Abstract
The study of cancer metabolism has evolved vastly beyond the remit of tumour proliferation and survival, with an unveiling of the ostensible role of ‘oncometabolites’ in tumorigenesis. Simply defined, oncometabolites are conventional metabolites that when aberrantly accumulated have pro-oncogenic functions. Their discovery has led us to revisit the original, dispelled Warburg hypothesis, first postulated in the 1950s, of aberrant metabolism as an aetiological determinant of cancer. As such, the identification of oncometabolites alongside their attractive utilisation in diagnostics and prognostics, as novel therapeutic targets and as biomarkers of disease, has been intensely sought after in oncology. To date, fumarate, succinate and 2-hydroxyglutarate have been characterised as bona fide oncometabolites. Renal cell carcinoma (RCC) is an established example of a cancer type with extensive metabolic reprogramming during tumour initiation and progression. With oncometabolites postulated to be rooted in the oncological origins and drivers of tumorigenesis, in combination with all three of these oncometabolites remarkably implicated in RCC, this timely review synthesises the literature to date on oncometabolites in RCC, their oncogenic mechanisms and the clinical impact oncometabolites may have in the management of RCC.
Introduction
Cancers of the kidney account for an estimated 2.2% of the global burden of all cancers, which translates into more than 400,000 new diagnoses worldwide in 20181. Renal cell carcinoma (RCC), a cancer of the kidney parenchyma is the most common solid tumour of the kidney and the most lethal of all urological malignancies2. Almost a third of all patients have metastatic dissemination at presentation and nearly half of all patients die from their disease1,2. RCC is increasingly recognised as a collection of renal cancer subtypes each with distinct histology, genetic and molecular alterations, different clinical courses, and therapeutic responses3–5. Recent single cell sequencing has shed light on the oncogenic events and cell of origin of this tumour type. Interestingly, the convoluted proximal tubular renal cell subtype was shown to be the likely common cell of origin in clear cell RCC (ccRCC) and type 1 papillary RCC (pRCC), suggesting that these tumours may arise from a common origin with divergent fates6. Recent studies have expanded upon the role of genomics in RCC tumour evolution7–9. Loss of chromosome 3p, a pathognomonic feature of ccRCC occurring in >90% of patients10,11, was typically found to be the initiating driver event in sporadic ccRCC, arising as early as childhood in as little as a few hundred cells, preceding cancer diagnosis by up to 3-5 decades7. The VHL gene alongside chromatin-modifying genes PBRM1, BAP1, and SETD2 are co-located in this lost chromosomal region and perhaps unsurprisingly, are the most prevalent somatic gene perturbations found in ccRCC, as patients are rendered vulnerable to complete (biallelic) inactivation of these genes during their lifetime7,10,12. Furthermore, this group have identified distinct evolutionary subtypes of ccRCC that correlate with clinical phenotypes and outcomes, which could be used to guide intervention and surveillance8,9. As genomic technology advances, the genetic perturbations implicated in ccRCC continue to expand and include somatic mutations in TERT7,13, PTEN10,12, MYC14,15, and mTOR12 signalling pathways as well as other numerous metabolic pathways10, which will be discussed further in this review alongside subtype-specific genetic perturbations such as mutations in fumarate hydratase (FH) in type 2 pRCC16.
RCC is increasingly recognised as a ‘disease of cell metabolism’. Before the advent of the ‘Omics’ era, at least twelve genes implicated in RCC pathogenesis were identified to have roles in fundamental metabolic processes 17,18. One classic example in RCC is the ability of VHL inactivation to rewire the normal metabolic adaptation response to oxygen deprivation. Inactivation of VHL in ccRCC leads to the aberrant accumulation of the transcription factors hypoxia-inducible factor (HIF)1α and HIF2α despite normoxia19,20, with resultant upregulation of pathways involved in glycolysis, fatty acid, and glycogen synthesis21–23. This example of the ‘metabolic reprogramming’ phenomenon observed in cancer cells, whereby metabolic adaptation facilitates the neoplasm’s capacity to meet its bioenergetic demands, such as uncontrolled proliferation and the acquisition of other hallmark traits of cancer, is now recognised as being fundamental in the malignant transformation of cells and also in the phenotypic evolution of tumours24,25. Interestingly, HIF has been found to be a common target for metabolic reprogramming in RCC by other genetic perturbations that affect FH26–28, succinate dehydrogenase (SDH)26,27,29, tuberous sclerosis complex30, and more recently, fructose-bisphosphatase 1 (FBP1)31. FBP1 gene encodes the key gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1), which was found to directly interact with HIFs to restrain its transcriptional activity. The discovery that FPB1 is ubiquitously suppressed in ccRCC further supports the characterisation of RCC as a metabolic disease31. HIF being a common denominator associated with multiple RCC subtypes highlights it as a potential key candidate for RCC therapies. Further corroboration was provided by the landmark TCGA integrated platform analyses studying the genome, transcriptome and proteome of more than 400 ccRCC tumours10. This study highlighted the extensive metabolic reprogramming captured in ccRCC involving the upregulation of fatty acid synthesis, pentose phosphate pathway, glutamine transporters, and downregulation of the tricarboxylic acid (TCA) cycle correlating to disease aggressiveness and worsened prognosis10 (Fig. 1). Furthermore, subtype-specific metabolic gene alterations correlating to disease aggression and patient survival were identified in subsequent TCGA studies across the three major RCC subtypes (clear cell, papillary and chromophobe) supporting the principle of subtype-specific management and providing potential subtype-specific targets for novel therapies5.
Figure 1. A simplified overview of key metabolic pathways studied in cancer metabolism, including the oncometabolite production pathways.
The key pathways are as summarised. Glycolysis, a series of pathways involved in glucose catabolism to pyruvate yielding intermediates for entry into PPP, lactate fermentation (‘anaerobic glycolysis’), TCA cycle and lipid synthesis. In oncology, the ‘Warburg effect’ (aerobic glycolysis) describes the upregulation of glycolysis observed in many cancers even in the presence of oxygen. In ccRCC, upregulation of glycolysis correlates with poor prognostic outcomes in patient10. Pentose Phosphate Pathway (PPP), is a branched pathway from glycolysis, provides reducing equivalents (NADPH) and precursors for nucleotide synthesis (building block of DNA/RNA). Upregulation of PPP correlates with aggressive ccRCC and poor prognostic outcomes in patient10. The TCA cycle is a series of reactions that fully oxidise carbohydrates, lipids and proteins and generates reducing equivalents (NADH) for the electron transport chain to generate ATP. TCA cycle intermediates provides a source of precursors for lipids and amino acid biosynthesis. Anaplerosis is the process of replenishing the TCA cycle intermediates. Downregulation of TCA cycle genes correlates with aggressive ccRCC and poor prognostic outcomes in patients10. Lipid Metabolism pathways, lipid synthesis is required for energy stores and synthesis of cell membrane components whereas lipid degradation (β-oxidation) is required for release of energy stores. Upregulation of fatty acid synthesis correlates with aggressive ccRCC and poor prognostic outcomes in patients10. Glutamine-derived reductive carboxylation, glutamine is metabolised to α-ketoglutarate for entry into the TCA cycle in a reversed flow of the canonical TCA cycle (orange arrows), citrate can be extracted for lipid synthesis. This is an essential metabolic pathway that supports the growth of cancer cells with mitochondrial defects such as FH-deficient RCC178. Oncometabolite production pathways, loss-of-function mutations in FH and SDH genes encoding the respective TCA cycle enzymes lead to an accumulation of fumarate and succinate. 2HG exists in two isoforms (D2HG/L2HG). D2HG is accumulated by gain-of-function neomorphic activity of IDH enzymes. L2HG is accumulated by promiscuous activity of MDH/LDH activity. Loss-of-function of the enzymes D-/L2HGDH which catalyse the oxidation of D-/L2HG to α-ketoglutarate also result in the accumulation of D2HG and L2HG respectively.
Abbreviations: αKGDH- α-ketoglutarate dehydrogenase, ACO2 – aconitase, CoA – coenzyme A, ATP- adenosine triphosphate, ccRCC – clear cell RCC, D2HG – D-2-hydroxyglutarate, D2HGDH – D-2HG dehydrogenase, FBP1 – fructose-1,6-bisphosphatase 1, FASN - fatty acid synthesis, FH- fumarate hydratase, GDH – glutamate dehydrogenase, GLS – glutaminase, IDH- isocitrate dehydrogenase, L2HG – L-2-hydroxyglutarate, L2HGDH – L2HG dehydrogenase, LDH – lactate dehydrogenase, MDH- malate dehydrogenase, RCC- renal cell carcinoma, SDH- succinate dehydrogenase, TCA – tricarboxylic acid
Key: Dashed arrows = multiple enzyme-catalysed reactions between these two metabolites, * = mutant form of enzyme, red crosses = loss-of-function
Given the metabolic nature of RCC and the emergence of metabolic reprogramming as a contemporary hallmark of cancer32, a surge in the field of metabolomics has rapidly developed over the last decade. In general, metabolomics encompasses the ability to globally detect the metabolites present in a system (cell, tissue or organism) under a given set of conditions33. The field of metabolomics enables the study of the final downstream product of the genome and crucially captures the underlying environmental influences and external perturbations of a system33,34. Particularly in the setting of cancer, the tumour microenvironment notoriously has a profound effect on metabolism35,36, therefore integrating the study of cancer metabolomics with other ‘omics’ studies enables a holistic approach to understanding cancer pathophysiology. The key metabolic pathways of interest in cancer, alongside a broad outline of these pathways are highlighted below with reference to RCC (Fig. 1).
A handful of metabolomic studies in RCC have been performed to date with the largest studies profiling a single cohort of 138 patients with ccRCC37,38. The metabolic profiles from these studies broadly corroborated several findings from the TCGA study of a network of metabolic shifts involving upregulation of glycolysis, pentose phosphate pathway, and glutamine uptake correlating with disease aggressiveness37,38. Integration of this metabolomic data with the TCGA dataset however highlighted a lack of linearity between enzyme expression and the corresponding catalysed metabolite levels37. This lack of linearity has been theorised to involve shunting of metabolites into alternate cancer-reprogrammed pathways37,39. As uncovered so far, physiological metabolism can be efficiently manipulated by RCC to provide the conditions needed for cancer cells to survive and proliferate. However, the identification of key genetic mutations in cancer cells encoding for enzymes in mitochondrial metabolism such as FH and SDH16,40,41 paved the way for the next chapter of cancer metabolism, the discovery and evolution of the oncometabolite paradigm. By definition, oncometabolites are conventional metabolites that when aberrantly accumulated have pro-oncogenic capabilities that can contribute to tumorigenesis, especially via epigenetic dysregulation, as well as influence tumour phenotype and progression. Interestingly, of the short list of established bona fide oncometabolites, the majority have been implicated in hereditary and sporadic subtypes of RCC. This observation coupled with the strong metabolic paradigm in RCC, alongside the aggressive metabolic reprogramming underpinning RCC tumour progression, commands attention to this growing area of cross-over research in oncology and metabolism. This review is timely in synthesising the literature to date of bona fide oncometabolites in RCC, their proposed mechanisms of action, and the clinical impact oncometabolites may have in management of this important disease process.
Inception of the oncometabolite paradigm
The inception of the oncometabolite paradigm predates its contemporary nomenclature into the literature, and one could argue it began with the once dispelled but now revived Warburg hypothesis of aberrant metabolism as an aetiological determinant of cancer42,43. Based on the observation of excessive fermentation of glucose in mammalian cancer cells irrespective of the presence of oxygen (the observed phenomena later coined the ‘Warburg effect’(Fig. 1).44), it was here that Warburg first postulated that this abnormal compensatory mechanism to counteract an irreversible injury of cellular respiration, induced cells into an undifferentiated state, giving rise to “cells that grow wildly- the cancer cells”42,43. In Warburg’s view, the prime cause of cancer was mitochondrial dysfunction. Shortly thereafter however, in light of the discovery of mutated oncogenes and tumour suppressor genes, Warburg’s hypothesis was soon dismissed and altered metabolism revised as a bystander effect secondary to these genetic perturbations identified in numerous cancers45–48. Warburg’s hypothesis however came full circle at the pinnacle of this era when several genetic perturbations in genes encoding two key enzymes in the TCA cycle, FH16 and SDH subunits40,41,49 were implicated in the development of Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) and hereditary Paraganglioma (PGL), respectively. Shortly after, seminal studies uncovered an unconventional and novel role, common to both fumarate and succinate, in deregulating the HIF pathway through direct inhibition of prolyl hydroxylases (PHD), enzymes involved in signalling HIF degradation26,27,29. Stabilisation of HIF1α/2α alongside upregulation of downstream HIF1 products such as VEGF and glucose transporter (GLUT1) were observed in these HLRCC tumours and SDH-deficient tumours, in the absence of VHL inactivation26,27, providing support for the role of these aberrant metabolite accumulations in creating the hypoxic signatures and highly vascularised phenotype that are characteristic of these tumours50–54. These studies proposed an alternative mechanism to creating the notorious ‘pseudo-hypoxic’ milieu in the absence of VHL inactivation, which has been well established in VHL-mutant disease and a characteristic tumour phenotype associated with RCC and tumour aggression55,56.
Dang et al however was the first group to coin the term ‘oncometabolite’ to describe the potential tumorigenic role of pathological accumulations of the metabolite D-2-Hydroxglutarate (D2HG)57. Under physiological settings, 2-hydroxyglutarate (2HG) exists in two natural isoforms L- and D- (L2HG and D2HG respectively), both are minor metabolic by-products produced via distinct biological mechanisms and are normally kept at unappreciable levels by conversion back to α-ketoglutarate (αKG) via the respective L-/D2HG dehydrogenase (L2HGDH/ D2HGDH) enzymes58–61. In Dang et al’s study, abnormally elevated D2HG levels (up to tens of μmol per gram of tissue)were expressed by>100-fold greater in patients with malignant gliomas harbouring a single mutant copy of the isocitrate dehydrogenase (IDH) 1 gene compared to malignant gliomas with wildtype IDH genes57. Wildtype IDH encodes the TCA cycle enzyme responsible for the reversible oxidative carboxylation of isocitrate to αKG. Mutation of the IDH1 gene confers a gain-of-function neomorphic activity of the IDH enzyme that catalyses the reduction of αKG to D2HG, leading to its accumulation57. Interestingly, in patients with inborn errors of 2HG metabolism, elevated levels of L2HG have been associated with brain tumours62,63 as well as one case of Wilm’s tumour (nephroblastoma)63,64, whereas D2HG accumulations in this cohort has not been associated with cancer65. Overall, these initial studies galvanized a myriad of ‘oncometabolite’-focussed studies that have expanded upon these findings, establishing a core group of bona fide oncometabolites. Currently, this consists of fumarate, succinate, L2HG and D2HG48,59,66,67. These oncometabolites are increasingly associated with numerous malignancies, including neuroendocrine tumours49,68,69, brain tumours70,71, haematological malignancies72,73, head and neck squamous cell carcinoma74, and our topic of interest, hereditary and sporadic forms of renal cell carcinoma (RCC)59,66,67.
