You are on page 1of 10

REVIEWS

ONCONEPHROLOGY

Metabolic reprogramming in clear cell


renal cell carcinoma
Hiromi I. Wettersten1, Omran Abu Aboud2, Primo N. Lara Jr3 and Robert H. Weiss2
Abstract | Research in many cancers has uncovered changes in metabolic pathways that control
tumour energetics and biosynthesis, so‑called metabolic reprogramming. Studies in clear cell
renal cell carcinoma (ccRCC) have been particularly revealing, leading to the concept that ccRCC
is a metabolic disease. ccRCC is generally accompanied by reprogramming of glucose and fatty
acid metabolism and of the tricarboxylic acid cycle. Metabolism of tryptophan, arginine and
glutamine is also reprogrammed in many ccRCCs, and these changes provide opportunities
for new therapeutic strategies, biomarkers and imaging modalities. In particular, metabolic
reprogramming facilitates the identification of novel and repurposed drugs that could potentially
be used to treat ccRCC, which when metastatic has currently limited long-term treatment
options. Further research and dissemination of these concepts to nephrologists and oncologists
will lead to clinical trials of therapeutics specifically targeted to tumour metabolism, rather than
generally toxic to all proliferating cells. Such novel agents are highly likely to be more effective
and to have far fewer adverse effects than existing drugs.

Tricarboxylic acid (TCA)


Cancer has historically been considered a disease of (TCA) cycle, glutamine metabolism, and tumour energet‑
cycle uncontrolled cell proliferation mediated by oncogenes, ics. Kidney cancer has, therefore, been aptly labelled a
A series of chemical reactions such as activating mutations in growth factor recep‑ ­metabolic disease4,10,11.
that comprises the oxidation tors1. However, since the initially puzzling observation The most common form of kidney cancer, clear cell
of acetyl-CoA to release stored
of enhanced glycolysis — an energetically inefficient renal cell carcinoma (ccRCC) (BOX 1), is an aggressive
energy.
process — occurring in aggressive tumours despite cancer that arises from the proximal tubular epithelium
normoxia2, a steady stream of discoveries has linked and, in its metastatic form, is associated with high mor‑
cancers to a variety of metabolic changes that are indi‑ tality12. In addition to the well-known and continually
rectly related to the more obvious phenotype of cell validated Warburg effect, reductive carboxylation occurs
1
University of California, proliferation. Several classical metabolic pathways are in many ccRCC cells13,14. This glutamine-­dependent
San Diego, Sanford increased, decreased or bypassed entirely in cancer cells pathway involves ‘backwards flow’ of the TCA cycle.
Consortium for Regenerative and many of these meta­bolic alterations, especially in Such unexpected aberrations of metabolism in ccRCC
Medicine, Room 4810,
cancers of the kidney3,4, have now been directly linked have provided new opportunities for imaging and new
2880 Torrey Pines Scenic
Drive, La Jolla, California to oncogenic mutations5–7 (TABLE 1). This research has targets for therapeutic intervention. In this Review, we
92037–0695, USA. led to the concept that altered metabolism, so‑called discuss the advantages of metabolic reprogramming for
2
Division of Nephrology, metabolic reprogramming, is associated with a ‘success‑ ccRCC cells and examine how exploitation of repro‑
University of California Davis, ful’ kidney cancer. Such reprogramming enables rap‑ grammed metabolism lends itself to novel opportu‑
Genome and Biomedical
Sciences Facility, Room 6311,
idly proliferating cancer cells to meet their basic needs nities for biomarkers, imaging and particularly new
451 Health Sciences Drive, for augmented levels of cellular building components, ­therapeutic paradigms.
Davis, California 95616, USA. including DNA and membrane constituents, as well
3
University of California Davis as high levels of molecules that regulate the enhanced Advantages of metabolic reprogramming
Comprehensive Cancer
tumour energetics. Independent of any proliferative advantage, cancer
Center, 4501 X Street,
Suite 3003, Sacramento, Kidney cancer is one of the most studied and perhaps cells with metabolic characteristics that enable them to
California 95817, USA. the exemplar of malignancies characterized by metabolic outcompete normal cells for the resources that support
Correspondence to R.H.W. reprogramming8,9. Genes that are mutated in kidney their reprogrammed metabolism have a clear survival
rhweiss@ucdavis.edu cancer are involved in a number of disparate pathways advantage, and hence such properties become propa­
doi:10.1038/nrneph.2017.59 that regulate various aspects of cellular metabolism, gated. For example, the initially puzzling increase in
Published online 8 May 2017 such as oxygen and/or iron sensing, the tricarboxylic acid glucose uptake and glycolysis that characterizes the

410 | JULY 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points building components21. Although metabolic reprogram‑


ming is advantageous for tumour cells, altered metabolic
• The incidence of kidney cancer, particularly clear cell renal cell carcinoma, is pathways can be exploited as a therapeutic strategy, for
increasing and knowledge of its pathophysiology is essential for nephrologists example by limiting the supply of nutrients on which the
• Reprogramming of metabolic pathways enables cancer cells to rapidly proliferate, cells depend22 or by devising compounds that modulate
survive in conditions of nutrient depletion and hypoxia, and evade the immune system the altered pathways23,24.
• Most forms of kidney cancer are associated with reprogramming of metabolic
pathways including oxygen sensing, the tricarboxylic acid cycle and the metabolism Reprogrammed metabolic pathways in ccRCC
of tryptophan, fatty acids, glucose, glutamine and arginine Metabolic reprogramming in ccRCC is most commonly
• Metabolic reprogramming provides opportunities for functional imaging approaches related to mutations in VHL, which occur in ~90% of
based on the altered pathways cases25. VHL is involved in oxygen and iron sensing
• Novel therapies for kidney cancers that target critical proteins or enzymes that are through the transcription factors hypoxia-inducible
involved in dysregulated metabolic pathways are being developed factor 1‑α (HIF‑1α) and HIF‑2α. In VHL-mutant dis‑
ease, constitutive activation of HIF-mediated metabolic
pathways leads to the activation of pathways that coun‑
Warburg effect might enable tumour cells to thrive in teract the effects of hypoxia in the normoxic (though
a nutrient-depleted state, while at the same time ena‑ progressively hypoxic) environment of ccRCC26. In addi‑
bling accelerated production of lipids and nucleotides tion, research involving genomic27, proteomic20,28 and
through the pentose phosphate pathway15, and providing metabo­lomic8,9 studies has identified a profound meta‑
tumour-intrinsic antioxidant activity through a switch to bolic shift in aggressive ccRCCs that involves the TCA,
glutamine metabolism9,16,17 (FIG. 1). In addition, activa‑ pentose phosphate, and phosphoinositide 3‑kinase
tion of the hypoxia pathway armamentarium, including (PI3K) ­pathways among others.
neoangiogenesis, even in the absence of hypoxia, ena‑
bles cancer cells to thrive as their surroundings become Tryptophan pathways
progressively more deprived of oxygen18. Such activa‑ Tryptophan is an essential amino acid with three pri‑
tion is seen in the majority of ccRCCs with mutations mary downstream pathways: the serotonin, indole­
in VHL, which encodes the Von Hippel-Lindau disease acetate and kynurenine pathways29,30. The majority of
tumour suppressor (VHL)19. The driver of the evolution tryptophan is catabolized through the kynurenine path‑
of dependence of some ccRCCs on extracellular amino way via the rate-limited enzyme activity of indoleam‑
acids, such as arginine20, is not obvious but it might ine 2,3‑dioxygenase (IDO)31. One of the most reported
be linked to enhanced synthesis of essential cellular effects of tryptophan metabolism is immunosuppression

Table 1 | Selected oncogenes and tumour suppressor genes that regulate metabolism
Gene Effects on metabolic pathways Relevance to RCC Refs
PTEN Inhibition of glycolysis through inactivation of AKT • Among patients with RCC, 2.6% have biallelic 117,118
loss and 16.6% have monoallelic loss of PTEN
• Loss of PTEN is associated with high stage and
grade of RCC
TSC1/2 Deficiency leads to the Warburg effect and Mutation is a risk factor for RCC 119,120
glutamine addiction through activation of mTOR
AKT Upregulation of glycolysis through activation of • AKT mutations are rare in RCC but AKT is 121–124
enzymes including hexokinase activated owing to loss of PTEN
• AKT inhibitors are being tested in clinical trials
for RCC
VHL Inhibition of the Warburg effect through Loss‑of‑function mutations found in >90% of 25,125
deactivation of HIF patients with RCC
p53 • Downregulation of glycolysis via deactivation p53 mutations are rare in RCC 126–129
of GLUT‑1/4 and upregulation of TIGAR
• Upregulation of glutamine metabolism via
increased transcription of glutaminase 2
LKB1 Upregulation of glycolysis and β‑oxidation and LKB1 activity is compromised in RCC (in vitro, 130–132
downregulation of lipid synthesis via activation in vivo and in patients)
of AMPK
Myc • Upregulation of the Warburg effect through • Often mutated and overexpressed in RCC 133–137
Warburg effect activation of hexokinase, LDHA and PDK1 • Activated by HIF‑2α
The phenomenon of cells • Upregulation of glutamine metabolism through • Myc overexpression induces RCC in mice
producing energy primarily by activation of glutaminase 1
glycolysis followed by lactate • Upregulation of lipid synthesis through activation
fermentation, rather than by of FAS and SCD1
glycolysis followed by the APMK, AMP-activated protein kinase; FAS, fatty acid synthase; GLUT, glucose transporter; HIF, hypoxia-inducible factor; LDHA,
tricarboxylic acid (TCA) cycle lactate dehydrogenase A; PDK1, pyruvate dehydrogenase kinase 1; RCC, renal cell carcinoma; SCD1, stearoyl-CoA desaturase 1;
in mitochondria. TIGAR, p53‑inducible glycolysis and apoptosis regulator.

