery; a new approach in nanomedicine, Adv.
sis, J. Immunol. (Baltim. Md.: 1950) 188 (2012) 4535–4542. Drug Deliv. Rev. 65 (2013) 348–356.
[246] K.D. Watt, R. Dierkhising, J.K. Heimbach, M.R. Charlton, Impact of sirolimus and ta- [274] C. Moser, M. Amacker, R. Zurbriggen, Influenza virosomes as a vaccine adjuvant
crolimus on mortality and graft loss in liver transplant recipients with or without and carrier system, Expert Rev. Vaccines 10 (2011) 437–446.
104 K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105

[275] I.A. Schaap, F. Eghiaian, A. des Georges, C. Veigel, Effect of envelope proteins [304] C. Wang, C. He, Z. Tong, X. Liu, B. Ren, F. Zeng, Combination of adsorption by porous
on the mechanical properties of influenza virus, J. Biol. Chem. 287 (2012) CaCO3 microparticles and encapsulation by polyelectrolyte multilayer films for
41078–41088. sustained drug delivery, Int. J. Pharm. 308 (2006) 160–167.
[276] E. Zamparo, D. Little, Immunogenicity and effectiveness of virosomal adjuvanted [305] J.A. Wolff, R.W. Malone, P. Williams, W. Chong, G. Acsadi, A. Jani, P.L. Felgner, Direct
vaccines against influenza: a brief review of their utility in the elderly population, gene transfer into mouse muscle in vivo, Science (New York, N.Y.) 247 (1990)
J. Prev. Med. Hyg. 52 (2011) 116–119. 1465–1468.
[277] C. Cinti, M. Taranta, I. Naldi, S. Grimaldi, Newly engineered magnetic erythrocytes [306] J.A. Wolff, V. Budker, The mechanism of naked DNA uptake and expression, Adv.
for sustained and targeted delivery of anti-cancer therapeutic compounds, PLoS Genet. 54 (2005) 3–20.
One 6 (2011), e17132. [307] E. Neumann, M. Schaefer-Ridder, Y. Wang, P.H. Hofschneider, Gene transfer into
[278] Y. Kaneda, Virosome: a novel vector to enable multi-modal strategies for cancer mouse lyoma cells by electroporation in high electric fields, EMBO J. 1 (1982)
therapy, Adv. Drug Deliv. Rev. 64 (2012) 730–738. 841–845.
[279] J.W. Yoo, D.J. Irvine, D.E. Discher, S. Mitragotri, Bio-inspired, bioengineered and bio- [308] J.F. Edd, L. Horowitz, R.V. Davalos, L.M. Mir, B. Rubinsky, In vivo results of a new
mimetic drug delivery carriers, Nat. Rev. Drug Discov. 10 (2011) 521–535. focal tissue ablation technique: irreversible electroporation, IEEE Trans. Biomed.
[280] M.A. Kay, J.C. Glorioso, L. Naldini, Viral vectors for gene therapy: the art of turning Eng. 53 (2006) 1409–1415.
infectious agents into vehicles of therapeutics, Nat. Med. 7 (2001) 33–40. [309] B. Rubinsky, G. Onik, P. Mikus, Irreversible electroporation: a new ablation modal-
[281] W.Z. Zhou, D.S. Hoon, S.K. Huang, S. Fujii, K. Hashimoto, R. Morishita, Y. Kaneda, ity–clinical implications, Technol. Cancer Res. Treat. 6 (2007) 37–48.
RNA melanoma vaccine: induction of antitumor immunity by human glycoprotein [310] Z.A. Levine, P.T. Vernier, Calcium and phosphatidylserine inhibit lipid electropore
100 mRNA immunization, Hum. Gene Ther. 10 (1999) 2719–2724. formation and reduce pore lifetime, J. Membr. Biol. 245 (2012) 599–610.