Pathogenesis of oncometabolite accumulation
Endogenous origins: tumour suppressors and oncogenes
Identification of loss-of-function mutations in genes encoding the key TCA cycle enzymes SDH and FH, which lead to the accumulation of succinate and fumarate respectively, as well as a gain-of-function mutation in IDH, which leads to the accumulation of D2HG, has led to appreciation of how these genetic perturbations act as tumour suppressor genes and oncogenes27,48,66,75. Both FH and SDH mutations in tumours follow the Knudson’s ‘two hit’ hypothesis of tumorigenesis76. In patients with heterozygous germline mutations for SDH or FH i.e. inheritance of one mutated allele, the loss of heterozygosity (LOH) i.e. loss of the remaining wildtype allele, seems to be the clinching factor in tumorigenesis48,66,75 and in both cases, converge on the predisposition to the development of PGL/ phaeochromocytomas (PCC)68,77–80. The SDH enzyme is composed of four subunits (SDHA, SDHB, SDHC and SDHD) as well as two assembly factors (SDHAF1 and SDHAF2), each encoded by distinct genes across multiple chromosomes81. Loss of heterozygosity in multiple subunits of SDH predisposes to a variety of cancers including SDH-deficient RCC, a very rare and aggressive disease5,78,79,82–84, whereas LOH in patients with heterozygous FH germline mutations predisposes to HLRCC16,85,86, an autosomal dominant hereditary cancer syndrome characterised by cutaneous and uterine leiomyomas and a highly aggressive form (type 2) of pRCC87. Interestingly, distinct clinical phenotypes are also observed in FH- and SDH-deficient diseases. Homozygous germline mutations in FH give rise to fumaric aciduria, a rare metabolic disease associated with infantile encephalopathy, brain malformations and neonatal polycythaemia without an associated cancer predisposition88,89, whereas homozygous germline mutations of SDHA cause severe neurological dysfunction and cardiomyopathy78. This divergence in the clinical phenotypes observed suggest that the ‘two-hit’ mutational timing and tissue-specific nature of mutations may be crucial to cancer predisposition. As such, it has been suggested that oncometabolites may be insufficient in themselves for oncogenic transformation59. Potentially confounding this notion is the finding that patients with inborn errors of metabolism such as fumarate aciduria often do not survive long enough90,91 for potential malignancies to manifest. An interesting point to discuss that currently eludes our knowledge is what drives or does not drive cancer in certain tissues upon LOH in an exquisitely-specific nature as evidenced by diseases such as HLRCC and SDH-deficient diseases. Current hypotheses have been recently discussed92, and, building upon this insight, we postulate a concept of ‘LOH tolerance’ in ‘permissive’ tissues that propagate tumourigenesis due to their capability to be more flexible (such as the ability to metabolically adapt and/or compensate as a result of these genetic perturbations e.g. reversal of the activity of the urea cycle enzyme arginosuccinate lyase (ASL) in FH-deficient cells which funnels accumulated fumarate into aberrant urea metabolism 93), whereas ‘LOH intolerance’ in a small proportion of cells and or tissues, results in lethality without further propagation or replication of these genetic perturbations and thus attenuating tumorigenesis in these tissues. Needless to say, further understanding into this current conundrum of what gives rise to distinctive patterns of tissue-specificity in cancer may reveal tissue-specific vulnerabilities that may impact greatly on future management of these clinically challenging diseases.
In contrast to SDH and FH, IDH1 and IDH2 genes, which encode the compartment-specific isoform of the IDH enzyme in the cytosol and mitochondrial respectively94, express a dominant pattern of oncogenic behaviour. Somatic mutations in only one copy of the IDH gene i.e. retention of one wildtype copy of IDH, were observed in patients in multiple cancers including gliomas71,95 and acute myeloid leukemia96, leading to the neomorphic gain-of-function activity in converting αKG into D2HG57. More recently, D2HG accumulation as a result of loss-of-function mutations in D2HGDH has been observed in a small subset of large B-cell lymphoma91, implicating both the synthesis and conversion of D2HG in its accumulation in cancer. Using the cBioPortal Database97, <1% of IDH1/2 mutations are found in large-scale cancer genomic studies of RCC such as the TCGA dataset97,98. Although 2HG was identified to be significantly accumulated in human ccRCC tissues, >90% of this was in the L2HG isoform99, suggesting D2HG is unlikely to have a significant role in RCC pathogenesis. Reduced expression of L2HGDH, an enzyme that catalyses the conversion of L2HG into αKG, was found to contribute to the accumulation of L2HG in patients with ccRCC99. LOH of the L2HGDH gene (which resides on chromosome 14q and noted to be a commonly deleted region in ccRCC100,101) correlated with reduced protein expression of L2HGDH and accumulation of 2HG, providing support that L2HGDH may also function as a tumour suppressor in RCC101. Furthermore, loss of L2HGDH conferred a worse prognosis in patients with ccRCC compared to those with L2HGDH, with preliminary metabolomic profiling suggesting that increasing levels of L2HG are associated with RCC tumour progression, further corroborating its role as an oncometabolite101.
More recently, mutations in several genes, αKG dehydrogenase (αKGDH)102, lipoic acid synthase (LIAS) 102 and lipoyltransferase-1 (LIPT1)103 have also been implicated in 2HG accumulation. These genes encode enzymes required for the proper functioning of the αKGDH-complex (αKGDHC), which catalyses the conversion of αKG to succinyl-coenzyme A in the TCA cycle. The truncated TCA cycle due to these mutations promoted the production of L2HG from accumulated αKG102,103, with evidence of downstream oncometabolite activity inhibiting PHDs leading to HIF stabilisation and HIF1-targeted gene activation including VEGF and GLUT1102. Interestingly, in patients with homozygous germline mutations of enzymes of lipoic acid synthesis, L2HG accumulations lead to suppression of PHD activity and subsequent HIF1 activation102. However as this is also a rare inborn errors of metabolism diseases and is generally lethal at a young age102,104 it may also preclude any potential oncometabolite-associated tumour development. Characterisation of heterozygous germline mutations in this setting may provide additional insight for the tumorigenic role of L2HG accumulation in αKGDH/LIAS mutations. Table 1. highlights the genetic mutations in oncometabolite-associated RCC subtypes, the clinical features and potential therapeutic strategies which will be discussed further in this review.
Table 1. Oncometabolite-associated RCC subtypes, clinical features and potential therapeutic strategies.
Oncometabolite | Gene mutation | Clinical features | Potential therapeutic strategies | References |
---|---|---|---|---|
Fumarate | FH (tumour suppressor) |
HLRCC-associated RCC (14-18% develop pRCC) Highly aggressive and early metastasis Early onset Bilateral Mainly papillary but also described as solid, tubulocystic, cribiform or cystic |
Arginine deprivation Haem oxygenase inhibition ABL1 inactivation Targeting TBK1/p65 axis GDH1 inhibition Glutaminase inhibition |
4,16,26,87,93,156,169,174,199,245 |
Succinate | SDHA SDHB (82%) SDHC SDHD SDHAF2 (tumour suppressors) |
SDH-deficient RCC (0.2% of all RCC) Early onset (mean age 37-46yo) Associated aggressive phenotype Bilateral RCC (26%) Associated with paraganglioma (25%) |
SIRT expression Exogenous glycine PC inhibition |
4,77–79,83,84,184,188,190–192,246–249 |
L2HG | L2HGDH (tumour suppressor) |
ccRCC Associated with Wilms’ tumour |
L2HGDH re-expression Glutaminase inhibition MDH2 inhibition |
64,99,101 |
Abbreviations: ABL1 - Abelson Murine Leukaemia viral oncogene homolog 1, ccRCC – clear cell RCC, FH- fumarate hydratase, GDH – glutamate dehydrogenase, HLRCC – Hereditary Leiomyomatosis and Renal Cell Cancer, L2HG – L-2-hydroxyglutarate, L2HGDH – L2HG dehydrogenase, MDH2 – malate dehydrogenase 2, PC – pyruvate carboxylase, pRCC – papillary RCC, RCC- renal cell carcinoma, SDHAF- succinate dehydrogenase assembly factor, SDH- succinate dehydrogenase, SIRT - silent mating type information regulation 2 homolog, TKB1- tank-binding kinase 1
Exogenous origins: environmental factors
Remarkably, in the absence of oncogenic mutations, oncometabolites have been demonstrated to accumulate in cells and to induce oncogenic transformation105. Identifying the environmental factors linked to oncometabolite accumulation may shed important light on how they impact or predispose individuals to cancer risk. Hypoxia-induced production of oncometabolites via “off-target”, substrate-promiscuous activity of the enzymes lactate dehydrogenase A (LDHA)106 and malate dehydrogenase (MDH)106,107 on glutamine-derived αKG23,106 results in L2HG accumulation, whereas promiscuous activity of D-3-phosphoglycerate dehydrogenase (PHGDH) catalyses the conversion of αKG to D2HG in IDH-wild type breast cancer cells108, demonstrating alternative pathways for 2HG accumulation in response to exogenous stimuli. In addition, acute ischaemic preconditioning in vivo resulted in 2HG accumulations in mouse myocardium109. Although modest accumulations of L2HG in hypoxic cells were observed compared to that in cancer cells23, hypoxia-induced L2HG accumulation was sufficient and necessary for exerting recognised ‘oncometabolite functions’ such as the repressive trimethylation of the histone protein, histone 3 lysine 9 (H3K9me3)106, in which upregulated levels were similarly observed in patients with IDH-mutant gliomas110. Histone methylation affects chromatin organisation and regulation of gene transcription110,111 and, in vitro, L2HG-induced hypermethylation of H3K9 has been demonstrated to block cellular differentiation in non-transformed astrocytes110, supporting its role in epigenetic regulation. Independent of hypoxia, acidic conditions have also been observed to drive L2HG accumulation, augmenting the promiscuous activity of LDH1/MDH2 activity as well as inhibiting the activity of L2HGDH in vitro112.
Succinate accumulation has also been observed in cancer cells grown under hypoxic conditions within a 3D tumour model113 and in animal models subjected to ischaemia-reperfusion injury in vivo 114–116. Succinate oxidation has been shown to contribute to cardiac injury at reperfusion114,116,117 via the generation of reactive oxidative species (ROS)114. However, rapid resolution of the accumulated succinate in these tissues back to baseline in this setting114,116,117 means chronic effects of succinate accumulation in tissues, analogous to SDH-deficient tumours, is challenging to study. Nevertheless, succinate accumulation observed in hypoxic retinas of rodents leads to an upregulation of angiogenic proteins such as VEGF in a HIF-independent manner via activation of the succinate receptor GPR91, suggesting alternative mechanisms for stimulating angiogenesis by succinate in this setting115. Of note, the hypoxic induction of oncometabolite accumulation may be propagated and amplified by the oncometabolites themselves as they can stabilise HIF expression through direct inhibition of the PHD enzymes involved in signalling HIF degradation19,26,27,29,29,118–120. In addition, succinate may also participate in an alternative positive-feedback system of reinforcing the HIF1α signalling loop121. HIF-dependent expression of micro-RNA210 (miR-210) in lung adenocarcinoma cells in vitro was demonstrated to target the SDHD subunit and impair SDH function, and the ensuing succinate accumulation in turn leads to HIF stabilisation through inhibition of PHDs perpetuating this hypoxic phenotype121.
Aside from hypoxia, mitochondrial dysfunction arising from glucose toxicity122–124 has been shown to result in oncometabolite production. Fumarate accumulation causing fumarate-dependent protein succination was observed in adipose tissues of hyperglycaemic mice124,125, analogous to the succination features exhibited by fumarate accumulation in cancer cells. Interestingly, a similar succination phenotype was also observed in the adipose tissue of obese, insulin-resistant, non-hyperglycaemic mice128, proposing other potential exogenous sources of oncometabolite production. Furthermore, evidence of succinate accumulation has been found in bone marrow stromal cells of diabetic mice, stimulating osteoclastogenesis through activation of the succinate receptor GPR91129. Of note, mitochondrial dysfunction has been linked to the metabolic syndrome, a distinct cluster of conditions including obesity, diabetes, hypertension, and hyperlipidaemia130, which in turn, has also been causally linked to RCC and are factors correlated with an increased risk of RCC131,132. This link provides a nidus for detailed investigations into the cross-talk between environment-induced oncometabolite accumulation and their tumorigenic role in RCC. In addition, infection has also been implicated in the exogenous production of oncometabolites, demonstrating stimulation of succinate accumulation in macrophages133,134, causing downstream HIF stabilisation and upregulation of HIF-targeted gene transcription such as interleukin 1β, a key pro-inflammatory signalling molecule133. Overall, further elucidation of the magnitude and mechanisms by which exogenous factors have on oncometabolite production and the subsequent sequalae, independent of oncogenic mutations, will be a critical step forwards in understanding and ameliorating the role of these factors in tumorigenesis and tumour evolution.
Mechanistic actions of oncometabolites in RCC
Oncometabolites exhibit a multitude of downstream pro-oncogenic functions. The functions that converge on a group of downstream pro-oncogenic pathways will be discussed first, followed by unique functions specific to each oncometabolite (Fig. 2).
Figure 2. Summary of the shared and individual oncometabolite downstream signalling pathways.
The oncometabolites fumarate, succinate, and 2HG and their production pathways are highlighted (red box). These oncometabolites converge on shared downstream effects through the inhibition of α-ketoglutarate dependent dioxygenases (bottom blue box). These oncometabolites also exhibit distinct, divergent downstream effects, which are summarised for each oncometabolite (top green box).
Abbreviations: αKG – alpha-ketoglutarate, 2SC – S-2-succino-cysteine, 5hmc -5-hydoxymethylcytosine, 5mc-methylcytosine, ABL1 - Abelson Murine Leukaemia viral oncogene homolog 1, CpG – cytosine-guanosine dinucleotide, D2HG – D-2-hydroxyglutarate, D2HGDH – D-2HG dehydrogenase, FH- fumarate hydratase, FTO – fat mass and obesity-associated protein, GLS – glutaminase, GPR91 – G-protein coupled receptor 91, Gpx1 – glutathione peroxidase 1, HIF- hypoxia-inducible factors, IDH- isocitrate dehydrogenase, KEAP1 = Kelch-like ECH-associated protein-1, KDM- histone demethylases, L2HG – L-2-hydroxyglutarate, L2HGDH – L2HG dehydrogenase, LDH – lactate dehydrogenase, MDH- malate dehydrogenase, mRNA -messenger RNA, mTOR – mammalian target of rapamycin, MYC - NFkB – nuclear factor kappa-light-chain-enhancer of activated B cells, Nrf2 -Nuclear factor (erythroid-derived 2)-like 2, OH – hydroxyl group, P – phosphorylated, PHD- prolyl hydroxylases, PTPN12 - protein tyrosine phosphatase non-receptor type 12, RCC- renal cell carcinoma, SDH- succinate dehydrogenase, TCA – tricarboxylic acid, TKB1- tank-binding kinase 1, TET – ten-eleven translocation enzymes, VEGF – vascular endothelial growth factor
Common downstream pro-oncogenic pathways
A characteristic trait shared between succinate, fumarate, and 2HG is their ability to competitively inhibit αKG-dependent dioxygenases (αKGDDs)66,135 through their structural similarity to αKG, an essential co-substrate for enzyme activity70,118. αKGDDs are a superfamily of enzymes involved in a diverse plethora of biological processes. The most studied αKGDDs related to oncometabolite signalling consist of PHDs, for which inhibition is involved in the induction of the ‘pseudohypoxic’ milieu27,48,66,67; the Jumonji C domain-containing histone lysine demethylases (KDM)70,110,118,137, and the ten-eleven translocation (TET) enzyme family of 5-methylcytosine (5mc) hydroxylases, involved in histone and DNA demethylation respectively70,110,118,137,138, in which characteristic hypermethylated phenotypes are associated with altered gene expressions including the regulators of epithelial-to-mesenchymal transition (EMT), a hallmark of tumour aggressiveness and metastatic progression137,139(Fig. 2).
