Cancer cells are markedly different from normal cells with regards to how their metabolic pathways are used to fuel cellular growth and survival. Two basic metabolites that exemplify these differences through increased uptake and altered metabolic usage are glucose and glutamine. These molecules can be catabolized to manufacture many of the building blocks required for active cell growth and proliferation. The alterations in the metabolic pathways necessary to sustain this growth have been linked to therapeutic resistance, a trait that is correlated with poor patient outcomes. By targeting the metabolic pathways that import, catabolize, and synthesize essential cellular components, drug-resistant cancer cells can often be resensitized to anticancer treatments. The specificity and efficacy of agents directed at the unique aspects of cancer metabolism are expected to be high; and may, when in used in combination with more traditional therapeutics, present a pathway to surmount resistance within tumors that no longer respond to current forms of treatment. Cancer Res; 73(9); 2709–17. ©2013 AACR.

One of the distinguishing features of cancer is an escape from the typical regulatory constraints that prevent rapid and uncontrolled cellular division. Many cellular modifications occur within the bioenergetic and metabolic pathways, driven by the energetic needs of a dividing cell. One of the earliest examples of altered metabolism was shown in Otto Warburg's seminal work, wherein Warburg observed that cancerous tissues use oxygen-independent methods for breaking down glucose (1, 2). While the Warburg effect is not yet fully understood, it seems to consistently arise in a wide variety of cancers with diverse genetic backgrounds.

For cells integrated into the vasculature, nutrients are continually available and cancer cells absorb these nutrients at a much higher rate than normal cells (3–5). Thus, the metabolic changes exhibited in cancer are a result of factors beyond pure energy production and instead work to support unconstrained cellular growth by generating the cellular resources that a rapidly dividing cell requires (6, 7). Nucleotide triphosphates and amino acids are necessary building blocks for genome replication and protein synthesis that occur as cells grow. Furthermore, growth and division both require expansion of the cellular, nuclear, and mitochondrial membranes, which necessitates increased fatty acid synthesis to support the production of the lipid bilayers. In addition to basic building blocks, other components necessary to fuel cellular growth (ATP and NADPH) or protect the growing cell (NADH and NAD+) are regulated through components of the aerobic glycolysis machinery and must have their levels carefully managed to ensure that the basic building blocks can be efficiently converted into new biomass (8).

Glucose and glutamine can be broken down into the raw materials necessary to drive tumor growth (6, 9). The metabolic shift and increased glucose catabolism observed in cancer cells results in increased production of the biosynthetic precursors consumed as a cell grows. With upregulated glucose import, a fraction of the glucose taken in will be used in biosynthesis pathways upstream of pyruvate or be converted to pyruvate and enter the TCA cycle to provide precursors for fatty acid and amino acid synthesis, whereas all remaining glucose is converted to lactate and exported from the cell (4). Glutamine catabolism follows a similar pattern: Glutamine uptake is enhanced, and a portion of the glutamine is catabolized to maintain cellular levels of basic biosynthetic precursors, but the majority of cellular glutamine is converted into lactate or alanine (4, 10, 11). In contrast with the catabolism of glucose, conversion of glutamine to lactate passes through the TCA cycle, which aids in the maintenance of the pool of mitochondrial carbons and NADPH. The remaining glutamine provides carbon and nitrogen sources for amino acid synthesis and contributes nitrogen in purine and pyrimidine base synthesis (11). The uptake and breakdown of glucose and glutamine represent the central pathway for fueling tumor growth and methods that target these pathways can have significant effects on cellular growth and viability.

