Purpose: The aim of this study was to investigate the expression patterns of glucose metabolism-related proteins and their clinicopathologic implications in adrenal cortical neoplasms (ACN) and pheochromocytoma (PCC). Methods: Immunohistochemical staining was performed to evaluate glucose metabolism-related proteins (GLUT1, CAIX, hexokinase II, G6PDH, PHGDH, and SHMT1) in 132 ACN cases (115 adrenal cortical adenoma [ACA] and 17 adrenal cortical carcinoma [ACC]) and 189 PCC cases. Results: Expression levels of GLUT1 in tumor cells ([T]; p < 0.001), GLUT1 in stromal cells ([S]; p < 0.001), G6PDH (p < 0.001), and SHMT1 (p = 0.002) were higher in ACN than in PCC. GLUT1 (T; p = 0.045) and PHGDH (p = 0.043) levels were higher in ACC than in ACA. In a univariate analysis of ACN, GLUT1 (T; p = 0.017), CAIX (S; p = 0.003), and PHGDH (p = 0.009) levels were correlated with a shorter overall survival (OS). GLUT1 (T; p = 0.001) and PHGDH (p < 0.001) were related to a shorter OS in PCC. GLUT1 (T) positivity (p = 0.043) in ACN predicted a poor OS in a multivariate Cox analysis. In PCC, high GAPP score (p = 0.026), GLUT1 (T; p = 0.002), and PHGDH (p < 0.001) were independent prognostic factors for poor OS. Conclusions: The adrenal gland tumors ACN and PCC had different expression patterns of glucose metabolism-related proteins (GLUT1, G6PDH, and SHMT1), with higher expression levels in ACN than in PCC. GLUT1 and PHGDH were significant prognostic factors in these adrenal neoplasms.

Adrenal gland tumors include adrenal cortical neoplasms (ACN) originating from the adrenal cortex and pheochromocytomas (PCC) originating from the adrenal medulla. ACN consist of adrenal cortical adenoma (ACA) and adrenal cortical carcinoma (ACC). ACA is often challenging to histologically differentiate from ACC [1], a rare and highly aggressive tumor for which effective targeted therapies are lacking [2]. Currently, all PCC cases are thought to have metastatic potential; previous categories of benign and malignant PCC have been eliminated [3]. Although the cells of origin of ACN and PCC are different, ACN and PCC may show histologically similar appearance; therefore, differential diagnosis is often required. In addition, since it is difficult to predict the biologic behavior of the tumors based on histological findings alone, combinations of various clinical, biochemical, and/or histological features are needed to predict the biologic behavior of adrenal neoplasms.

Tumor cells have different metabolic features from those of normal cells. Unlike normal cells, which produce energy by mitochondrial oxidative phosphorylation, cancer cells acquire energy by aerobic glycolysis, called the Warburg effect [4, 5]. This accelerated glycolysis is characterized by high rates of glucose consumption and lactate secretion in tumor tissues and is stimulated by various oncogenes. Deregulated cell metabolism is a crucial consequence of oncogenic mutations and a hallmark of cancer [6]. Glucose transporter 1 (GLUT1), hexokinase II, and carbonic anhydrase IX (CAIX) play important roles in this glycolytic process. GLUT1 is a glucose transporter, acting as a channel for glucose to enter the cell [7]. Hexokinase II is an enzyme that phosphorylates glucose in the cell to produce glucose-6-phosphate [8]. CAIX reversibly hydrates carbon dioxide and neutralizes acidification by lactate formed during glycolysis, allowing glycolysis to continue [9, 10]. Two major glucose catabolic pathways are associated with glycolysis. The pentose phosphate pathway links the synthesis of the nucleotide precursor ribose and nicotinamide adenine dinucleotide phosphate. In the pentose phosphate pathway, the oxidative branch produces nicotinamide adenine dinucleotide phosphate and ribonucleotides, and glucose-6-phosphate dehydrogenase (G6PDH) acts as an important enzyme [11]. The other is the glycine and serine metabolic pathway, which is a glycolytic intermediate metabolic pathway [12-15]. In the serine biosynthesis pathway, 3-phosphoglycerate generated during glycolysis is oxidized to 3-phosphohydroxypyruvate by phosphoglycerate dehydrogenase (PHGDH). Serine hydroxymethyltransferase (SHMT) causes the reversible conversion of serine and glycine.

We have previously identified differences in metabolic phenotypes among thyroid tumor subtypes [16-18]. Furthermore, several studies have shown that metabolism-related proteins may be predictors of cancer prognosis [19-21]. We hypothesized that adrenal gland tumors (ACA, ACC, and PCC) have different metabolic characteristics and that these properties could serve as prognostic factors in ACC and PCC, for which it is currently difficult to predict prognosis. In this study, we investigated the expression levels of glucose metabolism-related proteins in adrenal gland tumors and their clinicopathologic implications.