[282] K. Kamimura, T. Suda, G. Zhang, D. Liu, Advances in gene delivery systems, Pharm. [311] G. Pucihar, T. Kotnik, B. Valič, D. Miklavčič, Numerical determination of transmem-
Med. 25 (2011) 293–306. brane voltage induced on irregularly shaped cells, Ann. Biomed. Eng. 34 (2006)
[283] N. Nayerossadat, T. Maedeh, P.A. Ali, Viral and nonviral delivery systems for gene 642–652.
delivery, Adv. Biol. Res. 1 (2012) 27. [312] J. Teissié, M.P. Rols, An experimental evaluation of the critical potential difference
[284] A.P. Souza, L. Haut, A. Reyes-Sandoval, A.R. Pinto, Recombinant viruses as vaccines inducing cell membrane electropermeabilization, Biophys. J., 65 409-413.
against viral diseases, Braz. J. Med. Biol. Res. 38 (2005) 509–522 (… [et al.]). [313] G. Pucihar, T. Kotnik, J. Teissié, D. Miklavčič, Electropermeabilization of dense cell
[285] M. Morille, C. Passirani, A. Vonarbourg, A. Clavreul, J.P. Benoit, Progress in develop- suspensions, Eur. Biophys. J. 36 (2007) 173–185.
ing cationic vectors for non-viral systemic gene therapy against cancer, Biomate- [314] J. Gehl, [Investigational treatment of cancer using electrochemotherapy,
rials 29 (2008) 3477–3496. electrochemoimmunotherapy and electro-gene transfer], Ugeskr. Laeger 167
[286] P.L. Felgner, T.R. Gadek, M. Holm, R. Roman, H.W. Chan, M. Wenz, J.P. Northrop, (2005) 3156–3159.
G.M. Ringold, M. Danielsen, Lipofection: a highly efficient, lipid-mediated DNA- [315] F. Liu, L. Huang, A syringe electrode device for simultaneous injection of DNA and
transfection procedure, Proc. Natl. Acad. Sci. U. S. A. 84 (1987) 7413–7417. electrotransfer, Mol. Ther. 5 (2002) 323–328.
[287] G.J. Nabel, E.G. Nabel, Z.Y. Yang, B.A. Fox, G.E. Plautz, X. Gao, L. Huang, S. Shu, D. [316] I. Edhemovic, E.M. Gadzijev, E. Brecelj, D. Miklavcic, B. Kos, A. Zupanic, B. Mali, T.
Gordon, A.E. Chang, Direct gene transfer with DNA-liposome complexes in mela- Jarm, D. Pavliha, M. Marcan, G. Gasljevic, V. Gorjup, M. Music, T.P. Vavpotic, M.
noma: expression, biologic activity, and lack of toxicity in humans, Proc. Natl. Cemazar, M. Snoj, G. Sersa, Electrochemotherapy: a new technological approach
Acad. Sci. U. S. A. 90 (1993) 11307–11311. in treatment of metastases in the liver, Technol. Cancer Res. Treat. 10 (2011)
[288] C.J. Wheeler, P.L. Felgner, Y.J. Tsai, J. Marshall, L. Sukhu, S.G. Doh, J. Hartikka, J. 475–485.
Nietupski, M. Manthorpe, M. Nichols, M. Plewe, X. Liang, J. Norman, A. Smith, [317] M. Golzio, M.P. Rols, J. Teissie, In vitro and in vivo electric field-mediated perme-
S.H. Cheng, A novel cationic lipid greatly enhances plasmid DNA delivery and ex- abilization, gene transfer, and expression, Methods (San Diego, Calif.) 33 (2004)
pression in mouse lung, Proc. Natl. Acad. Sci. U. S. A. 93 (1996) 11454–11459. 126–135.
[289] J. Yoshida, M. Mizuno, M. Fujii, Y. Kajita, N. Nakahara, M. Hatano, R. Saito, M. [318] S. Zhu, D.A. Lee, S. Li, IL-12 and IL-27 sequential gene therapy via intramuscular
Nobayashi, T. Wakabayashi, Human gene therapy for malignant gliomas (glioblas- electroporation delivery for eliminating distal aggressive tumors, J. Immunol.
toma multiforme and anaplastic astrocytoma) by in vivo transduction with human (Baltim. Md.: 1950) 184 (2010) 2348–2354.
interferon beta gene using cationic liposomes, Hum. Gene Ther. 15 (2004) 77–86. [319] L.W. Matthiessen, H.H. Johannesen, H.W. Hendel, T. Moss, C. Kamby, J. Gehl,
[290] T.M. Allen, P.R. Cullis, Drug delivery systems: entering the mainstream, Science Electrochemotherapy for large cutaneous recurrence of breast cancer: a phase II
(New York, N.Y.) 303 (2004) 1818–1822. clinical trial, Acta Oncol. (Stockholm, Sweden) 51 (2012) 713–721.
