Você está na página 1de 17

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/257749942

Proteomics and metabolomics in cancer drug


development

Article in Expert Review of Proteomics October 2013


DOI: 10.1586/14789450.2013.840440 Source: PubMed

CITATIONS READS

7 148

2 authors:

Angelo D'Alessandro Lello Zolla


University of Colorado Tuscia University
181 PUBLICATIONS 2,024 CITATIONS 303 PUBLICATIONS 5,781 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Hypertonicity View project

metabolomics View project

All content following this page was uploaded by Angelo D'Alessandro on 10 July 2014.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
Review

Proteomics and metabolomics


in cancer drug development
5
Expert Rev. Proteomics 10(5), 000000 (2013)

Angelo DAlessandro In this review article, the main recent advancements in the field of proteomics and
and Lello Zolla* metabolomics and their application in cancer research are described. In the second part of
the review the main metabolic alterations observed in cancer cells are thoroughly dissected, 10
Department of Ecological and Biological
Sciences, University of Tuscia, Largo especially those involving anabolic pathways and NADPH-generating pathways, which
dellUniversita`, snc, 01100 Viterbo, Italy indirectly affect anabolic reactions, other than the maintenance of the redox poise.
*Author for correspondence: Alterations to mitochondrial pathways and thereby deriving oncometabolites are also detailed.
Tel.: +39 0761 357 100
Fax: +39 0761 357 630
The third section of the review is a discussion of how and to what extent (mutations to)
zolla@unitus.it tumor suppressors and oncogenes end up influencing cancer cell metabolism and cell fate, 15
either promoting survival and proliferation or autophagy and apoptosis. In the last section of
the review, an overview is provided of therapeutic strategies that make use of metabolic
reprogramming approaches.

KEYWORDS: cancer drug development mass spectrometry metabolism proteomics

upon mitochondrial oxidative phosphorylation


Metabolomics, the new/old hallmark of in terms of ATP production (~18-fold lower
cancer efficiency) [3]. However, since generalization of a 25
Human tumor pathogenesis is characterized by Warburg-like metabolism seems to be also appli-
the progressive accumulation of changes to nor- cable to many rapidly dividing embryonic tis-
mal cells, changes that make cells evolve to a sues, a tentative explanatory and evidence-based
neoplastic state through the gradual acquisition theory posits that aerobic glycolysis might have
of a series of hallmark capabilities. This multi- evolved to meet the elevated anabolic demand 30
step process utterly enables normal cells to (for biosynthetic purposes) and favor the uptake
become tumorigenic and, ultimately malig- and incorporation of nutrients into biomass by
nant [1]. While metabolic reprogramming has rapidly dividing cells [3].
only recently been included in the list of the so- Conversely, over generalizations should be
called hallmarks of cancer [1], echoes from the avoided as well, since tumor cells do not 35
last (at least) 60 years of research already sug- always display a Warburg-like metabolism.
gested a crucial correlation between chronic and Indeed, some tumors are characterized by two
uncontrolled cell proliferation and deregulated subpopulations of cancer cells, one consisting
metabolism [2]. The Warburg effect, named of glucose-dependent cells that secrete lactate
after Otto Warburg, the first researcher to docu- (Warburg-wise), while a second subpopulation 40
ment an exception to the Pasteur effect (inhibi- almost symbiotically relies upon the secreted
tion of glycolysis in presence of oxygen) in lactate to sustain their energy production via
highly proliferating cancer cells, is based upon the tricarboxylic acid cycle (TCA cycle, also
the appreciation of an increased glycolytic rate, known as Krebs cycle) [1].
at the expenses of mitochondrial metabolism During the last decade, molecular evidences 45
(preferentially exploited by normal differentiated have underpinned a role for genetic reprog-
cells for energy production purposes), even in ramming in the metabolic regulation observed
the presence of oxygen [2]. This phenomenon, in cancer cells, a phenomenon that is often
often referred to as aerobic glycolysis, was at accompanied by the preferential expression of
first deemed to be counterintuitive, since rapidly cancer-specific isoforms of certain metabolic 50
proliferating cells are supposed to have higher enzymes, or rather by peculiar and recurrent
energy requirements, while a strictly glycolytic cancer-associated mutations, especially in genes
metabolism is less efficient than one relying coding for TCA cycle enzymes [4]. In the light

www.expert-reviews.com 10.1586/14789450.2013.840440  2013 Informa UK Ltd ISSN 1478-9450 1


Review DAlessandro & Zolla

of these observations, Warburgs hypothesis was recently revived analyses. In SILAC, trypsin cleavage thus exposes C-terminally
55 and expanded, as it became to be considered one key target for labeled arginine or lysine, which allows relative quantitation of
therapeutic treatments [5]. The implementation of novel mass each digestion-generated peptide, except for the C-terminus
spectrometry (MS)-based metabolomics and proteomics peptide of the protein [7,9]. While SILAC was at first optimized 110
approaches has boosted this area of research, delivering promis- for unicellular model organisms [7,9], its experimental design
ing results and suggesting new avenues for further research makes it suitable for cell culture experiments and thus amena-
60 development in the field of cancer biology, as we will attempt ble for in vivo/ex vivo cancer research investigations. A direct
to review in this paper. evolution of SILAC is super-SILAC [10], a method that com-
bines a mixture of multiple SILAC-labeled cell lines. Indeed, 115
Mass spectrometry-based proteomics & metabolomics SILAC is a particularly accurate quantitative method, although
Cancer proteomics still represents a mainstay in cancer research until recently it was limited to cell lines or animals that could
since the dawn of the post-genomic era [6]. The underlying be metabolically labeled with heavy amino acids. This limita-
65 assumption is that proteins can be regarded as the effectors of tion of SILAC in studying patient tumor samples has been
cellular functions and thus, in biological terms, protein profil- overcome through the use of a mix of multiple SILAC-labeled 120
ing might be more informative than mRNA profiling [7]. cell lines as an internal standard, a technique called super-
Nevertheless, despite almost two decades of efforts, the ambi- SILAC [10]. This mix achieved superior quantification accuracy
tious agenda pursuing the complete annotation of the physio- compared with a single SILAC-labeled cell line, owing to the
70 logical role of all known genes still remains unfulfilled [8]. generation of hundreds of thousands of isotopically labeled
From a technical standpoint, recent technical advancements peptides in appropriate amounts to serve as internal standards 125
have opened new scenarios in the field of proteomics. While a for MS [11].
decade ago separative and quantitative proteomics approaches Isotope-coded affinity tags (ICAT) are one of the most rap-
mainly relied upon 2DE-based analyses and the implementa- idly expanding in vitro labeling techniques for protein quantita-
75 tion of HPLC-MS-based workflows was only auspicated, cur- tion. ICAT is based upon specific tagging of cysteine residues
rent proteomics analyses actually take advantage of quantitative with a reagent containing either eight or no deuterium atoms, 130
analyses via chromatography-MS approaches. Novel instru- along with a biotin group that enables affinity purification strat-
ments have been indeed implemented (both at the HPLC and egies to recover and enrich labeled peptides prior to MS analy-
MS levelthe interested reader is referred to reference [7] for sis [7]. One major limitation of this technique is that it can be
80 further details), as well as bioinformatics suites and tools, which only applied to those proteins that contain cysteine residues.
simplified quantitative analysis by allowing peak alignment, Isobaric tags for relative and absolute quantification 135
detection, protein identification and attribution of post- (iTRAQ) is another important in vitro labeling strategy. In
translational modifications (PTMs). iTRAQ, tagging requires a reporter group, a balance group and
a peptide reactive group. The reactive group binds the N-
Quantitative proteomics terminus and side-chain amines of peptides, while the reporter
85 Among quantitative proteomics approaches that have been (up to eight different labeling patterns are possible) and the 140
extensively applied in the field of cancer research, three main balance group are designed as to achieve isobaric balancing in
strategies have gained momentum: i) in vivo labeling with stable MS mode, and discriminating fragments in collision induced
isotopes; ii) in vitro labeling and iii) label-free approaches [7]. dissociation (CID) mode for relative quantitation on the basis
The basic concept behind labeling strategies is that a stable- of the relative abundance of different reporter groups [7].
90 isotope labeled peptide shares identical chemical features with Another labeling strategy for quantitative proteomics implies 145
its native (unlabeled) counterpart, which results in identical the use of isotopomer labels, referred to as tandem mass tags
behavior during chromatographic separation and mass spectro- (TMTs) [12]. TMTs are designed to ensure that identical pepti-
metric analysis, though it still allows them to be differentiated des labeled with different isotopomers exactly comigrate in all
owing to their mass difference. Relative abundances can be chromatographic separations. On the other hand, peptides
95 then grasped by measuring the ratio of signal intensities for the from different samples can be identified and relatively quanti- 150
labeled and unlabeled peptide pairs under different biological fied using CID-based analysis method. Relative abundance
conditions [7]. measurements made in the MS/MS mode using the new tags
Stable isotope labeling of amino acid in culture (SILAC), is are accurate and sensitive [12].
probably the most extensively adopted in vivo labeling Another strategy aims at quantifying protein abundances by
100 approach in cancer research [9]. The SILAC protocol envisages targeting a so-called proteotypic peptide (defined as an experi- 155
cell culturing in media containing either normal amino acids mentally observable peptide that uniquely identifies a specific
or amino acids labeled with heavy isotopes. The labeled amino protein or protein isoform [13]) through selected reaction moni-
acids are often lysine and arginine (with different combinations toring (SRM)-MS, which enables isolation and quantitative
of 13C, 15N, and 3H). The choice to label these basic amino assaying of the expected mass to charge ratio (against standards
105 acids stems from the broadly diffused adoption of trypsin as or in silico predicted values) [14]. Proteotypic standard peptides 160
the protease of choice upstream of HPLC-MS proteomics could be used as external references to determine calibration