Patient Selection

Formalin-fixed paraffin-embedded tissue samples from patients diagnosed with ACN and PCC in surgically resected adrenal glands at Severance Hospital from January 2000 to December 2012 were used. The study cohort consisted of 132 ACN (115 adrenal cortical adenoma [ACA] and 17 adrenal cortical carcinoma [ACC]) and 189 PCC cases. This study was approved by the Institutional Review Board of Severance Hospital. All cases were retrospectively reviewed by pathologists (K.J.S. and K.H.M.). For ACN, parameters corresponding to Weiss criteria were examined [22]. For PCC, parameters corresponding to the grading system for adrenal pheochromocytoma and paraganglioma (GAPP) were investigated [23]. Disease-free survival was calculated from the date of the first curative surgery to the date of the first locoregional or systemic relapse or death without any type of relapse. Overall survival (OS) was estimated from the date of the first curative operation to the date of the last follow-up or death from any cause. Clinical information was obtained from electronic medical records.

Tissue Microarray Analysis

Representative areas were selected on hematoxylin-eosin-stained slides. Three-millimeter core biopsies were taken from selected areas and placed in a 6 × 5 recipient block. More than 2 tissue cores were extracted from each case to minimize extraction bias.

Immunohistochemistry and Interpretation

Antibodies used for immunohistochemistry are listed in online suppl. Table 1 (see www.karger.com/doi/10.1159/000518208 for all online suppl. material). All immunohistochemistry was performed using an automatic immunohistochemistry staining device (Benchmark XT; Ventana Medical System, Tucson, AZ, USA). In brief, 5-µm-thick formalin-fixed paraffin-embedded tissue sections were transferred onto adhesive slides and dried at 62°C for 30 min. Standard heat epitope retrieval was performed for 30 min in ethylene diamine tetra-acetic acid, pH 8.0, using an autostainer.

Immunohistochemical markers were semiquantitatively evaluated according to previously reported methods [24]. Tumor cell and stromal cell staining was assessed as follows: 0, negative or weak immunostaining in <1% of the tumor/stroma; 1, focal expression in 1–10% of the tumor/stroma; 2, positive staining in 11–50% of the tumor/stroma; and 3, positive staining in 51–100% of the tumor/stroma. Scores of 0 and 1 were defined as negative, while 2 and 3 were defined as positive.

Statistical Analysis

Data were analyzed using SPSS for Windows, Version 22.0 (SPSS Inc., IBM Corporation, Chicago, IL, USA). For the determination of statistical significance, Student’s t tests and Fisher’s exact tests were used for continuous and categorical variables, respectively. For multiple comparisons, a corrected p value with the Bonferroni multiple comparison procedure was used. The threshold for statistical significance was set to p < 0.05. Kaplan-Meier survival curves and log-rank statistics were employed to evaluate the time to tumor recurrence and OS. A multivariate regression analysis was performed using the Cox proportional hazards model.

Basal Characteristics of Patients

ACC was characterized by a significantly larger tumor size (p < 0.001) and younger age (41.0 ± 25.1 years; p = 0.048) than those of ACA. Various factors, including Weiss criteria, also showed significant differences (online suppl. Table 2). The basal characteristics of PCC are presented in online suppl. Table 3.

Expression of Glucose Metabolism-Related Proteins in Adrenal Neoplasms

GLUT1 in tumor cells ([T]; p < 0.001), GLUT1 in stromal cells ([S]; p < 0.001), G6PDH (p < 0.001), and SHMT1 (p = 0.002) showed significant differences between ACN and PCC. ACN showed higher overall expression levels than those in PCC (Table 1; Fig. 1). In ACN, GLUT1 (T; p = 0.045) and PHGDH (p = 0.043) levels were significantly different between ACC and ACA. Expression was generally higher in ACC than in ACA (Table 2; Fig. 1).

Table 1.

Expression of glycolysis-related proteins in adrenal neoplasm

Expression of glycolysis-related proteins in adrenal neoplasm
Expression of glycolysis-related proteins in adrenal neoplasm
Table 2.

Expression of glycolysis-related proteins in adrenal cortical neoplasms

Expression of glycolysis-related proteins in adrenal cortical neoplasms
Expression of glycolysis-related proteins in adrenal cortical neoplasms
Fig. 1.

Expression of glucose metabolism-related proteins in adrenal neoplasms. GLUT1 in tumor cells and stromal cells, G6PDH, and SHMT1 show higher expression in adrenal cortical neoplasms than in PCC. ACC demonstrated higher levels of GLUT1 and PHGDH than those in ACA. GLUT1, glucose transporter 1; G6PDH, glucose-6-phosphate dehydrogenase; SHMT1, serine hydroxymethyltransferase 1; PHGDH, phosphoglycerate dehydrogenase; PCC, pheochromocytoma; ACC, adrenal cortical carcinoma; ACA, adrenal cortical adenoma.

Fig. 1.

Expression of glucose metabolism-related proteins in adrenal neoplasms. GLUT1 in tumor cells and stromal cells, G6PDH, and SHMT1 show higher expression in adrenal cortical neoplasms than in PCC. ACC demonstrated higher levels of GLUT1 and PHGDH than those in ACA. GLUT1, glucose transporter 1; G6PDH, glucose-6-phosphate dehydrogenase; SHMT1, serine hydroxymethyltransferase 1; PHGDH, phosphoglycerate dehydrogenase; PCC, pheochromocytoma; ACC, adrenal cortical carcinoma; ACA, adrenal cortical adenoma.