Although many aspects of tumor metabolism rely on the conserved metabolic pathways commonly used by rapidly proliferating cells, evidence indicates that there are facets of tumor metabolism that differ from nontumorigenic proliferating cells. Work investigating the M2 isoform of pyruvate kinase (PKM2) indicates that in cells with low pyruvate kinase activity, as it is in those expressing the PKM2 isoform, pyruvate can be formed from PEP through PGAM1 phosphorylation (12). This alternative pathway allows for a decoupling of ATP production from glucose metabolism, providing a way to prevent the allosteric inhibition of glycolysis pathway enzymes that occurs at high concentrations of ATP while still generating the downstream biomolecules necessary for cellular growth (12). Comparison of the gene expression profiles for oncogenic foci and induced pluripotent stem cells (iPSC) shows that whereas many pathways are modulated in a similar fashion, such as the upregulation of monosaccharide metabolic genes or downregulation of differentiation related genes, there are still distinct differences (13). The oncogenic foci upregulated a large variety of metabolic clusters—such as carbohydrate metabolism, sphingolipid metabolism, membrane lipid metabolism, and organophosphate metabolism—in addition to cellular stress and damage pathways, whereas the expression profile of the iPSC activated pluriopotency-related gene families (13). Exploiting the cancer-specific upregulation of stress pathways has already been shown to be an effective strategy for the development of highly selective anticancer treatments (14). Piperlongumine triggers cell death in cancerous cells but not in rapidly or slowly proliferating normal cells; murine tumor xenografts treated with piperlongumine displayed drastic response with no observed toxicity in normal mice (14). Similar specificity may be achievable through treatments targeting the dysregulated aspects of cancer metabolism.

Work linking dysregulated metabolism to drug resistance indicates that resistance may arise in part because altered metabolism can produce elevated ATP and NADPH levels (15, 16). Common mechanisms for generating chemoresistance are energy intensive and include enhanced DNA repair, misregulation of growth factor signaling, increased drug efflux, higher expression of antiapoptotic genes, or survival signaling pathways (17). Chemotherapies exert cytotoxic effects in part by inducing oxidative damage (18, 19), whereas increased glucose consumption gives rise to abundant NADPH, which contributes to therapeutic resistance (20, 21). The ability of deregulated metabolic pathways to protect cancer cells makes them attractive targets for improving chemotherapeutics (Supplementary Table S1).

Cancer cells depend on their altered metabolism to generate the necessary resources to fuel their growth, and the synthesis of these building blocks is dependent on the catabolism of glucose and glutamine. The uptake of glucose and glutamine is often increased through the upregulation of transporters, such as the GLUT family glucose transporters or the ASCT2 amino acid transporter. These are commonly targeted in an attempt to disrupt the dysregulated metabolism observed in cancer cells.

Glucose transport

The GLUT family proteins facilitate the uptake of extracellular glucose, and are often upregulated in cancer. Directly targeting these transporters is one way to attack tumor growth. In nude mice xenografts and cell culture, A549 lung cancer cells display lower GLUT1 expression and glucose uptake when treated with the irreversible GLUT1 inhibitor WZB117, resulting in an inhibition of growth that is synergistic with both cisplatin and paclitaxel (22). Exogenous ATP rescued cellular growth, indicating that the decrease in GLUT1 expression slowed tumor growth by limiting available ATP (22). In vitro WZB117 shows selectivity for cancer cells, whereas studies in animal models show relatively high efficacy and low toxicity, but treatment also induces transient hyperglycemia and a slight decrease in weight resulting from a loss of fat (22). Renal cell carcinomas lacking functional von Hippel–Lindau (VHL) tumor suppressor genes have been shown to be selectively targeted by an inhibitor of GLUT1, STF-31, whereas no effect has been observed on glycolysis in cells expressing the wild-type VHL gene (23). In vivo, STF-31–based inhibition of GLUT1 was able to impede glucose uptake and kill tumor cells with no toxicity for noncancerous cells (23). Although inhibition of glucose transporters varies in efficacy based on the tumor type when the strategy is used alone, it is effective when coupled with other drugs as part of a combinatorial strategy. The glucose transporters are critical for maintenance of multiple myeloma cells, but studies have shown that inhibition of glucose transporters can increase sensitivity to other chemotherapeutics. This can be observed in how the off-target inhibition of the GLUT4 transporter by the HIV protease inhibitor ritonavir increases doxorubicin sensitivity in multiple myeloma cell lines (24). Combinatorial treatment has also been an effective way to overcome chemoresistance. Hypoxia-inducible factor-1α (HIF-1α) enhances the expression of GLUT1 in hypoxic environments, a condition that frequently occurs in tumors. Hypoxia is well known to contribute to resistance to chemotherapeutics, yet inhibition of GLUT1 via phloretin overcomes this hypoxia-induced resistance when used in combination with daunorubicin (25). Whereas phloretin is a competitive inhibitor for glucose transporters, low concentrations have been shown to be effective on hypoxic cells—a property that could enhance the selectivity of this treatment (25). With data indicating that GLUT3 is upregulated in temozolomide-resistant cells, overcoming chemoresistance by targeting glucose transport coupled with treatment with standard chemotherapeutic agents may be an effective strategy against drug-resistant tumors (26).

