You are on page 1of 12

Metabolism_Layout 1 04/10/2011 17:39 Page 64

Drug Discovery

Targeting target
cancer metabolism
what is fuelling the resurgence?
Targeting the metabolic pathways of cancer is a hot topic for drug discovery.
The central dogma is that cancers have a higher demand for metabolic inputs
to aid proliferation and survival and that this could be an Achilles heal which
could be exploited therapeutically. This central metabolic facet of cancer has
been known for more than 50 years so what are the recent discoveries that
have fuelled the renewed investment in metabolic targets in cancer?

I
By Dr Neil P. Jones n recent years there has been resurgence in focus on cancer metabolism as a new approach to
interest in metabolism as a possible area for cancer treatment and has an active pipeline of
development of novel anti-cancer agents. This metabolism targets including isocitrate dehydroge-
has been fuelled by advances in technology and nase (IDH1/2)1 and Pyruvate kinase-M2 (PK-
understanding of cellular metabolism and how M2)2, targets which will be discussed in more
targeting this area could be of therapeutic benefit. detail later. Agios has also recently entered into a
The drivers behind these advances have come $130 million strategic alliance with Celgene to
from academic-led research pushing our under- explore novel drug targets in Cancer metabolism.
standing of cancer cell metabolism and the inter- Cornerstone Pharmaceutical was also founded
play between these pathways and other cancer through academic researchers (Paul Bingham and
related signalling events linked to changes in onco- Zuzana Zachar) with a focus on targeting cancer
gene or tumour suppressor gene status/function. metabolism and has a candidate drug, CPI-613,
In the past few years this spiralling interest has which targets pyruvate dehydrogenase3 currently
lead to a rapid increase in the number of publica- in multiple Phase I/II clinical trials. Another
tions on metabolism, multiple focused cancer biotech, Advanced Cancer Therapeutics, has
metabolism conferences and the renewed interest invested in research at the James Graham Brown
from the pharmaceutical industry in this area of Cancer Centre and the University of Louisville
anti-cancer research. Research Foundation to pursue cancer metabolism
This interest has lead to the establishment of targets for example; PFKFB3, a glycolytic regula-
several new companies focused on this area and to tor4 and choline kinase ␣ component of lipid
the generation of partnerships between the metabolism pathways5. Likewise AstraZeneca is
Pharma industry and research organisations to midway through a multi-project three-year
understand this complicated field and hunt for alliance with Cancer Research Technology, Cancer
new targets. For example, Agios Pharmaceuticals Research UK’s commercialisation and develop-
was founded by key academic experts in cancer, ment arm to work on a portfolio of targets select-
oncogenic signalling and metabolism research ed from Cancer Research UK’s biological research
(Lewis Cantley, Craig Thompson and Tak Mak) to in the emerging field of cancer metabolism.

64 Drug Discovery World Fall 2011


Metabolism_Layout 1 04/10/2011 17:39 Page 65

Drug Discovery

Figure 1
Targeting tumour metabolism.
Core metabolic pathways and
enzymes against which
inhibitors (shown in red) have
been reported. Key enzymes
are shown in green boxes and
key biosynthetic endpoints
listed in purple. Abbreviations:
GLUT – Glucose transporter;
HK2 – Hexokinase-2; PFK1 –
Phosphofructokinase 1;
PFKFB3 – 6-phosphofructo-2-
kinase/fructose-2,6-
biphosphatase 3; PK-M2 –
pyruvate kinase-M2; LDHa –
lactate dehydrogenase-A;
MCT1/4 – Monocarboxylate
transporter’s 1/4; AMPK –
AMP activated protein kinase;
G6PD – glucose-6-phosphate
dehydrogenase; TKT –
transketolase; TKTL1 –
transketolase like -1; ASCT2 –
solute carrier family 1,
member 5 [SLC1A5]; GLS1 –
Glutaminase 1; ME – malic
enzyme; FH – fumarate
hydratase; SDH – succinate
dehydrogenase; PDH –
pyruvate dehydrogenase; PDK1
– pyruvate dehydrogenase
kinase 1; ACLY – ATP citrate
lyase; ACACA – acetyl-CoA
Many potential drug targets within the metabol- cancer cells and rapidly proliferating normal cells carboxylase alpha; FASN –
ic field are currently being evaluated as potential require some of these adaptations for prolifera- fatty acid synthase; CA9/12 –
therapeutics in all stages of the drug development tion, however cancer cells must implement these carbonic anhydrase 9/12;
progress from early development, through pre-clin- processes in very different and often harsh or NHE1- Na+-H+ exchanger
ical development and into clinical trials although stressful environments where nutrients supplies [SLC9A1]
as yet no agent targeting a core metabolic pathway may be low and where redox balance, pH and
has been approved for cancer6. Figure 1 summaries oxygen levels may not be maintained. Cancer cells
some of the core metabolic pathways and some of have found ways to adapt to these dynamic situa-
the agents in development looking to target differ- tions and regulate their metabolic status in order
ent areas of cancer metabolism. In order to fully to survive, grow and even prosper.
understand why there are so many agents and The links between cancer and altered metabo-
pathways under consideration in cancer metabo- lism is not a new phenomenon. Otto Warburg, a
lism it is important to investigate the main drivers Nobel prize-winning scientist (for the discovery of
for targeting cancer metabolism. The rest of this cytochrome oxidase) noted more than 80 years ago
review will therefore consider why cancer metabo- that in tumour tissue slices ATP is generated from
lism could make an attractive area for therapeutic glucose via aerobic glycolysis which is an oxygen
intervention and how our understanding of these independent process rather than by oxygen
pathways and advances in technology/knowledge dependent oxidative phosphorylation even when
is driving the hunt for new therapeutics. oxygen is present7,8. This switch in ATP generation
which has been termed the Warburg Effect is ini-
Cancer metabolism and tially paradoxical as while ATP generation is more
the Warburg Effect rapid via glycolytic pathways, far less ATP is gen-
Dividing cells require ATP to maintain energy sta- erated (2 molecules ATP/molecule glucose) than via
tus, increased biosynthetic intermediates and oxidative phosphorylation (up to 36 molecules
maintenance of cellular redox status. In order to ATP/molecule glucose).
meet these needs carbohydrates, proteins, In more recent times this switch has been attrib-
nucleotide and lipid alterations are required. Both uted to the ability of glycolytic pathways to supply