Close modal

Correlations between Glucose Metabolism-Related Protein Expression Levels and Pathologic Parameters in Adrenal Neoplasms

In ACN, the expression of GLUT1 (T) was associated with increased mitosis (p = 0.007), and CAIX (S) expression was associated with venous invasion (p < 0.001; Fig. 2a, b). PHGDH expression was also related to increased mitosis (p = 0.001), a decreased clear cell proportion (p < 0.001), and venous invasion (p = 0.001; Fig. 2c–e). In PCC, CAIX (T) expression was associated with norepinephrine type (p = 0.002), and CAIX (S) expression was related to the nonzellballen pattern (p = 0.002; Fig. 2f, g).

Fig. 2.

Correlation between the expression levels of glucose metabolism-related proteins and pathologic parameters in adrenal neoplasms. GLUT1 (T) expression is associated with increased mitosis (a), and CAIX (S) expression is associated with venous invasion (b) in adrenal cortical neoplasms. PHGDH expression is also related to increased mitosis (c), decreased clear cell proportion (d), and venous invasion (e). CAIX (T) expression is associated with norepinephrine type (f), and CAIX (S) expression is related to the nonzellballen pattern (g) in pheochromocytoma. GLUT1, glucose transporter 1; CAIX, carbonic anhydrase IX; PHGDH, phosphoglycerate dehydrogenase.

Fig. 2.

Correlation between the expression levels of glucose metabolism-related proteins and pathologic parameters in adrenal neoplasms. GLUT1 (T) expression is associated with increased mitosis (a), and CAIX (S) expression is associated with venous invasion (b) in adrenal cortical neoplasms. PHGDH expression is also related to increased mitosis (c), decreased clear cell proportion (d), and venous invasion (e). CAIX (T) expression is associated with norepinephrine type (f), and CAIX (S) expression is related to the nonzellballen pattern (g) in pheochromocytoma. GLUT1, glucose transporter 1; CAIX, carbonic anhydrase IX; PHGDH, phosphoglycerate dehydrogenase.

Close modal

Impact of Glucose Metabolism-Related Proteins on Prognosis in Adrenal Neoplasms

A univariate analysis showed that GLUT1 (T; p = 0.017), CAIX (p = 0.003), and PHGDH (p = 0.009) in ACN were related to a shorter OS (online suppl. Table 4; Fig. 3a–c). In PCC, GLUT1 (T; p = 0.001) and PHGDH (p < 0.001) were associated with a shorter OS (online -suppl. Table 5; Fig. 3d, e).

Fig. 3.

Impact of the expression levels of glucose metabolism-related proteins on prognosis in adrenal neoplasms. Expression of GLUT1 (a), CAIX (b), and PHGDH (c) in adrenal cortical neoplasms was associated with shorter OS; positivity for GLUT1 (d) and PHGDH (e) was associated with shorter OS in pheochromocytoma. GLUT1, glucose transporter 1; CAIX, carbonic anhydrase IX; PHGDH, phosphoglycerate dehydrogenase.

Fig. 3.

Impact of the expression levels of glucose metabolism-related proteins on prognosis in adrenal neoplasms. Expression of GLUT1 (a), CAIX (b), and PHGDH (c) in adrenal cortical neoplasms was associated with shorter OS; positivity for GLUT1 (d) and PHGDH (e) was associated with shorter OS in pheochromocytoma. GLUT1, glucose transporter 1; CAIX, carbonic anhydrase IX; PHGDH, phosphoglycerate dehydrogenase.

Close modal

In a multivariate Cox analysis of OS, GLUT1 positivity was an independent predictor of poor prognosis in ACN (HR: 404.8, 95% CI: 1.204–136,128, p = 0.043; Table 3). In PCC, a high GAPP score (>3, HR: 35.190, 95% CI: 1.528–810.4, p = 0.026), GLUT1 positivity (HR: 10.875, 95% CI: 2.330–50.76, p = 0.002), and PHGDH positivity (HR: 52.229, 95% CI: 7.672–355.5, p < 0.001) were independent factors associated with shorter OS (Table 4).

Table 3.

Multivariate analysis of disease-free survival and overall-survival of patients with adrenal cortical neoplasms

Multivariate analysis of disease-free survival and overall-survival of patients with adrenal cortical neoplasms
Multivariate analysis of disease-free survival and overall-survival of patients with adrenal cortical neoplasms
Table 4.