2 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

curves, as resulting from the correlation of MS readings (either Further developments in the field of cancer redox proteomics
peak intensities or peak areas) in response to a variation in pep- are awaited in the next few years.
tide concentrations. However, SRM is not necessarily a synonym
165 for label free quantitation. Early evolutions of the SRM Phosphoproteomics
approach imply the use of isotopically labeled synthetic peptides Despite the significant body of accumulating knowledge about
(SPIKETIDES [15,16] or AQUA peptides) to enable absolute genes involved in the development of human cancer (at least 220
quantification of specific proteins in a targeted fashion. The clear 300 have been discovered so far), only a limited number of can-
advantage over non-labeled peptides is that isotopically labeled cer genes encode for proteins that are suitable targets for effective
170 proteotypic peptides can be directly spiked in the sample to be drugs. In this view, protein kinases (such as Abl tyrosine kinase)
used as an internal reference, which also helps coping with any are among the best eligible targets for small molecule inhibi-
untoward technical bias at the nano HPLC or MS level. tors [18,19]. Indeed, sustaining proliferative signaling is a key hall- 225
While labeling-based quantitative strategies are rather expen- mark of cancer, which is often achieved through kinase-triggered
sive and often time consuming (especially in vivo, which might phosphorylation cascades [1]. It is thus pivotal to further our
175 require four to six replication cycles of cultured cells to achieve understanding of the biological role of protein phosphorylations
full labeling), label free approaches are increasingly attracting a through the introduction of novel analytical strategies to enhance
great deal of interest, owing to the reduced costs and ease of their detection. Within this context, big strides have been 230
implementation to classic HPLC-MS proteomics workflows. recently made in the field of phosphoproteomics. Phosphoryla-
Recently introduced post hoc algorithms allow calculation of the tion (mostly of S/T, and to a lesser extent to Y amino acid resi-
180 so-called exponentially-modified protein abundance index dues) is a reversible PTM that plays important regulatory
(emPAI), where the number of identified peptides, normalized functions in cellular signaling pathways, which can influence cell
against the number of all the possibly identifiable peptides for growth, differentiation, invasion, metastasis and apoptosis [18,19]. 235
a given protein, is used as an indicator of the absolute protein Since protein phosphorylations are often sub-stoichiometric,
abundance on a logarithmic basis. Other indicators of absolute enrichment strategies are often necessary to enable determina-
185 protein abundances include spectral counting and peak tion of differential phosphorylation events. Enrichment
intensities [7]. strategies are either based upon affinity chromatography
(immobilized metal ion affinity chromatography), titanium 240
Redox proteomics dioxide, zirconium dioxide, calcium phosphonate precipitation
Deregulation of metabolism in cancer cells is also intercon- or strong cation exchange or immunoprecipitation strategies.
nected with increased susceptibility to, and exacerbation of, oxi- Detection via non-MS methods mainly involves antibody-based
190 dative stress (as it will be extensively described in the following approaches, while MS-based approaches rather rely on isotopic
paragraphs). The key approaches to redox proteomics have labeling (ICAT, iTRAQ or 32P/33P) and/or alternative (to colli- 245
been recently reviewed [14,17]. Redox proteomics is a recent sion induced dissociationCID) fragmentation strategies, such
branch of proteomics that is devoted to the determination and, as electron transfer dissociation (ETD), which favors generation
possibly, quantification of oxidative modifications to proteins of c and z ions upon peptide fragmentation (instead of b and y
195 (including protein carbonylation, oxidation/nitrosylation of ions, which are predominant in CID) [19].
thiol groups and nitrosylation of tyrosines).
Among all redox modifications, oxidation of thiol groups Glycoproteomics 250
might affect the functional activity of several key enzymes (such Evading growth suppression and activating invasion and meta-
as metabolic and redox-homeostasis-related enzymes, including stasis (two key hallmarks of cancer [1]) is mainly achieved by
200 glyceraldehyde 3-phosphate dehydrogenase and peroxiredoxin cancer cells through the modulation of membrane proteins,
2) [14,17]. Owing to the labile nature of thiol groups-targeting which allow cancer cells to bypass contact-triggered growth-
oxidative modifications, an experimental strategy envisages the inhibitory signaling. Glycosylation is one of the most common 255
temporary quenching of free thiols (by means of trichloroacetic PTMs, estimated to be found in over 50% of human pro-
acid-based acidification or through the use of cell permeable teins [20] and more than 80% of membrane proteins [21]. Most
205 reagents for thiol-groups, such as the alkylating agents iodoace- membrane biomarkers of cancer cells, which are amenable to
tamide or N-ethylmaleimide) and subsequent specific reduction antibody-based therapies, are glycosylated proteins. Other than
(also with dithiothreitol, sodium arsenite or dimedone) [14,17]. extracellular membrane proteins, secreted proteins (which can 260
On the other hand, commercially available antibodies can be be thus searched for in the patients body fluids) are often
now exploited to determine the extent of S-nitrosylations, via N-glycosylated in the endoplasmic reticulum or Golgi appara-
210 enabling antibody-based enrichment strategies (immunoprecipi- tus. It is thus small a wonder that protein glycosylation is
tation) or direct immunoassay detection (ELISA, western blot). increasingly attracting a great deal of interest in the field of
Other analytical approaches also include biotin labeling for the cancer research. Glycosylations can be further distinguished 265
enrichment of S-glutathionylated peptides or isotope labeling into: i) N-linked glycosylation, ii) O-linked glycosylation
(especially ICAT, see above) to enrich, determine and quantify and iii) C-glycosylations. In like fashion to phosphoproteomics,
215 S-oxidative modifications [14,17]. glycoproteomics approaches often rely upon preliminary

www.expert-reviews.com 3
Review DAlessandro & Zolla

enrichment strategies (lectin affinity or boronic acid chromatog- From proteomics to metabolomics: MALDI imaging &
270 raphy or immunoprecipitation) to selectively enrich those pro- MALDI-based metabolomics
teins bearing N-, O- or C-glycosylations. Glycosylated peptides Metabolomics is the global quantitative assessment of metabo- 325
can then be screened via the use of MS, either MALDI or ESI- lites (low molecular weight compounds below the 1.5 kDa
MS, the latter relying both on CID and ETD fragmentation threshold, including sugars, phosphate compounds, organic
modes [22]. acids, nucleosides, lipids and fatty acids or exogenous com-
275 Enriched glycosylated peptides can be thus released via enzy- pounds) in a biological system [31].
matic digestion (with PNGase F for N-glycosylations) and by While proteomics investigates the effectors influencing the 330
chemical methods (for O-glycan release) [20]. Alternative enzy- phenotype, metabolites are the phenotype itself. Indeed, the
matic digestion combinations can help further define the historical precursor to metabolomics can be traced back to early
arrangement of side-chain branches, the most challenging task clinical biochemistry approaches [32], while technical advance-
280 in glycoproteomics analyses to date [23]. ments in the field NMR and, subsequently, of MS [32,33] have
boosted the refinement of this old/new omics discipline. NMR 335
Imaging mass spectrometry was at first favored by machine accessibility, established data
One of the greatest advances over the last decade in the field of handling and the conservative nature of the analysis, which
MS-based cancer research has been the introduction of MALDI allows further testing downstream of NMR analyses on the
imaging approaches. Imaging MS is a molecular analytical tech- same samples. Conversely, MS has gradually complemented
285 nology that enables the simultaneous measurement of multiple and often replaced NMR owing to its higher sensitivity, which 340
analytes directly from intact tissue sections [24]. Histological fea- results in MS being less demanding in terms of minimum
tures of the sample, as gleaned through classic immunohisto- detectable concentrations of the analyte [33]. Also, MS-based
chemistry staining, can indeed be correlated with molecular metabolomics holds the potential to better discriminate metab-
species (proteins, peptides, lipids and metabolites) without the olites, thus improving coverage of the metabolome space, espe-
290 need for target-speci?c reagents such as antibodies. cially when performing upstream compound-class-specific 345
Imaging MS is based upon matrix spraying on suitably- chromatographic separations [32,33].
treated cryostat sections mounted on conductive indium titai- In likewise fashion to quantitative proteomics, quantitative
num oxide-coated slides. The preliminary treatment depends MS-based metabolomics can also rely upon SRM or multiple
on the molecular species under investigation (e.g., organic sol- reaction monitoring (MRM) approaches [3436], which allow
295 vents might interfere with on tissue lipid analyses). MALDI detection and absolute quantification of a compound and its 350
imaging can indeed be applied to determine molecular signa- fragments against a pure standard.
tures that are specific of a tumor tissue, while theoretically eas- However, most advanced metabolomics studies today rely
ing the individuation of tumor biomarkers and their upon post hoc alignment, peak detection and metabolite dis-
discrimination from tumor border biomarkers [25]. The possi- crimination without any a priori restriction: this approach also
300 bility to combine it with to routine immunohistochemical goes by the name of untargeted metabolomics [37] and is 355
approaches offers the opportunity to validate and complement already providing decisive insights into cancer biology. In par-
the information attainable from a biopsy while obtaining addi- ticular, a rather recent application of MS-based untargeted
tional complimentary proteomics/lipidomics/metabolomics-rele- metabolomics is based upon carbon flux during catabolism and
vant results. anabolism, via supplying 13C-labeled metabolic substrates
305 Indeed, imaging MS can be applied to obtain tissue profiles (mainly glucose and glutamine) [38]. This approach allows the 360
of proteins [26], peptides [27], lipids and metabolites [28] (includ- kinetics of energy fluxes to be monitored during molecular
ing drug metabolites, thus helping monitoring the efficiency of biology experiments on cell lines (e.g., upon induction or
a therapeutical treatment). Additionally, protocols have been silencing of an oncogene or tumor suppressor protein), thus
developed also to allow imaging analyses of formalin-fixed par- helping further refine the understanding of metabolic networks
310 affin-embedded tissue slices [29]. in normal and cancer cells. 365
One main limitation of imaging techniques is related to repro- Biomedical application of MALDI MS is technically suited
ducibility (mainly affected by matrix depositing issues), although to monitoring metabolic variations directly on tissue sections
recently introduced automatic sprayers dramatically abated tech- from biopsies, but it also allows the screening of whole body
nical variability. Other limitations include the difficulty of moni- tissue sections from model organisms (e.g., mice) while looking
315 toring high molecular weight compounds (especially proteins for the metabolites derived from catabolism of a specific drug 370
above the 5060 kDa threshold) and the constraints related to under testing. This is relevant in the context of cancer research
the relative abundances of molecular species (e.g., most abundant since one of the key steps in drug discovery is the determina-
compounds are often easily visible, while low abundance ones are tion of the areas targeted by a therapeutic (body
hardly detectable through this approach). accumulation) [39].
320 However, the flexibility of the method makes it suitable for MALDI-imaging has recently been applied in cancer metab- 375
targeted quantitative approaches (such as SRM to low molecu- olomics research, especially in lipidomics analyses, since lipids
lar weight compounds, such as drugs [30]) directly on tissue. are highly preponderant and easily detectable through imaging

4 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

approaches. One clear advantage with


MALDI imaging approaches is that a
380 molecule can be directly detected on tis- MCT Glut1 GSR GLY SLC38A
GSSG GSH
sue, with a lateral resolution down to GSS
1015 mm with commonly available Glucose GLCY
NADPH NADPH
instruments (while in secondary ion HXK GCLM
imaging MS it can be improved up to G6P GL6P 6PG Ru5P CYST
G6PDH PGLS PGD
385 >50 nm [40]). These advancements have GNPNAT1 GFPT1 GPI
TKT
RPE RPIA GLUT
GlcNAc-6P GlcN-6P F6P X5P R5P GLS
paved the way for a broader application PGM FBPase PFK TALDO1
GLTM
of MALDI-based proteomics and GlcNAc-1P ALDOA
FBP E4P G3P TKT

F6P GLUD1
metabolomics strategies [41]. UAP1
DHAP G3P S7P
UDP-GlcNAc TPI GAPDH
MALDI-based metabolomics, on cell SBP
BPG
390 lysates or tissue homogenates instead of PGK ALDOA NADPH
tissue sections, would have several SER PSER 3PHPYR 3PG E4P DHAP
PSP PSAT 3PGDH PGAM
advantages over routinely used MS- SHMT 2PG ACO ISOCIT IDH
ENO
based metabolomics platforms (such as GLY PEP CITR -KET
PKM
LC-MS), in that it would be amenable Pyruvate
OGDH
ACLY SUCC-CoA
395 to automatization and multiplexing, LDH
PDH
Acetyl-CoA SUCCLG1
especially in combination with robotic Lactate OAA SUCC
MDH SD
auto samplers. MAL FD FUM