Drug Discovery World Fall 2011 65


Metabolism_Layout 1 04/10/2011 17:39 Page 66

Drug Discovery

essential intermediate components and co-factors sis and in redox balance. The multi-step conversion
via branches off of the core glycolysis pathway of glutamine to lactate helps to explain high lactate
including via the pentose phosphate pathway levels in tumours even when glycolytic flux has
which supplies NADPH for redox balance and been slowed to generate key intermediates via
lipid metabolism and ribose-5-phosphate for branched pathways20,21 (Figure 1).
nucleotide synthesis and via the serine biosynthesis For both glucose and glutamine metabolism
pathway (Figure 1)9,10. Alongside this it has been improved imaging techniques coupled with
postulated that glycolytic adaption could be the enhanced methods to monitor metabolic flux and
result of adaptations to hypoxic conditions during identify metabolites (nmr and mass spectroscopy)
early tumour development and in order to generate has really enabled researchers to elucidate what is
ATP and metabolites at a higher rate when glucose happening to the key metabolic start points in can-
is not limiting11. What is clear is the Warburg shift cer cells compared with other tissues and help to
demands that tumour cells implement an abnor- understand how cancer cells adapt to use these
mally high rate of glucose uptake to meeting their effectively to maintain growth and survival22,23.
increased demands for biosynthesis, energy and The advent of technologies such as RNAi, along-
reducing equivalents. side advances in genomic and proteomic profiling,
The advent of Flurodexoyglucose-positron emis- metabolic modelling and improved access to
sion topography (18F-FDG-PET) imaging12, in tumour samples, have proved invaluable in allow-
which a radioactive glucose analogue is used to ing researches to really probe metabolic function-
assess glucose uptake, has confirmed that many ality in cancer. Coupled with the enhanced under-
tumour types have high glucose uptake and is now standing into the fates of glucose and glutamine
used as part of clinical diagnostic packages for this has driven advances in understanding the com-
tumours. 18F-FDG (2-dexoy-2-(18F)fluoro-D-glu- plex nature of metabolic pathways in cells and how
cose), first synthesised in the 1970s is a glucose these are deregulated in cancers24,25.
analogue which enters the cell in normal ways and
is phosphorylated like normal glucose to prevent it Cancer metabolism pathways: drivers
being released again but cannot then be further and their potential metabolic targets
processed by glycolytic pathways before radioac- In recent years what has really catapulted cancer
tive decay and so is a good reflection of glucose metabolism right back into the spotlight is the
uptake in the body13,14. This ability to monitor understanding of the mechanism by which meta-
glucose within tumours and surrounding tissues bolic adaptations are controlled and regulated in
and the clear increases in glucose uptake and utili- tumours by known oncogenic signalling mecha-
sation in tumours confirms aspects of Warburg’s nism25. Alongside this has been the discovery that
hypothesis and underlies the important of glucose within several metabolic components there are
metabolism in many cancers15,16. cancer-related mutations (ie IDH1/2)26,27 or can-
It has also been know for more than 50 years cer-specific isoforms (ie PK-M2)28 that are critical-
that many tumours have increased rates of gluta- ly linked to progression of certain tumour types.
mine uptake and consumption. In fact many cancer The ability to sequence large sample banks of
cells cannot survive without exogenous glutamine tumours and understand the data has unlocked
and display a glutamine addiction17. As was seen many of the secrets of different cancers and helped
with glucose, the initial assumption was that the us begin to understand what drives tumour forma-
metabolism of glutamine by tumours is inefficient. tion and progression with deregulation of cellular
However, recent studies have shown that gluta- energetic now recognised as one of the hallmarks
mine is a key initial substrate in many processes of cancer29.
essential for cancer cell maintenance and growth. It is now clear that many oncogenic
Products of glutamine metabolism have been (Myc18,30,31, PI3k/AKT32-34, Ras10,35) and
found to be essential for the generation of acetyl tumour suppressor proteins (p5336-38,
CoA (the starting block of lipid synthesis), for PTEN 39,40 , LKB1 41,42 ) directly affect the expres-
NADH generation (for lipid synthesis and redox sion, regulation and activity of key components of
balance), for glutathione synthesis (for redox bal- metabolic pathways and it is now believed that
ance) and for serine synthesis (for nucleotide and these tumourigenic alterations act in part to drive
protein synthesis)(Figure 1)18,19. A large propor- cancer progression via promoting metabolic adap-
tion of glutamine is also converted to lactate in a tation towards enhanced glucose and glutamine
process which generates NADPH an essential dependence (see Table 1). For example, Myc has
reducing equivalent in lipid and nucleotide synthe- been shown to upregulate glutaminolysis via

66 Drug Discovery World Fall 2011


Metabolism_Layout 1 04/10/2011 17:39 Page 67

Drug Discovery

increasing the expression key components of the cers


Table predominantly involves
1: Control of metabolic the by
processes transporters
oncogenes and tumour suppressor genes
glutamine metabolic pathway43 and to enhance
oxidative metabolism of glucose via increased
pyruvate dehydrogenase44 and lactate dehydroge-
nase expression/activity45. HIF1␣11,46-48, a tran-
scription factor which can be upregulated in
hypoxic conditions or by enhanced oncogenic (ie
Myc49,50) or decreased tumour suppressor (ie Von
Hippel-Lindau51) function is also known to
upregulate the expression of many metabolic
enzymes including glucose transporters
(GLUT448), metabolic regulators (PFKFB352,53,
PFKFB452, pyruvate dehydrogenase kinase
(PDK1)44,54) and glycolytic enzymes (hexokinase-
2 (HK2)44, PK-M255, lactate dehydrogenase
(LDHa)56, monocarboxylate transporter 4
(MCT4)57). This suggests that metabolic adapta-
tions also play a role in maintaining tumour
growth and survival in hypoxic conditions. Akt
also increases glycolysis by increasing glucose
transporter expression32 and facilitating hexoki-
nase translocation to the mitochondria where it
functions to initiate glycolytic flux 34,58. The
tumour suppressor protein p5359,60 has also been
shown to regulate expression of various glycolytic
proteins including upregulation of hexokinase and
of a protein called TIGAR which acts as a negative
regulator of glycolysis61,62. p53 also promotes
oxidative phosphorylation via upregulation of GLUT1 and GLUT4 for glucose65 and
SCO263 and suppresses glycolysis via expression ASCT2(SLC1A5) for glutamine66. GLUT1 and
of PTEN, a negative regulator of the PI3K path- GLUT4 have been found to be overexpressed in
way64. Therefore, while loss of p53 may drive many cancers and to be upregulated by ras and
acquisition of the glycolytic phenotype it will be myc signalling65,67. Currently there are no clear
important to understand metabolic regulation in inhibitors of GLUT1/4 published although the
p53 wild type and mutant tumours. natural dihydrochalcone, Phloretin is reported to
Understanding the interplay between oncogenes have GLUT inhibitory activity and be effective in
and tumour suppressors and metabolic pathways inducing apoptosis in in vivo cancer models68.
will be key to deciphering how metabolism inte- ASCT2 is also overexpressed in some tumours69
grates into tumour initiation and progression and and reported to be directly upregulated by
in looking for potential therapeutic targets with myc18,70 although as yet no clear inhibitors of this
clear clinical stratification. The rest of this review transporter have been reported. The oncogenic
will take a stripped-down look at metabolism and upregulation of glucose and/or glutamine trans-
introduce a couple of potential metabolism targets porters in cancer helps to explain how tumours
which are currently being extensively investigated adapt to facilitate their increased reliance on glu-
both by academia and industry. cose and/or glutamine and how tumours can accu-
mulate high levels of these metabolites.
The basic stages of glucose and The next step is the initial processing of these
glutamine metabolism in cancer imported metabolises by conversion into the first
At its simplest level the complex process of glucose metabolic product which can act as an entry sub-
and glutamine metabolism can be split into four strate into the main metabolic processing path-
key phases (Figure 2). The first phase is the uptake ways. This conversion acts to trap the imported
of these essential metabolic substrates into the cell metabolite within the cell and to commit it to fur-
via transporter proteins. The process actively ther processing into downstream intermediates as
imports high concentrations of glucose or gluta- well as shift equilibrium balances to allow the
mine into the cell ready for utilisation and in can- influx of more glucose or glutamine to meet the