Multivariate analysis of disease-free survival and overall-survival of patients with pheochromocytoma

Multivariate analysis of disease-free survival and overall-survival of patients with pheochromocytoma
Multivariate analysis of disease-free survival and overall-survival of patients with pheochromocytoma

We examined the expression of glucose metabolism-related proteins in adrenal gland neoplasms. Previous studies have reported that glycolysis-related proteins are expressed in PCC [25] and ACN [26], but direct comparisons are difficult owing to the lack of investigations of expression differences between these tumors. We initially found that the expression levels of glucose metabolism-related proteins (GLUT1, G6PDH, and SHMT1) were higher in ACN than in PCC, suggesting that the glycolytic process is generally more active in ACN. This result can be explained by the genetic background of PCC. At least 30% of PCC and paraganglioma (PGL) cases are now known to be hereditary, and mutations in genes encoding succinate dehydrogenase enzymes (SDH gene family) have been identified as the most common cause (14–20%), followed by mutations in VHL (9%) and RET (5%) [3]. In loss-of-function SDH mutants, mitochondrial function is impaired, and metabolic reprogramming occurs from mitochondrial oxidative phosphorylation to glycolysis [27], which is also demonstrated by high radiolabeled deoxy-2-[18F]fluoro-D-glucose (18F-FDG) uptake in SDH-mutated PCC in positron emission tomography (PET) studies [25, 28]. In a recent comprehensive genomic study, PCC/PGL was classified into 4 molecular subtypes, including a pseudohypoxia subtype. Many tumors of this subtype have germline mutations in Krebs cycle genes, SDH and IDH, and had disruptions in the hypoxia signaling pathway [29]. Sporadic tumors, accounting for the remaining 70% of PCC/PGL, mainly show alterations, such as NF1, HRAS, and ATRX mutations, corresponding to kinase signaling and Wnt-altered subtypes. In addition, the mechanism underlying increased FDG uptake in SDH-mutated PCC is explained by accelerated glucose phosphorylation, rather than by increased glucose transporter expression [25].

GLUT1 (T) and PHGDH levels were higher in ACC than in ACA, which can be explained by an increase in glycolysis by glucose uptake, one of the metabolic characteristics of malignant tumors [4, 5]. The levels of GLUT1, CAIX, and MCT4, key metabolism-related proteins, have been reported to be higher in ACC than in ACA [26]. FDG-PET studies have also shown higher FDG uptake in ACC than in ACA, suggesting that glucose metabolism is increased in ACC [30, 31]. CAIX and GLUT1 are part of the hypoxia inducible factor-1 (HIF-1) pathway and have been suggested as endogenous markers for hypoxia in solid tumors [32]. HIF accumulates in response to decreased cellular oxygen levels and regulates the adaptation of tumor cells to hypoxic stress. HIF-1α raises the activity of several glycolytic protein isoforms, including GLUT-1, CAIX, GAPDH, PGK1, and others, and directly upregulates the genes coding for glucose transporters such as GLUT-1 and the enzymes of the glycolytic pathway [33, 34].

We examined the expression levels of glucose metabolism-related proteins in stromal cells as well as in tumor cells. GLUT1 was more highly expressed in stromal cells in ACN compared to PCC. Metabolic interactions between cancer cells and cells in the tumor microenvironment allow metabolites to migrate from stromal cells to meet metabolic demands and maintain ATP production in cancer cells. The expression of these proteins in stromal cells might be explained by the reverse Warburg effect, which describes a 2-compartment model in which stromal cells are induced by cancer cells to undergo aerobic glycolysis and then transfer the products back to the cancer cells for utilization for mitochondrial oxidative phosphorylation [35]. Metabolism occurs by oxidative phosphorylation in functional mitochondria in tumor cells, whereas metabolism in stromal cells by glycolysis results in dysfunctional mitochondria resulting from increased autophagy activity in breast cancers [36, 37]. Similar metabolic coupling between stromal cells and cancer cells has been observed in various cancers, including ovarian, prostate, liver, colon, and pancreatic cancers [38-42]. Therefore, further research is needed to determine whether the reverse Warburg effect is one of the metabolic features in adrenal cortical neoplasms.

The expression levels of GLUT1 (T), CAIX (S), and PHGDH in ACN, as well as GLUT1 (T) and PHGDH in PCC, were associated with poor prognosis in univariate analysis. In multivariate analysis, GLUT1 (T) in ACN and high GAPP score, GLUT1 (T), and PHGDH in PCC were independent prognostic factors for OS. GLUT1 is a hyp-oxia-responsive glucose transporter, and its overexpression has been identified in various cancers, including ACC, stomach, urinary bladder, ovary, oral cavity, esophagus, pancreas, colorectum, lung, and gallbladder cancer, resulting in increased glucose uptake into the cytoplasm of tumor cells under hypoxic conditions; it is considered a poor prognostic factor [26, 43, 44]. PHGDH is overexpressed in a substantial fraction of human cancers, mainly as a result of genomic amplification of the PHGDH gene on chromosome 1p12; it catalyzes a growth-promoting metabolic pathway. PHGDH is associated with poor prognosis in pancreatic cancer [45], lung cancer [46, 47], and gastric cancer [48]. Previous studies have suggested that PHGDH is a potential therapeutic target in tumors in which metabolism is twisted toward de novo serine biosynthesis [49]. Furthermore, inhibiting GLUT1 [50-52], CAIX [53, 54], G6PDH [55], PHGDH [56-59], and SHMT [60, 61] limits tumor growth. Therefore, glucose metabolism-related proteins, such as GLUT1 and PHGDH, may be prognostic markers and are expected to be effective treatment targets in adrenal gland tumors in the near future.