Anabolism & the Warburg effect Figure 1. An overview of the main catabolic pathways in normal and proliferat-
The recent introduction of specific ing cells. Glycolysis, Krebs cycle, PPP, serine synthesis, hexosamine synthesis and gluta-
400 metabolomics analytical platforms helped thione homeostasis are included. Enzymes are indicated in light grey, according to their
elucidating metabolic fluxes in highly relative UniProt names.
PPP: Pentose phosphate pathway.
proliferating cells [42,43]. In line with
Kilburns observations [44], dating back to
four decades ago, highly proliferating cells do not have extremely species (ROS) in the form of superoxide anions and hydroxyl
405 higher energy demands (in terms of glucose metabolization) in radicals, which ends up fertilizing the tumor microenviron-
comparison to resting cells. This at least in part justifies the met- ment [46] and promotes the accumulation of further muta-
abolic choice of oxidizing glucose via glycolysis while depressing tions to oncogenes and tumor suppressors [47]. Of note, the 435
the TCA cycle in cancer cells. However, an elevated replication impairment in ROS modulation/production in mitochondria
rate is based upon the accumulation of building blocks to build is often accompanied by mutations to electron transport
410 up mass and cell constituents before replication. In this scenario, chain components [48].
metabolomics analyses contributed significant insights by demon- In parallel, most cancers share distinct features such as
strating how alternative metabolic pathways are indeed activated defects in certain mitochondrial enzymes, including isoci- 440
along with glycolysis, which promote anabolism by constantly trate dehydrogenase (IDH), fumarate dehydrogenase (FD)
providing key reducing coenzymes such as NADPH (that is piv- and succinate dehydrogenase (SD) [49,50]. Alterations to IDH
415 otal, e.g., in lipid synthesis) (FIGURE 1). In parallel to aerobic glycol- or isoform switching (IDH1 vs IDH2) promote the utiliza-
ysis, glucose utilization fuels the main NADPH-generating tion of a-ketoglutarate from glutamine metabolism via
pathway, the (oxidative phase of the) pentose phosphate pathway reductive carboxylation to isocitrate, to fuel acetyl-CoA pro- 445
(PPP). Over-activation of the PPP at the non-oxidative phase duction for fatty acid biosynthetic purposes [5153] or amino
fuels the generation of ribose phosphate substrates for nucleoside acid synthesis via oxaloacetate intermediates (FIGURE 2). Of
420 biosynthesis, another central step toward DNA replication in note, isocitrate to a-ketoglutarate conversion by cytosolic
proliferating cells. It is perhaps worthwhile to recall that IDH is associated with the production of NADPH, analo-
NADPH also plays a fundamental role in the recycling of oxi- gous to the conversion of malate (another TCA cycle inter- 450
dized glutathione (GSSG) back to the reduced form (GSH), mediate) to pyruvate by malic enzyme. Anomalies to
thus contributing substantially to the redox poise (FIGURE 1). IDH1 enzyme (R132H) result in 2-hydroxyglutarate pro-
425 In this complex metabolic scenario, mitochondria are not duction from a-ketoglutarate, which negatively affects a-
just innocent and inactive bystanders in cancer cells [45]. ketoglutarate-dependent dioxygenase enzyme activity and
Mitochondrial metabolism is indeed fueled by glutamine, promotes malignant progression of brain tumors and, in 455
which is one major nitrogen source for biosynthesis reactions particular, gliomas [54].
and carbon source (via glutamate-a-ketoglutarate intermedi- Anomalies to FD and SD results in the accumulation of
430 ate conversion) for the TCA cycle (FIGURE 1). At the same time, fumarate and succinate, respectively. These metabolites (along
mitochondrial activation fuels production of reactive oxygen with the aforementioned 2-hydroxyglutarate) have been recently

www.expert-reviews.com 5
Review DAlessandro & Zolla

Fatty acid view, fumarate and succinate have been recently referred to as
Synthesis oncometabolites. Also, HIF accumulation diverts IDH-
dependent reductive carboxylation fluxes toward fatty acid 470
Reductive carboxylation
anabolism [56], which further stresses the intertwinement of
ISOCIT IDH
metabolic deregulation with proliferative capacity (FIGURE 3) [57].
CITR -KET GLUT GLTM
Cell proliferation, tumor suppressors, oncogenes &
SUCC-CoA
Acetyl-CoA metabolism
OAA SUCC Metabolic deregulation in cancer cells is partly the cause and 475
mostly the effect of genetic deregulation, at the oncogene and
MAL FUM tumor suppressor level [58]. As described in the previous para-
Amino acid
Synthesis
graph, these metabolic adjustments serve to build up anabolic
products to pursue cell proliferation [59]. At the same time, they
Figure 2. Cancer is often associated with mutations to TCA promote deregulation of oxidative phosphorylation and mito- 480
cycle enzymes, or expression of specific isoforms. In the chondrial events, thus favoring ROS accumulation and altera-
case of IDH, this results in the promotion of reductive carboxyla- tions to the tumor microenvironment. Exacerbation of oxidative
tion from glutamine-derived a-ketoglutarate, instead of regular
stress promotes senescence-like phenomena in cancer cells, while
TCA cycle fluxing towards succinyl Co-A. This utterly results in
acetyl Co-A accumulation, which promotes fatty acid synthesis decreasing glucose uptake, deregulating matrix attachment [59].
and, thus, cell growth and proliferation. At the same time, cancer cells cope with the excess of ROS by 485
IDH: Isocitrate dehydrogenase; TCA: Tricarboxylic acid cycle. activating pro-survival pathways, through the deregulation of
specific oncogenes [60], often complementary to inactivating or
gain of function mutations to tumor suppressor proteins, such
460 found to play a key role as direct inhibitor of dioxygenase and as proteins from the p53 family (p53, p63 and p73), which
AQ2 prolyl hydrolase, enzymes that indirectly catalyze the degrada- normally act as the guardians of the genome stability. One para- 490
tion of the hypoxia-inducible factor (HIF) [5153]. This inhibi- digmatic example is indeed represented by mutations to p53 (e.
tory phenomenon is relevant since accumulation of HIF, which g., R175H and R273H [61]), resulting in cell survival, increased
normally occurs under hypoxia, mediates the activation of pyr- proliferation and promotion of invasiveness.
465 uvate dehydrogenase kinase (PDK1), which in turn inhibits
pyruvate dehydrogenase and thus hinders conversion of pyru- The double role of p53 family members
vate to acetyl-CoA and shunts pyruvate to lactate [55]. In this While early cancer investigation studies indicated that p53 and 495
retinoblastoma protein (RB) mutations (comprehensively
Glucose detected in the great majority of tumors) mainly resulted in the
loss of function of their tumor-suppressor activity [62], recent
study indicate how these proteins (especially p53) might play a
more complex role in modulating the balance of pro-survival 500
ISOCIT and pro-apoptotic signaling, a function that escapes regulation
CITR -KET upon the acquisition of specific mutations to these proteins [63].
Pyruvate
Tp53, for example, is now known to take part in metabolic
SUCC-CoA
Acetyl-CoA modulation at several levels [64]. Analogous roles are increas-
PDH
Lactate OAA SUCC ingly emerging for all the members of the p53 family [65]. For 505
MDH SD
PDK MAL FD FUM example, p53 can inhibit the expression of the glucose trans-
porters GLUT1 and GLUT4 [64], and can increase the expres-
HIF PDH
sion of Tp53-inducible glycolysis and apoptosis regulator
(TIGAR), a fructose-bisphosphatase that inhibits glycolysis by
Figure 3. Cancer is often associated to mutations to Krebs
cycle enzymes, including SD, FD and MDH. These mutations reducing cellular levels of fructose-2,6-bisphosphate and thus 510
end up blocking TCA cycle catabolic fluxes, and promote the promoting a shift backward to the PPP, a process that pro-
accumulation of the respective substrates of these enzymes, suc- motes NADPH accumulation and plays a role in anti-apoptotic
AQ3 cinate, fumarate ad malate. These oncometabolites inhibit prolyl signaling via ROS-damage protection and promotes anabolic
hydrolase activity, thereby indirectly resulting in HIF stabilization. pathways, as summarized in the previous paragraphs [6668]
In turn, this promotes PDK1 activity, an inhibitory enzyme of
pyruvate dehydrogenase. This results in increased lactic acid (FIGURE 4). Besides, p53-responsive elements are present in the 515
fermentation and decreased fluxes from glycolysis to the promoters of PGM and hexokinase II (HK2), which is sugges-
TCA cycle. tive of the fact that p53 can promote at least some steps in gly-
FD: Fumarate dehydrogenase; HIF: Hypoxia-inducible factor; colysis. Of note, under hypoxic conditions, mitochondrial
MDH: Malate dehydrogenase; PDK1: Pyruvate dehydrogenase kin- localization of TIGAR stimulates HK2 (often bound to mito-
ase; SD: Succinate dehydrogenase; TCA: Tricarboxylic acid cycle.
chondrial membrane in tumors) [66]. 520