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | JULY 2017 | 411


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | Renal cell carcinoma Abnormalities of lipid metabolism in ccRCC were


reported as early as 1987 (REF. 39). A study using gas
• Diagnosed in approximately 64,000 patients in the USA annually113 chromatographic methods found increased cholesterol
• One of the relatively few malignancies with an increasing incidence114 ester storage in the tumour tissues of patients with ccRCC
• Five histologic subtypes exist: clear cell, papillary type 1, papillary type 2, compared with normal kidney tissues39. Consistent
chromophobe and oncocytoma with this finding, mRNA and protein levels of SCD1
• Clear cell renal cell carcinoma is the most common histologic subtype are increased in ccRCC tissues and are required for the
• The disease is frequently asymptomatic115 and, in the nephrology clinic, is commonly growth and survival of ccRCC cells40, and increased l­ evels
diagnosed incidentally during workup of acute kidney injury or haematuria4 of FAS correlate with tumour aggressiveness and poor
• Associated with paraneoplastic syndromes, most of which are related to patient survival41. Furthermore, a metabolomics study
reprogrammed metabolism showed increased levels of long-chain fatty acids8 and a
• Early immunomodulating therapies showed minimal success, but newer targeted proteomics study showed decreased levels of enzymes
therapies and checkpoint inhibitors have improved survival116 involved in β‑oxidation in ccRCC tissues9,20. Together,
these findings suggest upregulation of lipid storage and
utilization of lipids for membrane synthesis in ccRCC.
as a result of tryptophan depletion32 and an increase in Interestingly, urinary and tissue metabolomics ­studies
immunosuppressive metabolites in the kynurenine found increased levels of fatty acyl carnitines and carni‑
pathway30. tine in samples from patients with ccRCC compared with
In ccRCC, the level of tryptophan is reduced, sug‑ normal controls, and these alterations correlated
gesting increased utilization9. Tissue levels of kynure‑ with kidney cancer grade9,42. The possibility exists that
nine and quinolinate are increased in ccRCC, whereas the increase in carnitine levels is due to down­regulation
the ­levels of enzymes that feed the serotonin and of β‑oxidation enzymes; however, precisely how acyl-­
indoleacetate pathways, including DOPA decarboxylase, carnitines and carnitine affect the ccRCC phenotype
mono­amine oxidase and aldehyde dehydrogenase 2, and cellular activity is not yet known. Given the lack of
are decreased, suggesting activation of the kynurenine biomarkers for detection of ccRCC and the non-invasive
pathway9 (FIG. 2). Moreover, immunohistochemistry procedure of urine collection, an increase in urinary car‑
studies have shown that levels of IDO1 are increased in nitines could (with further validation) potentially be a
the endothelial cells of ccRCC tumour tissues compared novel biomarker for ccRCC screening. Such a biomarker
to those in normal kidney tissues33,34, and a non-targeted would be particularly useful for patients who have risk
urine metabolomics analysis showed that quinolinate factors for ccRCC, such as smoking or obesity.
was the most increased urinary metabolite (of those
identified) in patients with ccRCC compared to healthy Glucose metabolism and HIF
individuals35. Given the immunosuppressive effects of In normal cells, glucose is a major source of pyruvate,
kynurenine and quinolinate, these studies suggest that which feeds the TCA cycle for energy production under
increased utilization of tryptophan to produce these normoxia. Under hypoxia, normal cells switch their
metabolites in ccRCC cells results in immunosuppres‑ energy production from the TCA cycle to lactate fer‑
sion that might enable enhanced tumour growth and mentation; increased levels of HIF-1α lead to induction
contribute to the low success rate of interferon‑α-based of lactate dehydrogenase (LDH), which catalyses the
immuno­therapies in this disease34. Despite these find‑ conversion of pyruvate to lactate43. By contrast, cancer
ings, extant studies lack definitive evidence to support cells predominantly produce energy by lactic acid fer‑
the i­ mmunosuppressive effect of tryptophan metabolism mentation, regardless of the oxygen level; this change in
in ccRCC. metabolism is known as the Warburg effect or aero­bic
glycolysis44,45. Interestingly, several studies have shown
Fatty acid oxidation and synthesis that pluripotent stem cells have higher levels of aero‑
Metabolism of fatty acids is regulated by their bic glycolysis than somatic cells46, indicating that the
­β‑oxidation and by their synthesis via fatty acid syn‑ Warburg effect is not unique to cancer cells but might be
thase (FAS) (FIG. 3). When utilized for the TCA cycle, a common phenomenon among undifferentiated cells.
fatty acids are first converted into fatty acyl-CoA and The Warburg effect was among the first demonstra‑
then into fatty acyl-carnitine, which is transferred into tions of metabolic reprogramming in cancer but is not
mitochondria by carnitine palmitoyltransferase. In mito‑ universal to all malignancies. For example, lung cancer
chondria, fatty acyl-carnitine is converted back into cells do not rely on the Warburg effect, indicating the
fatty acyl-CoA, which undergoes β‑oxidation to form complexity and heterogeneity of cancer metabolism47.
acetyl-coenzyme A (acetyl-CoA). This lengthy process is ccRCC does, however, seem to follow the classic Warburg
regulated by various enzymes, including acyl-CoA dehy‑ effect20,28 (FIG. 3). The levels of glucose transporter 1
drogenase, hydroxyacyl-CoA dehydrogenase and enoyl- (GLUT‑1) are increased in ccRCC tumours compared
CoA hydratase36. Acetyl-CoA is converted back into fatty to normal control tissues, suggesting increased uptake
acyl-CoA and then into fatty acids by FAS37. Fatty acids of glucose48. Moreover, metabolomic8,9, proteomic9,20 and
are elongated and desaturated by stearoyl-CoA desat‑ transcriptomic8 studies have shown increased levels of
urase (SCD1) to make unsaturated fatty acids as well glycolysis metabolites and enzymes, including phos‑
as triglycerides and phospholipids that are required for phoglycerate kinase, hexokinase, pyruvate kinase 2,
membrane synthesis38. and LDH‑A in ccRCC cells and tissues, suggesting

412 | JULY 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

↑ Lipid synthesis
↑ Tryptophan
metabolism Carnitine Fatty acid Cholesterol
ROS inhibition
Tryptophan
↓ β-oxidation
↑ Reductive GSH or GSSG
• Kynurenine carboxylation
• Quinolinate ↑ Glycolysis Glucose

TCA Urea
Immunosuppression Lactate Pyruvate cycle Glutamine cycle Arginine

Increased Decreased Unknown Levels increased Levels decreased Levels unknown

Figure 1 | Metabolic reprogramming in clear cell renal cell carcinoma (ccRCC). In ccRCC, aerobic glycolysis, carnitine
and lipid synthesis, reductive carboxylation, the glutathione/oxidized glutathione (GSH/GSSG) pathway, and tryptophan
Nature Reviews | Nephrology
metabolism are upregulated, whereas the urea cycle and energy production through the tricarboxylic acid (TCA) cycle is
downregulated. These changes are advantageous for ccRCC cells as they enable them to survive in conditions of nutrient
depletion and hypoxia, provide the cellular building blocks that are required for proliferation, and result in the production of
immunosuppressive (kynurenine and quinolinate) and antioxidant (GSH and GSSG) metabolites. ROS, reactive oxygen species.

