Understanding the genetic basis of cancer has led to therapies that target driver mutations and has helped match patients with more personalized drugs. Oncogenic mutations influence tumor metabolism, but other tumor characteristics can also contribute to their metabolic phenotypes. Comparison of isogenic lung and pancreas tumor models suggests that use of some metabolic pathways is defined by lineage rather than by driver mutation. Lung tumors catabolize circulating branched chain amino acids (BCAA) to extract nitrogen for nonessential amino acid and nucleotide synthesis, whereas pancreatic cancer obtains amino acids from catabolism of extracellular protein. These differences in amino acid metabolism translate into distinct pathway dependencies, as genetic disruption of the enzymes responsible for utilization of BCAA nitrogen limits the growth of lung tumors, but not pancreatic tumors. These data argue that some cancer metabolic phenotypes are defined by cancer tissue-of-origin and environment and that these features constrain the influence of genetic mutations on metabolism. A better understanding of the factors defining tumor nutrient utilization could be exploited to help improve cancer therapy. Cancer Res; 77(12); 3131–4. ©2017 AACR.

When the first chemotherapy was developed to treat cancer, new drugs were identified through a combination of happenstance, observation, and deductive reasoning. Promising compounds were refined in the clinic, with effective regimens determined empirically and patients selected based on having a tumor with a specific site-of-origin and histologic classification (1). Early success came in the treatment of hematologic malignancies, with drugs such as the antifolates achieving stunning, but ultimately temporary remissions in some leukemias (2). With a limited understanding of these diseases at a molecular level, or how the drugs worked, physicians developed combination therapy regimens based on nonoverlapping toxicities to achieve longer remissions and even cures, although many patients were pushed to the brink of death by these treatments (1).

In the last three decades, the discovery of oncogenes and tumor-suppressor genes resulted in an improved understanding of how genetic events cause cancer, and dramatically shifted the approaches used to develop new cancer therapies (3, 4). The realization that cancer relies on mutations to corrupt existing pathways controlling development, cell growth, and proliferation suggested these same pathways might be targeted to treat cancer (5, 6). This resulted in the current paradigm where identifying the genetic changes driving tumor development leads to interventions that target vulnerabilities created by those mutations.

In some cases, identification and targeting of genetic drivers have been successful. For example, fusion oncogenes such as BCR-ABL in chronic myelongenous leukemia and anaplastic lymphoma kinase tyrosine kinase receptor (ALK) fusions in lung adenocarcinoma present unique druggable targets that have been exploited clinically with great effect (7). Tyrosine kinase inhibitors have also been effective therapies in cancers driven by mutations that activate growth factor signaling (7). However, it has been challenging to extend this paradigm to treat all cancers in part because tumor-suppressor mutations play a significant role in driving tumorigenesis, but remain difficult to target therapeutically (8). Nevertheless, there is hope that the broad application of tumor sequencing, even if it fails to identify directly targetable mutations, can further personalize treatment approaches to benefit a broader swath of patients (6, 9).

An alternative approach to developing therapies is to target pathways activated downstream of driver mutations or pathways that enable tumor phenotypes. Cancer metabolism represents one such area, although to date the most successful drugs targeting metabolism are chemotherapies that inhibit pathways involved in nucleotide metabolism (10). Changes in metabolism represent some of the first differences described between cancer and normal tissues, dating to Otto Warburg's initial observations that cancer tissues increase glucose uptake and have a tendency to ferment glucose despite the presence of oxygen (11). More recent work has connected increased glucose metabolism to oncogene activation (12), setting the stage for numerous studies detailing the role of oncogenes and tumor suppressors in regulating cancer cell metabolism to promote the accumulation of biomass necessary for enhanced proliferation (13–15). Because drugs are lacking to specifically treat patients with tumors harboring many mutations in genes such KRAS and TP53, one hope is to identify other pathway dependencies in these cancers (16–18). Both KRAS and TP53 mutations result in metabolic changes, and efforts to target the downstream metabolic effects of these events have demonstrated preclinical potential and led to development of drugs in current clinical trials (19, 20). Yet there are also data to suggest that these approaches will not be successful in all KRAS-mutant tumors, raising the possibility that factors other than tumor genetics can contribute to cancer metabolic dependencies (21).