[291] X. Wang, L. Yang, Z.G. Chen, D.M. Shin, Application of nanotechnology in cancer [320] J.A. Kyte, L. Mu, S. Aamdal, G. Kvalheim, S. Dueland, M. Hauser, H.P. Gullestad, T.
therapy and imaging, CA Cancer J. Clin. 58 (2008) 97–110. Ryder, K. Lislerud, H. Hammerstad, G. Gaudernack, Phase I/II trial of melanoma
[292] M.S. Muthu, D.T. Leong, L. Mei, S.S. Feng, Nanotheranostics - application and further therapy with dendritic cells transfected with autologous tumor-mRNA, Cancer
development of nanomedicine strategies for advanced theranostics, Theranostics 4 Gene Ther. 13 (2006) 905–918.
(2014) 660–677. [321] A.I. Daud, R.C. DeConti, S. Andrews, P. Urbas, A.I. Riker, V.K. Sondak, P.N. Munster,
[293] V. Pavot, M. Berthet, J. Resseguier, S. Legaz, N. Handke, S.C. Gilbert, S. Paul, B. D.M. Sullivan, K.E. Ugen, J.L. Messina, R. Heller, Phase I trial of interleukin-12 plas-
Verrier, Poly(lactic acid) and poly(lactic-co-glycolic acid) particles as versatile car- mid electroporation in patients with metastatic melanoma, J. Clin. Oncol. Off. J. Am.
rier platforms for vaccine delivery, Nanomedicine (Lond. Engl.) 9 (2014) Soc. Clin. Oncol. 26 (2008) 5896–5903.
2703–2718. [322] D.G. Kassan, A.M. Lynch, M.J. Stiller, Physical enhancement of dermatologic drug
[294] N.H. Abu Samah, C.M. Heard, Enhanced in vitro transdermal delivery of caffeine delivery: iontophoresis and phonophoresis, J. Am. Acad. Dermatol. 34 (1996)
using a temperature- and pH-sensitive nanogel, poly(NIPAM-co-AAc), Int. J. 657–666.
Pharm. 453 (2013) 630–640. [323] Y. Taniyama, K. Tachibana, K. Hiraoka, M. Aoki, S. Yamamoto, K. Matsumoto, T.
[295] M. Han, M. Oba, N. Nishiyama, M.R. Kano, S. Kizaka-Kondoh, K. Kataoka, Enhanced Nakamura, T. Ogihara, Y. Kaneda, R. Morishita, Development of safe and effi-
percolation and gene expression in tumor hypoxia by PEGylated polyplex micelles, cient novel nonviral gene transfer using ultrasound: enhancement of transfec-
Mol. Ther. 17 (2009) 1404–1410. tion efficiency of naked plasmid DNA in skeletal muscle, Gene Ther. 9 (2002)
[296] D. Kim, Z.G. Gao, E.S. Lee, Y.H. Bae, In vivo evaluation of doxorubicin-loaded poly- 372–380.
meric micelles targeting folate receptors and early endosomal pH in drug-resistant [324] G. Casey, J.P. Cashman, D. Morrissey, M.C. Whelan, J.O. Larkin, D.M. Soden, M.
ovarian cancer, Mol. Pharm. 6 (2009) 1353–1362. Tangney, G.C. O'Sullivan, Sonoporation mediated immunogene therapy of solid tu-
[297] K. Hanai, T. Kurokawa, Y. Minakuchi, M. Maeda, S. Nagahara, T. Miyata, T. Ochiya, A. mors, Ultrasound Med. Biol. 36 (2010) 430–440.
Sano, Potential of atelocollagen-mediated systemic antisense therapeutics for in- [325] S. Hayashi, M. Mizuno, J. Yoshida, A. Nakao, Effect of sonoporation on cationic
flammatory disease, Hum. Gene Ther. 15 (2004) 263–272. liposome-mediated IFNbeta gene therapy for metastatic hepatic tumors of murine
[298] F. Takeshita, Y. Minakuchi, S. Nagahara, K. Honma, H. Sasaki, K. Hirai, T. Teratani, N. colon cancer, Cancer Gene Ther. 16 (2009) 638–643.
Namatame, Y. Yamamoto, K. Hanai, T. Kato, A. Sano, T. Ochiya, Efficient delivery of [326] K. Iwanaga, K. Tominaga, K. Yamamoto, M. Habu, H. Maeda, S. Akifusa, T. Tsujisawa,
small interfering RNA to bone-metastatic tumors by using atelocollagen in vivo, T. Okinaga, J. Fukuda, T. Nishihara, Local delivery system of cytotoxic agents to tu-
Proc. Natl. Acad. Sci. U. S. A. 102 (2005) 12177–12182. mors by focused sonoporation, Cancer Gene Ther. 14 (2007) 354–363.