A limitation of this study is that assessing the expression of enzymes or proteins via immunohistochemistry may not necessarily equate to their activity. Additional factors that may affect the enzymatic activity include posttranslational modification (e.g., addition of functional groups or proteins/peptides, chemical conversion, and structural changes), various inhibitors (e.g., competitive, uncompetitive, noncompetitive, suicide, and mixed), activators (e.g., allosteric, co-factors like vitamins, and nonvitamins/minerals), and environmental factors (e.g., changing pH). Therefore, it is necessary to conduct more in-depth research while considering these various factors as much as possible.

In summary, the expression levels of GLUT1, G6PDH, and SHMT1, which are glucose metabolism-related proteins, differ between ACN and PCC and are higher in ACN. In addition, GLUT1 and PHGDH are associated with poor prognosis in adrenal gland tumors.

This study was approved by the Institutional Review Board of Severance Hospital (4-2019-0902). Informed consent from patients was exempted by IRB. This study was exempt from obtaining written informed consent due to the retrospective nature of the study.

The authors declare no conflicts of interest.

No external funding source was used in this study.

E.K.K. participated in the design of the study and performed the statistical analysis and carried out the immunoassays. H.M.K. conducted the data collection and analysis. J.S.K. conceived the study and participated in its design and coordination and helped to draft the manuscript. All authors read and approved the final manuscript.

All data generated or analyzed during this study are included in this article and its online supplementary material. Further enquiries can be directed to the corresponding author.