The majority of metabolic changes in cancer have been characterized in culture; however, nutrient availability differs dramatically between culture, tissues, and even within tissues or tumors (22, 23). Indeed, altering the culture environment changes metabolic preferences (24–26) and environment can influence the metabolic phenotypes of cancer cells with differences observed for the same cells in culture and in tumors (21). These studies argue that understanding the metabolic constraints on cancer cells within the native tumor environment will be important for identifying metabolic drug targets.

Analysis of gene expression data from 22 different cancer types and paired normal tissue showed that cancer metabolic gene expression more closely resembles that of the parental tissue than it does of other cancers (27). These data argue that tumors adapt the normal metabolic program of their tissue-of-origin to support inappropriate cell proliferation. In fact, expressing different oncogenes in the same tissue (MYC and MET in the liver) as well as the same oncogene in different tissues (MYC in liver and lung) had distinct effects on glucose and glutamine metabolism, suggesting that both oncogene and tissue type contribute to the metabolic phenotype of cancers (28).

Elevated plasma branched chain amino acids (BCAA) were identified as an early event in pancreatic ductal adenocarcinoma (PDAC) in both mice and humans, a whole body metabolic change that results from increased muscle protein turnover in individuals with this cancer (29). To determine whether the BCAA elevation was unique to PDAC, isogenic mouse models of PDAC and non–small cell lung carcinoma (NSCLC) that are driven by both activating Kras mutations and Trp53 disruption were analyzed, and in contrast to mice with early PDAC, mice with early NSCLC had decreased levels of plasma BCAAs (30). Tracing the fate of isotope-labeled BCAAs in animals with lung or pancreatic cancer showed that NSCLC tumors took up more free BCAAs compared with normal lung tissue, whereas PDAC tumors had decreased BCAA uptake compared with normal pancreas. Unlike PDAC tumors, NSCLC tumors incorporated these amino acids into proteins and used BCAA-derived nitrogen for nonessential amino acid and nucleotide synthesis. In contrast, PDAC tumors rely on the uptake and metabolism of protein in their environment to acquire amino acids (31) as well as nutrients secreted by stromal cells (32, 33). These findings argue that some metabolic phenotypes are dictated by the tissue-of-origin or environment, outside the purview of the initiating driver mutations. For BCAA metabolism, the metabolic preferences between lung and pancreatic cancer were reflected in enzyme expression differences (30). Importantly, there was no selection against expression of genes allowing BCAA metabolism in PDAC tumors, as activation of BCAA metabolism in PDAC tumor cells had no effect of tumor growth. However, deletion of Bcat1 and Bcat2, enzymes involved in obtaining nitrogen from BCAAs, prevented NSCLC tumor formation in vivo, but had limited effect on PDAC tumor growth, arguing that some dependencies are hard-wired based on tumor lineage.

Although most cells in an animal have identical genomes, each tissue and its composite cell types serve a specific physiologic role for the organism. These distinctions and functionalities are conferred by the differential expression of genes and the regulation of gene products. Among these gene products are a subset of all metabolic enzymes, which define the metabolic network available to support cell activities. A corollary to this is that various nutrient levels will vary by location, and levels of available nutrients will also affect which metabolic pathways are utilized by different cell types (34). In other words, there is a reciprocal interplay between the intrinsic set of metabolic enzymes expressed and their regulation by cell signaling, and extrinsic factors such as nutrient availability, which together shape the unique metabolism of each tissue (Fig. 1).

Figure 1.

Tissue-of-origin constrains the metabolic network available to tumors. Each tissue expresses enzymes, which form a core metabolic network that is utilized to support the physiologic function of the tissue. This network is subject to regulation by cell signaling, as well as nutrient and metabolite levels that are influenced by the tissue environment. Most cells maintain some metabolic flexibility and employ additional pathways in response to stress. Oncogenic mutations reshape metabolism to support tumor growth, but their effects are limited by network constraints imposed by tissue-of-origin and by available nutrients.

Figure 1.

Tissue-of-origin constrains the metabolic network available to tumors. Each tissue expresses enzymes, which form a core metabolic network that is utilized to support the physiologic function of the tissue. This network is subject to regulation by cell signaling, as well as nutrient and metabolite levels that are influenced by the tissue environment. Most cells maintain some metabolic flexibility and employ additional pathways in response to stress. Oncogenic mutations reshape metabolism to support tumor growth, but their effects are limited by network constraints imposed by tissue-of-origin and by available nutrients.