Drug Discovery World Fall 2011 67


Metabolism_Layout 1 04/10/2011 17:39 Page 68

Drug Discovery

cell’s high demand for these metabolites. For glu- intermediates through metabolic cascades. This
cose, this initial step is conversion to glucose-6- process is not a liner and intermediates can be fur-
phosphate by the hexokinase’s of which hexoki- ther processed through branched pathways (ie tri-
nase 2 appears to predominant in many cancers71 carboxylic acid cycle, pentose phosphate pathway
and has shown to be regulated by both AKT58 and or serine biosynthesis pathway) to yield additional
mutant p5372. siRNA studies or use of non- products (see Figure 1). The net result of all the pro-
hydrolysable glucose mimics which have been cessing pathways of core metabolism is; the genera-
shown to inhibit Hexokinase-2 activity, suggest tion of biosynthetic intermediates for the manufac-
that modulating Hexokinase-2 activity could be of ture of lipids, proteins, nucleotides and complexes
therapeutic benefit in cancer73-75. Two of these sugars, the generation of energy in the form of ATP
potential hexokinase inhibitors; 2-doexyglucose76- and the generation of key co-factors, ie glutathione,
78 and 3-bromopyruvate71,74 have shown promis- NADH and NADPH, which are essential for the
ing anti-cancer activity in multiple pre-clinical functionality of many protein and have key roles in
models but as yet data from clinical trials has not protecting the cell from oxidative stress. Multiple
supported their use as clinical anti-cancer agents. proteins on these pathways have been shown to be
For glutamine the first step is conversion of glu- over expressed in cancer, dependent on oncogenic
tamine to glutamate by glutaminase proteins control or in inhibition studies (RNAi or tool com-
(GLS1/2)79. GLS1 expression has been shown to pounds) been shown to be involved in cell prolifer-
be increased in several tumour types and to be ation and/or survival mechanisms. Proteins which
under indirect control from Myc via myc-depend- are of potential interest as possible therapeutic tar-
ent regulation of miR23a/b levels31,80. Studies gets include the glycolytic enzymes 84 (eg
using siRNA technology81 or potential inhibitors Hexokinase-271, Phosphoglycerate kinase-185,86,
of GLS1 (DON82, 96883) have suggested that inhi- Phosphoglycerate mutase87,88 and Pyruvate kinase-
bition of this target could be of benefit in gluta- m228,89-91); the pentose phosphate proteins (eg
mine dependent tumour cells. Glucose-6-phosphate dehydrogenase92-94, transal-
The next phase of metabolite processing involves dolase95 and transketolase96-98) and lipid synthe-
the conversion of these metabolites into a series of sis/fatty acid metabolism targets (eg ATP citrate

Figure 2
Key phases of cancer cell
metabolism. Dashed boxes
represent the four key phases
of metabolism and illustrate
the key proteins, intermediates
or biosynthetic products
within each phase.
Abbreviations: GLUT –
Glucose transporter; ASCT2 –
solute carrier family 1;
member 5 [SLC1A5]; HK –
Hexokinase; GLS –
Glutaminase; MCT –
Monocarboxylate transporter

68 Drug Discovery World Fall 2011


Metabolism_Layout 1 04/10/2011 17:39 Page 69

Drug Discovery

Figure 3
Oncogenic regulation of
Pyruvate kinase M2 and ATP
production. The active
tetramer PK-M2 promotes
glycolytic flux and ATP
generation with a reduced
output to other biosynthetic
pathways. High ATP levels can
have a negative feedback effect
on this flux to reduce
additional ATP generation (left
panel). As a result of oncogenic
driven phosphorylation, PK-M2
switches to the low activity
dimeric form which results in
less ATP production, a slowing
of glycolytic flux and enhanced
biosynthesis to meet the
cancer cell biosynthetic
demands. In cancer cells,
pyruvate generation can also
be uncoupled from ATP
production by a novel
mechanism by which
phosphoenolpyruvate is
converted to pyruvate by
transference of a phosphate
group to histidine-11 of
phosphoglycerate mutase
(PGAM1). This process also
activates PGAM1 and so
lyase99,100, fatty acid synthase101,102, monoglyc- mechanisms. Understanding what these mecha- enhances flux at this point of
glycolysis (right panel).
eride lipase103,104 and carnitine palmitoyltrans- nisms could be may offer an attractive therapeutic Abbreviations: PGAM1 –
ferase 1C105). See 6,84,106 for more detailed reviews strategy as inhibiting ammonia removal could phosphoglycerate mutase 1;
of some of these. cause build up of this toxic product and thereby ENO – enolase; PKM2 –
The final process within centralised metabolism potentially kill the tumour cell. pyruvate kinase M2; LDHa –
is the export of end-point products/waste from the Therefore even in this stripped-down version of lactate dehydrogenase-A; X? –
Unknown phosphotransfer
cell in order to protect the cell from build up of metabolism it is clear that tumours are adapting to protein
potentially toxic components and also to modify maximise the usage of glucose and glutamine to
the extracellular vicinity around the cell which may promote survival and even growth in potential hos-
act to assist the cell’s establishment in this environ- tile environments and targeting these adaptions
ment. In cancer cells the main exported substance could be a therapeutic mechanism.
from glucose metabolism is lactate which is As understanding of cancer metabolism devel-
effluxed via the transporter proteins MCT1 and ops, potential therapeutic targets are identified
MCT457,66. Studies have found MCT transporters based on their roles in cancer metabolism coupled
to be overexpressed in multiple tumour types107- with cancer specific expression/isoforms, potential
110 and that chemical or genetic inhibition of MCT mutations and oncogenic control mechanisms.
function can reduce tumour growth suggesting that Targets which fit these profiles have been at the
molecular targeted therapy against MCT trans- forefront of the new push for cancer metabolism
porters could be a possible mechanism for target- drugs and examples include Pyruvate kinase M2
ing cancer metabolism57,111. Inhibitors targeting and Isocitrate dehydrogenase.
MCT1 have been successfully shown to affect in
vitro and in vivo tumour growth and a MCT1 Pyruvate kinase M2 (PKM2)
inhibitor designed by AstraZeneca (AZD-3965) is Pyruvate kinase (PK) catalyses the conversion of
about to enter clinical trials as part of the CR:UK phosphoenolpyruvate (PEP) into pyruvate in a rate
clinical development partnership. For glutamine limiting and ATP generating step within glycolysis
metabolism it is believed ammonia released during (Figure 3)2. There are multiple isoforms of PK of
the process of glutaminolysis diffuses into the which the muscle form is of key interest in cancer
extracellular environment by as yet undefined cells89. PKM1 is found in muscle and brain and is