As highlighted above, the initial evidence supporting the concept of oncometabolites was their role in inducing a pseudohypoxic milieu observed in SDH-deficient PGL/PCC and FH-deficient HLRCC26,27,29,120. Through direct inhibition of PHD19, hydroxylation of HIF1α/2α subunits by PHD is inactivated, ultimately culminating in aberrant HIF stabilisation with downstream upregulation of HIF-targeted genes such as VEGF and GLUT126,27. Notably, these tumours tend to exhibit intense vascularisation and hypoxic gene signatures in keeping with a pseudohypoxic tumour phenotype50–54. Interestingly, HIF1α/2α inactivation in SDHB-deficient osteosarcoma cells significantly impaired tumour growth in a mouse xenograft model140 whereas HIF1α/2α inactivation in a FH-deficient mouse model of renal cyst disease exacerbated or failed to ameliorate this phenotype respectively141. Overall, these studies highlight the complex role of HIF/pseudohypoxia in tumorigenesis suggesting it may be context dependent e.g. cell specific66. Whilst L2HG has also been shown to inactivate PHDs and aberrantly stabilise HIF1α102,142,143, the activity of D2HG on PHD remains contentious. Unexpected agonistic activity of D2HG on PHDs has been observed in vitro102,143, however these findings are challenged due to the non-enzymatic oxidation of D2HG to αKG activity observed in vitro144, which would provide the necessary co-substrate for PHD activation in this situation.
Oncometabolite also have a role in epigenetic alterations through direct inhibition of KDMs110,137 and TETs138, which are groups of enzymes responsible for histone and DNA demethylation respectively. This oncometabolite function leads to changes in chromatin structure and function that lead to hypermethylation phenotypes that alters the expression of a wide range of genes involved in cellular differentiation and acquisition of malignant features. Of note, the epigenetic effects of DNA and histone methylation on transcriptional activity are challenging to distinguish as they are often interdependent and inter-regulated145. Generally speaking, histone hypermethylation e.g. due to KDM inhibition, results in either transcriptional gene repression or activation (also known as repressive or active marks) depending on the type of histone residues and the number of methyl groups added66,145. DNA methylation at ‘CpG islands’ (clusters of dinucleotide sequence of a cytosine followed by a guanosine nucleotide in the 5’-3’ direction, often found in promoter regions upstream of transcription sites) usually represses downstream gene transcription66,145. Cytosine methylation in position 5, also known as 5-methylcytosine (5mC) undergoes oxidation by TET enzymes that convert 5mC to hydroxylated 5mc (5hmC)66,145. This reaction primes the cytosine to demethylation, generating unmethylated cytosine (5C). In general, global DNA hypomethylation, leading to inappropriate transcriptional activity and chromosomal instability, coupled with specific patterns of hypermethylated CpG promoter islands, especially upstream of tumour suppressor genes resulting in repressed expression, is characteristic of many tumour types145–147.
Several studies to date have investigated the oncometabolic effects on histone/DNA hypermethylation through inhibition of KDM and TETs in FH, SDH and IDH-mutant tumours70,110,118,137,142,148–150. SDH-deficient tissues from patients with PGL and PCC demonstrated high levels of repressive histone marks (H3K27me3)137. In addition, succinate accumulation in SDHB-knockout chromaffin cells137 and SDHB-knockdown murine ovarian cancer cells151, as well as 2HG accumulation in IDH1 mutant cells70,110, demonstrated KDM and TET inhibition with characteristic hypermethylation phenotypes associated with suppression of cellular differentiation110,137 and activation of EMT, through up- or downregulation of positive and negative EMT regulator genes respectively137,151. HLRCC-derived FH-deficient cells also elicited an EMT signature in keeping with SDH-deficient cells137, via fumarate-induced TET-mediated epigenetic suppression of miR-200, a short RNA molecule with tumour suppressive effects on EMT gene expression by modulating mRNA translation139. This EMT phenotypic switch induced by oncometabolite accumulation in FH- and SDH-mutant RCC tumours is no doubt a contributing factor to their clinically aggressive behaviour. In vivo, SDHA- and FH-silencing in mouse hepatocytes led to succinate and fumarate accumulation respectively, with evidence of KDM and TET inhibition and regulation of target gene expression118. DNA hypermethylation linking to repression of specific-lineage differentiation has also been observed in patients with IDH1/2 mutant chondrosarcoma149 and acute myeloid leukaemia148. Furthermore, accumulations of D2HG resulted in increased DNA methylation (5mc) with concurrent decreased DNA hydroxymethylation, indicating TET inhibition, was observed in human IDH1 glioma tissue70 and in ectopic expression of IDH1/2 mutations in various cell types, which blocked cellular differentiation148–150.
Identification of specific DNA hypermethylation patterns within a subset of colorectal cancers152 gave rise to the CpG island methylator phenotype (CIMP)-associated cancer subtypes, characterised by their extensive epigenomic aberrations and distinct biology150,152,153, which has been increasingly recognised in other malignancies including gliomas (G-CIMP) 150,153,154 and more recently, in a subset of type 2 papillary RCC (CIMP-RCC)5,155. Interestingly, G-CIMP tumours are tightly associated with IDH1 mutations150,154 and introduction of mutant-IDH1 into human primary astrocytes leads to an accumulation of D2HG, inhibition of TET and reproduced a DNA hypermethylation profile that mirrored the changes observed in G-CIMP150. The recent characterisation of CIMP-RCC has been associated with early-onset disease and perhaps unsurprisingly, germline or somatic mutations of the FH gene5,155. Given the role of oncometabolites in αKGDD inhibition, including on TET enzymes, it is plausible that fumarate accumulation may be causally linked to the hypermethylated state in CIMP-RCC. Given that CIMP-RCC conferred the worst prognosis of all the RCC subtypes5 and the highly aggressive nature of FH-deficient RCCs156–159, in combination with the ineffectiveness of current RCC therapies in advanced FH-deficient RCC157,160, understanding the molecular underpinnings of this disease process is warranted to find more effective strategies to treat these patients, such as the potential use of histone and DNA methylation inhibitors, which will be discussed in the next section.
Of note, several studies have demonstrated that oncometabolites have varying IC50 values (half maximal inhibitory concentration, a measure of the potency of a substance to inhibit a specific biology process/function by 50%) for different αKGDDs70,118,138,142 suggesting that oncometabolite type and accumulation levels may determine the precise nature of downstream oncogenic processes in a given cell. Beyond the common inhibition of αKGDDs, we have begun to appreciate the distinct biological functions of individual metabolites, those that are especially relevant to RCC tumorigenic capabilities will be discussed next.
Distinct downstream effects of oncometabolites
Fumarate
Fumarate has demonstrated the most versatility of the bona fide oncometabolites to date, impacting on oncogenic signalling, antioxidant response, and phenotype switching (Fig. 2). In addition to the direct inhibition of PHD enzymes that facilitate pseudohypoxia induction, fumarate has also been shown to drive a hypoxic phenotype on a transcriptional level through the non-canonical activation of NFkB, a family of transcription factors that can promote HIF1α transcription. This signalling pathway is dependent on fumarate activation of Tank-binding kinase 1 (TBK1), an enzyme that phosphorylates p65 (a subunit of NFkB) with subsequent NFkB activation. Furthermore, inhibition of TBK1/p65 axis in FH-deficient RCC cells blocked HIF1α expression and reduced cellular invasion in vitro, suggesting a novel target for treatment in FH-deficient RCC158. This finding supports the critical tumorigenic role of HIF1α and pseudohypoxia in aggressive RCC subtypes such as FH-deficient RCC (HLRCC)16,55,162. In a similar fashion, silencing HIF1α in HLRCC-derived (FH-deficient) RCC cell lines diminished the invasive properties of these cells163. Potentially contradicting this theory, the genetic inactivation of HIF1α/2α in Fh1 (murine FH)-deficient mice was shown to exacerbate, or failed to ameliorate, the renal cyst phenotype respectively, suggesting alternate mechanisms for oncogenesis in FH-deficient cells141.
An alternative candidate oncogenic pathway in FH-deficient disease via the stabilisation of the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) antioxidant pathway has been proposed141,157. A distinct feature of FH-deficient tumours is the ability of accumulated fumarate to modify a wide range of proteins126,164. The post-translational protein modification triggered by fumarate, known as succination, can impair protein function, and is caused by fumarate reacting with specific cysteine residues on proteins, producing S-2-succino-cysteine (2SC) residues126,164. A key target of succination is the protein Kelch-like ECH-associated protein-1 (KEAP1). Removing KEAP1’s repressive effects on the transcription factor Nrf2 results in upregulation of Nrf2-dependent genes involved in antioxidant pathways that regulates the cells ability to adapt to oxidative stress141,157. In keeping with this, Nrf2 and downstream Nrf2-targeted genes were found to be upregulated in HLRCC-derived type 2 pRCC tumour cells141, highlighting potential alternative targets for treatment strategies in this aggressive disease141,157. On the contrary, succination of the antioxidant glutathione in FH-deficient RCC cells, depletes the antioxidant capacity of these cells, rendering them susceptible to endogenous accumulation of ROS165 with subsequent stabilisation of HIF1α165 and induction of cellular senescence166. Senescence is a state of irreversible growth arrest linked to tumour suppressive activation and thought to be a protective phenotype against cancer167. Ablation of a key mediator of senescence, p21, in Fh1-deficient mice induced the transformation of benign renal cysts into hyperplastic lesions suggesting that this fumarate-induced senescent event needs to be overcome for renal tumorigenesis to proceed166. Although ROS in itself can activate the Nrf2 signalling pathway through KEAP1 inhibition168, fumarate-dependent succination of KEAP1 seems to be the predominant mechanism for Nrf2 activation in these FH-deficient cells165,166.
Furthermore, although not within the context of FH-deficiency, fumarate has also been found to bind directly to glutathione peroxidase 1 (Gpx1), activating this ROS scavenging enzyme in cancer cells with upregulated glutamate dehydrogenase 1 (GDH1) expression169. GDH1 maintains the levels of TCA cycle intermediates by catalysing the conversion of glutamate to αKG and subsequently to fumarate, where it can bind to Gpx1 and confer a proliferative advantage to cancer cells by regulating redox homeostasis169. Furthermore, GDH1 inhibition attenuated cancer cell proliferation and tumour growth in vivo169. It is plausible, given the nature of fumarate accumulation in FH-deficient tumours and that glutamine entry into the TCA cycle (via GDH1) is a dominant pathway in this setting170,171, GDH1 inhibition in the setting of FH-deficiency may also ameliorate these pro-tumoural effects. Nevertheless, these studies in FH-deficient cells demonstrate that they have highly adapted and intrinsic mechanisms to combat redox stress in a multi-layered approach that confers tumour survival. Tying the HIF and Nrf2 pathways together was the identification of the Abelson Murine Leukaemia viral oncogene homolog 1 (ABL1) upregulated in FH-deficient tumours160. Fumarate-mediated activation of ABL1 occurs via the suppression of the protein phosphatase PTPN12 via oxidative stress172, which leads to activation of the Nrf2 antioxidant pathway and the mTOR/HIF1α hypoxic signalling pathway in FH-deficient RCC cells160. Furthermore, ABL1 inhibition suppressed the invasion capacity and growth of these cells in vitro and in vivo160,172. As ABL1 is upstream of two major pathways implicated in FH-deficient tumours, this may suggest that multimodal treatment strategies i.e. targeting multiple pathways, may be beneficial in these diseases.
Another unique oncometabolic feature of fumarate is its ability to directly modulate cellular metabolism. Conventionally, fumarate participates in several major interlinked pathways such as the TCA cycle and the urea cycle93,173. Accumulation of fumarate in FH-deficient cells has been shown to reverse the activity of the urea cycle enzyme arginosuccinate lyase (ASL)93. Normally, argininosuccinate is produced from citrulline and aspartate via argininosuccinate synthetase (ASS) in the urea cycle, which is then converted into fumarate and arginine via argininosuccinate lyase (ASL). Reversal of ASL activity results in an accumulation of argininosuccinate and renders FH-deficient cells auxotrophic for arginine93. Expectedly, arginine depletion led to reduced cellular survival and proliferation in vitro93. In addition, the loss of FH leads to a complex metabolic rewiring pattern involving the diversion of increased glutamine uptake into the haem synthesis/degradation pathway, which critically sustains mitochondrial NADH levels and mitochondrial membrane potential174. Targeting this unique FH-deficient haem pathway, in particular the enzyme haem oxygenase 1, which catalyses the degradation of haem, thus rendered a selective synthetic lethality to FH-deficient cells, cleverly sparing normal (wildtype FH) tissues174. These two studies93,174 highlight how FH-specific liabilities can be meaningfully manipulated to provide novel strategies to treat FH-deficient tumours such as in patients with HLRCC. Perhaps unsurprisingly in RCC, given the multitude of ways in which fumarate lives up to its deserving oncometabolite status, an increased gene expression of FH is correlated with better survival outcomes10, and correspondingly, FH gene suppression correlates with very poor prognosis5,139,155. Furthermore, FH is found to be suppressed in a large subset of patients with ccRCC, which correlates with EMT and poor prognosis139. Therefore, identifying the pervasive sequalae of fumarate accumulation in these tumours can be utilised as a nidus for the development of more effective and targeted therapies that are required for the management of FH-deficient RCC.
Succinate
Besides the inhibitory role of succinate against αKGDDs shared with fumarate and 2HG, succinate also exhibits distinct oncometabolite features that may impact on the phenotype of SDH-deficient tumours. Activation of the succinate receptor GPR91 by high levels of succinate has been shown to upregulate angiogenic proteins including VEGF in a HIF-independent manner in hypoxic retinal ganglion cells of rodents115, and induce an angiogenic phenotype in human endothelial cells in vitro and in transgenic zebrafish in vivo115,175. Activation of this succinate/GPR91 signalling axis may also be an important pathway in tumour angiogenesis115,175 and highlights the ability of succinate to exhibit hormone-like traits. Through activation of the GPR91 pathway, elevated circulating levels of succinate have been implicated in renovascular hypertension via activation of the renin-angiotensin-system (RAS) in kidneys176,177. Interestingly, hyperglycaemia was also found to trigger this pathway, potentially implicating it in the pathophysiology of diabetic nephropathy178. The succinate/GPR91 signalling axis has also been implicated in the pathological hypertrophy of ischaemic cardiomyocytes179 and activation of fibrosis in ischaemic-induced liver damage180.
Succinate, similar to fumarate, has been linked to a role in the post-translational protein modification known as protein succinylation (different from fumarate-induced succination)181–183. However, as it has been observed that succinylation results from succinyl-CoA reacting with the lysine residues in proteins182,183, it is likely that succinate accumulation in SDH-deficiency, which can equilibrate with succinyl-CoA, is the underlying mechanism behind this link182. Interestingly, the accumulation of D2HG competitively inhibits SDH activity in IDH1-mutant fibrosarcoma cells, causing a hypersuccinylated phenotype and apoptosis resistance184, two established hallmarks of cancer24. Re-expression of the desuccinylase SIRT5, as well as glycine supplementation led to reversal of this hypersuccinylated phenotype and slowed oncogenic growth in vitro184. Mechanistically, glycine depletes the availability of succinyl-CoA to succinylate proteins, by condensing directly with succinyl-CoA to form 5-aminoevuilinc acid which enters the haem biosynthesis pathway184. Remarkably, type 2 pRCC tumours with FH-mutations were also found to be hypersuccinylated compared to FH-wildtype RCC184, demonstrating the likely convergence of oncometabolites onto this process. In addition, several key metabolic enzymes such as MDH and IDH2, as well as histones185 are found to be targets of protein succinylation182,186, possibly suggesting an autoregulatory role in metabolism and perturbation of the cellular epigenome, however these functional effects are yet to be fully elucidated133,186,187. Lastly, SDH-deficient cells have also been identified to exhibit dependency on pyruvate carboxylase (PC) to funnel pyruvate into the truncated TCA cycle for aspartate biosynthesis, an important precursor in sustaining cellular growth188,189. Furthermore, silencing PC expression attenuated SDH-deficient tumour growth in vivo in a mouse model188. This coupled with an increased mRNA expression of PC in a range of human SDH-deficient tumours including PC protein expression in SDH-deficient RCC highlights a potential target for synthetic lethality in SDH-deficient RCC188.