Close modal

Tissue identity with regards to metabolism, then, can be defined by the core set of expressed metabolic enzymes that form pathways cells rely on to support their function. When challenged with changes in the environment such as nutrient limitation or hypoxia, different tissues can engage adaptive metabolic programs, such as lipid and protein scavenging (24, 25, 35) to compensate, and these responses also contribute to the metabolic identity of normal and disease tissues. Metabolic flexibility, however, is not infinite, and extreme or prolonged changes in nutrient availability or intrinsic limitations of the cell to adapt to stress will constrain metabolism in all cells. In addition, the limits of adaptability are unlikely to be the same across all cells and tissues.

Mutations in oncogenes and tumor suppressors that lead to tumor initiation and progression will act on the metabolic network within the cell, and thus the effects of these mutations on metabolism are subject to a set of constraints that are defined at least in part by tissue lineage and environment. These growth-promoting mutations may alter metabolic pathway activity, such as increased glycolysis or nutrient scavenging (24, 25), but do not drive convergence of tumor metabolism to a common proliferative program as tumors more closely resemble their parental lineage (27). Only those pathways that remain available based on the given tissue-of-origin can contribute to the tumor metabolic network and use available nutrients to generate a tumor.

Although selection based on genetics remains the current paradigm to develop new cancer treatments, other factors may be better for identifying patients for drugs targeting metabolism. In fact, tumor genetics has not been a successful way to find patients likely to respond to chemotherapies that target nucleotide metabolism, and giving these drugs based on tissue-of-origin has been the standard practice for decades (36). Some newer agents targeting metabolism also act on cancers in a way that is agnostic to cancer genotypes (10). Consistent with this idea, the metabolic constraints imposed by tumor lineage and environment, in addition to mutation status (28, 30), should be considered when examining potential pathways for therapeutic targeting. Furthermore, identifying those metabolic pathways that are most constrained and/or less flexible within specific tumors is critical to achieve both efficacy and a therapeutic window in patients.

The overall set of intrinsic metabolic constraints imposed by the tissue-of-origin and altered growth signaling, together with the extrinsic metabolic constraints imposed by the environment, may be more apparent in vivo. This could be because the types and quantities of nutrients available to cancer cells in tissues are more restricted than they are in culture. Thus, developing cell culture systems that better reflect the nutrient limitations of tumors (37, 38) or studies of tumors in vivo (39) may be a more reliable way to identify cancer metabolic targets. Furthermore, whole body metabolic investigations may reveal potential targets outside the tumor itself when cooperation between nontumor tissues are involved.

Cancer genetics, tumor microenvironment, and tissue-of-origin can all contribute to tumor metabolic dependencies. Both the intrinsic, genetic nature and the extrinsic, nurturing environment define normal tissues and the tumors that arise from them. Understanding the precise contributions of each factor across different cancers holds the best hope for matching the right patients with drugs targeting metabolism and to extend this avenue of precision-medicine to a broad array of cancers.

M.G. Vander Heiden has ownership interest (including patents) in Agios Pharmaceuticals and is a consultant/advisory board member for Agios Pharmaceuticals and Aeglea Biotherapeutics. No potential conflicts of interest were disclosed by the other authors.

We thank members of the Vander Heiden Lab and in particular A.M. Hosios for edits and critical discussions on this article.

J.R. Mayers was supported by grant F30CA183474 from the NCI and grant T32 GM007753. M.G. Vander Heiden was supported by HHMI (Faculty Scholars Program), the Lustgarten Foundation, a Stand Up To Cancer Innovative Research Grant (Grant Number SU2C-AACR-IRG-09-16. Stand Up To Cancer is a program of the Entertainment Industry Foundation. Research grants are administered by the American Association for Cancer Research, the scientific partner of SU2C), the Ludwig Center at MIT, and the NIH (P30CA1405141, R01CA168653, and R01CA201276).