upregu­lation of glucose utilization for lactate fermenta‑ tissues8,9,20,28,62 (FIG. 3). Non-targeted metabolomic ana­lyses
tion, the sine qua non of the Warburg effect. On the other have shown increased levels of citrate and cis-­aconitate
hand, fructose‑1,6‑bisphosphatase 1 (FBP1), which in ccRCC tissue, whereas levels of fumarate and malate
antagonizes glycolysis, is depleted in ccRCC tumours, are decreased8,9,20,62. The increase in levels of citrate
and ectopic expression of FBP1 inhibited ccRCC tumour and cis-aconitate might be the result of upregulation of
growth in a xenograft model49. The levels of pyruvate reductive carboxylation for fatty acid synthesis. Tracing
carboxylase and pyruvate dehydrogenase, the enzymes of 13C-labelled glutamine showed utilization of glutamine
that catabolize pyruvate to feed the TCA cycle, are also for reductive carboxylation in VHL-deficient ccRCC cell
significantly decreased in ccRCC9, further suggesting lines and xenografts; HIF‑2α was sufficient to promote
that these tumours rely on lactate fermentation. this reductive TCA cycle and glutamine addiction63.
Interestingly, the increase in GLUT‑1 expression in The decrease in levels of fumarate and malate in
ccRCC tumours correlates with a decrease in the num‑ ccRCC tissue is likely due to a reduction in levels of suc‑
bers of infiltrating CD8+ T cells50, suggesting an additional cinate dehydrogenase8,9, which catabolizes succinate to
mechanism by which ccRCC might suppress the immune form fumarate. The levels of isocitrate dehydrogenase
system. This decrease in CD8+ T cells might be the result (IDH), which makes α‑ketoglutarate from isocitrate,
of increased lactate levels owing to GLUT‑1 induction as were also decreased in some grades of ccRCC tissues
lactate has been reported to inhibit T‑cell activity51. compared to adjacent normal tissues8,9. Gain‑of‑function
As VHL loss is a common occurrence in ccRCC, mutations in IDH1 and IDH2 result in increases in the
a substantial proportion of the current understanding of levels of an enantiomer of l-2‑hydroxyglutarate that is
this tumour type has been derived from the study of HIF an oncometabolite in ccRCC64.
biology. HIF‑α has profound effects on tumour metabo‑
lism and is the apparent driving force behind the Warburg Glutamine metabolism
effect in RCC. In VHL-deficient ccRCC, HIF-1α increases Glutamine has various physiological functions, includ‑
the expression of GLUT‑1, which promotes cellular glu‑ ing as a building block for protein synthesis, as a major
cose uptake52. HIF-1α also transcriptionally upregulates source for lipid synthesis and energy production, and as
genes that encode enzymes involved in glycolysis, such a precursor to the antioxidant molecule, glutathione65–67.
as hexokinase 1 and 2 and glyceraldehyde 3‑phosphate In the normal renal cortex, glutamine also regulates uri‑
dehydrogenase53,54. In addition, HIF-1α upregulates LDH nary pH by producing ammonia68. Several independent
expression and thus promotes the conversion of pyruvate studies have shown that glutamine utilization is increased
to lactate and shifts cellular metabolism away from the in ccRCC compared to normal kidney tissues8,9,63,69.
TCA cycle through the regulation of pyruvate dehydro‑ Although glutamine can be utilized in the reductive
genase55,56. HIF‑1α also regulates the expression of several carboxylation pathway to generate fatty acids and l-2‑­
microRNAs, including miR‑210 (REF. 57), which is over‑ hydroxyglutarate13,64, another function of glutamine
expressed in ccRCC58,59 and has been shown to down­ in ccRCC seems to be to feed the glutathione/oxidized
regulate mitochondrial respiration60. Selected antagonists ­glutathione pathway8,9 (FIG. 3).
of HIF‑2α have been developed24,61 and their therapeutic Glutathione is an antioxidant that converts hydrogen
Oncometabolite potential in ccRCC is discussed below. peroxide into water. Reduced glutathione is converted
A small molecule component to oxidized glutathione via a reaction that is regulated
of normal metabolism that TCA cycle and the electron transport chain by glutathione peroxidase. A combined proteomics and
on accumulation, results in
metabolic dysregulation and
In ccRCC tissues, the TCA cycle is downregulated metabolomics study in ccRCC showed increased levels
consequently primes cells for between succinate and malate and upregulated between of metabolites in the glutamine and glutathione/oxidized
progression to malignancy. citrate and α‑ketoglutarate compared to normal kidney glutathione pathways, including glutamine, glutamate,

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | JULY 2017 | 413


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Semi-essential amino acid glutathione, and oxidized glutathione 9. The  ­l evels hence these cells are auxotrophic for arginine77–79.
An amino acid that can only be of enzymes that inhibit use of glutamine for  the In biopsy samples from patients with ccRCC, ASS1 was
synthesized under specific glutathione/oxidized glutathione pathway (glutathione not expressed or was downregulated in tumour cells
metabolic conditions. Also S‑transferase and γ‑glutamyl transpeptidase) were but was highly expressed in normal proximal tubule
known as a conditionally
essential amino acid.
decreased, whereas levels of glutathione peroxidase 1 were cells22. This finding was confirmed in all ccRCC grades
increased, suggesting that ccRCC cells utilize glutamine in in a proteomic study20. The reason why some tumours
this pathway to scavenge reactive oxygen species (ROS), become arginine auxotrophic is not well understood.
essentially acting as an intrinsic antioxidant system that A possible explanation for the loss of ASS1 activity
enables cell survival. Indeed, upregulation of the gluta­ in some tumours might be cellular dedifferentiation
mine and glutathione/oxidized glutathione pathways during tumorigenesis22. Alternatively the lack of ASS1
correlates with high-grade, high-stage and metastasis might be a result of accumulation of aspartate, which
of ccRCC8,9. In some tumours and cancer cell lines23,70,71 is directed into pyrimidine and nucleotide synthesis in
including a human ccRCC cell line9,72, inhibition of gluta­ these tumours21. Consistent with this model, arginine
minase (which catalyses the conversion of glutamine deprivation inhibited tumour growth in the RENCA
to glutamate) or removal of glutamine from the culture mouse model of RCC22. This finding suggests a new
media resulted in a decrease in cell survival in vitro, sug‑ therapeutic strategy aimed at depletion of extracellular
gesting a dependence on exogenous glutamine (termed arginine, which is discussed below.
glutamine addiction) in these malignancies.
Functional imaging
Arginine reprogramming Glucose
The semi-essential amino acid arginine has a vital role in The current standard staging procedure for RCC
multiple metabolic pathways, including protein syn‑ employs computed tomography (CT) scanning. The
thesis and the production of nitric oxide, polyamines, high uptake of glucose by cancer cells (a signature of the
urea, creatine, nucleotides, proline, and glutamate73,74. Warburg effect) can be visualized in vivo using positron
Arginine is synthesized from citrulline in two steps of the emission tomography (PET) imaging with the glucose
urea cycle: citrulline and aspartate are first converted to analogue 18F-fluorodeoxyglucose (18F-FDG)80. This
argininosuccinate via the enzyme argininosuccinate syn‑ technique has revolutionized imaging for some malig‑
thase‑1 (ASS1), and argininosuccinate is then converted nancies81–83, but is problematic in RCC owing to variable
into arginine and fumarate by argininosuccinate lyase tumour uptake of 18F-FDG84 and its secretion into the
(FIG. 4). ASS1 is the rate-limiting enzyme for the conver‑ urinary system85, so is not commonly used for routine
sion of citrulline to arginine for ammonia detoxification staging of kidney tumours.
through the urea cycle in the liver and kidney cortex75. A meta-analysis of 158 articles published between
Regeneration of arginine from citrulline is, therefore, 2004 and 2015 suggested that in RCC, 18F-FDG PET
dependent on an adequate supply of active ASS1. has lower sensitivity for diagnosis of primary renal
The loss of ASS1 or its absence during oncogenesis masses but higher sensitivity for diagnosis of metasta‑
makes cells dependent on extracellular sources of argi‑ ses86 than enhanced CT. 18F-FDG PET could, therefore,
nine for survival, a state known as arginine auxotrophy76. potentially be used as a noninvasive pharmacodynamic
Several studies have reported that ASS1 is not expressed marker for the response to novel targeted anticancer
in a variety of epithelial and lymphoid tumours, and agents in patients with advanced (metastatic) RCC. To
improve the effectiveness of 18F-FDG tracer utilization in
advanced RCC, the relationship between the molecular
5-hydroxy-L-tryptophan Tryptophan Tryptamine features of RCC and standard uptake values obtained by
DDC
MAOA or
18
F-FDG PET was evaluated87–90. These studies showed
Epacadostat IDO*
MAOB that many RCC targeted therapies disrupt transcription
of GLUT‑1 and its translocation to the plasma mem‑
DDC N-formyl-kynurenine Indole-3-acetaldehyde brane to promote glucose utilization89. 18F-FDG PET
has also been evaluated as an indicator of prognosis and
ALDH2
of treatment response in patients with advanced RCC
KN QN
receiving targeted therapies, such as inhibitors of PI3K,
Serotonin Immunosuppressors Indoleacetate
AKT and mTOR90,91. The results suggest that ­18F-FDG
Increased Decreased Unknown PET is a useful pharmacodynamics biomarker for
­assessing the efficacy of these therapies.
Levels increased Levels decreased Levels unknown
Glutamine
Figure 2 | Altered tryptophan metabolism in clear cell renal cell carcinoma (ccRCC). As many ccRCCs are glutamine avid and glutamine
Nature Reviews | Nephrology
In ccRCC, upregulation of tryptophan metabolism through the kynurenine (KN) pathway
reprogramming has been identified in ccRCC, this
results in increased production of the immunosuppressive metabolites KN and
quinolinate (QN). This pathway can be inhibited using the indoleamine 2,3‑dioxygenase amino acid is now being exploited for novel PET-based
(IDO) inhibitor epacadostat. The levels of enzymes that feed the serotonin and imaging techniques. A series of publications suggest that
indoleacetate pathways of tryptophan metabolism, including DOPA decarboxylase
18
F-FDG negative tumours might utilize glutaminolysis
(DDC), monoamine oxidase A (MAOA) or MAOB and aldehyde dehydrogenase 2 preferentially to glycolysis92–95. Concomitantly, we and
(ALDH2), are decreased in ccRCC. *Upregulated in tumour endothelial cells. others have found that RCC is strongly glutamine avid