6 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

Other than p53, a transactivation-proficient isoform of field of cancer drug research are aimed at evaluating the effects 575
p73 has been recently shown to play a role in the promotion of starvation on cancer cells. One simple approach to promote
of a metabolic shift toward the PPP, via the upregulation of cancer cell starvation in vivo would be to prevent angiogenesis,
enzymes involved in the oxidative phase of the pentose phos- which would reduce the blood flux and thus oxygen and
525 phate shunt such as 6-phosphogluconolactonase [69]. nutrient delivery [78]. Anti-angiogenic therapy mainly relies
Tp53 also regulates the expression of sestrin proteins, which upon the administration of anti-VEGF-targeting monoclonal 580
activate AMPK to regulate growth and autophagy but also antibodies (e.g., bevacizumab).
function as antioxidants, protecting cells from hydrogen Within this framework, in vitro metabolomics experiments
peroxide-induced damage [65]. Furthermore, also in terms of have recently provided a clearer understanding of the mecha-
530 antioxidant defenses, p53 triggers the activation of glutathione nisms underlying the effects of starvation on cancer cells. Since
peroxidase, aldehyde dehydrogenase and tumor protein the main metabolic substrates for carbon and nitrogen build up 585
p53-inducible nuclear protein 1 (TP53INP1), all of them dis- in cancer cells are both glucose and glutamine, starvation
playing antioxidant functions. experiments have been so far performed through the supple-
However, it should not be forgotten that many p53-induci- mentation of cell media that are depleted of these two com-
535 ble proteins participate in apoptotic responses via the promo- pounds [7983]. A complex scenario emerged whereby glucose
tion of ROS production, including p53-induced gene might represent the main energy source for certain cell lines 590
3 (PIG3), proline oxidase, BAX, PUMA and p66SHC [65], as (such as head and neck squamous carcinoma cells [79]), while
well as of cytochrome C oxidase 2 [70]. only glutamine depletion actually triggered apoptosis in other
Tp53 further influences mitochondrial metabolism by pro- cell lines [83].
540 moting the expression of glutaminase 2, glutaminases being a Other than glucose and glutamine, cancer cells necessitate
family of enzymes responsible for glutamine to glutamate con- serine to support anabolism by providing precursors for biosyn- 595
version [71]. Analogous effects on glutaminase expression have thesis of proteins, nucleotides, creatine, porphyrins, phospholi-
been recently reported also for p63 [72]. In turn, glutamate is a pids and glutathione. Also, up-regulation of the serine synthesis
pivotal constituent of the tripeptide GSH, or rather it can be pathway occurs in some breast cancers [84,85]. It has been
545 further metabolized to a-ketoglutarate, that can either undergo recently demonstrated that p53-mediated cell responses to ser-
reductive carboxylation to isocitrate or further oxidation to ine starvation involve over-activation of the serine synthesis 600
succinyl-CoA via the TCA cycle. pathway. Besides, it promotes inhibition of glycolysis, since a
Energy or nutrient deprivation, extreme environmental con- rate-limiting enzyme, in particular, the specific and less efficient
ditions or Ca2+ release from the lumen of the endoplasmic cancer isoform, pyruvate kinase M2 (PKM2) [86], is allosteri-
550 reticulum (ER) results in the disruption of proper protein- cally activated by serine and thus, serine starvation, ends up
folding activity in this organelle, a condition that promotes the inhibiting glycolysis (FIGURE 4) [87]. This prompts two main 605
so-called ER-stress. ER-stress has been observed to influence effects: increase in TCA cycle fluxes to cope with the decreased
p53 stability by modulating its differential phosphorylation to ATP production, and an increase in PPP fluxes, to generate
serine 315 and serine 376, which promotes p53 localization in NADPH and thus cope with oxidative stress arising from the
555 the cytoplasm and its degradation [73,74]. Conversely, other elevation in TCA cycle-dependent energy production [85]. Of
p53 family members, p63 and p73, have been shown to pro- note, PKM2 expression in cancer cells results in relatively 610
mote ER stress and scotin (protein shisa-5) [75,76], suggesting an decreased glycolytic rates, which promotes accumulation of
intricate cross-talk among p53-family members, other than early glycolysis intermediates, including 3-phosphoglycerate, a
direct competition for p53 responsive elements or oligomeriza- precursor to serine de novo synthesis.
560 tion through direct binding. In this view, it is interesting to Serine metabolism is also at the crossroads between p53 and
note that almost pleiotropic metabolic effects are also expected starvation-induced autophagic responses [88]. Autophagy is a 615
for other p53 family members, since, for example, catabolic mechanism that promotes cell degradation of unneces-
TAp73 deletion reduces cellular ATP levels, oxygen consump- sary or dysfunctional cellular components through the lysoso-
tion and mitochondrial complex IV activity, with increased mal machinery to cope with the decrease in energy and
565 ROS production and oxidative stress sensitivity [77]. This phe- anabolic resources, and it is often regarded as: i) an alternative
nomenon involves the mitochondrial complex IV subunit cyto- option cells might choose to commit suicide, other than apop- 620
chrome C oxidase subunit 4 (Cox4i1), which is a direct tosis, ii) a cells defensive strategy upon cell damage or iii) a
TAp73 [77]. cells major adaptive (survival) strategy to cope with metabolic
stress, such as nutrient deprivation, or starvation in general.
Metabolic starvation experiments Starvation, the most extensively investigated inducer of auto-
570 On the basis of the evidences for modest energy demands, phagic responses, triggers the activation of AMPK [89,90], a kin- 625
albeit extreme substrate uptake, especially of glucose and gluta- ase that is activated by increased AMP/ATP ratios, and is
mine (we hereby purposely neglect uptake of lipids from the regulated to some extent by mammalian target of rapamycin
medium, which would deserve an entirely dedicated review) for (mTOR), a key molecular sensor for nutrient availability and a
anabolic and redox homeostasis purposes, new trends in the regulator of cell growth and proliferation [9195]. In particular,

www.expert-reviews.com 7
Review DAlessandro & Zolla

phosphatidylinositol (4,5)-biphosphate
(PIP2). In this way, PTEN modulates
GLUT1,
GLUT4 PIP3/PIP2 ratios and, indirectly, cell
Anabolism and Antioxidant responses
survival [96].
HXK
Glucose
NADPH NADPH
The list of oncogenes involved in meta- 655
bolic regulation is not only limited to HIF
G6P Oxidative phase PPP
and mTOR, but includes many other play-
F6P ers. One of those is MYC, which encodes
TIGAR Nucleoside a transcription factor c-Myc that links
Non-oxidative phase PPP
biosynthesis
p53 FBP altered cellular metabolism to tumorigene- 660
GLTM sis [97]. Indeed, c-Myc regulates genes
Bax
GLS2 involved in the biogenesis of ribosomes
GLUT and mitochondria, affects glucose and glu-
PUMA 3PG PEP
p66SHC Sestrins tamine metabolism, nucleotide metabolism
PKM2
Glutathione peroxidase
Cytochrome C oxidase 2 Pyruvate Lactate
Aldehyde dehydrogenase
and, along with E2F1, DNA replication 665
SER and miRNA expression [97]. Ectopic c-Myc
TP53INP1
cooperates with HIF to promote the
induction of a transcriptional program for
ROS
hypoxic adaptation, involving up-
Mitochondria regulation of glycolytic genes including lac- 670
Figure 4. Metabolic regulation by p53 at a glance. As a tumor suppressor, p53 is
tate dehydrogenase A, or the repression of
long known to promote apoptosis via enhancing pro-apoptotic factors and ROS- microRNAs (miRNAs) miR-23a/b to
generating mitochondrial metabolism. However, it recently emerged as a role for p53, as increase glutaminase protein expression
a pro-survival mediator under mild stress conditions. This phenomenon appears to involve and glutamine metabolism.
a TIGAR, a fructose-2,6-bisphosphatase that promotes accumulation of early glycolytic Recent flux-balance analyses have indi- 675
precursors thus boosting a diversion towards the PPP. The production of NADPH at the
non-oxidative phase of the PPP sustains anabolic and antioxidant processes. Evidences
cated a key role in glycolytic modulation
also indicate a role for p53 in the induction of anti-oxidant defenses (sestrins, glutathione (toward increase) and glutamine metabo-
peroxidase, aldehyde dehydrogenase, TP53INP1). At the same time, non-oxidative phase lism (towards reductive carboxylation) for
products fuel de novo nucleoside synthesis. Pro-survival effects mediated by p53 under another oncogene, KRAS [98,99]. In partic-
mild stress conditions appear to involve the homeostasis of serine metabolism (SER). ular, KRAS appears to be involved in the 680
PPP: Pentose phosphate pathway; ROS: Reactive oxygen species; TIGAR: Tp53-induced
glycolysis and apoptosis regulator; TP53INP1: Tumor protein p53-inducible nuclear
increase in glycolytic metabolism and
protein 1. channeling of glycolytic intermediates to
non-oxidative phase PPP reactions and
hexosamine biosynthesis (in turn promot-
630 autophagy initiation is associated with downregulation of ing protein glycosylation) [98]. Also, KRAS seems to promote an 685
mTOR complex 1 (mTORC1) activity. mTOR is a well- alternative pathway for glutamine metabolism to glutamate and
conserved serine/threonine kinase that belongs to the phosphoi- alternative downstream pathway. Such alternative route involving
nositide 3-kinase (PI3K)-related kinase family, and it plays an glutamate metabolism appears to be dependent on transami-
important role in the signaling network that controls growth nases, especially aspartate transaminases (GOT), which promote
635 and metabolism in response to environmental cues. The activa- glutamate and oxalacetate production from aspartate and a- 690
tion of mTORC1 (one of the two distinct multi-protein com- ketoglutarate (FIGURE 5). This results in oxaloacetate cytosolic accu-
plexes involving mTOR) requires glutamine and essential mulation (certain GOTs operate only in the cytosol), which
amino acids such as leucine, while it has been recently demon- prompts its conversion to malate (via malate dehydrogenase)
strated to also depend on availability of serine [87]. At the same and, through malic enzyme, to pyruvate, a reaction that concom-
640 time, Akt and AMPK communicate directly with mTORC1, itantly fuels NADPH production [99]. 695
by phosphorylating raptor (an mTORC1 component), leading Analogously, the tumor suppressor promyelocytic leukemia
to 14-3-3 binding and the allosteric inhibition of mTORC1 [94]. (PML) gene acted as both a negative regulator of PPARg coac-
Activation of mTORC1 promotes protein synthesis by phos- tivator 1A (PGC1A) acetylation and a potent activator of
phorylating the kinase S6K and the translation regulator 4E- PPAR signaling and fatty acid oxidation in breast cancer
645 BP1 and lipid biogenesis, via activation of SREBP and PPARg cells [100]. Finally, it is at least worth mentioning the long time 700
transcription factors [94,95]. The phosphatase and tensin homo- established role in metabolic regulation of the insulin-like
log (PTEN) interferes with the whole phosphoinositide 3-kin- growth factors (IGF-I and IGF-II) system [101].
ase (PI3K)/Akt/mTORC1 axis, by dephosphorylating MicroRNAs (miRNAs) are small RNA molecules that regu-
phosphatidylinositol (3,4,5)-trisphosphate (PIP3) at the 3 posi- late gene expression post-transcriptionally [97]. As anticipated in
650 tion of the phosphate of the inositol ring, thus producing the previous paragraphs, miRNA expression can be more or 705

8 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

less directly involved in metabolic mod-


ulation [102105]. Small RNAs may have
an intrinsic function in tumor suppres-
sion, since their levels are globally Cytosol
710 decreased in human cancers cells [88]. In CO2
NADPH NAD+
line with this, the transcription of some ME1 MDH1 GOT1
miRNA genes (such as miR-34) is regu- Lactate PYR MAL OAA ASP GLTM
GLS
lated by p53 [102]. Of note, miR-34a GLUT CYST
appears to be a key regulator of hepatic
715 lipid homeostasis. Together with GLY
miR-34, other miRNAs play a key role GSH
in metabolic modulation, including
miR-33a and miR-33b, which have a ctive
Redu tion
crucial role in controlling cholesterol x yla
carbo
720 and lipid metabolism in concert with
-KET
their host genes, the sterol-regulatory
element-binding protein (SREBP) tran- Acetyl-CoA TCA cycle