Drug Discovery World Fall 2011 69


Metabolism_Layout 1 04/10/2011 17:39 Page 70

Drug Discovery

reported to be constitutively active whereas PKM2 by PKM1 have shown that PKM2 is involved in
is present in embryonic and adult stem cells and tumour progression and that PKM2 expression con-
controlled by various regulator mechanisms. It has fers a tumourigenic advantage over PKM1 expres-
been widely reported that PKM2 is also over- sion89. However, it has also been shown that while
expressed in many tumour cells and a switch from PKM1 can efficiently promote glycolysis, PKM2 is
PKM1 to PKM2 expression in cancer has been pro- characteristically found in an inactive state and is
posed2,28,89. The PKM2 isoform is generated by inefficient in promoting glycolysis2,28,89,114. PKM2
alternative splicing of exon 10 on the PK gene, an exists in two possible conformations, an inactive
event shown to be under myc regulation suggesting dimer and more active tetramer (Figure 3).
a potential oncogenic driver for PKM2 expression Oncogenic tyrosine kinases (eg fibroblast growth
in cancers30,112. However, the PKM2 expression factor receptor kinase) have been found to promote
hypothesis has been recently questioned in a study the formation of the inactive dimer via the phos-
which used mass-spectroscopy to identify PK iso- phorylation of tyrosine 705 on PKM290,114. PKM2
forms and reported that PKM2 is expressed in nor- activity has also been shown to be negatively regu-
mal tissues as well as cancers and that PKM1 had lated by acetylation induced by high levels of glu-
low expression in cancers as well as in normal tis- cose115. This data collectively suggests that expres-
sue113. Although the ratio of PKM2 to PKM1 sion of PKM2 in cancer could actually decrease gly-
expression was similar between cancers and colytic flux. While this was initially thought to be
matched normal tissues, the actual amounts of counterintuitive, when considered in terms of the
each protein were much higher in the tumours sug- cancer cell’s metabolic needs, a mechanism which
gesting that both PKM2 and PKM1 were over- slows glycolysis is actually potential advantageous.
expressed in tumour samples113. By slowing glycolytic flux the cancer is able to
Studies in which PKM2 is knockdown or replaced obtain building block, co-factors and precursors by

What — You Don’t Dip and Read™?!


Do less and get more from your kinetic assays
r Screen up to 1000 kinetic interactions in 6 hours
r Analyze crude samples microfluidics-free in 96-
and 384-well plates
r Get ka, kd, KD data in minutes, not hours
or days
r Acquire real-time, SPR-quality kinetic data
without the cost
r Monitor protein, small molecule, DNA,
membrane protein and virus-like particle The
Oc tet® Platform
interactions

Octet Systems with Dip and Read assays.


Easy, worry-free kinetics.

fortebio.com
1-888-OCTET-75
Fast. Accurate. EASY.

70 Drug Discovery World Fall 2011


Metabolism_Layout 1 04/10/2011 17:39 Page 71

Drug Discovery

allowing glucose metabolites to enter subsidiary far larger sample sets (often more than 200 samples) Figure 4
pathways including pentose phosphate, serine and in multiple tumour types. Using these tech- Isocitrate dehydrogenase
mutations and the TCA cycle.
biosynthesis, hexcosamine and glycerol synthesis niques it has been found that 60-90% of secondary
In the normal mitochondrial
pathway which support cancer proliferation and gliomas (around 5% of primary gliomas)27,119 and TCA cycle IDH2/3 enzymes
survival. Another metabolic role for PKM2 is a pro- 12-18% of acute myeloid leukaemias have muta- convert isocitrate to ␣-
posed direct interaction and stabilisation of Hif1␣ tions in the oxidative phosphorylation/TCA cycle ketoglutarate (␣-KG) and
which in turn acts to promote glycolytic metabo- components IDH1 or IDH226,120,121. For gliomas IDH1 converts cytoplasmic
isocitrate to ␣-KG. However,
lism, angiogenesis and cancer progression55. An the common mutations are IDH1 Arg132 and IDH2
mutant IDH1/2 enzymes
alternative glycolytic route which bypasses PK in the Arg140 and Arg172 whereas for glioma most muta- (shown in red boxes) have a
conversion of PEP into Pyruvate has recently been tions are in the IDH2 protein1,27,120. It is also neomorphic enzyme capacity
identified88,116. This route uncouples ATP and worth noting that the vast majority of these muta- and convert ␣-KG into 2-
Pyruvate generation and could provide biosynthetic tions are heterozygous. hdyroxyglutarate (2-HG). 2-
HG is believed to inhibit ␣-KG
intermediates without potential feedback inhibition Mutations affecting the catalytic sites of IDH1
dependent processes including
of glycolysis from ATP accumulation (Figure 3). or 2 are thought to be functionally equivalent and TET1/2 methyltransferase and
The key question around PKM2 is whether acti- were initially reported to negatively affect IDH the histone demethylase
vators or inhibitors or PKM2 kinase activity would catalytic activity by reducing isocitrate binding KDMa2 leading to epigenetic
be the best strategy and if PK-M2 would really be and the ability to convert isocitrate into alpha- disregulation. 2HG may also
act to stabilise HIF1␣.
a cancer specific target. Recent publications have ketoglutarate (␣-KG). However, recent mass-spec-
Abbreviations: CS – citrate
shown that progress is being made in designing troscopy data has discovered IDH mutations synthase; ACO1/2 – Aconitase
tool compounds117,118 to test out the PKM2 exhibit an altered catalytic activity and convert ␣- 1/2; IDH – isocitrate
hypothesis and it will be interesting to monitor the KG (the product of wild type IDH proteins) to 2- dehydrogenase; mIDH –
outcome of studies with these and other PKM2 hydorxyglutarate (Figure 4)120-122. This altered mutant isocitrate
dehydrogenase; OGDH –
modulation agents to see if a clear rationale and metabolite is found to be 100-fold increased in
oxoglutarate (␣-ketoglutarate)
patient selection strategy can be defined for this glioma or AML patients with IDH mutations sug- dehydrogenase; SCS – succinyl-
complex metabolic target. gesting it could act as a clinical biomarker120. CoA synthetase; SDH –
Large scale analysis of DNA methylation in succinate dehydrogenase; FH –
Isocitrate dehydrogenase (IDH1/2) human gliomas provided a key insight into the Fumarate hydratase; MDH2 –
malate dehydrogenase 2; PDH
Advances in large scale sequencing technologies has role of this mutation in cancer. This study showed
– pyruvate dehydrogenase;
enabled more in-depth profiling of the genetics of that nearly all IDH1 and IDH2 mutations were GLS1 – glutaminase 1
multiple metabolism and oncogenic components in associated with a highly specific DNA methylation