As highlighted earlier, although SDH-deficient renal tumours represent a rare (0.2% of all RCCs) and recently recognised distinct RCC subtype (WHO 2016 Classification), overall it is a highly aggressive tumour with a younger onset of disease (mean age 37- 46years) with the majority of tumours likely to harbour SDHB mutations (82%), although mutations in all four subunits and the assembly factor SDHAF2 have been implicated as tumour suppressor genes in the pathogenesis of RCC79,83,84,190–192. Several common features in keeping with HLRCC renal tumours (highly aggressive, younger onset) makes SDH-deficient RCC as challenging to manage82,191. Although within the remit of ccRCC, increased gene expression of SDHB, SDHC, SDHD have been correlated with better patient survival outcomes10. In a similar manner, elucidating the common and individual sequalae of succinate accumulation in the setting of RCC will form the basis for future strategies in targeting this cohort.
2-hydroxyglutarate
The recent discovery and elucidation of L2HG accumulation in ccRCC99,101 highlights the relevance of distinguishing the tumorigenic role of this oncometabolite. As discussed, 2HG exists in two isoforms (L2HG/D2HG), produced by distinct biological mechanisms that are differentially upregulated in cancers and are found to exhibit distinct features beyond αKGDD enzyme inhibition59,67. Interestingly, studies of 2HG in leukaemic cells has yielded conflicting results. In multiple IDH-wildtype leukaemic cell lines, dose-dependent inhibition of cell proliferation and viability were demonstrated upon addition of D2HG193. However, exogenous D2HG added at a comparable concentration in similar IDH-wildtype cells demonstrated a contrasting phenotype of cell proliferation and leukaemic transformation in another study105. This disparity has been partly attributed to the discrepancy in in vitro conditions used in these experiments193. However, in support of an anti-tumoural effect, D2HG accumulation (either exogenous addition or endogenously through IDH mutation) demonstrated attenuated progression of the disease and increased survival in an in vivo xenograft model of leukaemia193. Mechanistically, D2HG competitively inhibits the fat mass and obesity-associated protein (FTO)193, the first identified mRNA demethylase and member of the αKGDD family193,194, which in turn downmodulates the expression of targeted genes, such as MYC, RARA and ASB2, normally involved in promoting cell growth and transformation193,195. Interestingly, these findings were also recapitulated in glioma cells as well as with exogenous L2HG193, suggesting a convergence in function of these 2HG isoforms as well as in phenotypic effects across multiple cancers. Furthermore, direct inhibition of ATP synthase and subsequent downregulation of mTOR signalling by D2HG accumulation in IDH1-mutant glioma cells in vitro and in vivo suggest growth suppressive functions of D2HG and further corroborates the anti-tumoural effects of D2HG196. This phenomenon may partially marry up the correlation between IDH-mutations in gliomas and improved patient prognosis196,197.
One rationale for the convincing simultaneous pro- and anti-tumoural roles of 2HG, is that its effects are contingent on the specific cancer and/or specific stage in tumour evolution (i.e. tumour initiation versus tumour progression)193,196. Supporting this notion, accumulations of D2HG and L2HG were observed in colorectal cell lines in the absence of IDH or D-/L2HGDH mutations198. Dissecting their individual roles in this setting revealed that D2HG, but not L2HG, was found to have pro-tumoural roles in EMT gene upregulation through KDM inhibition and subsequent histone hypermethylation, as well as in the acquisition of invasive and migratory phenotypes in these cells198. Furthermore, this phenotype was ameliorated by the addition of a glutaminase inhibitor198, signifying that D-/L2HG accumulation in this context is dependent on glutamine-derived anaplerotic flux. In keeping with these findings, colorectal cancer specimens demonstrated elevated D2HG levels, with increased levels of D2HG correlated with higher frequency of distant metastases198. Ascertaining the IC50 levels for these isoforms within this context may add insight into this disparity between D-/L2HG, which as discussed, shows variation between oncometabolites, target enzymes and experimental conditions66,70,118,138,142. Overall, these studies highlight that both 2HG isoforms can converge on a range of non-metabolic functions such as DNA/histone hypermethylation70,101,110,142,148, however also potentially diverges in respects to the pro- and anti-tumoural effects as well as the 2HG isoform implicated101,102,105,110,112,193,195,196,198. Thus, it would be imperative for future studies elucidating the role of 2HG isoforms in tumours and the potential clinical applications associated, to be cancer and tumour stage specific.
Clinical applications of oncometabolites in RCC
Unravelling the oncogenic identity of a small group of seemingly innocuous metabolites has given rise to the oncometabolite paradigm, whereby aberrant accumulations of member metabolites have demonstrated potent pro-oncogenic capabilities that impact on tumorigenesis, tumour phenotypes and progression. Naturally, a number of potential clinical applications utilising oncometabolites has surfaced as a result. This section highlights the areas in which oncometabolites may have a role in clinical practice (Fig. 3), particularly in regard to the management of RCC.
Figure 3. Overview of oncometabolite-associated pathways highlighting potential therapeutic targets.
This schematic provides an overview of the different stages in which oncometabolites can be exploited for therapeutic intervention, also highlighting the opportunity for multimodal or multi-layered synergistic approaches. This ranges from targeting the exogenous drivers of oncometabolite production (green arrow), to the nutrient sources and enzymatic perturbations involved in oncometabolite accumulation (red box), to the downstream enzymatic, epigenetic and phenotypic phenomena (blue and yellow boxes).
Abbreviations: αKG – alpha-ketoglutarate, 5hmc -5-hydoxymethylcytosine, 5mc-methylcytosine, CpG – cytosine-guanosine dinucleotide, D2HG – D-2-hydroxyglutarate, D2HGDH – D-2HG dehydrogenase, FH- fumarate hydratase, GLS – glutaminase, HIF- hypoxia-inducible factors, IDH- isocitrate dehydrogenase, KDM- histone demethylases, L2HG – L-2-hydroxyglutarate, L2HGDH – L2HG dehydrogenase, LDH – lactate dehydrogenase, MDH- malate dehydrogenase, OH – hydroxyl group, PHD- prolyl hydroxylases, RCC- renal cell carcinoma, SDH- succinate dehydrogenase, TET – ten-eleven translocation enzymes, VEGF – vascular endothelial growth factor
Key: Red dashed boxes = therapeutic strategies, * = mutant form of enzyme, red crosses = loss-of-function
Novel therapeutic targets
Multiple targets within oncometabolite-associated pathways for therapeutic intervention have been highlighted. These can broadly be divided into targeting oncometabolite accumulation i.e. production and/or degradation pathways, or targeting the downstream sequalae either in the broad sense e.g. DNA hypomethylation agents149, or specific pathways e.g. arginine deprivation in FH-deficient RCC93,199.
Targeting oncometabolite accumulation
Targeting the pathways that contribute to oncometabolite accumulation has shown promising results to date with evidence of translation into clinical practice200. The development of specific mutant-IDH1/2 inhibitors demonstrating reduction in D2HG levels with reversal of the DNA/histone hypermethylation profile and phenotypic reversal of the cellular differentiation block in leukaemia in pre-clinical studies is one such example105,201–203. Further supported by clinical studies200,204, this effort has resulted in the recent approval for their use in the management of IDH-mutant acute myeloid leukaemia200.
Another rapidly emerging area of interest is the use of glutaminase inhibitors in oncology. Cancer cells are long recognised to rely on glutamine as an essential fuel source and biosynthetic precursor to support the demands of rapid growth, survival and stress in cancer cells171. Glutamine has several fates, however the conversion of glutamine to αKG as an anaplerotic source is of particular relevance, in which the first step from glutamine to glutamate is catalysed by the enzyme glutaminase171. In addition, cancers with defective mitochondria such as FH- and SDH-deficient RCC, predominantly utilise glutamine-derived αKG in a reductive carboxylation manner (reversal of the canonical TCA cycle flow, Fig. 1), allowing these cells to bypass the truncated TCA cycle and replenish essential TCA cycle intermediates such as citrate, which is cleaved to form acetyl-coenzyme A for lipid biosynthesis170,171. In addition, glutamine-derived αKG appears to be the dominant source for 2HG production in several cancer subtypes including breast 205, chondrosarcomas 206, colorectal cancer , and RCC 101, as well as being the main source of fumarate in FH-deficient RCC cells170,171. As highlighted, L2HG is significantly accumulated in human ccRCC tissues99, partly attributable to the LOH of the L2HGDH gene99 as well as the promiscuous activity of MDH2 on predominantly glutamine-derived αKG101. Targeting the ‘production’ pathway of 2HG accumulation i.e. glutamine/MDH2 axis via pharmacological or genetic inhibition resulted in significantly reduced L2HG levels and suppression of the migratory phenotype in multiple RCC cell lines with restoration of epigenetic TET activity, as evidenced by elevated DNA 5hmc levels in vitro101. Moreover, glutaminase inhibition in vivo demonstrated suppression of tumour growth, adding to the evidence base that targeting glutamine in this setting profoundly affects L2HG accumulation with suppression of tumour phenotype. Of note, several other independent studies have investigated glutaminase inhibition in the wider context of RCC, including in VHL-mutant and VHL-wildtype RCC, also demonstrating tumour growth suppression in vivo15,207,208. These studies have facilitated the translation of glutaminase inhibitors into several phase 1/2 clinical studies either as monotherapy or in combination with approved therapeutic agents and have included FH- and SDH-deficient RCC subtypes, as well as in several metastatic RCC cohorts with early promising results209–211. Although it is highly unlikely that the effects of glutaminase inhibition in RCC are purely mediated through L2HG (as not all RCC tumours accumulate L2HG), given that the loss of L2HGDH and accumulation of L2HG confers a worse prognosis in patients with ccRCC101 and concurrently, upregulation of the glutamine transporter correlates with aggressive ccRCC and worse prognosis10, it would be of immense value to ascertain whether there is crossover talk, given the ability of L2HG to significantly modulate the epigenetic cell state, as well as determine whether glutaminase inhibition has a more profound effect in L2HG-associated RCC tumours given the oncometabolite gamut of capabilities. In addition to glutamine/MDH2 axis inhibition, genetic restoration of L2HGDH also demonstrated suppression of tumour growth in vivo101. These findings highlight several vulnerabilities on both sides of L2HG accumulation that can be exploited for the development of targeted therapies in L2HG-associated RCC. Establishing whether use of multiple approaches to reduce L2HG levels have a synergistic effect or not may impact on strategic management of this subset of RCC tumours. Of note, given that L2HG has no known physiological role58–61, specific targeting of L2HGDH may be preferable over MDH2 given that MDH has a physiological role and targeting this enzyme may lead to undesirable systemic effects.
Targeting downstream oncometabolite-sequelae
Potential therapeutic strategies have been developed to tackle both broad oncometabolite-induced pathways as well as cancer-specific liabilities, that is, oncometabolite-associated phenomena observed in specific cancer types (Fig. 3). The general convergence of oncometabolites on the inhibition of αKGDD enzymes led to an early and straightforward strategy of overcoming competitive inhibition by administering αKG in excess. Studies in SDH-deficient cancer cells and in RCC cells treated with exogenous fumarate, administration of αKG lead to a reversal of the HIF pseudohypoxic drive through restoration of PHD activity212,213 as well as reversal of DNA 5mc accumulation, indicative of TET activity restoration137. Dose-dependent suppression of HIF1α and VEGF protein levels by αKG were also observed in lung cancer cells and in hepatocellular carcinoma cells in vitro214,215. Furthermore, αKG administered to human colorectal cancer cells under hypoxic conditions led to PHD-induced destabilisation of HIF expression and furthermore, exhibited PHD-dependent hypoxic cell death216. In corroboration, αKG exhibited anti-tumoural effects in vivo, suppressing tumour growth and angiogenesis in a lung cancer xenograft model214. Overall, these preclinical studies suggest that utilising αKG, in a wide variety of cancer subtypes, can meaningfully reverse oncometabolite-induced αKGDD inhibition at a molecular and phenotypic level. As epigenetic dysregulation and pseudohypoxia drive are strongly implicated in the pathogenesis and progression of RCC, broad targeting of αKGDD combating both these elements, warrants further investigation in this setting. In a similar vein, targeting the HIF transcription factors, drivers of the mutual pseudohypoxic phenotype observed in oncometabolite-associated RCC subtypes, may be another promising therapeutic approach in this setting. Attenuation of tumour growth upon HIF2α inhibition was demonstrated in vitro and in multiple RCC tumourgraft models217,218. These studies helped lay the foundation for the first human studies and clinical trials using HIF2α inhibition in patients with locally advanced and metastatic ccRCC with promising early results demonstrating two-thirds of patients having complete/partial response or stable disease with HIF2α inhibition217,219. Given the robust capability of oncometabolites to induce the HIF-signalling pathway independent of VHL-deficient RCC, these studies will prove insightful in the management strategies for targeting rare but aggressive oncometabolite-associated RCCs.
The development of DNA hypomethylation agents, also known as DNA methyltransferase (DNMT) inhibitors for clinical practice, such as 5-azacitidine, have demonstrated to improve outcomes and delay transformation in patients with high-risk myelodysplastic syndrome220,221. As highlighted, the hypermethylation phenotype is a characteristic trait amongst oncometabolite-associated tumours and DNMT inhibitors have demonstrated potential in ameliorating these associated phenotypes137,149,222. Studies using low doses of DNMT inhibitors have demonstrated impairment in cell growth, reversal of the migratory phenotype and restoration of cell differentiation in a range of SDH-knockout and IDH1/2-mutant cells in vitro137,149,222, which is further evidenced by reversal of DNA methylation marks222. Furthermore, decitabine (a derivative of 5-azacitidine) demonstrated tumour growth suppression in IDH1-mutant glioma cells in vivo222. Whilst these studies provide a potential strategy for targeting DNA-related oncometabolite-induced epigenetic modifications, both DNA and histone methylation have a role in modulating transcriptional activity and therefore simultaneous targeting of multiple epigenetic modifiers may prove to be more strategic66. Of note, high doses of decitabine induced cytotoxicity in all cells137, therefore careful characterisation of the desired therapeutic window will also be important for future studies.
The recent elucidation of the role of L2HG in RCC epigenetic dysregulation has added more insight into this disease process and potential therapeutic strategies. Interrogation of the epigenetic effects demonstrated elevated levels of the trimethylated histone H3K27Me3 which corresponded with reduced levels of DNA 5hmc suggesting L2HG-induced KDM inhibition in RCC101. In conjunction, lowering L2HG levels in these cells leads to the re-expression of H3K27Me3 target genes as well as polycomb repressor complex 2 (PRC2) target genes, which encodes a histone methyltransferase responsible for the repressive trimethylation of H327Me3101,223. Inhibition of PRC2 via knockdown of the PRC2 catalytic subunit, enhancer of zeste homologue 2 gene, in RCC cells with high 2HG-levels, resulted in reduced H3K27Me3 levels as well as reduced migratory abilities101. Furthermore, knockdown of KDM6A, a known H3K27 demethylase, in L2HGDH-wildtype RCC phenocopied the enhanced migratory properties of elevated L2HG-RCC cells, implicating KDM6A as a specific target for L2HG in RCC101. Interestingly, mutations, predominantly somatic, in KDM6A (also referred to as UTX) have been identified in renal cancer 224,225, suggesting that chromatin remodelling via oncometabolites may recapitulate the effects of other epigenetic modifiers mutated in RCC. In other words, oncometabolites and chromatic modifiers may converge towards the same gene signature. Due to the identification of mutations in epigenetic regulators, such as KDM6A/UTX in renal cancer224,225, several studies have investigated the effects of DNMT inhibitors in renal cancer with encouraging results demonstrating growth inhibition226 in VHL-mutant and -wildtype RCC cell lines. In addition, re-expression of silenced genes was observed in a dose-dependent manner with DNMT inhibition in several RCC cell lines226,227, and moreover, re-expression of interferon (IFN) response genes in RCC cells via reversal of the gene silencing methylation by DNMT inhibitors augmented interferon-induced apoptosis in vitro228. In an early clinical study, combined DNMT inhibition and interferon therapy has demonstrated potential efficacy in the setting of metastatic RCC229. Overall, these studies demonstrate that targeting epigenetic modifiers in RCC has evidence of anti-tumoural effects that may also potentiate and synergise with other adjunctive therapies such as interferon therapy. Given that oncometabolites and other mutated epigenetic modifiers in RCC may converge towards the same gene signature, these studies are especially relevant to therapeutic tactics for targeting aggressive oncometabolite-associated RCC diseases.