1.
Erickson
LA
.
Challenges in surgical pathology of adrenocortical tumours
.
Histopathology
.
2018 Jan
;
72
(
1
):
82
96
. .
2.
Varghese
J
,
Habra
MA
.
Update on adrenocortical carcinoma management and future directions
.
Curr Opin Endocrinol Diabetes Obes
.
2017 Jun
;
24
(
3
):
208
14
. .
3.
Lloyd
RV
,
Osamura
RY
,
Klöppel
G
,
Rosai
J
, editors.
WHO classification of tumours of endocrine organs
. 4th ed.
Lyon
:
International Agency for Research on Cancer (IARC)
;
2017
.
4.
Warburg
O
.
On the origin of cancer cells
.
Science
.
1956 Feb 24
;
123
(
3191
):
309
14
. .
5.
Kim
JW
,
Dang
CV
.
Cancer’s molecular sweet tooth and the Warburg effect
.
Cancer Res
.
2006 Sep 15
;
66
(
18
):
8927
30
. .
6.
Hanahan
D
,
Weinberg
RA
.
Hallmarks of cancer: the next generation
.
Cell
.
2011 Mar 4
;
144
(
5
):
646
74
. .
7.
Olson
AL
,
Pessin
JE
.
Structure, function, and regulation of the mammalian facilitative glucose transporter gene family
.
Annu Rev Nutr
.
1996
;
16
:
235
56
. .
8.
Lehto
M
,
Xiang
K
,
Stoffel
M
,
Espinosa
R
 3rd
,
Groop
LC
,
Le Beau
MM
,
Human hexokinase II: localization of the polymorphic gene to chromosome 2
.
Diabetologia
.
1993 Dec
;
36
(
12
):
1299
302
. .
9.
Opavsky
R
,
Pastorekova
S
,
Zelnik
V
,
Gibadulinova
A
,
Stanbridge
EJ
,
Zavada
J
,
Human MN/CA9 gene, a novel member of the carbonic anhydrase family: structure and exon to protein domain relationships
.
Genomics
.
1996 May 1
;
33
(
3
):
480
7
.
10.
Becker
HM
.
Carbonic anhydrase IX and acid transport in cancer
.
Br J Cancer
.
2020 Jan
;
122
(
2
):
157
67
. .
11.
Kletzien
RF
,
Harris
PK
,
Foellmi
LA
.
Glucose-6-phosphate dehydrogenase: a “housekeeping” enzyme subject to tissue-specific regulation by hormones, nutrients, and oxidant stress
.
Faseb J
.
1994 Feb
;
8
(
2
):
174
81
. .
12.
Mullarky
E
,
Mattaini
KR
,
Vander Heiden
MG
,
Cantley
LC
,
Locasale
JW
.
PHGDH amplification and altered glucose metabolism in human melanoma
.
Pigment Cell Melanoma Res
.
2011 Dec
;
24
(
6
):
1112
5
. .
13.
Possemato
R
,
Marks
KM
,
Shaul
YD
,
Pacold
ME
,
Kim
D
,
Birsoy
K
,
Functional genomics reveal that the serine synthesis pathway is essential in breast cancer
.
Nature
.
2011 Aug 18
;
476
(
7360
):
346
50
. .
14.
Jain
M
,
Nilsson
R
,
Sharma
S
,
Madhusudhan
N
,
Kitami
T
,
Souza
AL
,
Metabolite profiling identifies a key role for glycine in rapid cancer cell proliferation
.
Science
.
2012 May 25
;
336
(
6084
):
1040
4
. .
15.
Zhang
WC
,
Shyh-Chang
N
,
Yang
H
,
Rai
A
,
Umashankar
S
,
Ma
S
,
Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis
.
Cell
.
2012 Jan 20
;
148
(
1–2
):
259
72
. .
16.
Sun
WY
,
Kim
HM
,
Jung
WH
,
Koo
JS
.
Expression of serine/glycine metabolism-related proteins is different according to the thyroid cancer subtype
.
J Transl Med
.
2016 Jun 8
;
14
(
1
):
168
. .
17.
Kim
HM
,
Koo
JS
.
Differential expression of glycolysis-related proteins in follicular neoplasms versus hurthle cell neoplasms: a retrospective analysis
.
Dis Markers
.
2017
;
2017
:
6230294
.
18.
Nahm
JH
,
Kim
HM
,
Koo
JS
.
Glycolysis-related protein expression in thyroid cancer
.
Tumour Biol
.
2017 Mar
;
39
(
3
):
1010428317695922
. .
19.
van Kuijk
SJ
,
Yaromina
A
,
Houben
R
,
Niemans
R
,
Lambin
P
,
Dubois
LJ
.
Prognostic significance of carbonic anhydrase IX expression in cancer patients: a meta-analysis
.
Front Oncol
.
2016
;
6
:
69
. .
20.
Chen
X
,
Lu
P
,
Zhou
S
,
Zhang
L
,
Zhao
JH
,
Tang
JH
.
Predictive value of glucose transporter-1 and glucose transporter-3 for survival of cancer patients: a meta-analysis
.
Oncotarget
.
2017 Feb 21
;
8
(
8
):
13206
13
. .
21.
Gomez-Cebrian
N
,
Rojas-Benedicto
A
,
Albors-Vaquer
A
,
Lopez-Guerrero
JA
,
Pineda-Lucena
A
,
Puchades-Carrasco
L
.
Metabolomics contributions to the discovery of prostate cancer biomarkers
.
Metabolites
.
2019 Mar 8
;
9
(
3
):
48
.
22.
Weiss
LM
.
Comparative histologic study of 43 metastasizing and nonmetastasizing adrenocortical tumors
.
Am J Surg Pathol
.
1984 Mar
;
8
(
3
):
163
9
. .
23.
Kimura
N
,
Takayanagi
R
,
Takizawa
N
,
Itagaki
E
,
Katabami
T
,
Kakoi
N
,
Pathological grading for predicting metastasis in phaeochromocytoma and paraganglioma
.
Endocr Relat Cancer
.
2014 Jun
;
21
(
3
):
405
14
. .
24.