TCA
scription factors [104]. Other metabolic OAA
miRNAs, such as miR-103 and
Mitochondrion
725 miR-107, regulate insulin and glucose
homeostasis [104], while Figure 5. Glutamine metabolism can follow different fates in normal and cancer
miR-143 regulates hexokinase-2 expres- cells. Glutamine can be converted to glutamate (via glutaminase enzymesGLS), and thus
sion in cancer cells [105]. AQ4 be metabolized to ketoglutarate, whereby it enters TCA cycle or rather promotes reduc-
tive carboxylation. In parallel, glutamate can represent a building block of the tripeptide
Induction of apoptosis via gene glutathione (GSH). An alternative route involves the malate-aspartate shuttle, a pathway
that involves glutamine-derived aspartate towards the accumulation of oxaloacetate and
730 therapy & metabolic malate intermediates in the cytosol. This pathway is relevant in that NADPH is produced
reprogramming to sustain anabolic and anti-oxidant pathways.
In the previous paragraphs we have sum- GSH: Glutathione; TCA: Tricarboxylic acid cycle.
marized how cancer cells often suffer
from mutations that provide a competi-
735 tive edge over normal cells in terms of biomass accumulation and laboratory testing or already under clinical evaluation (since 760
proliferative capacity. Starting from Warburgs initial hypothesis, they are based on already commercialized drugs).
latest research has revealed that metabolic reprogramming occurs
as a consequence of mutations in cancer genes and alterations in Targeting glycolysis to modulate the Warburg effect
cellular signaling. Thus, we described how these mutations end Substantiation of the Warburg effect derives from the increased
740 up promoting certain pathways (glycolysis, PPP, nucleoside and glucose uptake by cancer cells and increased lactate production
fatty acid biosynthesis, NADPH-generating reactions) at the via glycolysis, even in presence of oxygen. On this ground, 765
expenses of others (above all, oxidative phosphorylation). Appreci- these characteristics have fostered the concept of new classes of
ation of these phenomena was a step forward from the canonic drugs that can be used either to monitor cancer cell metabo-
conception of the Warburg effect and opened new avenues for lism via state of the art diagnostic tools or to make it amenable
745 future developments in the field of cancer treatment [106]. There- to therapeutic interventions [111114].
fore, untuning the metabolic machine [99] might represent the For example, since tumors consume higher levels of glu- 770
new trend in cancer therapies, which might surprisingly be cose, clinicians have long been able to monitor tumor uptake
boosted by decades of research in the field of metabolism-related of a fluorine radioisotope of glucose, 18F-deoxyglucose, by
diseases, including diabetes and drugs for diabetes treatment (such FDG-PET. This technique has proven its usefulness in deter-
750 as metformin) [107]. mining the cancer stage, to identify metastatic sites and mon-
Other metabolic reprogramming strategies envisage the pro- itor treatment effectiveness. In parallel, a correlation has been 775
motion of oxidative stress via mitochondrial uncoupling [108]. observed between the initial degree of FDG-PET positivity
Another approach is related to caloric restriction, ketogenic diets and the overall patient outcome across cancer types and
and modulation of circulating nutrient levels through enzymes subtypes [111].
755 such as asparaginase, these strategies imply that tumor cells are Therapeutic intervention based upon drugs that target glyco-
more demanding in terms of nutrients in comparison to normal lytic enzyme activities are currently under evaluation. Among 780
cells [106110]. the possible targets, hexokinase (HXK), phosphofructokinase
In this section, we will briefly describe the main metabolic (PFK), glyceraldehyde 3-phosphate dehydrogenase (GADPH)
reprogramming strategies [105109] that are currently either under and lactate dehydrogenase (LDH) represent ideal therapeutic

www.expert-reviews.com 9
Review DAlessandro & Zolla

targets, since chemical inhibitors are already known (though recent strategy to promote autophagic responses relies upon the
785 they are often not extremely specific). administration of cannabinoid receptor agonists [120].
Known inhibitors of hexokinase include 2-deoxyglucose, Starvation (via nutrient deprivation) of cancer cells has been 840
3-bromopyruvate, 5-thioglucose and mannoheptulose. In par- proposed as a viable strategy to tackle cancer cell prolifera-
ticular, 3-bromopyruvate is a strong alkylating agent toward tion [121,122], especially when used in combination with
the free SH groups of cysteine residues in proteins, which chemotherapy [123,124].
790 might thus also affect those enzymes with thiol groups in their
active sites (such as GAPDH). Old drugs, new benefits
Lonidamine is known to inhibit only the mitochondria- Drug discovery is an extremely complicated, expensive and, 845
bound hexokinase, which is a distinctive feature of cancer cells most of the time a challenging (if not discouraging) area of
(please, refer to the previous paragraphs) [111114]. research. Patenting, testing and commercializing new effective
795 Use of a PFK inhibition strategy implies the suppression of drugs is a prohibitive task even for multi-national companies,
the PFKB3 isozyme, which controls the cellular level of fruc- which need to invest resources (in terms of funds and trained
tose-2,6-bisphosphate and thus affects the glycolytic flow by personnel) for more than two decades, before (when lucky) 850
allosterically-modulating PFK activity. receiving the final approval by US FDA. A record of currently
Known inhibitors of GAPDH include a-chlorohydrin, available drugs, the latest version of DrugBank (release 2.0),
800 ornidazole and iodoacetate, as well as the pentovalent includes a list of approximately 4900 drug entries, in which are
arsenate [111]. enlisted both FDA-approved small molecule and biotech
LDH-A can be knocked down in tumor cells by shRNAs, drugs [125]. Since metabolic diseases have long been investi- 855
thereby stimulating energy fluxes from pyruvate to mitochon- gated, especially those involving deregulation of glucose homeo-
dria, which in turn promotes mitochondrial uncoupling stasis, such as diabetes, a long list of currently commercialized
805 (ROS production, accumulation of pro-oxidant intermedi- drugs already exists that might be amenable for cancer treat-
ates) in those tumors bearing mutation of mitochondrial ment, in the light of the revisited role of the Warburg effect
enzymes. Targeting LDH might represent a cancer-cell sup- and metabolic reprogramming in cancer progression. Bigua- 860
pressing preferential strategy, while it could be less toxic to nides (such as metformin) belong to this category of old drugs
normal cells [111]. with potential new benefits. Biguanides were first isolated in
810 Other therapeutical interventions might involve the use of 1920 from the French lilac Galega officinalis, which was known
oxythiamine (a thiamine antagonist that inhibits transketolase to contain an agent that reduced the frequent urination associ-
and pyruvate dehydrogenase) or glufosfamide. Glufosfamide is ated with diabetes. Biguanides have step up to the spotlight for 865
a conjugate of glucose (highly consumed by cancer cells) and their ability to suppress liver gluconeogenesis, which is believed
ifosfamide, an alkylating agent with cytotoxic effects. Of note, to occur through activation of hepatic AMP-activated protein
815 glufosfamide is uptaken via the SAAT1 glucose transporter, kinase (AMPK) signaling [107]. It was but in recent years that
which is overexpressed in cancer cells [111114]. the antitumor effect of metformin could be observed, an effect
Owing to its pro-glycolytic potential, HIF-1-targeting drugs that is mediated by the activation of AMPK and thereby mod- 870
should be included, to a certain extent, in the same category of ulating the AMPK/mTOR pathways. At higher doses (than
glycolysis inhibitors [115]. Several classes of drugs have been physiologically achievable in vivo), metformin appears to
820 designed and tested over the years, targeting HIF transcription, directly affect mitochondrial oxidative phosphorylation in
synthesis, stability, heterodimerization, DNA-binding activity cancer cells.
or targets downstream to HIF-controlled signaling [115].
Targeting NAD-metabolism has recently emerged as a Targeting fatty acid synthesis & metabolism 875
potential therapeutic approach to tackle cancer cell prolifera- Owing to their highly proliferating nature, tumor cells need to
825 tion, since NAD undergoes crucial changes in cancer cells, build up new membrane to favor replication into daughter
whereby its use in transcription, DNA repair, cell cycle progres- cells. In this view, fatty acid synthesis and uptake from the
sion, apoptosis and metabolism processes is deregulated in medium (tumor microenvironment) are two key pathways that
comparison to normal cells [116]. have attracted a great deal of interest at least during the last 880
10 years [126]. Targeting fatty acid synthesis involves promoting
Targeting autophagy lipid lowering PPAR-pathways (via fenofibrate, also decreasing
830 Although autophagy can result in the suppression of tumor local angiogenesis) [127] or rather by addressing fatty acid oxida-
development, it may also mirror an extreme and desperate tion (FAO) [126,128]. While most cancer researchers focused on
attempt of the cancer cells to survive. Owing to the complexity glycolysis, glutaminolysis and fatty acid synthesis, the role of 885
of autophagic responses in mediating cancer survival/suppres- fatty acid oxidation in cancer cell metabolic transformation has
sion, several strategies have been proposed to tackle autophagy not been hitherto carefully examined [129].
835 over the last few years [117119]. FAO is inhibited by oxidative stress and, though indirectly,
Treatments with rapamycin (targeting mTOR), for example, it might contribute to counteracting ROS accumulation in can-
induce glucose starvation-like effects in cancer cell [119]. Another cer cells [129]. Indeed, FAO generates one molecule of acetyl 890

10 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

CoA in each oxidation cycle and two in the last cycle. Acetyl HSPs in anticancer vaccines, exploiting their ability to act as 945
CoA enters the Krebs cycle, and combines with oxaloacetate to immunological adjuvants [135].
give rise to citrate. As described in the previous paragraphs,
IDH-mediated cytosolic conversion of isocitrate to a- Expert commentary
895 ketoglutarate also produces cytosolic NADPH (for anabolic Big strides have been made over the last decade in the biologi-
and antioxidant purposes). This is the same pathway (though cal understanding of the phenomena underpinning cancer
in the opposite direction), according to which fatty acid synthe- metabolism deregulation. This accumulating body of laboratory 950
sis under hypoxic conditions (or when mitochondrial respira- science has paved the way for designing new therapeutical strat-
tion is limited) might rely upon glutamine-glutamate-a- egies and re-discovering of older drugs, with metabolism-
900 ketoglutarate generating reactions, thereby driving reductive regulatory aptitude.
carboxylation toward the accumulation of acetyl CoA, as a Mass spectrometry-based proteomics and metabolomics have
building block for fatty acid synthesis and elongation [130]. been at the core of these basic science advancements, which 955
It is also worthwhile recalling that glutamine-derived gluta- will undoubtedly translate into actual pharmacological applica-
mate generates NADPH when converted to a-ketoglutarate by tions within the next decades. Without the broader distribution
905 glutamate dehydrogenase (GLUD1). Also, since glutamine- of highly sensitive and accurate MS instruments and new quan-
derived a-ketoglutarate might fuel acetyl CoA accumulation titative (isotope labeling-based) strategies, cancer research would
and thus fatty acid synthesis, glutamine might represent a key have hardly had any chance to set even one single step into the 960
therapeutic target (e.g., at the glutaminase level, through DON deep forest of metabolic intricacies that we described in the
and azaserine) also when attempting to tackle lipid previous sections. Many molecular biologists have revised their
910 synthesis [131]. positions, firmly standing on a reductionistic ground, while
Potential pharmacological targets to inhibit FAO are repre- starting to complement classic molecular biology experimental
sented by carnitine palmitoyl transferase (CPT1), the rate- approaches (to put it cursorily, knock out one gene, induce 965
limiting enzyme in FAO, and 3-ketoacylthiolase (3-KAT), another gene, knock down another) with emerging
which catalyzes the final step in FAO [129,132] and ATP-citrate omics disciplines, theoretically encompassing the whole pro-
915 lyase (ACLY), a cytosolic enzyme that catalyzes the generation teome and metabolome (within the current capabilities of the
of acetyl CoA from citrate [133]. applied technique).
Metabolism-targeting compounds already include a broad 970
Targeting protein markers of cancer list of patented drugs and food derived nutrient/pharmaceuti-
In the present paper we mainly focused on metabolites or pro- cal-like molecules, also known as nutraceuticals [137]. Revisiting
tein targets mainly related to cancer metabolism. However, the Warburg effect with proteomics and metabolomics tools
920 there is a long list of emerging molecular markers of cancer has revealed that we might already have an old answer (com-
that is continuously expanding [134]. mercialized drugs) for a new, compelling question (tackling 975
One of the most promising classes of protein biomarkers cancer proliferation via metabolic reprogramming).
that are currently undergoing clinical testing is represented by
molecular chaperones of the heat shock protein (HSP) family. Five-year view
925 A wide range of human cancers is accompanied by overex- Further advancements will soon be achieved through the imple-
pression of HSPs, which are implicated in tumor cell prolifera- mentation of in silico prediction tools, based upon machine
tion, differentiation, invasion, metastasis, death and recognition learning algorithms, a wind of change that embraces the 980
by the immune system [135]. HSPs are useful biomarkers for broader concept of systems biology [138,139]. Based upon bioin-
carcinogenesis in some tissues and might be used as a valid formatic models, computer predictions will also help predicting
930 indicators of the degree of differentiation and the aggressiveness untoward effects (scarce efficiency, scarce specificity) of in vitro
of some cancers. Serum levels of HSP and HSP-specific anti- designed drugs [140]. Improvements in drug specificity and
bodies in cancer patients might help tumor diagnosis and be selectivity at the design phase will make it amenable to target 985
related to prognosis of specific cancers. Overexpression of cancer specific enzyme isoforms or mutations (PKM2, mito-
HSP27, for example, is associated with poor prognosis in gas- chondrial HXK2, monocarboxylate transporters MCT4 for lac-
935 tric, liver and prostate carcinoma and osteosarcomas [135]. On tate secretion [141]), by exploiting the concept of synthetic
the other hand, HSP70 [136] is correlated with poor prognosis lethality. As summarized by Kaelin [142], two genes are syn-
in breast, endometrial, uterine cervical and bladder carcinomas. thetic lethal if mutation of either alone is compatible with via- 990
HSPs might interfere with therapetuic treatments and/or bility but mutation of both leads to death. Therefore, it could
induce resistance to chemotherapy in breast cancer, leukemia be possible to target a gene that is synthetic lethal to a cancer-
940 patients and osteosarcomas. Two main strategies have been pro- relevant mutation, as to kill only cancer cells while sparing nor-
posed to target HSPs, depending on whether they are related mal cells [142].
to an improved or worsened prognosis of a specific cancer. Finally, as we hope it emerged from this paper, cancer cells 995
These strategies include: i) pharmacological modification of display an intricate network of intertwined and mutually-
HSP expression or molecular chaperone activity and ii) use of compensating metabolic pathways. Altering one node of the