The discovery of individual oncometabolite properties permits the development of novel therapeutic methods to manipulate these pathways for amelioration of pro-tumoural effects. As highlighted throughout this review, there are numerous targets and strategies that are oncometabolite- and cancer-specific, that provides the basis for further translational studies. Table 1 provides a summary of the potential therapeutic targets for oncometabolite-associated RCC subtypes as discussed in this review. Whilst not novel, targeting the modifiable exogenous factors implicated in oncometabolite accumulation may ameliorate pro-tumoural effects of oncometabolites, particularly given the examples of dose-dependent effects on oncometabolite-induced downstream pathways highlighted this review. Although not fully elucidated, hyperglycaemic-induced oncometabolite production has the most compelling evidence in eliciting phenotypes analogous to those observed in oncometabolite-associated cancer cells124–126,164 and thus studying the role of antidiabetic therapies such as metformin, may be of interest in oncometabolite-associated tumours. Of note, metformin is currently in oncological clinical trials in patients with breast and prostate cancer, although the data is challenging, it has demonstrated a degree of anti-tumoural activity with recognised roles in modulating numerous metabolic pathways 39,230.
Biomarkers of disease
Oncometabolite-associated metabolic imaging
Oncometabolites accumulate to millimolar levels in the tissue. The specific accumulation of these metabolites could be exploited to detect tumour masses using multiple metabolic imaging modalities. One recent advance has been in the development of hyperpolarised magnetic resonance imaging (hpMRI)231,232. Administration of isotopically labelled 13C-glutamine in an IDH1/2-mutant chondrosarcoma xenograft mouse model with hpMRI enables visualisation of glutamine conversion to 2HG in real-time206. More strikingly, hpMRI was able to capture the suppression of 2HG accumulation in response to IDH inhibition206. A similar study performed recently in a ccRCC xenograft model utilised labelled 13C-pyruvate to visualise the metabolic response of the glycolytic flux to lactate in response to mTOR inhibitors233. Capitalising on the unique oncometabolite properties in RCC, hpMRI has multiple potential applications. It can facilitate the diagnoses of oncometabolite-associated RCC subtypes such as L2HG-associated RCC, whilst concomitantly conferring prognostic information as well i.e. L2HG is associated with poorer patient prognosis and tumour progression101. Furthermore, dynamic assessment of oncometabolite levels in this setting could be used as biomarkers of therapeutic efficacy, and by elucidating the IC50 for the oncometabolites and αKGDDs70,118,138,142 implicated in RCC, hpMRI could provide a means of monitoring progression or recurrence of the disease. In the wider context, given the basis of RCC as a metabolic disease process with extensive metabolic reprogramming associated with tumour progression, utilising hpMRI alongside selective tracers to identify malignant metabolic signatures, would facilitate more robust diagnoses of small and/or indeterminate renal lesions. The largely non-invasive and safe, non-ionising radioactive nature of hpMRI makes this a very attractive tool for development and translation into clinical practice. Similarly, other imaging modalities may also prove valuable in the diagnosis and management of oncometabolite-associated RCC, such as proton magnetic resonance spectroscopy (1H-MRS) and Positron Emission Tomography (PET) imaging. Successful detection using 1H-MRS of the oncometabolites succinate in patients with a variety of SDH-deficient tumours234,235, and 2HG in IDH-mutant gliomas236–238 has seen transition of 1H-MRS into clinical practice including in disease monitoring of IDH-mutant gliomas237. Although 1H-MRS has been explored in the setting of RCC patients to assess the general metabolic profile239, capitalising on the knowledge that the majority of oncometabolites have been implicated in RCC provides strong evidence for further investigation and may hold promise for patients with rare and aggressive RCC subtypes such as in SDH-deficiency. In addition, a recent pre-clinical study capitalising on glutamine reliance in several RCC subtypes, have demonstrated the ability to dynamically assess ccRCC metabolism in vivo using PET imaging with the radiotracer 18F-(2S,4R)4-fluoroglutamine (18F-FGln)207. In particular, this may also be a potential method of diagnosing and staging RCCs as well as stratifying patients that are likely to respond to glutaminase inhibition207.
Optimising surgical oncology
Oncometabolite-associated biomarkers may also prove indispensable for optimising surgical oncology. Intraoperative mass spectrometry of the oncometabolite 2HG has been used to guide brain tumour resections with promising results240. Identifying the presence of oncometabolites at tumour resection margins or “molecular margins” identifies the presence of tumour cells, thus providing a straightforward guide for the need for further resections240. More so, it provides the metabolite information within minutes and concurrently yields relevant information about genotype, tumour classification and potentially prognosis240. With multiple oncometabolites implicated in numerous RCC subtypes including in ccRCC99,101 and partial nephrectomies the gold-standard treatment for localised RCC241, utilising these methods may help optimise surgical margins in patients with oncometabolite-associated RCC undergoing partial nephrectomies. This may prove to be of great benefit as positive surgical margins have been demonstrated to correlate with tumour recurrence242. Furthermore, given the highly aggressive phenotypes of FH- and SDH-deficient RCCs in which a significant proportion present with bilateral disease, utilising intraoperative mass spectrometry concurrently may assist in meticulous tumour resections to help preserve renal function in these patients.
Cancer-specific oncometabolite-associated biomarkers
Finally, unique oncometabolite properties such as post-translational modifications of proteins and metabolic rewiring can be exploited for use as diagnostic or prognostic biomarkers. Capitalising on the ability of fumarate to induce protein succination (2SC), detection of the distinct 2SC protein modification signature on immunohistochemistry or cyst staining signifies fumarate accumulation and has been corroborated to be a robust diagnostic biomarker of FH deficiency, such as in HLRCC patients with ramifications for genetic testing 126,243. In addition, metabolomic analyses of urine from Fh1-deficient mice and growth media of FH-deficient cells revealed consistently elevated levels of argininosuccinate as a result of fumarate-induced reversal of ASL activity, raising its potential as a urinary biomarker for the early detection of FH-deficient renal cancer93. Although this biomarker awaits validation, the non-invasive nature of sampling, the specificity to FH-deficiency metabolism and the straightforward detection methods make this an exciting and attractive diagnostic biomarker.
Conclusion
Although in its infancy, the oncometabolite paradigm has been gathering momentum over the last decade with a firm movement away from the traditional view of metabolism as a simple by-product of genetic perturbations that occur in cancer. A growing body of evidence has substantiated the roles of a small group of seemingly innocuous metabolites that when aberrantly accumulated are transformed into oncometabolites that possess a plethora of capabilities that can contribute to tumorigenesis and tumour progression. Given that RCC is an established metabolic disease process, it is of no surprise that multiple oncometabolites are implicated in renal cancer. In general, oncometabolites in RCC exert significant effects on chromatin remodelling and epigenetic dysregulation leading to characteristic hypermethylated phenotypes, inducing an EMT phenotypic switch and the propagation of a pseudohypoxic signature contributing to the aggressive features of these RCC subtypes. Furthermore, by elucidating the roles of oncometabolites, it permits the exploitation of these molecules and their associated signalling pathways for multiple clinical applications such as the development of novel targets or as biomarkers of disease.
Key points.
Oncometabolites are aberrantly accumulated metabolites that possess pro-oncogenic capabilities that contribute to tumorigenesis via epigenetic dysregulation and can influence tumour progression through phenotypic switches such as EMT.
L-2HG, fumarate and succinate are bona fide RCC oncometabolites. Exploitation of these oncometabolites and their downstream signalling effects are attractive targets for novel therapies and as biomarkers of disease.
Chromatin remodelling via oncometabolites may recapitulate the effects of other epigenetic modifiers mutated in RCC thus converging on the same gene signature. Identification of these pathways involved will influence treatment strategy.
Elucidation of the exogenous factors that give rise to oncometabolite production such as hyperglycaemia may prove to be a synergistic strategy in reducing oncometabolite levels and their subsequent sequelae.
Acknowledgements
C.Y. is funded by the Wellcome Trust and The Urology Foundation. C.F. is supported by the Medical Research Council, grant MRC_MC_UU_12022/6.
Footnotes
Competing interests statement
The authors declare no competing interests.
Contributions
C.Y. and C.F. conceptualised the review. C.Y. wrote the manuscript. G.D.S: contributed to the development of the clinical focus. G.D.S. and C.F. contribute to the final editing of the review. All authors discussed the content of the review.
References
- 1.Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Bhatt JR, Finelli A. Landmarks in the diagnosis and treatment of renal cell carcinoma. Nature Reviews Urology. 2014;11:517–525. doi: 10.1038/nrurol.2014.194. [DOI] [PubMed] [Google Scholar]
- 3.Linehan WM, Walther MM, Zbar B. The genetic basis of cancer of the kidney. J Urol. 2003;170:2163–2172. doi: 10.1097/01.ju.0000096060.92397.ed. [DOI] [PubMed] [Google Scholar]
- 4.Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs—Part A: Renal, Penile, and Testicular Tumours. European Urology. 2016;70:93–105. doi: 10.1016/j.eururo.2016.02.029. [DOI] [PubMed] [Google Scholar]
- 5.Ricketts CJ, et al. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma. Cell Reports. 2018;0 doi: 10.1016/j.celrep.2018.03.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Young MD, et al. Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science. 2018;361:594–599. doi: 10.1126/science.aat1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mitchell TJ, et al. Timing the Landmark Events in the Evolution of Clear Cell Renal Cell Cancer: TRACERx Renal. Cell. 2018;173:611–623.e17. doi: 10.1016/j.cell.2018.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Turajlic S, et al. Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal. Cell. 2018;173:595–610.e11. doi: 10.1016/j.cell.2018.03.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Turajlic S, et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell. 2018;173:581–594.e12. doi: 10.1016/j.cell.2018.03.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499:43–49. doi: 10.1038/nature12222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zbar B, Brauch H, Talmadge C, Linehan M. Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma. Nature. 1987;327:721. doi: 10.1038/327721a0. [DOI] [PubMed] [Google Scholar]
- 12.Sato Y, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nature Genetics. 2013;45:860–867. doi: 10.1038/ng.2699. [DOI] [PubMed] [Google Scholar]
- 13.Hosen I, et al. TERT promoter mutations in clear cell renal cell carcinoma. International Journal of Cancer. 2015;136:2448–2452. doi: 10.1002/ijc.29279. [DOI] [PubMed] [Google Scholar]
- 14.Tang S-W, et al. MYC pathway is activated in clear cell renal cell carcinoma and essential for proliferation of clear cell renal cell carcinoma cells. Cancer Lett. 2009;273:35–43. doi: 10.1016/j.canlet.2008.07.038. [DOI] [PubMed] [Google Scholar]
- 15.Shroff EH, et al. MYC oncogene overexpression drives renal cell carcinoma in a mouse model through glutamine metabolism. Proc Natl Acad Sci U S A. 2015;112:6539–6544. doi: 10.1073/pnas.1507228112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tomlinson IPM, et al. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet. 2002;30:406–410. doi: 10.1038/ng849. [DOI] [PubMed] [Google Scholar]
- 17.Linehan WM, Ricketts CJ. The metabolic basis of kidney cancer. Semin Cancer Biol. 2013;23:46–55. doi: 10.1016/j.semcancer.2012.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Linehan WM, Srinivasan R, Schmidt LS. The genetic basis of kidney cancer: a metabolic disease. Nat Rev Urol. 2010;7:277–285. doi: 10.1038/nrurol.2010.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jaakkola P, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 2001;292:468–472. doi: 10.1126/science.1059796. [DOI] [PubMed] [Google Scholar]
- 20.Maxwell PH, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–275. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
- 21.Pescador N, et al. Hypoxia Promotes Glycogen Accumulation through Hypoxia Inducible Factor (HIF)-Mediated Induction of Glycogen Synthase 1. PLoS One. 2010;5 doi: 10.1371/journal.pone.0009644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Semenza GL. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest. 2013;123:3664–3671. doi: 10.1172/JCI67230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wise DR, et al. Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of -ketoglutarate to citrate to support cell growth and viability. Proceedings of the National Academy of Sciences. 2011;108:19611–19616. doi: 10.1073/pnas.1117773108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hanahan D, Weinberg RA. Hallmarks of Cancer: The Next Generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 25.Pavlova NN, Thompson CB. The Emerging Hallmarks of Cancer Metabolism. Cell Metabolism. 2016;23:27–47. doi: 10.1016/j.cmet.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Isaacs JS, et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: Novel role of fumarate in regulation of HIF stability. Cancer Cell. 2005;8:143–153. doi: 10.1016/j.ccr.2005.06.017. [DOI] [PubMed] [Google Scholar]
- 27.Pollard PJ, et al. Accumulation of Krebs cycle intermediates and over-expression of HIF1α in tumours which result from germline FH and SDH mutations. Hum Mol Genet. 2005;14:2231–2239. doi: 10.1093/hmg/ddi227. [DOI] [PubMed] [Google Scholar]
- 28.Sudarshan S, et al. Reduced expression of fumarate hydratase in clear cell renal cancer mediates HIF-2α accumulation and promotes migration and invasion. PLoS ONE. 2011;6:e21037. doi: 10.1371/journal.pone.0021037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Selak MA, et al. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell. 2005;7:77–85. doi: 10.1016/j.ccr.2004.11.022. [DOI] [PubMed] [Google Scholar]
- 30.Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG. TSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell. 2003;4:147–158. doi: 10.1016/s1535-6108(03)00187-9. [DOI] [PubMed] [Google Scholar]