Drug Discovery World Fall 2011 71


Metabolism_Layout 1 04/10/2011 17:39 Page 72

Drug Discovery

profile corresponding to the oligodendrocyte sub- olism enzymes in cancer have been identified, it is
type of glioma26. Similar changes in DNA methy- likely that in many tumours the impact of metabo-
lation profiling were also seen in human AML lism targets will be a complex interplay between
samples. A decrease in ␣-KG levels (probably due expression, regulation, flux and oncogenic
to heterodimerisation between ␣-KG producing changes. It will also be important to consider that
WT IDH and ␣-KG metabolising mutant IDH within metabolism pathway there are plenty of
[Figure 3]) coupled with an increase in 2-HG lev- opportunities for tumour cells to evade metabolic
els is reported to block epigenetic events including blocks through bypass pathways and redundancy,
histone demethylase and TET1/2 (hydroxylases so understanding key nodal points and possible
which usually produce 5-hydorxymethylcytosine) combination strategies or synthetic lethality
activity and are believed to be the major mecha- approaches will be essential. Likewise the interplay
nism by which IDH mutants function in tumours1. between metabolism targets and current
2HG has also been reported to stabilise HIF1␣ chemotherapies (many of which create increased
which can regulate many metabolic components demand for biosynthetic intermediates as the
and also promote VEGF signalling, a driver of tumour attempts to survive these agents) could be
tumour angiogenesis123. an important therapeutic strategy to consider as
Therefore, IDH mutants represent an attractive more metabolism targeting agents reach clinical
target for targeted therapy as they show a unique trials in the future.
cancer specific function and generate a potential Pharma is rapidly adapting to these needs by the
cancer biomarker in 2HG for disease stratification. way it is approaching this area with ties to leading
However, one intriguing recent piece of evidence is academic experts in cancer metabolism and utili-
that in IDH mutant appear to have slightly pro- sation of existing expertise against metabolic dis-
longed survival which adds more complexity to orders for which many companies already have a
this intriguing target124. platform. It is hoped that with this approach and
the renewed interest in cancer metabolism the way
Future challenges in targeting is paved for a new generation of cancer metabo-
cancer metabolism lism therapeutics. DDW
The challenges around targeting metabolism will
involve a clear understanding of how a cancer cell
differs in its metabolism to that of a rapidly pro-
liferating normal cell. It has long been known the
neurological cells also have high glucose demands
and also a reliance on other metabolites linked to
some of the canonical metabolism pathways and
linked to cancer, for example glutamine and ser-
ine. For example, it has been shown in normal T-
lymphocytes125 and astrocytes126 that siRNA
depletion of metabolic enzymes such as PFKFB3
decreases the proliferation rates of these ‘normal’
cell lines127,128. Therefore it will be important to
fully understand the metabolic drivers by which a
cancer cells may differ from normal cells and also
the potential toxicity risk associated with target-
ing metabolism.
Another challenge will be around patient strati-
fication/selection. While many tumours may dis- Dr Neil P Jones is a Principal Target Validation
play high glucose or glutamine uptake/utilisation, Scientist at Cancer Research Technology and
this alone is likely to be insufficient as a predictive involved in the drug discovery alliance between
marker for therapeutic potential of an anti-metab- Cancer Research Technology and AstraZeneca to
olism agent. It will be important to fully under- identify new cancer therapies targeting cancer
stand how known oncogenic drivers of cancer (ie metabolism. He received his PhD at the University
myc, ras, PI3K) or loss of tumour suppressor genes of Southampton in the Cell and Molecular
(ie p53, PTEN, LKB1) impact on metabolic flux Bioscience Laboratory and has previously worked
and/or regulation of key metabolic points in differ- in lipid signalling pathways and cancer at the
ent tumour types. While a few mutations in metab- Institute of Cancer Research, London.