www.expert-reviews.com 11
Review DAlessandro & Zolla

network might result in the compensation effect promoted by Financial & competing interests disclosure
the so-called plasticity of the metabolic network itself. There- A D Alessandro and L Zolla are supported by funds from the Italian
1000 fore, omics/systems biology-wise efforts should be pursued to National Blood Centre (Centro Nazionale SangueCNSIstituto Superiore 1010
unveil as many pathways as possible, and to gain an improved SanitaRome, Italy). The authors have no relevant affiliations or finan-
understanding of the role of already known, albeit underinvesti- cial involvement with any organization or entity with a financial interest
gated ones, such as the folate and mevalonate pathways (both in or financial conflict with the subject matter or materials discussed in
producing NADPH at some steps of the cycles). The final the manuscript. This includes employment, consultancies, honoraria, stock
1005 goal will be to determine, case by case, cancer by cancer, the ownership or options, expert testimony, grants or patents received or pend- 1015
most suitable targets for multi-targeted pharmaceutical ing or royalties.
interventions [143]. No writing assistance was utilized in the production of this manuscript.

1020 Key issues


The introduction of label-based and label-free quantitative proteomics and metabolomics analyses revived the field of
cancer metabolism.
Eighty years after Warburgs early observations, cancer metabolism is now deemed to be one key hallmark of cancer.
Most of the cancer-specific frequent mutations to tumor suppressors and oncogenes have the potential to affect cancer
1025 cells metabolism.
These changes often promote glycolysis at the expenses of oxidative phosphorylation in terms of energy production, other than anabolic
and NADPH-generating pathways. However, mitochondrial metabolism still plays a key role in producing oncometabolites with regula-
tory effects on key oncogenes, other than providing substrates for anabolic reactions (especially fatty acid de novo synthesis) and contri-
buting to the redox poise (via NADPH generation).
1030 Starvation promotes autophagy, which could provide a therapeutical intervention opportunity for some cancers. Induction of autophagy
might represent a viable alternative to induction of apoptosis.
Small molecule inhibitors of most of the key enzymes involved in the pathways described above are already available and deserve
further testing to understand their specificity and effectiveness. Cancer isoform-targeting (synthetic lethality) specific drugs must also
be designed as well.
1035 Among the thousands of commercially available drugs, those having a validated effect in the treatment of metabolic diseases may also
be suitable for the treatment of some cancers.

References 4 Frezza C, Pollard PJ, Gottlieb E. Inborn cell culture (SILAC) as a tool for prostate
and acquired metabolic defects in cancer. cancer research. Mol. Cell. Proteomics 3(7),
Papers of special note have been highlighted as:
J. Mol. Med. (Berl). 89(3), 213220 (2011). 729735 (2004).
of interest
5 DAlessandro A, Zolla L. Metabolomics and 10 Geiger T, Cox J, Ostasiewicz P,
of considerable interest
cancer drug discovery: let the cells do the Wisniewski JR, Mann M. Super-SILAC mix
1 Hanahan D, Weinberg RA. Hallmarks of talking. Drug Discov. Today 17(12), 39 for quantitative proteomics of human tumor
cancer: the next generation. Cell 144(5), (2012). tissue. Nat. Methods 7(5), 383385 (2010).
646674 (2011).
6 Bichsel VE, Liotta LA, Petricoin III EF. 11 Geiger T, Wisniewski JR, Cox J et al. Use
An instant classic on cancer biology, Cancer proteomics: from biomarker of stable isotope labeling by amino acids
whereby hallmarks of cancer are discovery to signal pathway profiling. Cancer in cell culture as a spike-in standard in
summarized and described into J. 7, 6978 (2001). quantitative proteomics. Nat. Protoc. 6,
details. 7 Bachi A, Bonaldi T. Quantitative proteomics 147157 (2011).
2 Warburg O. On the origin of cancer cells. as a new piece of the system biology puzzle. 12 Thompson A, Schafer J, Kuhn K et al.
Science 123, 309314(1956). J. Proteomics 71(3), 357367 (2008). Tandem mass tags: a novel quantification
Warburgs decade long observations are Quantitative proteomics approaches are strategy for comparative analysis of complex
summarized in this paper. reviewed and described into detail in this protein mixtures by MS/MS. Anal. Chem.
75(8), 18951904 (2003).
3 Vander Heiden MG, Cantley LC, paper.
Thompson CB. Understanding the Warburg 13 Kuster B, Schirle M, Mallick P,
8 Legrain P, Aebersold R, Archakov A et al.
effect: the metabolic requirements of Aebersold R. Scoring proteomes with
The human proteome project: current state
cell proliferation. Science 324, 10291033 proteotypic peptide probes. Nat. Rev. Mol.
and future direction. Mol. Cell. Proteomics
(2009) Cell. Biol. 6(7), 577583 (2005).
10(7), M111.009993 (2011).
14 DAlessandro A, Rinalducci S, Zolla L.
A thorough review encompassing metabolic 9 Everley PA, Krijgsveld J, Zetter BR,
Redox proteomics and drug development.
reprogramming events in cancer cells from Gygi SP. Quantitative cancer proteomics:
J. Proteomics 74(12), 25752595 (2011).
a Warburg-wise perspective. stable isotope labeling with amino acids in