- 31.Li B, et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature. 2014;513:251–255. doi: 10.1038/nature13557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell. 2012;21:297–308. doi: 10.1016/j.ccr.2012.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Goodacre R, Vaidyanathan S, Dunn WB, Harrigan GG, Kell DB. Metabolomics by numbers: acquiring and understanding global metabolite data. Trends in Biotechnology. 2004;22:245–252. doi: 10.1016/j.tibtech.2004.03.007. [DOI] [PubMed] [Google Scholar]
- 34.Horgan RP, Kenny LC. ‘Omic’ technologies: genomics, transcriptomics, proteomics and metabolomics: The Obstetrician & Gynaecologist. The Obstetrician & Gynaecologist. 2011;13:189–195. [Google Scholar]
- 35.Gouirand V, Guillaumond F, Vasseur S. Influence of the Tumor Microenvironment on Cancer Cells Metabolic Reprogramming. Front Oncol. 2018;8 doi: 10.3389/fonc.2018.00117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yang LV. Tumor Microenvironment and Metabolism. Int J Mol Sci. 2017;18 doi: 10.3390/ijms18122729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hakimi AA, et al. An Integrated Metabolic Atlas of Clear Cell Renal Cell Carcinoma. Cancer Cell. 2016;29:104–116. doi: 10.1016/j.ccell.2015.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wettersten HI, et al. Grade-Dependent Metabolic Reprogramming in Kidney Cancer Revealed by Combined Proteomics and Metabolomics Analysis. Cancer Res. 2015;75:2541–2552. doi: 10.1158/0008-5472.CAN-14-1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.van der Mijn JC, Panka DJ, Geissler AK, Verheul HenkM, Mier JW. Novel drugs that target the metabolic reprogramming in renal cell cancer. Cancer & Metabolism. 2016;4:14. doi: 10.1186/s40170-016-0154-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Astuti D, et al. Gene mutations in the succinate dehydrogenase subunit SDHB cause susceptibility to familial pheochromocytoma and to familial paraganglioma. Am J Hum Genet. 2001;69:49–54. doi: 10.1086/321282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Niemann S, Müller U. Mutations in SDHC cause autosomal dominant paraganglioma, type 3. Nat Genet. 2000;26:268–270. doi: 10.1038/81551. [DOI] [PubMed] [Google Scholar]
- 42.Warburg O. The Metabolism of Carcinoma Cells. The Journal of Cancer Research. 1925;9:148–163. [Google Scholar]
- 43.Warburg O. On the origin of cancer cells. Science. 1956;123:309–314. doi: 10.1126/science.123.3191.309. [DOI] [PubMed] [Google Scholar]
- 44.Racker E. Bioenergetics and the problem of tumor growth. Am Sci. 1972;60:56–63. [PubMed] [Google Scholar]
- 45.Liberti MV, Locasale JW. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem Sci. 2016;41:211–218. doi: 10.1016/j.tibs.2015.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Haber DA, Fearon ER. The promise of cancer genetics. The Lancet. 1998;351:SII1–SII8. doi: 10.1016/s0140-6736(98)90326-9. [DOI] [PubMed] [Google Scholar]
- 47.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;235:1492–1495. doi: 10.1126/science.3103217. [DOI] [PubMed] [Google Scholar]
- 48.Frezza C, Pollard PJ, Gottlieb E. Inborn and acquired metabolic defects in cancer. J Mol Med. 2011;89:213–220. doi: 10.1007/s00109-011-0728-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Baysal BE, et al. Mutations in SDHD, a Mitochondrial Complex II Gene, in Hereditary Paraganglioma. Science. 2000;287:848–851. doi: 10.1126/science.287.5454.848. [DOI] [PubMed] [Google Scholar]
- 50.Pollard P, et al. Evidence of increased microvessel density and activation of the hypoxia pathway in tumours from the hereditary leiomyomatosis and renal cell cancer syndrome. J Pathol. 2005;205:41–49. doi: 10.1002/path.1686. [DOI] [PubMed] [Google Scholar]
- 51.Cervera AM, Apostolova N, Crespo FL, Mata M, McCreath KJ. Cells silenced for SDHB expression display characteristic features of the tumor phenotype. Cancer Res. 2008;68:4058–4067. doi: 10.1158/0008-5472.CAN-07-5580. [DOI] [PubMed] [Google Scholar]
- 52.Dahia PLM, et al. A HIF1α Regulatory Loop Links Hypoxia and Mitochondrial Signals in Pheochromocytomas. PLoS Genet. 2005;1 doi: 10.1371/journal.pgen.0010008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.López-Jiménez E, et al. Research resource: Transcriptional profiling reveals different pseudohypoxic signatures in SDHB and VHL-related pheochromocytomas. Mol Endocrinol. 2010;24:2382–2391. doi: 10.1210/me.2010-0256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Vanharanta S, et al. Distinct expression profile in fumarate-hydratase-deficient uterine fibroids. Hum Mol Genet. 2006;15:97–103. doi: 10.1093/hmg/ddi431. [DOI] [PubMed] [Google Scholar]
- 55.Mohlin S, Wigerup C, Jögi A, Påhlman S. Hypoxia, pseudohypoxia and cellular differentiation. Exp Cell Res. 2017;356:192–196. doi: 10.1016/j.yexcr.2017.03.007. [DOI] [PubMed] [Google Scholar]
- 56.Wigerup C, Påhlman S, Bexell D. Therapeutic targeting of hypoxia and hypoxia-inducible factors in cancer. Pharmacology & Therapeutics. 2016;164:152–169. doi: 10.1016/j.pharmthera.2016.04.009. [DOI] [PubMed] [Google Scholar]
- 57.Dang L, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:739–744. doi: 10.1038/nature08617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Achouri Y, et al. Identification of a dehydrogenase acting on D-2-hydroxyglutarate. Biochem J. 2004;381:35–42. doi: 10.1042/BJ20031933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Collins RRJ, Patel K, Putnam WC, Kapur P, Rakheja D. Oncometabolites: A New Paradigm for Oncology, Metabolism, and the Clinical Laboratory. Clin Chem. 2017;63:1812–1820. doi: 10.1373/clinchem.2016.267666. [DOI] [PubMed] [Google Scholar]
- 60.Linster CL, Van Schaftingen E, Hanson AD. Metabolite damage and its repair or pre-emption. Nature Chemical Biology. 2013;9:72–80. doi: 10.1038/nchembio.1141. [DOI] [PubMed] [Google Scholar]
- 61.Struys EA. 2-Hydroxyglutarate is not a metabolite; d-2-hydroxyglutarate and l-2-hydroxyglutarate are! Proc Natl Acad Sci U S A. 2013;110:E4939. doi: 10.1073/pnas.1318777110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Moroni I, et al. L-2-hydroxyglutaric aciduria and brain malignant tumors: a predisposing condition? Neurology. 2004;62:1882–1884. doi: 10.1212/01.wnl.0000125335.21381.87. [DOI] [PubMed] [Google Scholar]
- 63.Aghili M, Zahedi F, Rafiee E. Hydroxyglutaric aciduria and malignant brain tumor: a case report and literature review. J Neurooncol. 2009;91:233–236. doi: 10.1007/s11060-008-9706-2. [DOI] [PubMed] [Google Scholar]
- 64.Rogers RE, et al. Wilms tumor in a child with L-2-hydroxyglutaric aciduria. Pediatr Dev Pathol. 2010;13:408–411. doi: 10.2350/09-12-0768-CR.1. [DOI] [PubMed] [Google Scholar]
- 65.Kranendijk M, et al. IDH2 Mutations in Patients with d-2-Hydroxyglutaric Aciduria. Science. 2010;330:336–336. doi: 10.1126/science.1192632. [DOI] [PubMed] [Google Scholar]
- 66.Morin A, Letouzé E, Gimenez-Roqueplo A-P, Favier J. Oncometabolites-driven tumorigenesis: From genetics to targeted therapy. International Journal of Cancer. 2014;135:2237–2248. doi: 10.1002/ijc.29080. [DOI] [PubMed] [Google Scholar]
- 67.Sciacovelli M, Frezza C. Oncometabolites: Unconventional triggers of oncogenic signalling cascades. Free Radic Biol Med. 2016;100:175–181. doi: 10.1016/j.freeradbiomed.2016.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Castro-Vega LJ, et al. Germline mutations in FH confer predisposition to malignant pheochromocytomas and paragangliomas. Hum Mol Genet. 2014;23:2440–2446. doi: 10.1093/hmg/ddt639. [DOI] [PubMed] [Google Scholar]
- 69.Kaelin WG. SDH5 mutations and familial paraganglioma: somewhere Warburg is smiling. Cancer Cell. 2009;16:180–182. doi: 10.1016/j.ccr.2009.08.013. [DOI] [PubMed] [Google Scholar]
- 70.Xu W, et al. Oncometabolite 2-Hydroxyglutarate Is a Competitive Inhibitor of α-Ketoglutarate-Dependent Dioxygenases. Cancer Cell. 2011;19:17–30. doi: 10.1016/j.ccr.2010.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yan H, et al. IDH1 and IDH2 Mutations in Gliomas. N Engl J Med. 2009;360:765–773. doi: 10.1056/NEJMoa0808710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Gross S, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010;207:339–344. doi: 10.1084/jem.20092506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ward PS, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010;17:225–234. doi: 10.1016/j.ccr.2010.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mukherjee PK, et al. Metabolomic analysis identifies differentially produced oral metabolites, including the oncometabolite 2-hydroxyglutarate, in patients with head and neck squamous cell carcinoma. BBA Clinical. 2017;7:8–15. doi: 10.1016/j.bbacli.2016.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Gottlieb E, Tomlinson IPM. Mitochondrial tumour suppressors: a genetic and biochemical update. Nature Reviews Cancer. 2005;5:857–866. doi: 10.1038/nrc1737. [DOI] [PubMed] [Google Scholar]
- 76.Knudson AG. Mutation and Cancer: Statistical Study of Retinoblastoma. PNAS. 1971;68:820–823. doi: 10.1073/pnas.68.4.820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Neumann HPH, et al. Distinct clinical features of paraganglioma syndromes associated with SDHB and SDHD gene mutations. JAMA. 2004;292:943–951. doi: 10.1001/jama.292.8.943. [DOI] [PubMed] [Google Scholar]
- 78.Ni Y, et al. Germline Mutations and Variants in the Succinate Dehydrogenase Genes in Cowden and Cowden-like Syndromes. The American Journal of Human Genetics. 2008;83:261–268. doi: 10.1016/j.ajhg.2008.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Malinoc A, et al. Biallelic inactivation of the SDHC gene in renal carcinoma associated with paraganglioma syndrome type 3. Endocr Relat Cancer. 2012;19:283–290. doi: 10.1530/ERC-11-0324. [DOI] [PubMed] [Google Scholar]
- 80.Clark GR, et al. Germline FH Mutations Presenting With Pheochromocytoma. J Clin Endocrinol Metab. 2014;99:E2046–E2050. doi: 10.1210/jc.2014-1659. [DOI] [PubMed] [Google Scholar]
- 81.Vranken JGV, Na U, Winge DR, Rutter J. Protein-mediated assembly of succinate dehydrogenase and its cofactors. Critical Reviews in Biochemistry and Molecular Biology. 2015;50:168–180. doi: 10.3109/10409238.2014.990556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lee C-H, et al. Persistent Severe Hyperlactatemia and Metabolic Derangement in Lethal SDHB-Mutated Metastatic Kidney Cancer: Clinical Challenges and Examples of Extreme Warburg Effect. Precision Oncology. 2017 doi: 10.1200/PO.16.00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.McEvoy CR, et al. SDH-deficient renal cell carcinoma associated with biallelic mutation in succinate dehydrogenase A: comprehensive genetic profiling and its relation to therapy response. npj Precision Oncology. 2018;2:9. doi: 10.1038/s41698-018-0053-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vanharanta S, et al. Early-Onset Renal Cell Carcinoma as a Novel Extraparaganglial Component of SDHB-Associated Heritable Paraganglioma. Am J Hum Genet. 2004;74:153–159. doi: 10.1086/381054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Alam NA, et al. Genetic and functional analyses of FH mutations in multiple cutaneous and uterine leiomyomatosis, hereditary leiomyomatosis and renal cancer, and fumarate hydratase deficiency. Hum Mol Genet. 2003;12:1241–1252. doi: 10.1093/hmg/ddg148. [DOI] [PubMed] [Google Scholar]
- 86.Lehtonen HJ, et al. Increased risk of cancer in patients with fumarate hydratase germline mutation. J Med Genet. 2006;43:523–526. doi: 10.1136/jmg.2005.036400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Linehan WM, Rouault TA. Molecular pathways: Fumarate hydratase-deficient kidney cancer--targeting the Warburg effect in cancer. Clin Cancer Res. 2013;19:3345–3352. doi: 10.1158/1078-0432.CCR-13-0304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kerrigan JF, Aleck KA, Tarby TJ, Bird CR, Heidenreich RA. Fumaric aciduria: Clinical and imaging features. Annals of Neurology. 2000;47:583–588. [PubMed] [Google Scholar]
- 89.Allegri G, et al. Fumaric aciduria: an overview and the first Brazilian case report. J Inherit Metab Dis. 2010;33:411–419. doi: 10.1007/s10545-010-9134-2. [DOI] [PubMed] [Google Scholar]
- 90.Loeffen J, Smeets R, Voit T, Hoffmann G, Smeitink J. Fumarase deficiency presenting with periventricular cysts. J Inherit Metab Dis. 2005;28:799–800. doi: 10.1007/s10545-005-0044-7. [DOI] [PubMed] [Google Scholar]
- 91.Lin A-P, et al. D2HGDH regulates alpha-ketoglutarate levels and dioxygenase function by modulating IDH2. Nat Commun. 2015;6:7768. doi: 10.1038/ncomms8768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Haigis KM, Cichowski K, Elledge SJ. Tissue-specificity in cancer: The rule, not the exception. Science. 2019;363:1150–1151. doi: 10.1126/science.aaw3472. [DOI] [PubMed] [Google Scholar]
- 93.Zheng L, et al. Reversed argininosuccinate lyase activity in fumarate hydratase-deficient cancer cells. Cancer Metab. 2013;1:12. doi: 10.1186/2049-3002-1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wallace DC. Mitochondria and cancer. Nature Reviews Cancer. 2012;12:685–698. doi: 10.1038/nrc3365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Parsons DW, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008;321:1807–1812. doi: 10.1126/science.1164382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Marcucci G, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28:2348–2355. doi: 10.1200/JCO.2009.27.3730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Gao J, et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci Signal. 2013;6:pl1. doi: 10.1126/scisignal.2004088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lee JH, et al. IDH1 R132C mutation is detected in clear cell hepatocellular carcinoma by pyrosequencing. World J Surg Oncol. 2017;15 doi: 10.1186/s12957-017-1144-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Shim E-H, et al. l-2-Hydroxyglutarate: An Epigenetic Modifier and Putative Oncometabolite in Renal Cancer. Cancer Discov. 2014;4:1290–1298. doi: 10.1158/2159-8290.CD-13-0696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Mitchell TJ, Rossi SH, Klatte T, Stewart GD. Genomics and clinical correlates of renal cell carcinoma. World J Urol. 2018;36:1899–1911. doi: 10.1007/s00345-018-2429-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Shelar S, et al. Biochemical and Epigenetic Insights into L-2-Hydroxyglutarate, a Potential Therapeutic Target in Renal Cancer. Clin Cancer Res. 2018 doi: 10.1158/1078-0432.CCR-18-1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Burr SP, et al. Mitochondrial Protein Lipoylation and the 2-Oxoglutarate Dehydrogenase Complex Controls HIF1α Stability in Aerobic Conditions. Cell Metabolism. 2016;24:740–752. doi: 10.1016/j.cmet.2016.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ni M, et al. Functional Assessment of Lipoyltransferase-1 Deficiency in Cells, Mice, and Humans. Cell Rep. 2019;27:1376–1386.e6. doi: 10.1016/j.celrep.2019.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Baker PR, et al. Variant non ketotic hyperglycinemia is caused by mutations in LIAS, BOLA3 and the novel gene GLRX5. Brain. 2014;137:366–379. doi: 10.1093/brain/awt328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Losman J-A, et al. (R)-2-Hydroxyglutarate Is Sufficient to Promote Leukemogenesis and Its Effects Are Reversible. Science. 2013;339:1621–1625. doi: 10.1126/science.1231677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Intlekofer AM, et al. Hypoxia Induces Production of L-2-Hydroxyglutarate. Cell Metab. 2015;22:304–311. doi: 10.1016/j.cmet.2015.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Oldham WM, Clish CB, Yang Y, Loscalzo J. Hypoxia-Mediated Increases in L-2-hydroxyglutarate Coordinate the Metabolic Response to Reductive Stress. Cell Metab. 2015;22:291–303. doi: 10.1016/j.cmet.2015.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Fan J, et al. Human Phosphoglycerate Dehydrogenase Produces the Oncometabolite d-2-Hydroxyglutarate. ACS Chem Biol. 2015;10:510–516. doi: 10.1021/cb500683c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Nadtochiy SM, et al. Metabolomic profiling of the heart during acute ischemic preconditioning reveals a role for SIRT1 in rapid cardioprotective metabolic adaptation. J Mol Cell Cardiol. 2015;88:64–72. doi: 10.1016/j.yjmcc.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Lu C, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474–478. doi: 10.1038/nature10860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Berger SL. The complex language of chromatin regulation during transcription. Nature. 2007;447:407–412. doi: 10.1038/nature05915. [DOI] [PubMed] [Google Scholar]