Glutamine transport

Like glucose transport, upregulation of glutamine transporters is commonly observed in cancer cells (11). For many cancers, disruption of the constant uptake of glutamine can lead to outcomes more drastic than expected for pure amino acid starvation (27–30). In hepatocellular carcinoma, silencing of the glutamine transporter ASCT2 by inducible antisense RNA results in cellular death within 48 hours (31). The importance of glutamine transporters for cellular survival is further shown by the effect of selective estrogen receptor modulators (SERM), such as tamoxifen and raloxifene, on ER cell lines and cell lines derived from estrogen-insensitive tissues (32). Both tamoxifen and raloxifene can inhibit the glutamine transporter ASCT2 and lower cellular uptake of glutamine, resulting in growth inhibition and apoptosis in cell lines that are estrogen insensitive (32). Finally, inhibition of the SLC6A14 amino acid transporter by α-methyl-dl-tryptophan (α-MT) induces phenotypes associated with amino acid starvation, such as mTOR inhibition and activation of autophagy, but when coupled with an inhibitor of autophagy, the combined treatment triggers apoptosis (33). Although α-MT is only effective in the subset of cells expressing the SLC6A14 transporter, which is not expressed in all cancer types, it is highly selective for malignant cells, as SLC6A14 seems not to be expressed in nonmalignant cells (33). Both glucose and glutamine transporters play a key role in providing cancerous cells with resources, and inhibiting them results in slower cell growth or cellular death, even in cells that have been shown to be refractory to other chemotherapeutic treatments.

Targeting the resource import machinery that fuels dysregulated metabolism can be an effective strategy for cancer therapeutics. Once the basic resources have entered the cell, a wide variety of components within the biosynthetic cellular machinery are required to synthesize the biologic precursors that support continued cellular division. Later in this review we discuss selected elements within the biosynthetic pathways that have shown promise in sensitizing chemoresistant cells.

Glycolysis

Hexokinase catalyzes the first step in the catabolism of glucose, a key step in the glycolytic pathway. Two small-molecule inhibitors, 2-deoxy-d-glucose (2-DG) and 3-bromopyruvate (3-BP), target hexokinase and to disrupt the early steps of glycolysis and have shown promise as chemotherapeutic agents.

2-DG is a glucose analogue that cannot be fully processed by the glycolytic enzymes, resulting in an inhibition of glycolysis after it is phosphorylated by hexokinase. We have shown that 2-DG can effectively induce apoptosis in alveolar rhabdomyosarcoma, whereas hypoxic tumor cells are known to be sensitive to 2-DG treatment (34–36). The use of 2-DG as a solo agent is limited because of the high concentration necessary to compete with glycolytic pathway products, but it has been effective as part of a combinatorial treatment regimen. The chemotherapeutic agents doxorubicin and paclitaxel significantly slowed tumor growth and prolonged survival of mice with osteosarcoma and non–small cell lung xenografts, respectively, when mice were cotreated with 2-DG (36). We have also shown that trastuzumab-resistant breast cancer cells can be resensitized to trastuzumab through the addition of 2-DG (37). Finally, Bcl-2 family antagonists are only effective in certain cell varieties, but highly resistant leukemias are sensitized and undergo rapid apoptosis when Bcl-2 antagonist treatment is preceded by glycolysis inhibitor 2-DG (38).