12 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

15 Ang CS, Rothacker J, Patsiouras H, imaging protocol for in situ characterization 39 van Kampen JJ, Burgers PC, de Groot R,
Gibbs P, Burgess AW, Nice EC. Use of of tryptic peptide identity and distribution Gruters RA, Luider TM. Biomedical
multiple reaction monitoring for multiplex in formalin-fixed tissue. Rapid Commun. application of MALDI mass spectrometry
analysis of colorectal cancer-associated Mass Spectrom. 27(6), 655670 (2013). for small-molecule analysis. Mass Spectrom.
proteins in human feces. Electrophoresis 28 Monteiro MS, Carvalho M, Bastos ML, Rev. 30(1), 101120 (2011).
32(15), 19261938 (2011). de Pinho PG. Metabolomics analysis for 40 Klitzing HA, Weber PK, Kraft ML.
16 Schnatbaum K, Zerweck J, Nehmer J, biomarker discovery: advances and challenges. Secondary ion mass spectrometry imaging
Wenschuh H, Schutkowski M, Reimer U. Curr. Med. Chem. 20(2), 257271 (2013). of biological membranes at high spatial
SpikeTidesproteotypic peptides for 29 Casadonte R, Caprioli RM. Proteomic resolution. Methods Mol. Biol.
large-scale MS-based proteomics. Nature analysis of formalin-fixed paraffin-embedded 950, 483501 (2013).
Methods 8 (Application note) (2011). tissue by MALDI imaging mass 41 Issaq HJ, Fox SD, Chan KC, Veenstra TD.
17 Bachi A, Dalle-Donne I, Scaloni A. Redox spectrometry. Nat. Protoc. 6(11), Global proteomics and metabolomics in
proteomics: chemical principles, 16951709 (2011). cancer biomarker discovery. J. Sep. Sci.
methodological approaches and biological/ 30 Picotti P, Aebersold R. Selected reaction 34(24), 34843492 (2011).
biomedical promises. Chem. Rev. 113(1), monitoring-based proteomics: workflows, 42 Dang CV. Links between metabolism
596698 (2013). potential, pitfalls and future directions. Nat. and cancer. Genes Dev. 26(9), 877890
The most thorough review on redox Methods 9(6):555566 (2012). (2012).
proteomics to date. 31 Putri SP, Yamamoto S, Tsugawa H, 43 Lopez-Lazaro M. The warburg effect: why
18 Ashman K, Villar EL. Phosphoproteomics Fukusaki E. Current metabolomics: and how do cancer cells activate glycolysis
and cancer research. Clin. Transl. Oncol. technological advances. J. Biosci. Bioeng. in the presence of oxygen? Anticancer Agents
11(6), 356362 (2009). 116(1), 916 (2013). Med. Chem. 8(3), 305312 (2008).
19 Engholm-Keller K, Larsen MR. 32 DAlessandro A, Giardina B, Gevi F, 44 Kilburn DG, Lilly MD, Webb FC. The
Technologies and challenges in large-scale Timperio AM, Zolla L. Clinical energetics of mammalian cell growth. J. Cell
phosphoproteomics. Proteomics 13(6), metabolomics: the next stage of clinical Sci. 4(3), 645654 (1969).
910931 (2013). biochemistry. Blood Transfus. 10(2), s1924 45 Frezza C, Gottlieb E. Mitochondria in
20 Kim EH, Misek DE. (2012). cancer: not just innocent bystanders. Semin.
Glycoproteomics-based identification of Summarizes the history of metabolomics Cancer Biol. 19, 411 (2009).
cancer biomarkers. Int. J. Proteomics and clinical biochemistry. 46 Martinez-Outschoorn UE, Lin Z,
2011, 601937 (2011). 33 Milne SB, Mathews TP, Myers DS, Trimmer C et al. Cancer cells metabolically
21 Gupta R, Brunak S. Prediction of Ivanova PT, Brown HA. Sum of the parts: "fertilize" the tumor microenvironment with
glycosylation across the human proteome mass spectrometry-based metabolomics. hydrogen peroxide, driving the Warburg
and the correlation to protein function. Pac. Biochemistry 52(22), 38293840 (2013). effect: implications for PET imaging of
Symp. Biocomput. 7, 310322 (2002). 34 DAlessandro A, Gevi F, Zolla L. A robust human tumors. Cell Cycle 10(15),
22 Pan S, Chen R, Aebersold R, Brentnall TA. high resolution reversed-phase HPLC 25042520 (2011).
Mass spectrometry based glycoproteomics strategy to investigate various metabolic 47 Cardaci S, Ciriolo MR. TCA Cycle Defects
from a proteomics perspective. Mol. Cell. species in different biological models. Mol. and cancer: when metabolism tunes redox
Proteomics 10(1), R110.003251 (2011). Biosyst. 7(4), 10241032 (2011). state. Int. J. Cell Biol. 2012, 161837 (2012).
23 Chen R, Jiang X, Sun D et al. 35 Guo B, Chen B, Liu A, Zhu W, Yao S. 48 Guzy RD, Hoyos B, Robin E et al.
Glycoproteomics analysis of human liver Liquid chromatography-mass spectrometric Mitochondrial complex III is required for
tissue by combination of multiple enzyme multiple reaction monitoring-based hypoxia-induced ROS production and
digestion and hydrazide chemistry. strategies for expanding targeted profiling cellular oxygen sensing. Cell Metab.
J. Proteome Res. 8(2), 651661 (2009). towards quantitative metabolomics. Curr. 1, 401408 (2005).
24 Schwamborn K, Caprioli RM. MALDI Drug Metab. 13(9), 12261243 (2012). 49 Frezza C, Tennant DA, Gottlieb E.
imaging mass spectrometrypainting 36 DAlessandro A, Gevi F, Zolla L. Targeted IDH1 mutations in gliomas: when an
molecular pictures. Mol. Oncol. 4(6), mass spectrometry-based metabolomic enzyme loses its grip. Cancer Cell
529538 (2010). profiling through multiple reaction 17, 79 (2010).
25 Schone C, Hofler H, Walch A. MALDI monitoring of liver and other biological 50 Pollard PJ, Briere JJ, Alam NA et al.
imaging mass spectrometry in cancer matrices. Methods Mol. Biol. Accumulation of Krebs cycle intermediates
research: Combining proteomic profiling 909, 279294 (2012). and over-expression of HIF1alpha in
and histological evaluation. Clin. Biochem. 37 Tautenhahn R, Cho K, Uritboonthai W, tumours which result from germline FH
46(6), 539545 (2013). Zhu Z, Patti GJ, Siuzdak G. An accelerated and SDH mutations. Hum. Mol. Genet.
26 Franck J, Longuespee R, Wisztorski M et al. workflow for untargeted metabolomics using 14, 22312239 (2005).
MALDI mass spectrometry imaging of the METLIN database. Nat. Biotechnol. 30(9), 51 Yang M, Soga T, Pollard PJ, Adam J. The
proteins exceeding 30,000 daltons. Med. Sci. 826828 (2012). emerging role of fumarate as an
Monit. 16(9), BR293299 (2010). 38 Crown SB, Ahn WS, Antoniewicz MR. oncometabolite. Front. Oncol. 2, 85 (2012).
27 Gustafsson OJ, Eddes JS, Meding S, Rational design of 13C-labeling experiments 52 Selak MA, Armour SM, MacKenzie ED
McColl SR, Oehler MK, Hoffmann P. for metabolic flux analysis in mammalian et al. Succinate links TCA cycle dysfunction
Matrix-assisted laser desorption/ionization cells. BMC Syst. Biol. 6, 4357 (2012). to oncogenesis by inhibiting HIF-alpha

www.expert-reviews.com 13
Review DAlessandro & Zolla

prolyl hydroxylase. Cancer Cell 65 Berkers CR, Maddocks OD, Cheung EC, 77 Rufini A, Niklison-Chirou MV, Inoue S
7, 7785 (2005). Mor I, Vousden KH. Metabolic Regulation et al. TAp73 depletion accelerates aging
53 Hewitson KS, Lienard BM, by p53 Family Members. Cell. Metab. through metabolic dysregulation. Genes Dev.
McDonough MA et al. Structural and doi:10.1016/j.cmet.2013.06.019 (2013) 26(18), 20092014 (2012).
mechanistic studies on the inhibition of the (Epub ahead of print). 78 Zulato E, Curtarello M, Nardo G,
hypoxiainducible transcription factor 66 Cheung EC, Ludwig RL, Vousden KH. Indraccolo S. Metabolic effects of
hydroxylases by tricarboxylic acid cycle Mitochondrial localization of TIGAR under anti-angiogenic therapy in tumors. Biochimie
intermediates. J. Biol. Chem. hypoxia stimulates HK2 and lowers ROS 94(4), 925931 (2012).
282, 32933301 (2007). and cell death. Proc. Natl Acad. Sci. USA 79 Wyld L, Tomlinson M, Reed MW,
54 Navis A, Niclou S, Fack F et al. Increased 109(50), 2049120496 (2012). Brown NJ. Aminolaevulinic acid-induced
mitochondrial activity in a novel 67 Bensaad K, Cheung EC, Vousden KH. photodynamic therapy: cellular responses to
IDH1-R132H mutant human Modulation of intracellular ROS levels by glucose starvation. Br. J. Cancer 86(8),
oligodendroglioma xenograft model: in situ TIGAR controls autophagy. EMBO J. 13431347 (2002).
detection of 2-HG and a-KG. Acta 28(19), 30153026 (2009). 80 Sandulache VC, Ow TJ, Pickering CR et al.
Neuropathol. Commun. 1(1), 18 (2013). 68 Wanka C, Steinbach JP, Rieger J. Glucose, not glutamine, is the dominant
55 Maxwell PH, Pugh CW, Ratcliffe PJ. Tp53-induced glycolysis and apoptosis energy source required for proliferation and
Activation of the HIF pathway in cancer. regulator (TIGAR) protects glioma cells survival of head and neck squamous carcinoma
Curr. Opin. Genet. Dev. from starvation-induced cell death by cells. Cancer 117(13), 29262938 (2011).
11, 293299 (2001). up-regulating respiration and improving 81 Ogiso Y, Tomida A, Kim HD, Tsuruo T.
56 Wise DR, Ward PS, Shay JE et al. Hypoxia cellular redox homeostasis. J. Biol. Chem. Glucose starvation and hypoxia induce
promotes isocitrate dehydrogenase- 287(40), 3343633446 (2012). nuclear accumulation of proteasome in
dependent carboxylation of 69 DAlessandro A, Marrocco C, Rinalducci S cancer cells. Biochem. Biophys. Res. Commun.
alpha-ketoglutarate to citrate to support cell et al. Analysis of TAp73-dependent 258(2), 448452 (1999).
growth and viability. Proc. Natl Acad. Sci. signaling via Omics technologies. 82 Berardinis RJ, Mancuso A, Daikhin E et al.
USA 108, 1961119616 (2011). J. Proteome Res. 12(9), 42074220 (2013) Beyond aerobic glycolysis: transformed cells
57 Vance DE, Vance JE. Physiological 70 Wanka C, Brucker DP, Bahr O et al. can engage in glutamine metabolism that
consequences of disruption of mammalian Synthesis of cytochrome C oxidase 2: exceeds the requirement for protein and
phospholipid biosynthetic genes. J. Lipid a p53-dependent metabolic regulator that nucleotide synthesis. Proc. Natl Acad. Sci.
Res. 50, S132 (2009). promotes respiratory function and protects USA 104, 1934519350 (2007).
58 Levine AJ, Puzio-Kuter AM. The control of glioma and colon cancer cells from 83 Yuneva M, Zamboni N, Oefner P,
the metabolic switch in cancers by hypoxia-induced cell death. Oncogene Sachidanandam R, Lazebnik Y. Deficiency
oncogenes and tumor suppressor genes. 31(33), 37643776 (2012). in glutamine but not glucose induces
Science 330, 13401344 (2010). 71 Hu W, Zhang C, Wu R, Sun Y, Levine A, MYC-dependent apoptosis in human cells.
59 Schafer ZT, Grassian AR, Song L et al. Feng Z. Glutaminase 2, a novel p53 target J. Cell. Biol. 178(1), 93105 (2007).
Antioxidant and oncogene rescue of gene regulating energy metabolism and 84 McCarthy N. Metabolism: Sensitivity to
metabolic defects caused by loss of matrix antioxidant function. Proc. Natl Acad. Sci. serine starvation. Nat. Rev. Cancer 13(2), 77
attachment. Nature 461, 109113 (2009). USA 107(16), 74557460 (2010). (2013).
60 Vander Heiden MG, Lunt SY, Dayton TL 72 Giacobbe A, Bongiorno-Borbone L, 85 Maddocks OD, Berkers CR, Mason SM
et al. Metabolic pathway alterations that Bernassola F et al. p63 regulates glutaminase et al. Serine starvation induces stress and
support cell proliferation. Cold Spring Harb. 2 expression. Cell Cycle 12(9), 13951405 p53-dependent metabolic remodelling in
Symp. Quant. Biol. 76, 325334 (2011). (2013). cancer cells. Nature 493(7433), 542546
Describes the importance of metabolic 73 Stavridi ES, Halazonetis TD. p53 and stress (2013).
reprogramming in cancer cells and its role in the ER. Genes Dev. 18(3), 241244 This elegant paper describes the linkage
(2004). between serine starvation responses,
in sustaining anabolic reactions.
74 Pluquet O, Qu LK, Baltzis D, metabolic reprogramming (increased PPP
61 Muller PA, Vousden KH, Norman JC.
Koromilas AE. Endoplasmic reticulum stress fluxes and maintenance of the redox poise,
p53 and its mutants in tumor cell migration
accelerates p53 degradation by the activation of the serine synthesis pathway
and invasion. J. Cell Biol. 192(2), 209218
cooperative actions of Hdm2 and glycogen and Krebs cycle) and the most important
(2011).
synthase kinase 3beta. Mol. Cell. Biol. tumor suppressor, Tp53.
62 Sherr CJ, McCormick F. The RB and 25(21), 93929405 (2005).
p53 pathways in cancer. Cancer Cell 2(2), 86 Christofk HR, Vander Heiden MG,
75 Zocchi L, Bourdon JC, Codispoti A et al. Harris MH et al. The M2 splice isoform of
103112 (2002).
Scotin: A new p63 target gene expressed pyruvate kinase is important for cancer
63 Yeung SJ, Pan J, Lee MH. Roles of p53, during epidermal differentiation. Biochem.
MYC and HIF-1 in regulating glycolysis - metabolism and tumour growth. Nature
Biophys. Res. Commun. 367(2), 271276 452, 230233 (2008).
the seventh hallmark of cancer. Cell. Mol. (2008).
Life Sci. 65, 39813999 (2008). 87 Chaneton B, Hillmann P, Zheng L et al.
76 Terrinoni A, Ranalli M, Cadot B et al. Serine is a natural ligand and allosteric
64 Vousden KH, Ryan KM. p53 and p73 alpha is capable of inducing scotin and
metabolism. Nat. Rev. Cancer activator of pyruvate kinase M2. Nature
ER stress. Oncogene 23(20), 37213725 491(7424), 458462 (2012).
9, 691700 (2009). (2004).