1.
DeVita
VT
 Jr.
,
Chu
E.
A history of cancer chemotherapy
.
Cancer Res
2008
;
68
:
8643
53
.
2.
Farber
S
,
Diamond
LK
. 
Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid
.
N Engl J Med
1948
;
238
:
787
93
.
3.
Hanahan
D
,
Weinberg
RA
. 
The hallmarks of cancer
.
Cell
2000
;
100
:
57
70
.
4.
Notta
F
,
Chan-Seng-Yue
M
,
Lemire
M
,
Li
Y
,
Wilson
GW
,
Connor
AA
, et al
A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns
.
Nature
2016
;
538
:
378
82
.
5.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: The next generation
.
Cell
2011
;
144
:
646
74
.
6.
Garraway
LA.
Genomics-driven oncology: Framework for an emerging paradigm
.
J Clin Oncol
2013
;
31
:
1806
14
.
7.
Roychowdhury
S
,
Chinnaiyan
AM
. 
Translating genomics for precision cancer medicine
.
Annu Rev Genomics Hum Genet
2014
;
15
:
395
415
.
8.
Guo
XE
,
Ngo
B
,
Modrek
AS
,
Lee
WH
. 
Targeting tumor suppressor networks for cancer therapeutics
.
Curr Drug Targets
2014
;
15
:
2
16
.
9.
Grossman
RL
,
Heath
AP
,
Ferretti
V
,
Varmus
HE
,
Lowy
DR
,
Kibbe
WA
, et al
Toward a shared vision for cancer genomic data
.
N Engl J Med
2016
;
375
:
1109
12
.
10.
Martinez-Outschoorn
UE
,
Peiris-Pages
M
,
Pestell
RG
,
Sotgia
F
,
Lisanti
MP
. 
Cancer metabolism: A therapeutic perspective
.
Nat Rev Clin Oncol
2016
;
14
:
11
31
.
11.
Warburg
O
,
Posener
K
. 
Ueber den Stoffwechsel der Carcinomzelle
.
Die Naturwissenschaften
1924
;
12
:
1131
7
.
12.
Flier
JS
,
Mueckler
MM
,
Usher
P
,
Lodish
HF
. 
Elevated levels of glucose transport and transporter messenger RNA are induced by ras or src oncogenes
.
Science
1987
;
235
:
1492
5
.
13.
White
E.
Exploiting the bad eating habits of Ras-driven cancers
.
Genes Develop
2013
;
27
:
2065
71
14.
Dang
CV
. 
MYC on the path to cancer
.
Cell
2012
;
149
:
22
35
.
15.
Maddocks
O
,
Vousden
KH
. 
Metabolic regulation by p53
.
J Mol Med
2011
;
89
:
237
45
.
16.
Vasan
N
,
Boyer
JL
,
Herbst
RS
. 
A RAS renaissance: emerging targeted therapies for KRAS-mutated non-small cell lung cancer
.
Clin Cancer Res
2014
;
20
:
3921
30
.
17.
Patricelli
MP
,
Janes
MR
,
Li
LS
,
Hansen
R
,
Peters
U
,
Kessler
LV
, et al
Selective inhibition of oncogenic KRAS Output with small molecules targeting the inactive state
.
Cancer Discov
2016
;
6
:
316
29
.
18.
Muller
PA
,
Vousden
KH
. 
Mutant p53 in cancer: New functions and therapeutic opportunities
.
Cancer Cell
2014
;
25
:
304
17
.
19.
Gross
MI
,
Demo
SD
,
Dennison
JB
,
Chen
L
,
Chernov-Rogan
T
,
Goyal
B
, et al
Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer
.
Mol Cancer Ther
2014
;
13
:
890
901
.
20.
Son
J
,
Lyssiotis
CA
,
Ying
H
,
Wang
X
,
Hua
S
,
Ligorio
M
, et al
Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway
.
Nature
2013
;
496
:
101
5
.
21.
Davidson
SM
,
Papagiannakopoulos
T
,
Olenchock
BA
,
Heyman
JE
,
Keibler
MA
,
Luengo
A
, et al
Environment impacts the metabolic dependencies of ras-driven non-small cell lung cancer
.
Cell Metab
2016
;
23
:
517
28
.
22.
Mayers
JR
,
Vander Heiden
MG
. 
Famine versus feast: Understanding the metabolism of tumors in vivo
.
Trends Biochem Sci
2015
;
40
:
130
40
.
23.
Hensley
CT
,
Faubert
B
,
Yuan
Q
,
Lev-Cohain