72 Drug Discovery World Fall 2011


Metabolism_Layout 1 04/10/2011 17:39 Page 73

Drug Discovery

References 20 Berardi, MJ and Fantin, VR. Survival of the 39 Yuan, TL and Cantley, LC. PI3K pathway
1 Yen, KE et al. Cancer-associated IDH fittest: metabolic adaptations in cancer. Curr alterations in cancer: variations on a theme.
mutations: biomarker and therapeutic Opin Genet Dev. 21(1): p. 59-66. Oncogene, 2008. 27(41): p. 5497-510.
opportunities. Oncogene. 29(49): p. 6409-17. 21 DeBerardinis, RJ et al. Beyond aerobic 40 Kalaany, NY and Sabatini, DM. Tumours
2 Gupta, V and Bamezai, RN. Human pyruvate glycolysis: transformed cells can engage in with PI3K activation are resistant to dietary
kinase M2: a multifunctional protein. Protein Sci. glutamine metabolism that exceeds the restriction. Nature, 2009. 458(7239): p. 725-31.
19(11): p. 2031-44. requirement for protein and nucleotide 41 Shaw, RJ et al. The LKB1 tumor suppressor
3 Zachar, Z et al. Non-redox-active lipoate synthesis. Proc Natl Acad Sci U S A, 2007. negatively regulates mTOR signaling. Cancer
derivates disrupt cancer cell mitochondrial 104(49): p. 19345-50. Cell, 2004. 6(1): p. 91-9.
metabolism and are potent anticancer agents in 22 Plathow, C and Weber, WA. Tumor cell 42 Shaw, RJ et al. The tumor suppressor LKB1
vivo. J Mol Med (Berl). metabolism imaging. J Nucl Med, 2008. 49 Suppl kinase directly activates AMP-activated kinase
4 Clem, B et al. Small-molecule inhibition of 6- 2: p. 43S-63S. and regulates apoptosis in response to energy
phosphofructo-2-kinase activity suppresses 23 Bohndiek, SE and Brindle, KM. Imaging and stress. Proc Natl Acad Sci U S A, 2004. 101(10):
glycolytic flux and tumor growth. Mol Cancer ‘omic’ methods for the molecular diagnosis of p. 3329-35.
Ther, 2008. 7(1): p. 110-20. cancer. Expert Rev Mol Diagn. 10(4): p. 417-34. 43 Wise, DR et al. Myc regulates a
5 Yalcin, A et al. Selective inhibition of choline 24 Tennant, DA et al. Metabolic transformation transcriptional program that stimulates
kinase simultaneously attenuates MAPK and in cancer. Carcinogenesis, 2009. 30(8): p. 1269-80. mitochondrial glutaminolysis and leads to
PI3K/AKT signaling. Oncogene. 29(1): p. 139-49. 25 Cairns, RA, Harris, IS and Mak, TW. glutamine addiction. Proc Natl Acad Sci U S A,
6 Vander Heiden, MG. Targeting cancer Regulation of cancer cell metabolism. Nat Rev 2008. 105(48): p. 18782-7.
metabolism: a therapeutic window opens. Nat Cancer. 11(2): p. 85-95. 44 Kim, JW et al. Hypoxia-inducible factor 1
Rev Drug Discov. 10(9): p. 671-84. 26 Figueroa, ME et al. Leukemic IDH1 and IDH2 and dysregulated c-Myc cooperatively induce
7 Warburg, O, Wind, F and Negelein, E. The mutations result in a hypermethylation vascular endothelial growth factor and
Metabolism of Tumors in the Body. J Gen phenotype, disrupt TET2 function, and impair metabolic switches hexokinase 2 and pyruvate
Physiol, 1927. 8(6): p. 519-30. hematopoietic differentiation. Cancer Cell. dehydrogenase kinase 1. Mol Cell Biol, 2007.
8 Koppenol, WH, Bounds, PL and Dang, CV. 18(6): p. 553-67. 27(21): p. 7381-93.
Otto Warburg’s contributions to current 27 Yan, H et al. IDH1 and IDH2 mutations in 45 Shim, H et al. c-Myc transactivation of LDH-
concepts of cancer metabolism. Nat Rev gliomas. N Engl J Med, 2009. 360(8): p. 765-73. A: implications for tumor metabolism and
Cancer. 11(5): p. 325-37. 28 Mazurek, S. Pyruvate kinase type M2: a key growth. Proc Natl Acad Sci U S A, 1997. 94(13):
9 DeBerardinis, RJ et al. The biology of cancer: regulator within the tumour metabolome and a p. 6658-63.
metabolic reprogramming fuels cell growth and tool for metabolic profiling of tumours. Ernst 46 Semenza, GL. Regulation of Metabolism by
proliferation. Cell Metab, 2008. 7(1): p. 11-20. Schering Found Symp Proc, 2007(4): p. 99-124. Hypoxia-Inducible Factor 1. Cold Spring Harb
10 Vander Heiden, MG, Cantley, LC and 29 Hanahan, D and Weinberg, RA. Hallmarks of Symp Quant Biol.
Thompson, CB. Understanding the Warburg cancer: the next generation. Cell. 144(5): 47 Lum, JJ et al. The transcription factor HIF-1␣
effect: the metabolic requirements of cell p. 646-74. plays a critical role in the growth factor-
proliferation. Science, 2009. 324(5930): 30 Collier, JJ et al. c-Myc is required for the dependent regulation of both aerobic and
p. 1029-33. glucose-mediated induction of metabolic anaerobic glycolysis. Genes Dev, 2007. 21(9):
11 Dang, CV. The interplay between MYC and enzyme genes. J Biol Chem, 2003. 278(8): p. 1037-49.
HIF in the Warburg effect. Ernst Schering Found p. 6588-95. 48 Semenza, GL. HIF-1: upstream and
Symp Proc, 2007(4): p. 35-53. 31 Dang, CV, Le, A and Gao, P. MYC-induced downstream of cancer metabolism. Curr Opin
12 Reivich, M et al. Measurement of local cancer cell energy metabolism and therapeutic Genet Dev. 20(1): p. 51-6.
cerebral glucose metabolism in man with 18F-2- opportunities. Clin Cancer Res, 2009. 15(21): 49 Gordan, JD, Thompson, CB and Simon, MC.
fluoro-2-deoxy-d-glucose. Acta Neurol Scand p. 6479-83. HIF and c-Myc: sibling rivals for control of
Suppl, 1977. 64: p. 190-1. 32 Elstrom, RL et al. Akt stimulates aerobic cancer cell metabolism and proliferation. Cancer
13 Busk, M et al. Inhibition of tumor lactate glycolysis in cancer cells. Cancer Res, 2004. Cell, 2007. 12(2): p. 108-13.
oxidation: consequences for the tumor 64(11): p. 3892-9. 50 Podar, K and Anderson, KC. A therapeutic
microenvironment. Radiother Oncol. 99(3): 33 Fan, Y, Dickman, KG and Zong, WX. Akt and role for targeting c-Myc/Hif-1-dependent
p. 404-11. c-Myc differentially activate cellular metabolic signaling pathways. Cell Cycle. 9(9): p. 1722-8.
14 Fowler, JS and Ido, T. Initial and subsequent programs and prime cells to bioenergetic 51 Kaelin, WG, Jr. The von Hippel-Lindau
approach for the synthesis of 18FDG. Semin inhibition. J Biol Chem. 285(10): p. 7324-33. tumour suppressor protein: O2 sensing and
Nucl Med, 2002. 32(1): p. 6-12. 34 Robey, RB and Hay, N. Is Akt the “Warburg cancer. Nat Rev Cancer, 2008. 8(11): p. 865-73.
15 Shani, J et al. Distribution of 18F-5- kinase”?-Akt-energy metabolism interactions 52 Bobarykina, AY et al. Hypoxic regulation of
fluorouracil in tumor-bearing mice and rats. Int J and oncogenesis. Semin Cancer Biol, 2009. PFKFB-3 and PFKFB-4 gene expression in
Nucl Med Biol, 1978. 5(1): p. 19-28. 19(1): p. 25-31. gastric and pancreatic cancer cell lines and
16 Zhu, A, Lee, D and Shim, H. Metabolic 35 Furuta, E et al. Metabolic genes in cancer: expression of PFKFB genes in gastric cancers.
positron emission tomography imaging in cancer their roles in tumor progression and clinical Acta Biochim Pol, 2006. 53(4): p. 789-99.
detection and therapy response. Semin Oncol. implications. Biochim Biophys Acta. 1805(2): p. 53 Obach, M et al. 6-Phosphofructo-2-kinase
38(1): p. 55-69. 141-52. (pfkfb3) gene promoter contains hypoxia-
17 Dang, CV. Glutaminolysis: supplying carbon 36 Bensaad, K and Vousden, KH. p53: new roles inducible factor-1 binding sites necessary for
or nitrogen or both for cancer cells? Cell Cycle. in metabolism. Trends Cell Biol, 2007. 17(6): transactivation in response to hypoxia. J Biol
9(19): p. 3884-6. p. 286-91. Chem, 2004. 279(51): p. 53562-70.
18 Dang, CV. Rethinking the Warburg effect 37 Cheung, EC and Vousden, KH. The role of 54 Koukourakis, MI et al. Pyruvate
with Myc micromanaging glutamine metabolism. p53 in glucose metabolism. Curr Opin Cell Biol. dehydrogenase and pyruvate dehydrogenase
Cancer Res. 70(3): p. 859-62. 22(2): p. 186-91. kinase expression in non small cell lung cancer
19 Wise, DR and Thompson, CB. Glutamine 38 Gottlieb, E and Vousden, KH. p53 regulation and tumor-associated stroma. Neoplasia, 2005.
addiction: a new therapeutic target in cancer. of metabolic pathways. Cold Spring Harb 7(1): p. 1-6.
Trends Biochem Sci. 35(8): p. 427-33. Perspect Biol. 2(4): p. a001040. Continued on page 74