Henry
LR
,
Lee
HO
,
Lee
JS
,
Klein-Szanto
A
,
Watts
P
,
Ross
EA
,
Clinical implications of fibroblast activation protein in patients with colon cancer
.
Clin Cancer Res
.
2007 Mar 15
;
13
(
6
):
1736
41
. .
25.
van Berkel
A
,
Rao
JU
,
Kusters
B
,
Demir
T
,
Visser
E
,
Mensenkamp
AR
,
Correlation between in vivo 18F-FDG PET and immunohistochemical markers of glucose uptake and metabolism in pheochromocytoma and paraganglioma
.
J Nucl Med
.
2014 Aug
;
55
(
8
):
1253
9
. .
26.
Pinheiro
C
,
Granja
S
,
Longatto-Filho
A
,
Faria
AM
,
Fragoso
MC
,
Lovisolo
SM
,
Metabolic reprogramming: a new relevant pathway in adult adrenocortical tumors
.
Oncotarget
.
2015 Dec 29
;
6
(
42
):
44403
21
. .
27.
Kluckova
K
,
Tennant
DA
.
Metabolic implications of hypoxia and pseudohypoxia in pheochromocytoma and paraganglioma
.
Cell Tissue Res
.
2018 May
;
372
(
2
):
367
78
. .
28.
van Berkel
A
,
Vriens
D
,
Visser
E
,
Janssen
M
,
Gotthardt
M
,
Hermus
A
,
Metabolic subtyping of pheochromocytoma and paraganglioma by (18)F-FDG pharmacokinetics using dynamic PET/CT scanning
.
J Nucl Med
.
2019
;
60
(
6
):
745
51
.
29.
Fishbein
L
,
Leshchiner
I
,
Walter
V
,
Danilova
L
,
Robertson
AG
,
Johnson
AR
,
Comprehensive molecular characterization of pheochromocytoma and paraganglioma
.
Cancer Cell
.
2017 Feb 13
;
31
(
2
):
181
93
. .
30.
Groussin
L
,
Bonardel
G
,
Silvéra
S
,
Tissier
F
,
Coste
J
,
Abiven
G
,
18F-Fluorodeoxyglucose positron emission tomography for the diagnosis of adrenocortical tumors: a prospective study in 77 operated patients
.
J Clin Endocrinol Metab
.
2009 May
;
94
(
5
):
1713
22
. .
31.
Nunes
ML
,
Rault
A
,
Teynie
J
,
Valli
N
,
Guyot
M
,
Gaye
D
,
18F-FDG PET for the identification of adrenocortical carcinomas among indeterminate adrenal tumors at computed tomography scanning
.
World J Surg
.
2010 Jul
;
34
(
7
):
1506
10
. .
32.
Hoskin
PJ
,
Sibtain
A
,
Daley
FM
,
Wilson
GD
.
GLUT1 and CAIX as intrinsic markers of hypoxia in bladder cancer: relationship with vascularity and proliferation as predictors of outcome of ARCON
.
Br J Cancer
.
2003 Oct 6
;
89
(
7
):
1290
7
. .
33.
Semenza
GL
.
HIF-1: upstream and downstream of cancer metabolism
.
Curr Opin Genet Dev
.
2010 Feb
;
20
(
1
):
51
6
. .
34.
Al Tameemi
W
,
Dale
TP
,
Al-Jumaily
RMK
,
Forsyth
NR
.
Hypoxia-modified cancer cell metabolism
.
Front Cell Dev Biol
.
2019
;
7
:
4
. .
35.
Wilde
L
,
Roche
M
,
Domingo-Vidal
M
,
Tanson
K
,
Philp
N
,
Curry
J
,
Metabolic coupling and the reverse Warburg effect in cancer: implications for novel biomarker and anticancer agent development
.
Semin Oncol
.
2017 Jun
;
44
(
3
):
198
203
. .
36.
Pavlides
S
,
Whitaker-Menezes
D
,
Castello-Cros
R
,
Flomenberg
N
,
Witkiewicz
AK
,
Frank
PG
,
The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma
.
Cell Cycle
.
2009 Dec
;
8
(
23
):
3984
4001
. .
37.
Witkiewicz
AK
,
Whitaker-Menezes
D
,
Dasgupta
A
,
Philp
NJ
,
Lin
Z
,
Gandara
R
,
Using the “reverse Warburg effect” to identify high-risk breast cancer patients: stromal MCT4 predicts poor clinical outcome in triple-negative breast cancers
.
Cell Cycle
.
2012 Mar 15
;
11
(
6
):
1108
17
. .
38.
Koukourakis
MI
,
Giatromanolaki
A
,
Harris
AL
,
Sivridis
E
.
Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma
.
Cancer Res
.
2006 Jan 15
;
66
(
2
):
632
7
. .
39.
Nieman
KM
,
Kenny
HA
,
Penicka
CV
,
Ladanyi
A
,
Buell-Gutbrod
R
,
Zillhardt
MR
,
Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth
.
Nat Med
.
2011
;
17
(
11
):
1498
503
. .
40.
Fiaschi
T
,
Marini
A
,
Giannoni
E
,
Taddei
ML
,
Gandellini
P
,
De Donatis
A
,
Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay
.
Cancer Res
.
2012 Oct 1
;
72
(
19
):
5130
40
. .
41.
Huang
D
,
Li
T
,
Wang
L
,
Zhang
L
,
Yan
R
,
Li
K
,
Hepatocellular carcinoma redirects to ketolysis for progression under nutrition deprivation stress
.
Cell Res
.
2016 Oct
;
26
(
10
):
1112
30
. .
42.
Sousa
CM
,
Biancur
DE
,
Wang
X
,
Halbrook
CJ
,
Sherman
MH
,
Zhang
L
,
Pancreatic stellate cells support tumour metabolism through autophagic alanine secretion
.
Nature
.
2016 Aug 25
;
536
(
7617
):
479
83
. .
43.
Fenske
W
,
Völker
HU
,
Adam
P
,
Hahner
S
,
Johanssen
S
,
Wortmann
S
,