- 112.Nadtochiy SM, et al. Acidic pH Is a Metabolic Switch for 2-Hydroxyglutarate Generation and Signaling. J Biol Chem. 2016;291:20188–20197. doi: 10.1074/jbc.M116.738799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Rodenhizer D, et al. A 3D engineered tumour for spatial snap-shot analysis of cell metabolism and phenotype in hypoxic gradients. Nat Mater. 2016;15:227–234. doi: 10.1038/nmat4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Chouchani ET, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014;515:431–435. doi: 10.1038/nature13909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sapieha P, et al. The succinate receptor GPR91 in neurons has a major role in retinal angiogenesis. Nature Medicine. 2008;14:1067–1076. doi: 10.1038/nm.1873. [DOI] [PubMed] [Google Scholar]
- 116.Zhang J, et al. Accumulation of Succinate in Cardiac Ischemia Primarily Occurs via Canonical Krebs Cycle Activity. Cell Rep. 2018;23:2617–2628. doi: 10.1016/j.celrep.2018.04.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kohlhauer Matthias, et al. Metabolomic Profiling in Acute ST–Segment-Elevation Myocardial Infarction Identifies Succinate as an Early Marker of Human Ischemia–Reperfusion Injury. Journal of the American Heart Association. 7:e007546. doi: 10.1161/JAHA.117.007546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Xiao M, et al. Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 2012;26:1326–1338. doi: 10.1101/gad.191056.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.O’Flaherty L, et al. Dysregulation of hypoxia pathways in fumarate hydratase-deficient cells is independent of defective mitochondrial metabolism. Hum Mol Genet. 2010;19:3844–3851. doi: 10.1093/hmg/ddq305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Selak MA, Durán RV, Gottlieb E. Redox stress is not essential for the pseudo-hypoxic phenotype of succinate dehydrogenase deficient cells. Biochimica et Biophysica Acta (BBA) - Bioenergetics. 2006;1757:567–572. doi: 10.1016/j.bbabio.2006.05.015. [DOI] [PubMed] [Google Scholar]
- 121.Puisségur M-P, et al. miR-210 is overexpressed in late stages of lung cancer and mediates mitochondrial alterations associated with modulation of HIF-1 activity. Cell Death Differ. 2011;18:465–478. doi: 10.1038/cdd.2010.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Gao C-L, et al. Mitochondrial dysfunction is induced by high levels of glucose and free fatty acids in 3T3-L1 adipocytes. Mol Cell Endocrinol. 2010;320:25–33. doi: 10.1016/j.mce.2010.01.039. [DOI] [PubMed] [Google Scholar]
- 123.Blatnik M, Frizzell N, Thorpe SR, Baynes JW. Inactivation of Glyceraldehyde-3-Phosphate Dehydrogenase by Fumarate in Diabetes: Formation of S-(2-Succinyl)Cysteine, a Novel Chemical Modification of Protein and Possible Biomarker of Mitochondrial Stress. Diabetes. 2008;57:41–49. doi: 10.2337/db07-0838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Frizzell N, Thomas SA, Carson JA, Baynes JW. Mitochondrial stress causes increased succination of proteins in adipocytes in response to glucotoxicity. Biochemical Journal. 2012;445:247–254. doi: 10.1042/BJ20112142. [DOI] [PubMed] [Google Scholar]
- 125.Blatnik M, Thorpe SR, Baynes JW. Succination of Proteins by Fumarate. Annals of the New York Academy of Sciences. 2008;1126:272–275. doi: 10.1196/annals.1433.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Bardella Chiara, et al. Aberrant succination of proteins in fumarate hydratase-deficient mice and HLRCC patients is a robust biomarker of mutation status. The Journal of Pathology. 2011;225:4–11. doi: 10.1002/path.2932. [DOI] [PubMed] [Google Scholar]
- 127.Yang M, et al. The Succinated Proteome of FH-Mutant Tumours. Metabolites. 2014;4:640–654. doi: 10.3390/metabo4030640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Thomas SA, Storey KB, Baynes JW, Frizzell N. Tissue Distribution of S-(2-Succino)cysteine (2SC), a Biomarker of Mitochondrial Stress in Obesity and Diabetes. Obesity. 2012;20:263–269. doi: 10.1038/oby.2011.340. [DOI] [PubMed] [Google Scholar]
- 129.Guo Y, et al. Succinate and its G-protein-coupled receptor stimulates osteoclastogenesis. Nat Commun. 2017;8 doi: 10.1038/ncomms15621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mitchell T, Darley-Usmar V. Metabolic syndrome and mitochondrial dysfunction: insights from pre-clinical studies with a mitochondrially targeted antioxidant. Free Radic Biol Med. 2012;52:838–840. doi: 10.1016/j.freeradbiomed.2011.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhang G-M, Zhu Y, Ye D-W. Metabolic syndrome and renal cell carcinoma. World J Surg Oncol. 2014;12:236. doi: 10.1186/1477-7819-12-236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Häggström C, et al. Metabolic Factors Associated with Risk of Renal Cell Carcinoma. PLOS ONE. 2013;8:e57475. doi: 10.1371/journal.pone.0057475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tannahill GM, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496:238–242. doi: 10.1038/nature11986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Mills EL, et al. Succinate Dehydrogenase Supports Metabolic Repurposing of Mitochondria to Drive Inflammatory Macrophages. Cell. 2016;167:457–470.e13. doi: 10.1016/j.cell.2016.08.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kaelin WG. Cancer and Altered Metabolism: Potential Importance of Hypoxia-Inducible Factor and 2-Oxoglutarate-Dependent Dioxygenases. Cold Spring Harb Symp Quant Biol. 2011;76:335–345. doi: 10.1101/sqb.2011.76.010975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Loenarz C, Schofield CJ. Expanding chemical biology of 2-oxoglutarate oxygenases. Nature Chemical Biology. 2008;4:152–156. doi: 10.1038/nchembio0308-152. [DOI] [PubMed] [Google Scholar]
- 137.Letouzé E, et al. SDH Mutations Establish a Hypermethylator Phenotype in Paraganglioma. Cancer Cell. 2013;23:739–752. doi: 10.1016/j.ccr.2013.04.018. [DOI] [PubMed] [Google Scholar]
- 138.Laukka T, et al. Fumarate and Succinate Regulate Expression of Hypoxia-inducible Genes via TET Enzymes. J Biol Chem. 2016;291:4256–4265. doi: 10.1074/jbc.M115.688762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sciacovelli M, et al. Fumarate is an epigenetic modifier that elicits epithelial-to-mesenchymal transition. Nature. 2016;537:544–547. doi: 10.1038/nature19353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Guzy RD, Sharma B, Bell E, Chandel NS, Schumacker PT. Loss of the SdhB, but Not the SdhA, Subunit of Complex II Triggers Reactive Oxygen Species-Dependent Hypoxia-Inducible Factor Activation and Tumorigenesis. Mol Cell Biol. 2008;28:718–731. doi: 10.1128/MCB.01338-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Adam J, et al. Renal Cyst Formation in Fh1-Deficient Mice Is Independent of the Hif/Phd Pathway: Roles for Fumarate in KEAP1 Succination and Nrf2 Signaling. Cancer Cell. 2011;20:524–537. doi: 10.1016/j.ccr.2011.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Chowdhury R, et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO reports. 2011;12:463–469. doi: 10.1038/embor.2011.43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Koivunen P, et al. Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation. Nature. 2012;483:484–488. doi: 10.1038/nature10898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Tarhonskaya H, et al. Non-enzymatic chemistry enables 2-hydroxyglutarate-mediated activation of 2-oxoglutarate oxygenases. Nature Communications. 2014;5:3423. doi: 10.1038/ncomms4423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cedar H, Bergman Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet. 2009;10:295–304. doi: 10.1038/nrg2540. [DOI] [PubMed] [Google Scholar]
- 146.Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet. 2006;7:21–33. doi: 10.1038/nrg1748. [DOI] [PubMed] [Google Scholar]
- 147.Jones PA, Baylin SB. The Epigenomics of Cancer. Cell. 2007;128:683–692. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Figueroa ME, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18:553–567. doi: 10.1016/j.ccr.2010.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lu C, et al. Induction of sarcomas by mutant IDH2. Genes Dev. 2013;27:1986–1998. doi: 10.1101/gad.226753.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Turcan S, et al. IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature. 2012;483:479–483. doi: 10.1038/nature10866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Aspuria P-JP, et al. Succinate dehydrogenase inhibition leads to epithelial-mesenchymal transition and reprogrammed carbon metabolism. Cancer Metab. 2014;2 doi: 10.1186/2049-3002-2-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ahuja N, et al. Association between CpG island methylation and microsatellite instability in colorectal cancer. Cancer Res. 1997;57:3370–3374. [PubMed] [Google Scholar]
- 153.Issa J-P, et al. CpG island methylator phenotype in cancer. Nature Reviews Cancer. 2004;4:988–993. doi: 10.1038/nrc1507. [DOI] [PubMed] [Google Scholar]
- 154.Noushmehr H, et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell. 2010;17:510–522. doi: 10.1016/j.ccr.2010.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Cancer Genome Atlas Research Network et al. Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma. N Engl J Med. 2016;374:135–145. doi: 10.1056/NEJMoa1505917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Chan MMY, et al. Cascade Fumarate Hydratase mutation screening allows early detection of kidney tumour: a case report. BMC Med Genet. 2017;18 doi: 10.1186/s12881-017-0436-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Ooi A, et al. An Antioxidant Response Phenotype Shared between Hereditary and Sporadic Type 2 Papillary Renal Cell Carcinoma. Cancer Cell. 2011;20:511–523. doi: 10.1016/j.ccr.2011.08.024. [DOI] [PubMed] [Google Scholar]
- 158.Shanmugasundaram K, et al. The oncometabolite fumarate promotes pseudohypoxia through noncanonical activation of NF-κB signaling. J Biol Chem. 2014;289:24691–24699. doi: 10.1074/jbc.M114.568162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Schmidt LS, Linehan WM. Hereditary leiomyomatosis and renal cell carcinoma. Int J Nephrol Renovasc Dis. 2014;7:253–260. doi: 10.2147/IJNRD.S42097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sourbier C, et al. Targeting ABL1-Mediated Oxidative Stress Adaptation in Fumarate Hydratase-Deficient Cancer. Cancer Cell. 2014;26:840–850. doi: 10.1016/j.ccell.2014.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.van Uden P, Kenneth NS, Rocha S. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem J. 2008;412:477–484. doi: 10.1042/BJ20080476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Bratslavsky G, Sudarshan S, Neckers L, Linehan WM. Pseudohypoxic Pathways in Renal Cell Carcinoma. Clinical Cancer Research. 2007;13:4667–4671. doi: 10.1158/1078-0432.CCR-06-2510. [DOI] [PubMed] [Google Scholar]
- 163.Tong W-H, et al. The glycolytic shift in fumarate-hydratase-deficient kidney cancer lowers AMPK levels, increases anabolic propensities and lowers cellular iron levels. Cancer Cell. 2011;20:315–327. doi: 10.1016/j.ccr.2011.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Yang M, et al. The Succinated Proteome of FH-Mutant Tumours. Metabolites. 2014;4:640–654. doi: 10.3390/metabo4030640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Sullivan LB, et al. The Proto-oncometabolite Fumarate Binds Glutathione to Amplify ROS-Dependent Signaling. Molecular Cell. 2013;51:236–248. doi: 10.1016/j.molcel.2013.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Zheng L, et al. Fumarate induces redox-dependent senescence by modifying glutathione metabolism. Nature Communications. 2015;6:6001. doi: 10.1038/ncomms7001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.van Deursen JM. The role of senescent cells in ageing. Nature. 2014;509:439–446. doi: 10.1038/nature13193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Fourquet S, Guerois R, Biard D, Toledano MB. Activation of NRF2 by nitrosative agents and H2O2 involves KEAP1 disulfide formation. J Biol Chem. 2010;285:8463–8471. doi: 10.1074/jbc.M109.051714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Jin L, et al. Glutamate Dehydrogenase 1 Signals through Antioxidant Glutathione Peroxidase 1 to Regulate Redox Homeostasis and Tumor Growth. Cancer Cell. 2015;27:257–270. doi: 10.1016/j.ccell.2014.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mullen AR, et al. Reductive carboxylation supports growth in tumour cells with defective mitochondria. Nature. 2012;481:385–388. doi: 10.1038/nature10642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Hensley CT, Wasti AT, DeBerardinis RJ. Glutamine and cancer: cell biology, physiology, and clinical opportunities. J Clin Invest. 2013;123:3678–3684. doi: 10.1172/JCI69600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Xu Y, et al. Pathologic Oxidation of PTPN12 Underlies ABL1 Phosphorylation in Hereditary Leiomyomatosis and Renal Cell Carcinoma. Cancer Res. 2018;78:6539–6548. doi: 10.1158/0008-5472.CAN-18-0901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Shambaugh GE. Urea biosynthesis I. The urea cycle and relationships to the citric acid cycle. Am J Clin Nutr. 1977;30:2083–2087. doi: 10.1093/ajcn/30.12.2083. [DOI] [PubMed] [Google Scholar]
- 174.Frezza C, et al. Haem oxygenase is synthetically lethal with the tumour suppressor fumarate hydratase. Nature. 2011;477:225–228. doi: 10.1038/nature10363. [DOI] [PubMed] [Google Scholar]
- 175.Mu X, et al. Oncometabolite succinate promotes angiogenesis by upregulating VEGF expression through GPR91-mediated STAT3 and ERK activation. Oncotarget. 2017;8:13174–13185. doi: 10.18632/oncotarget.14485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Baumbach L, Leyssac PP, Skinner SL. Studies on renin release from isolated superfused glomeruli: effects of temperature, urea, ouabain and ethacrynic acid. J Physiol. 1976;258:243–256. doi: 10.1113/jphysiol.1976.sp011417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.He W, et al. Citric acid cycle intermediates as ligands for orphan G-protein-coupled receptors. Nature. 2004;429:188–193. doi: 10.1038/nature02488. [DOI] [PubMed] [Google Scholar]
- 178.Toma I, et al. Succinate receptor GPR91 provides a direct link between high glucose levels and renin release in murine and rabbit kidney. J Clin Invest. 2008;118:2526–2534. doi: 10.1172/JCI33293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Aguiar CJ, et al. Succinate causes pathological cardiomyocyte hypertrophy through GPR91 activation. Cell Communication and Signaling. 2014;12:78. doi: 10.1186/s12964-014-0078-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Correa PRAV, et al. Succinate is a paracrine signal for liver damage. J Hepatol. 2007;47:262–269. doi: 10.1016/j.jhep.2007.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Jiang S, Yan W. Succinate in the cancer-immune cycle. Cancer Lett. 2017;390:45–47. doi: 10.1016/j.canlet.2017.01.019. [DOI] [PubMed] [Google Scholar]