414 | JULY 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Glucose

GLUT-1

• Triglycerides
Glucose Cholesterol Unsaturated FA
↑ Glycolysis • Phopholipids
Mitochondria
HK
• Elongation
SCD1
• Desaturation
G6P
Carnitine ↓ β-oxidation
GPI
Short chain FA
Fatty acyl-CoA Acylcarnitines Fatty acyl-CoA VLCAD
F6P Long chain FA
PDK1 • HADH • MCAD
• GAPDH • PGK
• PFKP • ENO • SCEH • ACAT1 Fatty acid
TVB-2640 FAS
• ALDO • PKM2 synthesis
PDH Acetyl-CoA
Acetyl-CoA Fatty acyl-CoA
Pyruvate

PC Oxaloacetate Citrate Citrate


HIF-α LDH-A
Malate Cis-aconitate
PT2385 Lactate TCA CB-839
cycle ↑ Reductive
Fumarate Isocitrate carboxylation
↑ Lactate fermentation
SDHA or IDH GLS
SDHB
Succinate α-KG α-KG Glutamate Glutamine

Increased Levels increased • GST


• GGT
Decreased Levels decreased GPX1
Unknown Levels unknown ROS inhibition GSSG GSH

Figure 3 | Altered energy and glutamine metabolism in clear cell renal cell carcinoma (ccRCC). In ccRCC, aerobic
Nature Reviews | Nephrology
glycolysis, lactate fermentation, carnitine and lipid synthesis, reductive carboxylation and the glutathione/oxidized
glutathione (GSH/GSSG) pathway are upregulated, whereas energy production through the tricarboxylic acid (TCA) cycle
is downregulated. These altered metabolic pathways provide opportunities for therapy: increased lactate fermentation
can be targeted using the hypoxia-inducible factor α (HIF‑α) inhibitor PT2385; fatty acid synthesis can be inhibited using
the fatty acid synthase (FAS) inhibitor TVB‑2640; and glutamine metabolism can be inhibited using the glutaminase (GLS)
inhibitor CB‑839. ACAT1, acetyl-CoA acetyltransferase 1; α‑KG, α‑ketoglutarate; ALDO, aldolase; ENO, enolase; FA, fatty
acid; F6P, fructose 6‑phosphate; GAPDH, glyceraldehyde 3‑phosphate dehydrogenase; GGT, γ‑glutamyl transpeptidase;
GLUT‑1, glucose transporter 1; GPI, glucose-6‑phosphate isomerase; GPX1, glutathione peroxidase; GST, glutathione
S‑transferase; G6P, glucose-6‑phosphate; HADH, hydroxyacyl-CoA dehydrogenase; HK, hexokinase; IDH, isocitrate
dehydrogenase; LDH‑A, lactate dehydrogenase A; MCAD, medium-chain specific acyl-CoA dehydrogenase; PC, pyruvate
carboxylase; PDH, pyruvate dehydrogenase; PDK1, pyruvate dehydrogenase kinase 1; PFKP, phosphofructokinase;
PGK, phosphoglycerate kinase; PKM2, pyruvate kinase 2; ROS, reactive oxygen species; SCD1, stearoyl-CoA desaturase‑1;
SCEH, short-chain enoyl-CoA hydratase; SDH, succinate dehydrogenase; VLCAD, very long-chain specific acyl-CoA
dehydrogenase.

and possibly even glutamine addicted9,17. As a result HIF pathway


of this work, the glutamine analogue 4-18F-(2S,4R)- Utilization of the HIF pathway for imaging has also
fluoroglutamine was developed and shown to be taken been investigated, capitalizing on HIF-1α stabilization
up by cancer cells96. and degradation as well as its interaction with VHL98,99.
PET imaging has been used to show glutamine uptake These efforts as well as the first‑in‑kind use of glutamine
in animal models and in patients with glioma97. Moreover, metabolic reprogramming for real-time patient imaging
glutamine uptake correlated with disease progression in sets the stage for further exploitation of reprogrammed
these patients. In ccRCC, we used PET scanning to show metabolic pathways in patient care.
glutamine uptake in vitro and in several mouse xenograft
models (O.A.A. and R.H.W, unpublished observations). Therapeutic approaches
Imaging based on the glutamine reprogramming of RCC Disruption of VHL through mutation, deletion or methy­
could potentially be used for RCC staging, patient selec‑ lation is widely recognized as the most fundamental and
tion, and real-time monitoring of glutaminase inhibition critical molecular alteration in ccRCC19. This disruption
as a novel therapy for this disease. leads to deregulation of downstream oxygen-sensing

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | JULY 2017 | 415


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Extracellular Mitochondria to resistance owing to the development of binding site


Ornithine Citruline mutations in HIF‑2α and second site suppressor muta‑
OCT tions in HIF‑1β. The study investigators also identi­
fied a gene signature of HIF‑2α‑driven tumours that
could potentially be used to select patients who might
Urea cycle bene­it from HIF‑2α–targeted therapy24. Clinical trials
Ornithine Citruline of the PT2399 analogue, PT2385 (an orally bioavailable
ASL inhibitors ­formulation), are ongoing in patients with ccRCC102.
Urea Aspartate
Arginase ASS1
H20 Glutaminase inhibitors
ASL
Arginine Arginine Argininoscuccinate Another promising approach is direct targeting of
enzymes involved in tumour metabolism, such as
Increased
glutaminase (FIG. 3). In ccRCC, glutaminase activity is
ADI-PEG20