Drug Discovery World Fall 2011 73


Metabolism_Layout 1 04/10/2011 17:39 Page 74

Drug Discovery

Continued from page 73 72 Mathupala, SP, Heese, C and Pedersen, PL. 89 Christofk, HR et al. The M2 splice isoform of
55 Luo, W and Semenza, GL. Pyruvate kinase Glucose catabolism in cancer cells. The type II pyruvate kinase is important for cancer
M2 regulates glucose metabolism by functioning hexokinase promoter contains functionally metabolism and tumour growth. Nature, 2008.
as a coactivator for hypoxia-inducible factor 1 in active response elements for the tumor 452(7184): p. 230-3.
cancer cells. Oncotarget. 2(7): p. 551-6. suppressor p53. J Biol Chem, 1997. 272(36): 90 Christofk, HR et al. Pyruvate kinase M2 is a
56 Firth, JD, Ebert, BL and Ratcliffe, PJ. Hypoxic p. 22776-80. phosphotyrosine-binding protein. Nature, 2008.
regulation of lactate dehydrogenase A. 73 Kurtoglu, M, Maher, JC and Lampidis, TJ. 452(7184): p. 181-6.
Interaction between hypoxia-inducible factor 1 Differential toxic mechanisms of 2-deoxy-D- 91 Hitosugi, T et al. Tyrosine phosphorylation
and cAMP response elements. J Biol Chem, glucose versus 2-fluorodeoxy-D-glucose in inhibits PKM2 to promote the Warburg effect
1995. 270(36): p. 21021-7. hypoxic and normoxic tumor cells. Antioxid and tumor growth. Sci Signal, 2009. 2(97): p. ra73.
57 Izumi, H et al. Monocarboxylate transporters 1 Redox Signal, 2007. 9(9): p. 1383-90. 92 Cosentino, C, Grieco, D and Costanzo, V.
and 4 are involved in the invasion activity of human 74 Lampidis, TJ et al. Efficacy of 2-halogen ATM activates the pentose phosphate pathway
lung cancer cells. Cancer Sci. 102(5): p. 1007-13. substituted D-glucose analogs in blocking promoting anti-oxidant defence and DNA
58 Majewski, N et al. Hexokinase-mitochondria glycolysis and killing “hypoxic tumor cells”. repair. EMBO J. 30(3): p. 546-55.
interaction mediated by Akt is required to Cancer Chemother Pharmacol, 2006. 58(6): 93 Jiang, P et al. p53 regulates biosynthesis
inhibit apoptosis in the presence or absence of p. 725-34. through direct inactivation of glucose-6-
Bax and Bak. Mol Cell, 2004. 16(5): p. 819-30. 75 Maher, JC et al. Hypoxia-inducible factor-1 phosphate dehydrogenase. Nat Cell Biol. 13(3):
59 Vousden, KH. Functions of p53 in metabolism confers resistance to the glycolytic inhibitor 2- p. 310-6.
and invasion. Biochem Soc Trans, 2009. 37(Pt 3): deoxy-D-glucose. Mol Cancer Ther, 2007. 6(2): 94 Kruger, A and Ralser, M. ATM is a redox
p. 511-7. p. 732-41. sensor linking genome stability and carbon
60 Vousden, KH and Ryan, KM. p53 and 76 Singh, D et al. Optimizing cancer metabolism. Sci Signal. 4(167): p. pe17.
metabolism. Nat Rev Cancer, 2009. 9(10): radiotherapy with 2-deoxy-d-glucose dose 95 Perl, A et al. Oxidative stress, inflammation
p. 691-700. escalation studies in patients with glioblastoma and carcinogenesis are controlled through the
61 Bensaad, K. Cheung, EC and Vousden, KH. multiforme. Strahlenther Onkol, 2005. 181(8): pentose phosphate pathway by transaldolase.
Modulation of intracellular ROS levels by TIGAR p. 507-14. Trends Mol Med. 17(7): p. 395-403.
controls autophagy. EMBO J, 2009. 28(19): 77 Dwarakanath, BS et al. Clinical studies for 96 Coy, JF et al. Mutations in the transketolase-
p. 3015-26. improving radiotherapy with 2-deoxy-D-glucose: like gene TKTL1: clinical implications for
62 Bensaad, K et al. TIGAR, a p53-inducible present status and future prospects. J Cancer neurodegenerative diseases, diabetes and cancer.
regulator of glycolysis and apoptosis. Cell, 2006. Res Ther, 2009. 5 Suppl 1: p. S21-6. Clin Lab, 2005. 51(5-6): p. 257-73.
126(1): p. 107-20. 78 Prasanna,VK et al. Differential responses of 97 Langbein, S et al. Expression of transketolase
63 Matoba, S et al. p53 regulates mitochondrial tumors and normal brain to the combined TKTL1 predicts colon and urothelial cancer
respiration. Science, 2006. 312(5780): p. 1650-3. treatment of 2-DG and radiation in glioablastoma. patient survival: Warburg effect reinterpreted. Br
64 Stambolic, V et al. Regulation of PTEN J Cancer Res Ther, 2009. 5 Suppl 1: p. S44-7. J Cancer, 2006. 94(4): p. 578-85.
transcription by p53. Mol Cell, 2001. 8(2): 79 Dang, CV. MYC, microRNAs and glutamine 98 Liu, H et al. Fructose induces transketolase
p. 317-25. addiction in cancers. Cell Cycle, 2009. 8(20): flux to promote pancreatic cancer growth.
65 Macheda, ML, Rogers, S and Best, JD. p. 3243-5. Cancer Res. 70(15): p. 6368-76.
Molecular and cellular regulation of glucose 80 Gao, P et al. c-Myc suppression of miR-23a/b 99 Bauer, DE et al. ATP citrate lyase is an important
transporter (GLUT) proteins in cancer. J Cell enhances mitochondrial glutaminase expression component of cell growth and transformation.
Physiol, 2005. 202(3): p. 654-62. and glutamine metabolism. Nature, 2009. Oncogene, 2005. 24(41): p. 6314-22.
66 Ganapathy, V, Thangaraju, M and Prasad, PD. 458(7239): p. 762-5. 100 Hanai, JI et al. Inhibition of lung cancer
Nutrient transporters in cancer: relevance to 81 Seltzer, MJ et al. Inhibition of glutaminase growth: ATP citrate lyase knockdown and statin
Warburg hypothesis and beyond. Pharmacol preferentially slows growth of glioma cells with treatment leads to dual blockade of mitogen-
Ther, 2009. 121(1): p. 29-40. mutant IDH1. Cancer Res. 70(22): p. 8981-7. activated protein kinase (MAPK) and
67 Maher, JC et al. Differential sensitivity to 2- 82 Catane, R et al. Azaserine, DON, and phosphatidylinositol-3- kinase (PI3K)/AKT
deoxy-D-glucose between two pancreatic cell azotomycin: three diazo analogs of L-glutamine pathways. J Cell Physiol.
lines correlates with GLUT-1 expression. with clinical antitumor activity. Cancer Treat 101 Kuhajda, FP. Fatty acid synthase and cancer:
Pancreas, 2005. 30(2): p. e34-9. Rep, 1979. 63(6): p. 1033-8. new application of an old pathway. Cancer Res,
68 Wu, CH et al. In vitro and in vivo study of 83 Wang, JB et al. Targeting mitochondrial 2006. 66(12): p. 5977-80.
phloretin-induced apoptosis in human liver glutaminase activity inhibits oncogenic 102 Zhan, Y et al. Control of cell growth and
cancer cells involving inhibition of type II transformation. Cancer Cell. 18(3): p. 207-19. survival by enzymes of the fatty acid synthesis
glucose transporter. Int J Cancer, 2009. 124(9): 84 Pelicano, H et al. Glycolysis inhibition for pathway in HCT-116 colon cancer cells. Clin
p. 2210-9. anticancer treatment. Oncogene, 2006. 25(34): Cancer Res, 2008. 14(18): p. 5735-42.
69 Witte, D et al. Overexpression of the neutral p. 4633-46. 103 Nomura, DK et al. Monoacylglycerol lipase
amino acid transporter ASCT2 in human 85 Wang, J et al. A glycolytic mechanism regulates a fatty acid network that promotes
colorectal adenocarcinoma. Anticancer Res, regulating an angiogenic switch in prostate cancer pathogenesis. Cell. 140(1): p. 49-61.
2002. 22(5): p. 2555-7. cancer. Cancer Res, 2007. 67(1): p. 149-59. 104 Ye, L et al. Monoacylglycerol lipase (MAGL)
70 Kaadige, MR, Elgort, MG and Ayer, DE. 86 Zieker, D et al. Phosphoglycerate kinase 1 a knockdown inhibits tumor cells growth in
Coordination of glucose and glutamine promoting enzyme for peritoneal dissemination colorectal cancer. Cancer Lett. 307(1): p. 6-17.
utilization by an expanded Myc network. Transcr. in gastric cancer. Int J Cancer. 126(6): p. 1513-20. 105 Zaugg, K et al. Carnitine
1(1): p. 36-40. 87 Ren, F et al. Quantitative proteomics palmitoyltransferase 1C promotes cell survival
71 Pedersen, PL. Warburg, me and Hexokinase identification of phosphoglycerate mutase 1 as a and tumor growth under conditions of
2: Multiple discoveries of key molecular events novel therapeutic target in hepatocellular metabolic stress. Genes Dev. 25(10): p. 1041-51.
underlying one of cancer’s most common carcinoma. Mol Cancer. 9: p. 81. 106 Tennant, DA, Duran, RV and Gottlieb, E.
phenotypes, the “Warburg Effect”, i.e., elevated 88 Vander Heiden, MG et al. Evidence for an Targeting metabolic transformation for cancer
glycolysis in the presence of oxygen. J Bioenerg alternative glycolytic pathway in rapidly therapy. Nat Rev Cancer. 10(4): p. 267-77.
Biomembr, 2007. 39(3): p. 211-22. proliferating cells. Science. 329(5998): p. 1492-9. Continued on page 75