14 Expert Rev. Proteomics 10(5), (2013)


Proteomics, metabolomics & drug development Review

88 Ye J, Mancuso A, Tong X et al. Pyruvate 102 Sassen S, Miska EA, Caldas C. MicroRNA: 116 Chiarugi A, Dolle C, Felici R, Ziegler M.
kinase M2 promotes de novo serine implications for cancer. Virchows. Arch. The NAD metabolomea key determinant
synthesis to sustain mTORC1 activity and 452(1), 110 (2008). of cancer cell biology. Nat. Rev. Cancer
cell proliferation. Proc. Natl Acad. Sci. USA 103 Jansson MD, Lund AH. MicroRNA and 12(11), 741752 (2012).
109(18), 69046909 (2012). cancer. Mol. Oncol. 6(6), 590610 (2012). 117 Amaravadi RK, Lippincott-Schwartz J,
89 Dandapani M, Hardie DG. AMPK: 104 Rottiers V, Naar AM. MicroRNAs in Yin XM et al. Principles and current
opposing the metabolic changes in both metabolism and metabolic disorders. Nat. strategies for targeting autophagy for cancer
tumour cells and inflammatory cells? Rev. Mol. Cell Biol. 13(4):239250 (2012). treatment. Clin. Cancer Res.
Biochem. Soc. Trans. 41(2), 687693 17, 654666 (2011).
105 Peschiaroli A, Giacobbe A, Formosa A et al.
(2013). 118 Bursch W, Karwan A, Mayer M et al. Cell
miR-143 regulates hexokinase 2 expression
90 Li L, Chen Y, Gibson SB. in cancer cells. Oncogene 32(6):797802 death and autophagy: cytokines, drugs, and
Starvation-induced autophagy is regulated (2013). nutritional factors. Toxicology 254(3),
by mitochondrial reactive oxygen species 147157 (2008).
106 Soga T. Cancer metabolism: key players in
leading to AMPK activation. Cell. Signal. 119 Peng T, Golub TR, Sabatini DM. The
metabolic reprogramming. Cancer Sci.
25(1), 5065 (2013). immunosuppressant rapamycin mimics a
104(3), 275281 (2013).
91 Kupiec M, Weisman R. TOR links starvation-like signal distinct from amino
107 Birsoy K, Sabatini DM, Possemato R.
starvation responses to telomere length acid and glucose deprivation. Mol. Cell Biol.
Untuning the tumor metabolic machine:
maintenance. Cell Cycle 11(12), 22682271 22(15), 55755584 (2002).
targeting cancer metabolism: a bedside
(2012). 120 Dando I, Donadelli M, Costanzo C et al.
lesson. Nat. Med. 18(7), 10221023 (2012).
92 Lamming DW, Ye L, Sabatini DM, Cannabinoids inhibit energetic metabolism
Baur JA. Rapalogs and mTOR inhibitors as This paper reviews the main concepts and induce AMPK-dependent autophagy in
anti-aging therapeutics. J. Clin. Invest. behind cancer metabolism reprogramming. pancreatic cancer cells. Cell Death Dis.
123(3), 980989 (2013). 108 Samudio I, Fiegl M, Andreeff M. 4, e664 (2013).
93 Efeyan A, Zoncu R, Sabatini DM. Amino Mitochondrial uncoupling and the Warburg 121 Lee C, Raffaghello L, Longo VD.
acids and mTORC1: from lysosomes to effect: molecular basis for the Starvation, detoxification, and multidrug
disease. Trends Mol. Med. 18(9), 524533 reprogramming of cancer cell metabolism. resistance in cancer therapy. Drug Resist.
(2012). Cancer Res. 69(6), 21632166 (2009). Updat. 15(12), 114122 (2012).
94 Laplante M, Sabatini DM. mTOR signaling 109 Tennant DA, Duran RV, Gottlieb E. 122 DiPaola RS, Dvorzhinski D, Thalasila A
in growth control and disease. Cell 149(2), Targeting metabolic transformation for et al. Therapeutic starvation and autophagy
274293 (2012). cancer therapy. Nat. Rev. Cancer in prostate cancer: a new paradigm for
10, 267277 (2010). targeting metabolism in cancer therapy.
95 Laplante M, Sabatini DM. mTOR
Signaling. Cold Spring Harb. Perspect. Biol. 110 DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Prostate 68(16), 17431752 (2008).
4(2) a011593 (2012). Thompson CB. The biology of cancer: 123 Raffaghello L, Lee C, Safdie FM et al.
metabolic reprogramming fuels cell growth and Starvation-dependent differential stress
96 Sun H, Lesche R, Li DM et al. PTEN proliferation. Cell Metab. 7, 1120 (2008).
modulates cell cycle progression and cell resistance protects normal but not cancer cells
survival by regulating phosphatidylinositol 111 Chen Z, Lu W, Garcia-Prieto C, Huang P. against high-dose chemotherapy. Proc. Natl
3,4,5,-trisphosphate and Akt/protein kinase The Warburg effect and its cancer Acad. Sci. USA 105(24), 82158220 (2008).
B signaling pathway. Proc. Natl Acad. Sci. therapeutic implications. J. Bioenerg. 124 Awale S, Lu J, Kalauni SK et al.
USA 96(11), 61996204 (1999). Biomembr. 39(3), 267274 (2007). Identification of arctigenin as an antitumor
97 Dang CV, Le A, Gao P. MYC-induced The paper summarizes the main strategies agent having the ability to eliminate the
cancer cell energy metabolism and available to tackle cancer metabolism via tolerance of cancer cells to nutrient
therapeutic opportunities. Clin. Cancer Res. drug-mediated glycolysis inhibition. starvation. Cancer Res. 66(3), 17511757
15(21), 64796483 (2009). 112 Feron O. Pyruvate into lactate and back: (2006).
98 Ying H, Kimmelman AC, Lyssiotis CA et al. from the Warburg effect to symbiotic 125 Wishart DS, Knox C, Guo AC et al.
Oncogenic Kras maintains pancreatic tumors energy fuel exchange in cancer cells. DrugBank: a knowledgebase for drugs, drug
through regulation of anabolic glucose Radiother. Oncol. 92(3), 329333 (2009). actions and drug targets. Nucleic Acids Res.
metabolism. Cell 149(3), 656670 (2012). 113 Granchi C, Minutolo F. Anticancer agents 36, D901D906 (2008).
99 Son J, Lyssiotis CA, Ying H et al. Glutamine that counteract tumor glycolysis. 126 Kuhajda FP, Jenner K, Wood FD et al.
supports pancreatic cancer growth through a ChemMedChem 7(8), 13181350 (2012). Fatty acid synthesis: a potential selective
KRAS-regulated metabolic pathway. Nature 114 Madhok BM, Yeluri S, Perry SL, target for antineoplastic therapy. Proc. Natl
496(7443), 101105 (2013). Hughes TA, Jayne DG. Targeting glucose Acad. Sci. USA 91, 63796383 (1994).
100 Carracedo A, Weiss D, Leliaert AK et al. metabolism: an emerging concept for 127 Panigrahy D, Kaipainen A, Huang S et al.
A metabolic prosurvival role for PML in anticancer therapy. Am. J. Clin. Oncol. PPARalpha agonist fenofibrate suppresses
breast cancer. J. Clin. Invest. 34(6), 628635 (2011). tumor growth through direct and indirect
122, 30883100 (2012). 115 Semenza GL. Hypoxia-inducible factors: angiogenesis inhibition. Proc. Natl Acad. Sci.
mediators of cancer progression and targets U.S.A. 105(3), 985990 (2008).
101 Binoux M. The IGF system in metabolism
regulation. Diabete. Metab. 21(5), 330337 for cancer therapy. Trends Pharmacol. Sci. 128 Samudio I, Harmancey R, Fiegl M et al.
(1995). 33, 207214 (2012). Pharmacologic inhibition of fatty acid

www.expert-reviews.com 15
Review DAlessandro & Zolla

oxidation sensitizes human leukemia cells to metabolism. Cancer Res. 72(15), 37093714 multiple drug treatments for metabolic
apoptosis induction. J. Clin. Invest. (2012). diseases and cancer. BMC Syst. Biol. 6(1),
120, 142156 (2010). 134 Sidransky D. Emerging molecular markers 115 (2012).
129 Carracedo A, Cantley LC, Pandolfi PP. of cancer. Nat. Rev. Cancer 2(3), 210219 140 Chang R, Xie L, Bourne P, Palsson B,
Cancer metabolism: fatty acid oxidation in (2002). Dunbrack R. Drug off-target effects
the limelight. Nat. Rev. Cancer 13(4), 135 Ciocca DR, Calderwood SK. Heat shock predicted using structural analysis in
227232 (2013). proteins in cancer: diagnostic, prognostic, the context of a metabolic network model.
130 Anastasiou D, Cantley LC. Breathless cancer predictive, and treatment implications. Cell PLoS Comput. Biol. 6, e1000938 (2010).
cells get fat on glutamine. Cell Res. 22(3), Stress. Chaperones. 10(2), 86103 (2005). 141 Draoui N, Feron O. Lactate shuttles at a
443446 (2012). 136 Shu CW, Huang CM. HSP70s: from glance: from physiological paradigms to
131 Wise DR, Thompson CB. Glutamine tumor transformation to cancer therapy. anti-cancer treatments. Dis. Model. Mech.
addiction: a new therapeutic target in Clin. Med. Oncol. 2, 335345 (2008). 4(6), 727732 (2011).
cancer. Trends Biochem. Sci. 35(8), 427433 137 DAlessandro A, Zolla L. We are what we 142 Kaelin WG Jr. The concept of synthetic
(2010). eat: food safety and proteomics. J. Proteome lethality in the context of anticancer
132 Middleton B. The mitochondrial long-chain Res. 11(1), 2636 (2012). therapy. Nat. Rev. Cancer 5, 689698
trifunctional enzyme: (2005).
138 Folger O, Jerby L, Frezza C, Gottlieb E,
2-enoyl-CoA hydratase, Ruppin E, Shlomi T. Predicting selective 143 Lehar J, Krueger AS, Avery W et al.
3-hydroxyacyl-CoA dehydrogenase and drug targets in cancer through metabolic Synergistic drug combinations tend to
3-oxoacyl-CoA thiolase. Biochem. Soc. Trans. networks. Mol. Syst. Biol. 7, 501 (2011). improve therapeutically relevant
22(2), 427431 (2002). selectivity. Nat. Biotechnol. 27, 659666
139 Facchetti G, Zampieri M, Altafini C.
133 Zaidi N, Swinnen JV, Smans K. (2009).
Predicting and characterizing selective
ATP-citrate lyase: a key player in cancer

16 Expert Rev. Proteomics 10(5), (2013)

View publication stats

Você também pode gostar