Fumarate
Decreased
thought to restore the TCA cycle in the context of the
Unknown
• Polyamines • Nitric oxide Warburg effect23,70. This restored pathway provides cellu‑
Levels increased
• Proline and • Nucleotides lar building blocks for rapidly proliferating ccRCC cells.
glutamate • Urea Levels decreased
Citruline • Creatine Levels unknown
In a clinical trial, the small molecule inhibitor of glu‑
Cytosol taminase, CB‑839, either alone or in combination with
Figure 4 | Altered arginine metabolism in clear cell renal cell carcinoma (ccRCC). the mTOR inhibitor everolimus, showed encouraging
Nature Reviews | Nephrology clinical activity in patients with RCC; nine of 15 (60%)
Arginine is synthesized from citrulline in two steps of the urea cycle. Argininosuccinate
synthase‑1 (ASS1) and argininosuccinate lyase (ASL) regulate intracellular arginine evaluable patients who received twice-daily therapy
levels. As ASS1 levels are markedly decreased in all grades of ccRCC, the tumour cells are showed radiographic partial response or stable disease
dependent on extracellular sources of arginine for their survival. Extracellular arginine at a median follow-up of 4.6 months72.
can be depleted using the pegylated form of arginine deaminase enzyme (ADI‑PEG20),
which converts arginine to citruline. ASL inhibitors can also be used for targeted therapy FAS inhibitors
of ccRCC. OCT, ornithine carbamoyl transferase.
FAS is another metabolic enzyme for which drug devel‑
opment efforts are coalescing. In RCC, FAS overexpres‑
sion is associated with tumour aggressiveness and poor
pathways, causing accumulation of HIF‑1α and sub‑ prognosis41. Interestingly, reports suggest that among
sequent upregulation of hypoxia-response genes100, patients with metastatic RCC, those who are obese
resulting in a clinical phenotype characterized by florid have lower FAS expression in their tumours and this
vasculature principally modulated by vascular endothe‑ reduction potentially contributes to an overall better
lial growth factor (VEGF) signalling pathways. In the prognosis for obese patients compared to those who are
past decade, remarkable advances have been made in not obese103. In lung, ovarian, prostate, and pancreatic
ccRCC drug development, primarily focusing on tar‑ tumour xenografts, FAS inhibition in combination with
geting either VEGF receptors (VEGFR) using kinase paclitaxel or docetaxel promoted cell death, disrupted
inhibitors or the VEGF ligand using mono­clonal anti‑ lipid rafts in cell membranes, and abrogated key signal‑
bodies. These advances led to FDA approval of several ling pathways104. A clinical trial of the novel FAS inhibi­
inhibitors of VEGFR (sunitinib, pazopanib, sorafenib, tor TVB‑2640 in patients with advanced stage solid
axitinib, cabozantinib and lenvantinib) and VEGF tumours is underway105.
(bevacizumab). These angiogenesis inhibitors were
only modestly effective in ccRCC, however, and often IDO inhibitors
characterized by off-target effects and chronic irrita­ Immunotherapy provides another avenue through
tive toxicities such as fatigue and rash101. Furthermore, which inhibitors of metabolic pathways have poten‑
the metabolic basis of ccRCC is only partly addressed tial clinical applications106. Among the most clinically
by targeting the terminal phenotype driven by VEGF. advanced approaches are inhibitors of IDO, which
Subsequent encouraging efforts in drug development has a role in one of many T‑cell immune checkpoints
have attempted to exploit ccRCC metabolic reprogram‑ relevant to cancer biology. As mentioned above, IDO
ming by targeting critical proteins or enzymes involved serves in the rate-limiting step of tryptophan catabolism
in dysregulated metabolic pathways. through the kynurenine pathway. This pathway leads to
tryptophan depletion in the local tumour microenviron‑
HIF‑2α antagonists ment, resulting in suppression of antitumour T cells34.
Among the most promising approaches to target meta­ Inhibition of IDO prevents this immunosuppressive
bolic reprogramming in ccRCC is the development of effect and e­ nables T‑cell activation34.
selective HIF‑2α antagonists24,61 such as the small mol‑ One of the IDO inhibitors currently being tested in
ecule inhibitor, PT2399, which was identified using a RCC is the selective IDO1‑targeted inhibitor epacadostat
structure-based design approach24. In preclinical stud‑ (FIG. 2). This orally bioavailable formulation enhances the
ies, PT2399 was found to dissociate the HIF‑2 hetero­ lytic ability of tumour-antigen-specific T cells in preclin‑
dimer (HIF-2α–HIF-1β) in ccRCC cells, resulting in ical models107. A clinical trial combining epacadostat
inhibition of tumour growth in the majority of cell lines with the PD‑1 inhibitor pembrolizumab is ongoing in
tested24. Interestingly, prolonged PT2399 treatment led patients with several types of malignancies, including

416 | JULY 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

ccRCC108. Early results from a cohort of patients with Conclusions and future perspectives
melanoma in this trial showed encouraging activity; in Kidney cancer is a disease of aberrant cell cycle progres‑
those with treatment-naive advanced melanoma (n = 19) sion that critically involves reprogramming of classical
the disease control rate was 74%, whereas the overall metabolic pathways that are important for the produc‑
Response Evaluation Criteria In Solid Tumours response tion of energy and cellular components as well as for the
rate was 58%. control of immune surveillance. For this reason, RCC can
be described as a metabolic disease, and all components
Arginine depletion of clinical management, from imaging to pathology to
The pegylated form of arginine deaminase (ADI‑PEG20) therapeutics can in theory capitalize on these repro‑
can be used to deplete arginine through catalytic deam‑ grammed pathways. Such new approaches are currently
ination of arginine to citrulline22,109 (FIG. 4). Cells that being ­evaluated and are close to fruition.
express normal levels of ASS1, such as normal kidney In the future, we foresee an era in which malignancies,
parenchyma cells, can recycle citrulline back to arginine, including RCC, will be classified on the basis of enhance‑
whereas cells that lack ASS1, such as ccRCC cells, are not ment or alteration of metabolic pathways in addition
able produce arginine from citrulline. ADI‑PEG20 can, to the site of occurrence or microscopic pathology111.
therefore, exert antitumour activity by limiting arginine Tumour heterogeneity must be considered, however, and
availability in tumour cells without any adverse effect is particularly relevant in RCC112. Separate regions of indi‑
on the surrounding renal parenchyma. Clinical trials of vidual tumours likely have distinct metabolic reprogram‑
ADI‑PEG20 are underway in hepatocellular carcinoma, ming and understanding of this variation is required to
acute myeloid leukaemia, non-small cell lung cancer, enable the development of successful treatments. Despite
non-Hodgkin lymphoma, breast carcinoma, melanoma this heterogeneity, therapeutics specifically targeted to
and mesothelioma110. Although arginine deprivation tumour metabolism, rather than toxic to all proliferating
using ADI‑PEG20 seems to be a promising strategy cells, are highly likely to be more effective, and have far
for treating ASS1‑deficient tumours such as ccRCC, fewer adverse effects than existing therapies. Metabolic
the effectiveness of this therapy could potentially be reprogramming is rapidly being translated into clinical
limited by the ability of some tumours to re‑express advances and we anticipate the development of additional
ASS1 (REF. 22). novel therapies for renal cancer in the near future.