[299] T. Kushibiki, K. Matsumoto, T. Nakamura, Y. Tabata, Suppression of tumor metasta- [327] R.F. Lopez, J.E. Seto, D. Blankschtein, R. Langer, Enhancing the transdermal delivery
sis by NK4 plasmid DNA released from cationized gelatin, Gene Ther. 11 (2004) of rigid nanoparticles using the simultaneous application of ultrasound and sodium
1205–1214. lauryl sulfate, Biomaterials 32 (2011) 933–941.
[300] P. Erbacher, S. Zou, T. Bettinger, A.M. Steffan, J.S. Remy, Chitosan-based vector/DNA [328] B.E. Polat, D. Hart, R. Langer, D. Blankschtein, Ultrasound-mediated transdermal
complexes for gene delivery: biophysical characteristics and transfection ability, drug delivery: mechanisms, scope, and emerging trends, J. Control. Release 152
Pharm. Res. 15 (1998) 1332–1339. (2011) 330–348.
[301] K.A. Howard, U.L. Rahbek, X. Liu, C.K. Damgaard, S.Z. Glud, M.O. Andersen, M.B. [329] V. Budker, G. Zhang, I. Danko, P. Williams, J. Wolff, The efficient expression of intra-
Hovgaard, A. Schmitz, J.R. Nyengaard, F. Besenbacher, J. Kjems, RNA interference vascularly delivered DNA in rat muscle, Gene Ther. 5 (1998) 272–276.
in vitro and in vivo using a novel chitosan/siRNA nanoparticle system, Mol. Ther. [330] G. Zhang, V. Budker, J.A. Wolff, High levels of foreign gene expression in hepato-
14 (2006) 476–484. cytes after tail vein injections of naked plasmid DNA, Hum. Gene Ther. 10 (1999)
[302] O. Boussif, F. Lezoualc'h, M.A. Zanta, M.D. Mergny, D. Scherman, B. Demeneix, J.P. 1735–1737.
Behr, A versatile vector for gene and oligonucleotide transfer into cells in culture [331] F. Liu, Y. Song, D. Liu, Hydrodynamics-based transfection in animals by systemic
and in vivo: polyethylenimine, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 7297–7301. administration of plasmid DNA, Gene Ther. 6 (1999) 1258–1266.
[303] A. Alshamsan, S. Hamdy, J. Samuel, A.O. El-Kadi, A. Lavasanifar, H. Uludag, The [332] T. Suda, D. Liu, Hydrodynamic gene delivery: its principles and applications, Mol.
induction of tumor apoptosis in B16 melanoma following STAT3 siRNA Ther. 15 (2007) 2063–2069.
delivery with a lipid-substituted polyethylenimine, Biomaterials 31 (2010) [333] H. Yazawa, T. Murakami, H.M. Li, T. Back, K. Kurosaka, Y. Suzuki, L. Shorts, Y.
1420–1428. Akiyama, K. Maruyama, E. Parsoneault, R.H. Wiltrout, M. Watanabe,
K.S. Gill et al. / Biochimica et Biophysica Acta 1866 (2016) 87–105 105

Hydrodynamics-based gene delivery of naked DNA encoding fetal liver kinase-1 [335] N. Mozzillo, E. Simeone, L. Benedetto, M. Curvietto, D. Giannarelli, G.
gene effectively suppresses the growth of pre-existing tumors, Cancer Gene Gentilcore, R. Camerlingo, M. Capone, G. Madonna, L. Festino, C. Caraco, G.
Ther. 13 (2006) 993–1001. Di Monta, U. Marone, M. Di Marzo, A.M. Grimaldi, S. Mori, G. Ciliberto, P.A.
[334] Z.C. Neal, M.K. Bates, M.R. Albertini, H. Herweijer, Hydrodynamic limb vein delivery Ascierto, Assessing a novel immuno-oncology-based combination therapy:
of a xenogeneic DNA cancer vaccine effectively induces antitumor immunity, Mol. ipilimumab plus electrochemotherapy, Oncoimmunology 4 (2015),
Ther. 15 (2007) 422–430. e1008842.

You might also like