Glucose transporter GLUT1 expression is an stage-independent predictor of clinical outcome in adrenocortical carcinoma
.
Endocr Relat Cancer
.
2009 Sep
;
16
(
3
):
919
28
. .
44.
Zhao
ZX
,
Lu
LW
,
Qiu
J
,
Li
QP
,
Xu
F
,
Liu
BJ
,
Glucose transporter-1 as an independent prognostic marker for cancer: a meta-analysis
.
Oncotarget
.
2018 Jan 5
;
9
(
2
):
2728
38
. .
45.
Song
Z
,
Feng
C
,
Lu
Y
,
Lin
Y
,
Dong
C
.
PHGDH is an independent prognosis marker and contributes cell proliferation, migration and invasion in human pancreatic cancer
.
Gene
.
2018 Feb 5
;
642
:
43
50
. .
46.
Zhu
J
,
Ma
J
,
Wang
X
,
Ma
T
,
Zhang
S
,
Wang
W
,
High expression of PHGDH predicts poor prognosis in non-small cell lung cancer
.
Transl Oncol
.
2016 Dec
;
9
(
6
):
592
9
. .
47.
Zhang
B
,
Zheng
A
,
Hydbring
P
,
Ambroise
G
,
Ouchida
AT
,
Goiny
M
,
PHGDH defines a metabolic subtype in lung adenocarcinomas with poor prognosis
.
Cell Rep
.
2017 Jun 13
;
19
(
11
):
2289
303
. .
48.
Xian
Y
,
Zhang
S
,
Wang
X
,
Qin
J
,
Wang
W
,
Wu
H
.
Phosphoglycerate dehydrogenase is a novel predictor for poor prognosis in gastric cancer
.
Onco Targets Ther
.
2016
;
9
:
5553
60
. .
49.
Mullen
AR
,
DeBerardinis
RJ
.
Genetically-defined metabolic reprogramming in cancer
.
Trends Endocrinol Metab
.
2012 Nov
;
23
(
11
):
552
9
. .
50.
Melstrom
LG
,
Salabat
MR
,
Ding
XZ
,
Milam
BM
,
Strouch
M
,
Pelling
JC
,
Apigenin inhibits the GLUT-1 glucose transporter and the phosphoinositide 3-kinase/Akt pathway in human pancreatic cancer cells
.
Pancreas
.
2008 Nov
;
37
(
4
):
426
31
. .
51.
Zhou
SH
,
Fan
J
,
Chen
XM
,
Cheng
KJ
,
Wang
SQ
.
Inhibition of cell proliferation and glucose uptake in human laryngeal carcinoma cells by antisense oligonucleotides against glucose transporter-1
.
Head Neck
.
2009 Dec
;
31
(
12
):
1624
33
. .
52.
Bao
YY
,
Zhou
SH
,
Fan
J
,
Wang
QY
.
Anticancer mechanism of apigenin and the implications of GLUT-1 expression in head and neck cancers
.
Future Oncol
.
2013 Sep
;
9
(
9
):
1353
64
. .
53.
Ahlskog
JK
,
Dumelin
CE
,
Trüssel
S
,
Mårlind
J
,
Neri
D
.
In vivo targeting of tumor-associated carbonic anhydrases using acetazolamide derivatives
.
Bioorg Med Chem Lett
.
2009 Aug 15
;
19
(
16
):
4851
6
. .
54.
Lock
FE
,
McDonald
PC
,
Lou
Y
,
Serrano
I
,
Chafe
SC
,
Ostlund
C
,
Targeting carbonic anhydrase IX depletes breast cancer stem cells within the hypoxic niche
.
Oncogene
.
2013 Oct 31
;
32
(
44
):
5210
9
. .
55.
Catanzaro
D
,
Gaude
E
,
Orso
G
,
Giordano
C
,
Guzzo
G
,
Rasola
A
,
Inhibition of glucose-6-phosphate dehydrogenase sensitizes cisplatin-resistant cells to death
.
Oncotarget
.
2015 Oct 6
;
6
(
30
):
30102
14
. .
56.
Jing
Z
,
Heng
W
,
Xia
L
,
Ning
W
,
Yafei
Q
,
Yao
Z
,
Downregulation of phosphoglycerate dehydrogenase inhibits proliferation and enhances cisplatin sensitivity in cervical adenocarcinoma cells by regulating Bcl-2 and caspase-3
.
Cancer Biol Ther
.
2015
;
16
(
4
):
541
8
. .
57.
Mullarky
E
,
Lucki
NC
,
Beheshti Zavareh
R
,
Anglin
JL
,
Gomes
AP
,
Nicolay
BN
,
Identification of a small molecule inhibitor of 3-phosphoglycerate dehydrogenase to target serine biosynthesis in cancers
.
Proc Natl Acad Sci U S A
.
2016 Feb 16
;
113
(
7
):
1778
83
. .
58.
Wang
Q
,
Liberti
MV
,
Liu
P
,
Deng
X
,
Liu
Y
,
Locasale
JW
,
Rational design of selective allosteric inhibitors of PHGDH and serine synthesis with anti-tumor activity
.
Cell Chem Biol
.
2017 Jan 19
;
24
(
1
):
55
65
. .
59.
Unterlass
JE
,
Baslé
A
,
Blackburn
TJ
,
Tucker
J
,
Cano
C
,
Noble
MEM
,
Validating and enabling phosphoglycerate dehydrogenase (PHGDH) as a target for fragment-based drug discovery in PHGDH-amplified breast cancer
.
Oncotarget
.
2018 Mar 2
;
9
(
17
):
13139
53
. .
60.
Paone
A
,
Marani
M
,
Fiascarelli
A
,
Rinaldo
S
,
Giardina
G
,
Contestabile
R
,
SHMT1 knockdown induces apoptosis in lung cancer cells by causing uracil misincorporation
.
Cell Death Dis
.
2014 Nov 20
;
5
:
e1525
. .
61.
Ducker
GS
,
Ghergurovich
JM
,
Mainolfi
N
,
Suri
V
,
Jeong
SK
,
Hsin-Jung Li
S
,
Human SHMT inhibitors reveal defective glycine import as a targetable metabolic vulnerability of diffuse large B-cell lymphoma
.
Proc Natl Acad Sci U S A
.
2017 Oct 24
;
114
(
43
):
11404
9
. .
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.