N
,
Jin
E
,
Kim
J
, et al
Metabolic heterogeneity in human lung tumors
.
Cell
2016
;
164
:
681
94
.
24.
Kamphorst
JJ
,
Cross
JR
,
Fan
J
,
de Stanchina
E
,
Mathew
R
,
White
EP
, et al
Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids
.
Proc Natl Acad Sci USA
2013
;
110
:
8882
7
.
25.
Commisso
C
,
Davidson
SM
,
Soydaner-Azeloglu
RG
,
Parker
SJ
,
Kamphorst
JJ
,
Hackett
S
, et al
Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells
.
Nature
2013
;
497
:
633
7
.
26.
Palm
W
,
Park
Y
,
Wright
K
,
Pavlova
NN
,
Tuveson
DA
,
Thompson
CB
. 
The utilization of extracellular proteins as nutrients is suppressed by mTORC1
.
Cell
2015
;
162
:
259
70
.
27.
Hu
J
,
Locasale
JW
,
Bielas
JH
,
O'Sullivan
J
,
Sheahan
K
,
Cantley
LC
, et al
Heterogeneity of tumor-induced gene expression changes in the human metabolic network
.
Nat Biotechnol
2013
;
31
:
522
9
.
28.
Yuneva
MO
,
Fan
TWM
,
Allen
TD
,
Higashi
RM
,
Ferraris
DV
,
Tsukamoto
T
, et al
The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type
.
Cell Metab
2012
;
15
:
157
70
.
29.
Mayers
JR
,
Wu
C
,
Clish
CB
,
Kraft
P
,
Torrence
ME
,
Fiske
BP
, et al
Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development
.
Nat Med
2014
;
20
:
1193
8
.
30.
Mayers
JR
,
Torrence
ME
,
Danai
LV
,
Papagiannakopoulos
T
,
Davidson
SM
,
Bauer
MR
, et al
Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers
.
Science
2016
;
353
:
1161
5
.
31.
Davidson
SM
,
Jonas
O
,
Keibler
MA
,
Hou
HW
,
Luengo
A
,
Mayers
JR
, et al
Direct evidence for cancer-cell-autonomous extracellular protein catabolism in pancreatic tumors
.
Nat Med
2017
;
23
:
235
41
.
32.
Sousa
CM
,
Biancur
DE
,
Wang
X
,
Halbrook
CJ
,
Sherman
MH
,
Zhang
L
, et al
Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion
.
Nature
2016
;
536
:
479
83
.
33.
Zhao
H
,
Yang
L
,
Baddour
J
,
Achreja
A
,
Bernard
V
,
Moss
T
, et al
Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism
.
Elife
2016
;
5
:
e10250
.
34.
Puchowicz
MA
,
Bederman
IR
,
Comte
B
,
Yang
D
,
David
F
,
Stone
E
, et al
Zonation of acetate labeling across the liver: Implications for studies of lipogenesis by MIDA
.
Am J Physiol
1999
;
277
:
E1022
7
.
35.
Kamphorst
JJ
,
Nofal
M
,
Commisso
C
,
Hackett
SR
,
Lu
W
,
Grabocka
E
, et al
Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein
.
Cancer Res
2015
;
75
:
544
53
.
36.
Wilson
PM
,
Danenberg
PV
,
Johnston
PG
,
Lenz
HJ
,
Ladner
RD
. 
Standing the test of time: Targeting thymidylate biosynthesis in cancer therapy
.
Nat Rev Clin Oncol
2014
;
11
:
282
98
.
37.
Birsoy
K
,
Possemato
R
,
Lorbeer
FK
,
Bayraktar
EC
,
Thiru
P
,
Yucel
B
, et al
Metabolic determinants of cancer cell sensitivity to glucose limitation and biguanides
.
Nature
2014
;
508
:
108
12
.
38.
Tardito
S
,
Oudin
A
,
Ahmed
SU
,
Fack
F
,
Keunen
O
,
Zheng
L
, et al
Glutamine synthetase activity fuels nucleotide biosynthesis and supports growth of glutamine-restricted glioblastoma
.
Nat Cell Biol
2015
;
17
:
1556
68
.
39.
Possemato
R
,
Marks
KM
,
Shaul
YD
,
Pacold
ME
,
Kim
D
,
Birsoy
K
, et al
Functional genomics reveal that the serine synthesis pathway is essential in breast cancer
.
Nature
2011
;
476
:
346
50
.