1. Cline, M. J., Slamon, D. J. & Lipsick, J. S. Oncogenes: carcinoma: memorial sloan-kettering cancer center 24. Chen, W. et al. Targeting renal cell carcinoma with a
implications for the diagnosis and treatment of cancer. experience. Clin. Cancer Res. 10, 6302s–6303s (2004). HIF‑2 antagonist. Nature 539, 112–117 (2016).
Ann. Intern. Med. 101, 223–233 (1984). 13. Metallo, C. M. et al. Reductive glutamine metabolism Demonstration of HIF‑2 antagonism as a possible
2. Warburg, O. On the origin of cancer cells. Science 123, by IDH1 mediates lipogenesis under hypoxia. Nature therapeutic approach in RCC.
309–314 (1956). 481, 380–384 (2012). 25. Nickerson, M. L. et al. Improved identification of von
The classic work of Warburg demonstrating the 14. Mullen, A. R. et al. Reductive carboxylation supports Hippel–Lindau gene alterations in clear cell renal
eponymous aerobic glycolysis effect. growth in tumour cells with defective mitochondria. tumors. Clin. Cancer Res. 14, 4726–4734 (2008).
3. Weiss, R. H. & Lin, P.‑Y. Kidney cancer: identification of Nature 481, 385–388 (2012). 26. Kaelin, W. G. Jr. The von Hippel–Lindau tumor
novel targets for therapy. Kidney Int. 69, 224–232 Introducing the concept of reductive carboxylation suppressor gene and kidney cancer. Clin. Cancer Res.
(2006). in ccRCC. 10, 6290s–6295s (2004).
4. Hu, S. L. et al. The nephrologist’s tumor: basic biology 15. Jiang, P., Du, W. & Wu, M. Regulation of the pentose 27. Cancer Genome Atlas Research Network.
and management of renal cell carcinoma. J. Am. Soc. phosphate pathway in cancer. Protein Cell 5, 592–602 Comprehensive molecular characterization of clear cell
Nephrol. 27, 2227–2237 (2016). (2014). renal cell carcinoma. Nature 499, 43–49 (2013).
A discussion of what practicing and research 16. Sayin, V. I. et al. Antioxidants accelerate lung cancer The Cancer Genome Atlas transcriptomic description
nephrologists need to know about kidney cancer. progression in mice. Sci. Transl Med. 6, 221ra15 of ccRCC.
5. Cantor, J. R. & Sabatini, D. M. Cancer cell metabolism: (2014). 28. Perroud, B. et al. Pathway analysis of kidney cancer
one hallmark, many faces. Cancer Discov. 2, 881–898 Discussion of the possibility that antioxidants can using proteomics and metabolic profiling. Mol. Cancer
(2012). worsen cancer; this effect might be related to 5, 64 (2006).
A detailed description of the concept of metabolic glutamine reprogramming to the glutathione 29. Schaechter, J. D. & Wurtman, R. J. Serotonin release
reprogramming in cancer. pathway. varies with brain tryptophan levels. Brain Res. 532,
6. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: 17. Wise, D. R. & Thompson, C. B. Glutamine addiction: 203–210 (1990).
the next generation. Cell 144, 646–674 (2011). a new therapeutic target in cancer. Trends Biochem. 30. Fallarino, F. et al. T cell apoptosis by tryptophan
7. van der Mijn, J. C., Panka, D. J., Geissler, A. K., Sci. 35, 427–433 (2010). catabolism. Cell Death Differ. 9, 1069–1077 (2002).
Verheul, H. M. & Mier, J. W. Novel drugs that target A concise discussion of glutamine reprogramming 31. Shimizu, T., Nomiyama, S., Hirata, F. & Hayaishi, O.
the metabolic reprogramming in renal cell cancer. in cancer. Indoleamine 2,3‑dioxygenase. Purification and some
Cancer Metab. 4, 14 (2016). 18. Baldewijns, M. M. et al. VHL and HIF signalling in renal properties. J. Biol. Chem. 253, 4700–4706 (1978).
8. Hakimi, A. A. et al. An integrated metabolic atlas cell carcinogenesis. J. Pathol. 221, 125–138 (2010). 32. Lee, G. K. et al. Tryptophan deprivation sensitizes
of clear cell renal cell carcinoma. Cancer Cell 29, 19. Bausch, B. et al. Renal cancer in von Hippel–Lindau activated T cells to apoptosis prior to cell division.
104–116 (2016).
 disease and related syndromes. Nat. Rev. Nephrol. 9, Immunology 107, 452–460 (2002).
Use of ccRCC transcriptomics and metabolomics 529–538 (2013). 33. Riesenberg, R. et al. Expression of indoleamine
to develop a visualization tool and demonstration 20. Perroud, B., Ishimaru, T., Borowsky, A. D. & Weiss, R. H. 2,3‑dioxygenase in tumor endothelial cells correlates
of discordance between the transcriptome and Grade-dependent proteomics characterization of kidney with long-term survival of patients with renal cell
metabolome in this disease. cancer. Mol. Cell. Proteomics 8, 971–985 (2008). carcinoma. Clin. Cancer Res. 13, 6993–7002 (2007).
9. Wettersten, H. I. et al. Grade-dependent metabolic 21. Rabinovich, S. et al. Diversion of aspartate in 34. Trott, J. F. et al. Inhibiting tryptophan metabolism
reprogramming in kidney cancer revealed by combined ASS1‑deficient tumours fosters de novo pyrimidine enhances interferon therapy in kidney cancer.
proteomics and metabolomics analysis. Cancer Res. synthesis. Nature 527, 379–383 (2015). Oncotarget 7, 66540–66557 (2016).
75, 2541–2552 (2015). 22. Yoon, C. Y. et al. Renal cell carcinoma does not express The first description of the use of a modulator of
Use of metabolomics and proteomics to argininosuccinate synthetase and is highly sensitive tryptophan reprogramming in a murine homograft
demonstrate metabolic reprogramming in ccRCC to arginine deprivation via arginine deiminase. RCC model.
and to identify new drug targets. Int. J. Cancer 120, 897–905 (2007). 35. Kim, K. et al. Urine metabolomic analysis identifies
10. Linehan, W. M. & Ricketts, C. J. The metabolic basis The first description of the use of ADI-PEG to target potential biomarkers and pathogenic pathways in
of kidney cancer. Semin. Cancer Biol. 23, 46–55 arginine reprogramming in a murine homograft RCC kidney cancer. OMICS 15, 293–303 (2011).
(2013). model. The first non-targeted urine metabolomics study
11. Linehan, W. M., Srinivasan, R. & Schmidt, L. S. 23. Gross, M. I. et al. Antitumor activity of the glutaminase in RCC, which identified immune modulators as
The genetic basis of kidney cancer: a metabolic disease. inhibitor CB‑839 in triple-negative breast cancer. possible biomarkers of ccRCC.
Nat. Rev. Urol. 7, 277–285 (2010). Mol. Cancer Ther. 13, 890–901 (2014). 36. Carracedo, A., Cantley, L. C. & Pandolfi, P. P. Cancer
12. Motzer, R. J., Bacik, J. & Mazumdar, M. Prognostic The first description of the use of a glutaminase metabolism: fatty acid oxidation in the limelight.
factors for survival of patients with stage IV renal cell inhibitor in cancer. Nat. Rev. Cancer 13, 227–232 (2013).