74 Drug Discovery World Fall 2011


Metabolism_Layout 1 04/10/2011 17:39 Page 75

Drug Discovery

Continued from page 74 124 Christensen, BC et al. DNA methylation,


107 Pertega-Gomes, N et al. Monocarboxylate isocitrate dehydrogenase mutation, and survival
transporter 4 (MCT4) and CD147 in glioma. J Natl Cancer Inst. 103(2): p. 143-53.
overexpression is associated with poor prognosis 125 Colombo, SL et al. Anaphase-promoting
in prostate cancer. BMC Cancer. 11: p. 312. complex/cyclosome-Cdh1 coordinates glycolysis
108 Pinheiro, C et al. Monocarboxylate and glutaminolysis with transition to S phase in
transporter 1 is up-regulated in basal-like breast human T lymphocytes. Proc Natl Acad Sci U S A.
carcinoma. Histopathology. 56(7): p. 860-7. 107(44): p. 18868-73.
109 Pinheiro, C et al. Increasing expression of 126 Herrero-Mendez, A et al. The bioenergetic
monocarboxylate transporters 1 and 4 along and antioxidant status of neurons is controlled
progression to invasive cervical carcinoma. Int J by continuous degradation of a key glycolytic
Gynecol Pathol, 2008. 27(4): p. 568-74. enzyme by APC/C-Cdh1. Nat Cell Biol, 2009.
110 Pinheiro, C et al. Increased expression of 11(6): p. 747-52.
monocarboxylate transporters 1, 2, and 4 in 127 Almeida, A, Bolanos, JP and Moncada, S. E3
colorectal carcinomas. Virchows Arch, 2008. ubiquitin ligase APC/C-Cdh1 accounts for the
452(2): p. 139-46. Warburg effect by linking glycolysis to cell
111 Kennedy, KM and Dewhirst, MW. Tumor proliferation. Proc Natl Acad Sci U S A. 107(2):
metabolism of lactate: the influence and p. 738-41.
therapeutic potential for MCT and CD147 128 Bolanos, JP, Almeida, A and Moncada, S.
regulation. Future Oncol. 6(1): p. 127-48. Glycolysis: a bioenergetic or a survival pathway?
112 David, CJ et al. HnRNP proteins Trends Biochem Sci. 35(3): p. 145-9.
controlled by c-Myc deregulate pyruvate kinase
mRNA splicing in cancer. Nature. 463(7279):
p. 364-8.
113 Bluemlein, K et al. No evidence for a shift
in pyruvate kinase PKM1 to PKM2 expression
during tumorigenesis. Oncotarget. 2(5):
p. 393-400.
114 Dang, CV. PKM2 tyrosine phosphorylation
and glutamine metabolism signal a different view
of the Warburg effect. Sci Signal, 2009. 2(97):
p. pe75.
115 Lv, L et al. Acetylation targets the M2
isoform of pyruvate kinase for degradation
through chaperone-mediated autophagy and
promotes tumor growth. Mol Cell. 42(6):
p. 719-30.
116 Locasale, JW. Vander Heiden, MG and
Cantley, LC. Rewiring of glycolysis in cancer cell
metabolism. Cell Cycle. 9(21): p. 4253.
117 Vander Heiden, MG et al. Identification of
small molecule inhibitors of pyruvate kinase M2.
Biochem Pharmacol. 79(8): p. 1118-24.
118 Boxer, MB et al. Identification of activators
for the M2 isoform of human pyruvate kinase
Version 3.
119 Frezza, C, Tennant, DA and Gottlieb, E.
IDH1 mutations in gliomas: when an enzyme
loses its grip. Cancer Cell. 17(1): p. 7-9.
120 Gross, S et al. Cancer-associated metabolite
2-hydroxyglutarate accumulates in acute
myelogenous leukemia with isocitrate
dehydrogenase 1 and 2 mutations. J Exp Med.
207(2): p. 339-44.
121 Ward, PS et al. The common feature of
leukemia-associated IDH1 and IDH2 mutations
is a neomorphic enzyme activity converting
alpha-ketoglutarate to 2-hydroxyglutarate.
Cancer Cell. 17(3): p. 225-34.
122 Dang, L et al. Cancer-associated IDH1
mutations produce 2-hydroxyglutarate. Nature,
2009. 462(7274): p. 739-44.
123 Zhao, S et al. Glioma-derived mutations in
IDH1 dominantly inhibit IDH1 catalytic activity
and induce HIF-1␣. Science, 2009. 324(5924): p.
261-5.

Drug Discovery World Fall 2011 75

You might also like