- 182.Mills E, O’Neill LAJ. Succinate: a metabolic signal in inflammation. Trends in Cell Biology. 2014;24:313–320. doi: 10.1016/j.tcb.2013.11.008. [DOI] [PubMed] [Google Scholar]
- 183.Zhang Z, et al. Identification of lysine succinylation as a new post-translational modification. Nat Chem Biol. 2011;7:58–63. doi: 10.1038/nchembio.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Li F, et al. NADP+-IDH Mutations Promote Hypersuccinylation that Impairs Mitochondria Respiration and Induces Apoptosis Resistance. Molecular Cell. 2015;60:661–675. doi: 10.1016/j.molcel.2015.10.017. [DOI] [PubMed] [Google Scholar]
- 185.Xie Z, et al. Lysine succinylation and lysine malonylation in histones. Mol Cell Proteomics. 2012;11:100–107. doi: 10.1074/mcp.M111.015875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Park J, et al. SIRT5-Mediated Lysine Desuccinylation Impacts Diverse Metabolic Pathways. Molecular Cell. 2013;50:919–930. doi: 10.1016/j.molcel.2013.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Frezza C. Mitochondrial metabolites: undercover signalling molecules. Interface Focus. 2017;7 doi: 10.1098/rsfs.2016.0100. 20160100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Cardaci S, et al. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat Cell Biol. 2015;17:1317–1326. doi: 10.1038/ncb3233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Lussey-Lepoutre C, et al. Loss of succinate dehydrogenase activity results in dependency on pyruvate carboxylation for cellular anabolism. Nat Commun. 2015;6:8784. doi: 10.1038/ncomms9784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Gill AJ, et al. Succinate Dehydrogenase (SDH)-deficient Renal Carcinoma: A Morphologically Distinct Entity. Am J Surg Pathol. 2014;38:1588–1602. doi: 10.1097/PAS.0000000000000292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Ricketts CJ, et al. Succinate dehydrogenase kidney cancer: an aggressive example of the Warburg effect in cancer. J Urol. 2012;188:2063–2071. doi: 10.1016/j.juro.2012.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Williamson SR, et al. Succinate dehydrogenase-deficient renal cell carcinoma: detailed characterization of 11 tumors defining a unique subtype of renal cell carcinoma. Modern Pathology. 2015;28:80–94. doi: 10.1038/modpathol.2014.86. [DOI] [PubMed] [Google Scholar]
- 193.Su R, et al. R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m6A/MYC/CEBPA Signaling. Cell. 2018;172:90–105.e23. doi: 10.1016/j.cell.2017.11.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Jia G, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7:885–887. doi: 10.1038/nchembio.687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Li Z, et al. FTO Plays an Oncogenic Role in Acute Myeloid Leukemia as a N6-Methyladenosine RNA Demethylase. Cancer Cell. 2017;31:127–141. doi: 10.1016/j.ccell.2016.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Fu X, et al. 2-Hydroxyglutarate Inhibits ATP Synthase and mTOR Signaling. Cell Metabolism. 2015;22:508–515. doi: 10.1016/j.cmet.2015.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Cancer Genome Atlas Research Network et al. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas. N Engl J Med. 2015;372:2481–2498. doi: 10.1056/NEJMoa1402121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Colvin H, et al. Oncometabolite D-2-Hydroxyglurate Directly Induces Epithelial-Mesenchymal Transition and is Associated with Distant Metastasis in Colorectal Cancer. Scientific Reports. 2016;6 doi: 10.1038/srep36289. 36289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Adam J, et al. A Role for Cytosolic Fumarate Hydratase in Urea Cycle Metabolism and Renal Neoplasia. Cell Rep. 2013;3:1440–1448. doi: 10.1016/j.celrep.2013.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Nassereddine S, Lap CJ, Haroun F, Tabbara I. The role of mutant IDH1 and IDH2 inhibitors in the treatment of acute myeloid leukemia. Ann Hematol. 2017;96:1983–1991. doi: 10.1007/s00277-017-3161-0. [DOI] [PubMed] [Google Scholar]
- 201.Wang F, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 2013;340:622–626. doi: 10.1126/science.1234769. [DOI] [PubMed] [Google Scholar]
- 202.Kernytsky A, et al. IDH2 mutation-induced histone and DNA hypermethylation is progressively reversed by small-molecule inhibition. Blood. 2015;125:296–303. doi: 10.1182/blood-2013-10-533604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Kats LM, et al. A pharmacogenomic approach validates AG-221 as an effective and on-target therapy in IDH2 mutant AML. Leukemia. 2017;31:1466–1470. doi: 10.1038/leu.2017.84. [DOI] [PubMed] [Google Scholar]
- 204.Birendra KC, DiNardo CD. Evidence for Clinical Differentiation and Differentiation Syndrome in Patients With Acute Myeloid Leukemia and IDH1 Mutations Treated With the Targeted Mutant IDH1 Inhibitor, AG-120. Clin Lymphoma Myeloma Leuk. 2016;16:460–465. doi: 10.1016/j.clml.2016.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Terunuma A, et al. MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. J Clin Invest. 2014;124:398–412. doi: 10.1172/JCI71180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Salamanca-Cardona L, et al. In Vivo Imaging of Glutamine Metabolism to the Oncometabolite 2-Hydroxyglutarate in IDH1/2 Mutant Tumors. Cell Metab. 2017;26:830–841.e3. doi: 10.1016/j.cmet.2017.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Abu Aboud O, et al. Glutamine Addiction in Kidney Cancer Suppresses Oxidative Stress and Can Be Exploited for Real-Time Imaging. Cancer Res. 2017;77:6746–6758. doi: 10.1158/0008-5472.CAN-17-0930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Gameiro PA, et al. In Vivo HIF-Mediated Reductive Carboxylation Is Regulated by Citrate Levels and Sensitizes VHL-Deficient Cells to Glutamine Deprivation. Cell Metab. 2013;17:372–385. doi: 10.1016/j.cmet.2013.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Raczka AM, Reynolds PA. Glutaminase inhibition in renal cell carcinoma therapy. Cancer Drug Resistance. 2019 doi: 10.20517/cdr.2018.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Song M, Kim S-H, Im CY, Hwang H-J. Recent Development of Small Molecule Glutaminase Inhibitors. Curr Top Med Chem. 2018;18:432–443. doi: 10.2174/1568026618666180525100830. [DOI] [PubMed] [Google Scholar]
- 211.Tannir NM, et al. CANTATA: A randomized phase 2 study of CB-839 in combination with cabozantinib vs. placebo with cabozantinib in patients with advanced/metastatic renal cell carcinoma. JCO. 2018;36:TPS4601–TPS4601. [Google Scholar]
- 212.Brière J-J, et al. Mitochondrial succinate is instrumental for HIF1alpha nuclear translocation in SDHA-mutant fibroblasts under normoxic conditions. Hum Mol Genet. 2005;14:3263–3269. doi: 10.1093/hmg/ddi359. [DOI] [PubMed] [Google Scholar]
- 213.MacKenzie ED, et al. Cell-permeating alpha-ketoglutarate derivatives alleviate pseudohypoxia in succinate dehydrogenase-deficient cells. Mol Cell Biol. 2007;27:3282–3289. doi: 10.1128/MCB.01927-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Matsumoto K, et al. Antitumor effects of 2-oxoglutarate through inhibition of angiogenesis in a murine tumor model. Cancer Science. 2009;100:1639–1647. doi: 10.1111/j.1349-7006.2009.01249.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Matsumoto K, et al. 2-Oxoglutarate downregulates expression of vascular endothelial growth factor and erythropoietin through decreasing hypoxia-inducible factor-1alpha and inhibits angiogenesis. J Cell Physiol. 2006;209:333–340. doi: 10.1002/jcp.20733. [DOI] [PubMed] [Google Scholar]
- 216.Tennant DA, et al. Reactivating HIF prolyl hydroxylases under hypoxia results in metabolic catastrophe and cell death. Oncogene. 2009;28:4009–4021. doi: 10.1038/onc.2009.250. [DOI] [PubMed] [Google Scholar]
- 217.Chen W, et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature. 2016;539:112–117. doi: 10.1038/nature19796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Cho H, et al. On-target efficacy of a HIF-2α antagonist in preclinical kidney cancer models. Nature. 2016;539:107–111. doi: 10.1038/nature19795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Courtney KD, et al. Phase I Dose-Escalation Trial of PT2385, a First-in-Class Hypoxia-Inducible Factor-2α Antagonist in Patients With Previously Treated Advanced Clear Cell Renal Cell Carcinoma. J Clin Oncol. 2018;36:867–874. doi: 10.1200/JCO.2017.74.2627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Fenaux P, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10:223–232. doi: 10.1016/S1470-2045(09)70003-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Gurion R, et al. 5-azacitidine prolongs overall survival in patients with myelodysplastic syndrome - a systematic review and meta-analysis. Haematologica. 2010;95:303–310. doi: 10.3324/haematol.2009.010611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Turcan S, et al. Efficient induction of differentiation and growth inhibition in IDH1 mutant glioma cells by the DNMT Inhibitor Decitabine. Oncotarget. 2013;4:1729–1736. doi: 10.18632/oncotarget.1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Yoo KH, Hennighausen L. EZH2 methyltransferase and H3K27 methylation in breast cancer. Int J Biol Sci. 2012;8:59–65. doi: 10.7150/ijbs.8.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Dalgliesh GL, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature. 2010;463:360–363. doi: 10.1038/nature08672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.van Haaften G, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet. 2009;41:521–523. doi: 10.1038/ng.349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Hu CY, et al. Kidney cancer is characterized by aberrant methylation of tissue-specific enhancers that are prognostic for overall survival. Clin Cancer Res. 2014;20:4349–4360. doi: 10.1158/1078-0432.CCR-14-0494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.To KKW, Zhan Z, Bates SE. Aberrant Promoter Methylation of the ABCG2 Gene in Renal Carcinoma. Mol Cell Biol. 2006;26:8572–8585. doi: 10.1128/MCB.00650-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Reu FJ, et al. Overcoming resistance to interferon-induced apoptosis of renal carcinoma and melanoma cells by DNA demethylation. J Clin Oncol. 2006;24:3771–3779. doi: 10.1200/JCO.2005.03.4074. [DOI] [PubMed] [Google Scholar]
- 229.Amato RJ, et al. MG98, a second-generation DNMT1 inhibitor, in the treatment of advanced renal cell carcinoma. Cancer Invest. 2012;30:415–421. doi: 10.3109/07357907.2012.675381. [DOI] [PubMed] [Google Scholar]
- 230.Vancura A, Bu P, Bhagwat M, Zeng J, Vancurova I. Metformin as an Anticancer Agent. Trends in Pharmacological Sciences. 2018;39:867–878. doi: 10.1016/j.tips.2018.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Brindle KM. Imaging metabolism with hyperpolarized (13)C-labeled cell substrates. J Am Chem Soc. 2015;137:6418–6427. doi: 10.1021/jacs.5b03300. [DOI] [PubMed] [Google Scholar]
- 232.Skinner JG, et al. Metabolic and Molecular Imaging with Hyperpolarised Tracers. Mol Imaging Biol. 2018;20:902–918. doi: 10.1007/s11307-018-1265-0. [DOI] [PubMed] [Google Scholar]
- 233.Dong Y, et al. Hyperpolarized MRI visualizes Warburg effects and predicts treatment response to mTOR inhibitors in patient-derived ccRCC xenograft models. Cancer Research. 2018 doi: 10.1158/0008-5472.CAN-18-2231. canres.2231.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Lussey-Lepoutre C, et al. In Vivo Detection of Succinate by Magnetic Resonance Spectroscopy as a Hallmark of SDHx Mutations in Paraganglioma. Clin Cancer Res. 2016;22:1120–1129. doi: 10.1158/1078-0432.CCR-15-1576. [DOI] [PubMed] [Google Scholar]
- 235.Casey RT, et al. Translating in vivo metabolomic analysis of succinate dehydrogenase deficient tumours into clinical utility. JCO Precis Oncol. 2018;2:1–12. doi: 10.1200/PO.17.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Bisdas S, et al. MR spectroscopy for in vivo assessment of the oncometabolite 2-hydroxyglutarate and its effects on cellular metabolism in human brain gliomas at 9.4T. J Magn Reson Imaging. 2016;44:823–833. doi: 10.1002/jmri.25221. [DOI] [PubMed] [Google Scholar]
- 237.de la Fuente MI, et al. Integration of 2-hydroxyglutarate-proton magnetic resonance spectroscopy into clinical practice for disease monitoring in isocitrate dehydrogenase-mutant glioma. Neuro Oncol. 2016;18:283–290. doi: 10.1093/neuonc/nov307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Kim H, Kim S, Lee HH, Heo H. In-Vivo Proton Magnetic Resonance Spectroscopy of 2-Hydroxyglutarate in Isocitrate Dehydrogenase-Mutated Gliomas: A Technical Review for Neuroradiologists. Korean J Radiol. 2016;17:620–632. doi: 10.3348/kjr.2016.17.5.620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Süllentrop F, Hahn J, Moka D. In Vitro and In Vivo 1H-MR Spectroscopic Examination of the Renal Cell Carcinoma. Int J Biomed Sci. 2012;8:94–108. [PMC free article] [PubMed] [Google Scholar]
- 240.Santagata S, et al. Intraoperative mass spectrometry mapping of an onco-metabolite to guide brain tumor surgery. Proc Natl Acad Sci USA. 2014;111:11121–11126. doi: 10.1073/pnas.1404724111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Ljungberg B, et al. EAU GUIDELINES ON RENAL CELL CARCINOMA. 21 [Google Scholar]
- 242.Petros FG, et al. Oncologic outcomes of patients with positive surgical margin after partial nephrectomy: a 25-year single institution experience. World J Urol. 2018:1–9. doi: 10.1007/s00345-018-2241-7. [DOI] [PubMed] [Google Scholar]
- 243.Maxwell PH. Seeing the smoking gun: a sensitive and specific method to visualize loss of the tumour suppressor, fumarate hydratase, in human tissues. J Pathol. 2011;225:1–3. doi: 10.1002/path.2950. [DOI] [PubMed] [Google Scholar]
- 244.Hino O, Kobayashi T, Mourning, Alfred G. Knudson: the two-hit hypothesis, tumor suppressor genes, and the tuberous sclerosis complex. Cancer Sci. 2017;108:5–11. doi: 10.1111/cas.13116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Yang M, Soga T, Pollard PJ, Adam J. The emerging role of fumarate as an oncometabolite. Front Oncol. 2012;2:85. doi: 10.3389/fonc.2012.00085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Gupta S, et al. Primary Renal Paragangliomas and Renal Neoplasia Associated with Pheochromocytoma/Paraganglioma: Analysis of von Hippel-Lindau (VHL), Succinate Dehydrogenase (SDHX) and Transmembrane Protein 127 (TMEM127) Endocr Pathol. 2017;28:253–268. doi: 10.1007/s12022-017-9489-0. [DOI] [PubMed] [Google Scholar]
- 247.Ozluk Y, et al. Renal carcinoma associated with a novel succinate dehydrogenase A mutation: a case report and review of literature of a rare subtype of renal carcinoma. Hum Pathol. 2015;46:1951–1955. doi: 10.1016/j.humpath.2015.07.027. [DOI] [PubMed] [Google Scholar]
- 248.Papathomas TG, et al. Non-pheochromocytoma (PCC)/paraganglioma (PGL) tumors in patients with succinate dehydrogenase-related PCC-PGL syndromes: a clinicopathological and molecular analysis. Eur J Endocrinol. 2014;170:1–12. doi: 10.1530/EJE-13-0623. [DOI] [PubMed] [Google Scholar]
- 249.Ricketts CJ, et al. Tumor risks and genotype-phenotype-proteotype analysis in 358 patients with germline mutations in SDHB and SDHD. Human Mutation. 2010;31:41–51. doi: 10.1002/humu.21136. [DOI] [PubMed] [Google Scholar]