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | JULY 2017 | 417


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

37. Wakil, S. J. Fatty acid synthase, a proficient sensitizes VHL-deficient cells to glutamine deprivation. 88. Kayani, I. et al. Sequential FDG-PET/CT as a biomarker
multifunctional enzyme. Biochemistry 28, 4523–4530 Cell Metab. 17, 372–385 (2013). of response to Sunitinib in metastatic clear cell renal
(1989). 64. Shim, E. H. et al. L-2‑hydroxyglutarate: an epigenetic cancer. Clin. Cancer Res. 17, 6021–6028 (2011).
38. Enoch, H. G., Catala, A. & Strittmatter, P. Mechanism modifier and putative oncometabolite in renal cancer. 89. Ma, W. W. et al. [18F]fluorodeoxyglucose positron
of rat liver microsomal stearyl-CoA desaturase. Cancer Discov. 4, 1290–1298 (2014). emission tomography correlates with Akt pathway
Studies of the substrate specificity, enzyme–substrate An early demonstration of a newly identified activity but is not predictive of clinical outcome during
interactions, and the function of lipid. J. Biol. Chem. oncometabolite active in RCC. mTOR inhibitor therapy. J. Clin. Oncol. 27, 2697–2704
251, 5095–5103 (1976). 65. Kanehisa, M., Furumichi, M., Tanabe, M., Sato, Y. & (2009).
39. Gebhard, R. L. et al. Abnormal cholesterol metabolism Morishima, K. KEGG: new perspectives on genomes, 90. Mizuno, T. et al. Clinically significant association
in renal clear cell carcinoma. J. Lipid Res. 28, pathways, diseases and drugs. Nucleic Acids Res. 45, between the maximum standardized uptake value on
1177–1184 (1987). D353–D361 (2017). 18F‑FDG PET and expression of phosphorylated Akt
40. von Roemeling, C. A. et al. Stearoyl-CoA desaturase 1 is 66. Kanehisa, M., Sato, Y., Kawashima, M., Furumichi, M. and S6 kinase for prediction of the biological
a novel molecular therapeutic target for clear cell renal & Tanabe, M. KEGG as a reference resource for gene characteristics of renal cell cancer. BMC Cancer 15,
cell carcinoma. Clin. Cancer Res. 19, 2368–2380 and protein annotation. Nucleic Acids Res. 44, 1097 (2015).
(2013). D457–D462 (2016). 91. Nakaigawa, N. et al. FDG PET/CT as a prognostic
41. Horiguchi, A. et al. Fatty acid synthase over 67. Kanehisa, M. & Goto, S. KEGG: kyoto encyclopedia of biomarker in the era of molecular-targeting therapies:
expression is an indicator of tumor aggressiveness and genes and genomes. Nucleic Acids Res. 28, 27–30 max SUVmax predicts survival of patients with
poor prognosis in renal cell carcinoma. J. Urol. 180, (2000). advanced renal cell carcinoma. BMC Cancer 16, 67
1137–1140 (2008). 68. Kamm, D. E. & Strope, G. L. The effects of acidosis (2016).
42. Ganti, S. et al. Urinary acylcarnitines are altered in and alkalosis on the metabolism of glutamine and 92. Hambardzumyan, D., Amankulor, N. M., Helmy, K. Y.,
kidney cancer. Int. J. Cancer 130, 2791–2800 (2012). glutamate in renal cortex slices. J. Clin. Invest. 51, Becher, O. J. & Holland, E. C. Modeling adult gliomas
43. Weidemann, A. & Johnson, R. S. Biology of 1251–1263 (1972). using RCAS/t‑va technology. Transl Oncol. 2, 89–95
HIF‑1alpha. Cell Death Differ. 15, 621–627 (2008). 69. Gatto, F., Miess, H., Schulze, A. & Nielsen, J. Flux (2009).
44. Warburg, O., Wind, F. & Negelein, E. The metabolism balance analysis predicts essential genes in clear cell 93. Mills, C. D. M1 and M2 macrophages: oracles of health
of tumors in the body. J. Gen. Physiol. 8, 519–530 renal cell carcinoma metabolism. Sci. Rep. 5, 10738 and disease. Crit. Rev. Immunol. 32, 463–488 (2012).
(1927). (2015). 94. Wu, C., Li, F., Niu, G. & Chen, X. PET imaging of
45. Ward, P. S. & Thompson, C. B. Metabolic 70. Chakrabarti, G. et al. Targeting glutamine metabolism inflammation biomarkers. Theranostics 3, 448–466
reprogramming: a cancer hallmark even warburg did sensitizes pancreatic cancer to PARP-driven metabolic (2013).
not anticipate. Cancer Cell 21, 297–308 (2012). catastrophe induced by ss‑lapachone. Cancer Metab. 95. Carman, A. J., Mills, J. H., Krenz, A., Kim, D. G. &
46. Wu, J., Ocampo, A. & Izpisua Belmonte, J. C. Cellular 3, 12 (2015). Bynoe, M. S. Adenosine receptor signaling modulates
metabolism and induced pluripotency. Cell 166, 71. Jacque, N. et al. Targeting glutaminolysis has permeability of the blood–brain barrier. J. Neurosci.
1371–1385 (2016). antileukemic activity in acute myeloid leukemia and 31, 13272–13280 (2011).
47. Sellers, K. et al. Pyruvate carboxylase is critical for synergizes with BCL‑2 inhibition. Blood 126, 96. Lieberman, B. P. et al. PET imaging of glutaminolysis in
non-small-cell lung cancer proliferation. J. Clin. Invest. 1346–1356 (2015). tumors by 18F-(2S,4R)4‑fluoroglutamine. J. Nucl. Med.
125, 687–698 (2015). 72. Meric-Bernstam, F. et al. Phase 1 study of CB‑839, 52, 1947–1955 (2011).
48. Ozcan, A., Shen, S. S., Zhai, Q. J. & Truong, L. D. a small molecule inhibitor of glutaminase (GLS), alone First description of the potential use of
Expression of GLUT1 in primary renal tumors: and in combination with everolimus (E) in patients (pts) glutamine-PET for evaluation of cancer metabolism.
morphologic and biologic implications. Am. J. Clin. with renal cell cancer (RCC) [abstract]. J. Clin. Oncol. 97. Venneti, S. et al. Glutamine-based PET imaging
Pathol. 128, 245–254 (2007). 34 (Suppl.), 4568 (2016). facilitates enhanced metabolic evaluation of gliomas
49. Li, B. et al. Fructose‑1,6‑bisphosphatase opposes 73. Wu, G. & Morris, S. M. Jr. Arginine metabolism: nitric in vivo. Sci. Transl Med. 7, 274ra17 (2015).
renal carcinoma progression. Nature 513, 251–255 oxide and beyond. Biochem. J. 336, 1–17 (1998). 98. Choi, C. Y. et al. Molecular imaging of hypoxia-
(2014). 74. Husson, A., Brasse-Lagnel, C., Fairand, A., Renouf, S. inducible factor 1 alpha and von Hippel–Lindau
50. Singer, K. et al. Warburg phenotype in renal cell & Lavoinne, A. Argininosuccinate synthetase from the interaction in mice. Mol. Imaging 7, 139–146 (2008).
carcinoma: high expression of glucose-transporter 1 urea cycle to the citrulline–NO cycle. Eur. J. Biochem. 99. Moroz, E. et al. Real-time imaging of HIF‑1alpha
(GLUT‑1) correlates with low CD8+ T‑cell infiltration in 270, 1887–1899 (2003). stabilization and degradation. PLoS ONE 4, e5077
the tumor. Int. J. Cancer 128, 2085–2095 (2011). 75. Haines, R. J., Pendleton, L. C. & Eichler, D. C. (2009).
51. Fischer, K. et al. Inhibitory effect of tumor cell-derived Argininosuccinate synthase: at the center of arginine 100. Riazalhosseini, Y. & Lathrop, M. Precision medicine
lactic acid on human T cells. Blood 109, 3812–3819 metabolism. Int. J. Biochem. Mol. Biol. 2, 8–23 (2011). from the renal cancer genome. Nat. Rev. Nephrol. 12,
(2007). 76. Delage, B. et al. Arginine deprivation and 655–666 (2016).
52. Mandriota, S. J. et al. HIF activation identifies early argininosuccinate synthetase expression in the 101. Schmidinger, M. Understanding and managing
lesions in VHL kidneys: evidence for site-specific tumor treatment of cancer. Int. J. Cancer 126, 2762–2772 toxicities of vascular endothelial growth factor (VEGF)
suppressor function in the nephron. Cancer Cell 1, (2010). inhibitors. EJC Suppl. 11, 172–191 (2013).
459–468 (2002). 77. Qiu, F. et al. Arginine starvation impairs mitochondrial 102. US National Library of Medicine. ClinicalTrials.gov
53. Semenza, G. L. Regulation of cancer cell metabolism respiratory function in ASS1‑deficient breast cancer https://clinicaltrials.gov/ct2/show/NCT02293980
by hypoxia-inducible factor 1. Semin. Cancer Biol. 19, cells. Sci. Signal. 7, ra31 (2014). (2017).
12–16 (2009). 78. Allen, M. D. et al. Prognostic and therapeutic impact 103. Gupta, S. Obesity: the fat advantage. Nature 537,
54. Semenza, G. L. HIF‑1: upstream and downstream of of argininosuccinate synthetase 1 control in bladder S100–S102 (2016).
cancer metabolism. Curr. Opin. Genet. Dev. 20, cancer as monitored longitudinally by PET imaging. 104. Heuer, T. S. et al. FASN inhibition and taxane
51–56 (2010). Cancer Res. 74, 896–907 (2014). treatment combine to enhance anti-tumor efficacy in
55. Papandreou, I., Cairns, R. A., Fontana, L., Lim, A. L. 79. Kobayashi, E. et al. Reduced argininosuccinate diverse xenograft tumor models throgh disruption of
& Denko, N. C. HIF‑1 mediates adaptation to hypoxia synthetase is a predictive biomarker for the tubulin palmitoylation and microtubule organization
by actively downregulating mitochondrial oxygen development of pulmonary metastasis in patients with and FASN inhibition-mediated effects on oncogenic
consumption. Cell Metab. 3, 187–197 (2006). osteosarcoma. Mol. Cancer Ther. 9, 535–544 (2010). signaling and gene expression. EBioMedicine 16,
56. Kim, J. W., Tchernyshyov, I., Semenza, G. L. & 80. Weber, W. A., Schwaiger, M. & Avril, N. Quantitative 51–62 (2017).
Dang, C. V. HIF‑1‑mediated expression of pyruvate assessment of tumor metabolism using FDG-PET 105. US National Library of Medicine. ClinicalTrials.gov
dehydrogenase kinase: a metabolic switch required imaging. Nucl. Med. Biol. 27, 683–687 (2000). https://clinicaltrials.gov/ct2/show/NCT02223247
for cellular adaptation to hypoxia. Cell Metab. 3, 81. Shankar, L. K. et al. Consensus recommendations for (2017).
177–185 (2006). the use of 18F‑FDG PET as an indicator of therapeutic 106. Sheridan, C. IDO inhibitors move center stage in
57. Huang, X. et al. Hypoxia-inducible mir‑210 regulates response in patients in National Cancer Institute trials. immuno-oncology. Nat. Biotechnol. 33, 321–322
normoxic gene expression involved in tumor initiation. J. Nucl. Med. 47, 1059–1066 (2006). (2015).
Mol. Cell 35, 856–867 (2009). 82. Tunariu, N., Kaye, S. B. & Desouza, N. M. Functional 107. Jochems, C. et al. The IDO1 selective inhibitor
58. Valera, V. A., Walter, B. A., Linehan, W. M. & imaging: what evidence is there for its utility in clinical epacadostat enhances dendritic cell immunogenicity
Merino, M. J. Regulatory effects of microRNA‑92 trials of targeted therapies? Br. J. Cancer 106, 619–628 and lytic ability of tumor antigen-specific T cells.
(miR‑92) on VHL gene expression and the hypoxic (2012). Oncotarget 7, 37762–37772 (2016).
activation of miR‑210 in clear cell renal cell carcinoma. 83. Juweid, M. E. & Cheson, B. D. Positron-emission 108. Incyte. Press releases. Incyte http://phx.corporate-ir.
J. Cancer 2, 515–526 (2011). tomography and assessment of cancer therapy. net/phoenix.zhtml?c=69764&p=irol-newsArticle_
59. White, N. M. et al. miRNA profiling in metastatic renal N. Engl. J. Med. 354, 496–507 (2006). print&ID=2210055 (2017).
cell carcinoma reveals a tumour-suppressor effect for 84. Robey, I. F. et al. Regulation of the Warburg effect in 109. Yoon, J. K., Frankel, A. E., Feun, L. G., Ekmekcioglu, S.
miR‑215. Br. J. Cancer 105, 1741–1749 (2011). early-passage breast cancer cells. Neoplasia (New & Kim, K. B. Arginine deprivation therapy for
60. Ivan, M. & Huang, X. miR‑210: fine-tuning the hypoxic York, N.Y.) 10, 745–756 (2008). malignant melanoma. Clin. Pharmacol. 5, 11–19
response. Adv. Exp. Med. Biol. 772, 205–227 (2014). 85. Gofrit, O. N. & Orevi, M. Diagnostic challenges in (2013).
61. Cho, H. et al. On‑target efficacy of a HIF‑2alpha kidney cancer: a systematic review of the role of 110. US National Library of Medicine. ClinicalTrials.gov
antagonist in preclinical kidney cancer models. Nature PET/CT. J. Urol. 196, 648–657 (2016). https://clinicaltrials.gov/ct2/results?term=ADI-
539, 107–111 (2016). 86. Aide, N. et al. Efficiency of [18F]FDG PET in PEG20+&Search=Search (2017).
A classic paper showing the link between HIF‑1 and characterising renal cancer and detecting distant 111. Peyser, N. D. & Grandis, J. R. Cancer genomics: spot
VHL and its role in RCC. metastases: a comparison with CT. Eur. J. Nucl. Med. the difference. Nature 541, 162–163 (2017).
62. Catchpole, G. et al. Metabolic profiling reveals key Mol. Imaging 30, 1236–1245 (2003). 112. Gerlinger, M. et al. Intratumor heterogeneity and
metabolic features of renal cell carcinoma. J. Cell. Mol. 87. Ueno, D. et al. Early assessment by FDG-PET/CT of branched evolution revealed by multiregion
Med. 15, 109–118 (2011). patients with advanced renal cell carcinoma treated sequencing. N. Engl. J. Med. 366, 883–892 (2012).
63. Gameiro, P. A. et al. In vivo HIF-mediated reductive with tyrosine kinase inhibitors is predictive of disease Discussion of the occurrence of intratumoral
carboxylation is regulated by citrate levels and course. BMC Cancer 12, 162 (2012). heterogeneity in ccRCC.

418 | JULY 2017 | VOLUME 13 www.nature.com/nrneph


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

113. Xu, K. Y. & Wu, S. Update on the treatment of 123. Toschi, A., Lee, E., Gadir, N., Ohh, M. & Foster, D. A. 133. Shim, H. et al. c‑Myc transactivation of LDH‑A:
metastatic clear cell and non-clear cell renal cell Differential dependence of hypoxia-inducible factors 1 implications for tumor metabolism and growth.
carcinoma. Biomark. Res. 3, 5 (2015). alpha and 2 alpha on mTORC1 and mTORC2. J. Biol. Proc. Natl Acad. Sci. USA 94, 6658–6663 (1997).
114. King, S. C., Pollack, L. A., Li, J., King, J. B. & Chem. 283, 34495–34499 (2008). 134. Kim, J. W., Gao, P., Liu, Y. C., Semenza, G. L. &
Master, V. A. Continued increase in incidence of renal 124. Park, J. Y., Lin, P. Y. & Weiss, R. H. Targeting the PI3K– Dang, C. V. Hypoxia-inducible factor 1 and
cell carcinoma, especially in young patients and high Akt pathway in kidney cancer. Expert Rev. Anticancer dysregulated c‑Myc cooperatively induce vascular
grade disease: United States 2001 to 2010. J. Urol. Ther. 7, 863–870 (2007). endothelial growth factor and metabolic switches
191, 1665–1670 (2014). 125. Ohh, M. et al. Ubiquitination of hypoxia-inducible hexokinase 2 and pyruvate dehydrogenase kinase 1.
Discussion of potential reasons for the increase in factor requires direct binding to the beta-domain of Mol. Cell. Biol. 27, 7381–7393 (2007).
RCC incidence in the USA. the von Hippel–Lindau protein. Nat. Cell Biol. 2, 135. Gao, P. et al. c‑Myc suppression of miR‑23a/b
115. Tosaka, A. et al. Incidence and properties of renal 423–427 (2000). enhances mitochondrial glutaminase expression and
masses and asymptomatic renal cell carcinoma 126. Schwartzenberg-Bar-Yoseph, F., Armoni, M. & glutamine metabolism. Nature 458, 762–765
detected by abdominal ultrasonography. J. Urol. 144, Karnieli, E. The tumor suppressor p53 down-regulates (2009).
1097–1099 (1990). glucose transporters GLUT1 and GLUT4 gene 136. Shroff, E. H. et al. MYC oncogene overexpression
116. Quinn, D. I. & Lara, P. N. Jr. Renal-cell cancer— expression. Cancer Res. 64, 2627–2633 (2004). drives renal cell carcinoma in a mouse model through
targeting an immune checkpoint or multiple kinases. 127. Bensaad, K. et al. TIGAR, a p53‑inducible regulator of glutamine metabolism. Proc. Natl Acad. Sci. USA 112,
N. Engl. J. Med. 373, 1872–1874 (2015). glycolysis and apoptosis. Cell 126, 107–120 (2006). 6539–6544 (2015).
117. Sumitomo, M. et al. Synergy in tumor suppression by 128. Suzuki, S. et al. Phosphate-activated glutaminase Discussion of the interaction of c‑Myc with
direct interaction of neutral endopeptidase with PTEN. (GLS2), a p53‑inducible regulator of glutamine glutamine reprogramming in RCC.
Cancer Cell 5, 67–78 (2004). metabolism and reactive oxygen species. Proc. Natl 137. Gordan, J. D. et al. HIF-alpha effects on c‑Myc
118. Lee, H. J. et al. Prognostic significance of biallelic loss Acad. Sci. USA 107, 7461–7466 (2010). distinguish two subtypes of sporadic VHL-deficient
of PTEN in clear cell renal cell carcinoma. J. Urol. 192, 129. Gurova, K. V., Hill, J. E., Razorenova, O. V., clear cell renal carcinoma. Cancer Cell 14, 435–446
940–946 (2014). Chumakov, P. M. & Gudkov, A. V. p53 pathway in renal (2008).
119. Inoki, K., Li, Y., Zhu, T., Wu, J. & Guan, K. L. TSC2 is cell carcinoma is repressed by a dominant mechanism.
phosphorylated and inhibited by Akt and suppresses Cancer Res. 64, 1951–1958 (2004). Author contributions
mTOR signalling. Nat. Cell Biol. 4, 648–657 (2002). 130. Shaw, R. J. et al. The tumor suppressor LKB1 kinase All authors researched the data for the article, made substan-
120. Bjornsson, J., Short, M. P., Kwiatkowski, D. J. & directly activates AMP-activated kinase and regulates tial contributions to discussions of the content, wrote the
Henske, E. P. Tuberous sclerosis-associated renal cell apoptosis in response to energy stress. Proc. Natl article and reviewed or edited the manuscript before
carcinoma. Clinical, pathological, and genetic features. Acad. Sci. USA 101, 3329–3335 (2004). submission.
Am. J. Pathol. 149, 1201–1208 (1996). 131. Yalniz, Z. et al. Novel mutations and role of the LKB1
121. Elstrom, R. L. et al. Akt stimulates aerobic glycolysis gene as a tumor suppressor in renal cell carcinoma. Competing interests statement
in cancer cells. Cancer Res. 64, 3892–3899 (2004). Tumour Biol. 35, 12361–12368 (2014). The authors declare no competing interests.
122. Miyamoto, S., Murphy, A. N. & Brown, J. H. Akt 132. Duivenvoorden, W. C. et al. Underexpression of tumour
mediates mitochondrial protection in cardiomyocytes suppressor LKB1 in clear cell renal cell carcinoma is Publisher’s note
through phosphorylation of mitochondrial common and confers growth advantage in vitro and Springer Nature remains neutral with regard to jurisdictional
hexokinase‑II. Cell Death Differ. 15, 521–529 (2008). in vivo. Br. J. Cancer 108, 327–333 (2013). claims in published maps and institutional affiliations.

NATURE REVIEWS | NEPHROLOGY VOLUME 13 | JULY 2017 | 419


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like