A second small-molecule glycolysis inhibitor that has shown promise as a chemotherapeutic agent is the pyruvic acid derivative 3-BP, which also targets hexokinase. Supplying extra ATP exogenously can induce chemoresistance in colon cancer cells, whereas ATP-depleting factors such as 3-BP have been shown to help induce sensitivity to cell lines selected for resistance to chemotherapeutic agents such as oxaliplatin and 5-fluorouracil (16). Studies show that ATP-binding cassette (ABC) transporter-based resistance to daunorubicin or mitoxantrone is also highly dependent on cellular ATP levels (39). Daunorubicin or mitoxantrone treatment in vitro (human myeloma, myelogenous leukemia, and liver carcinoma cell lines) or in vivo (murine xenografts) showed suppressed resistance, increased cytotoxicity, and slowed subcutaneous tumor growth when coupled with 3-BP (39). Studies in animal models have shown a lack of in vivo toxicity and an inability to cross the blood brain–barrier, but as a competitive inhibitor, the concentrations necessary for therapeutic effect as a solo agent may be prohibitive. The success of glycolysis inhibitors such as 2-DG and 3-BP predicts that other inhibitors targeting components in these pathways will also have a high likelihood to produce drug-sensitizing effects.

PKM2 is preferentially expressed in many cancers and favors glycolysis (12, 40). Inhibitors of PKM2 have shown highly selective antitumor activity in animal and human xenografts tumor models (41–45). Recent work also shows that reducing PKM2 activity via CD44 knockdown results in lower cellular glutathione levels, decreased glucose uptake, lactate production, and ATP production while increasing reactive oxygen species (ROS) levels and also enhancing cisplatin sensitivity (20). As decreased activity of PKM2 helps overcome hypoxia-induced drug resistance, small-molecule compounds that modify PKM2 activity—such as TLN-232 (inhibitor) and 2-Oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamides (activator)—may also be effective in lowering resistance to current chemotherapeutic treatments (41, 46). The TLN-232 inhibitor has exhibited high selectivity even at low doses for cancer cells in vitro and in in vivo animal models, and although TLN-232 treatment does trigger a transient weight loss, it is well tolerated with no toxicity observed even at high doses (41, 46).

Glutamine catabolism

Glutaminase catalyzes the first step in the catabolism of glutamine, hydrolyzing it into glutamate and ammonia. Inhibition of glutaminase can deprive the cell of biosynthetic precursors needed for rapid growth, evidenced by the reduction of in vitro growth of glioblastoma cells containing an isocitrate dehydrogenase 1 mutation common to many gliomas and acute myelogenous leukemias observed when glutaminase activity was reduced through siRNA or BPTES treatment (30). Glutamine analogues often cause in vivo toxicity, lack specificity, or are generally ineffective, whereas inhibitors of glutaminase—such as BPTES—provide an opportunity to realize the benefits of glutamine deprivation in a way that minimizes the clinical downsides (30). The small-molecule inhibitor 968 prevents Rho GTPase-mediated cellular transformation by inhibiting glutaminase, an essential component in Rho GTPase-mediated transformation, yet elicited no effect on the growth or morphology of normal cells (47). In addition to blocking cellular transformation, 968 also showed the ability to inhibit the in vivo growth of lymphoma murine xenografts (47). Glutaminolysis is also integrated into the amino acid biosynthesis pathways by activating mTORC1 signaling and fueling cellular growth, which also could induce mTORC1-mediated resistance to common chemotherapeutic agents such as cisplatin in malignant AFP-producing gastric carcinomas (48, 49). Inhibition of mTORC1 with the dual phosphoinositide 3-kinase (PI3K)/mTOR inhibitor NVP-BEZ235 can resensitize Jurkat cells with induced vincristine resistance, clearly showing that reducing mTORC1 activity can lead to therapeutic sensitivity in resistant cells (50). NVP-BEZ235 exhibits minimal effects on normal cells in vitro but shows good oral bioavailability and low toxicity in vivo (50). Inhibiting glutaminolysis is predicted to have a similar sensitizing effect, through the reduction of mTOR signaling and depletion of the cellular pool of available resources. Glutamine is a critical resource for dysregulated cellular growth, and inhibition of glutamine breakdown by targeting glutaminase merits further exploration as an antitumorigenic and therapeutic potentiating strategy. The important role of mTOR and PI3K pathways in cancer has been covered exhaustively elsewhere (51–63).

Lactate production and export

Lactate dehydrogenase (LDH) is a key glycolytic enzyme, as it converts pyruvate into lactate and provides a pathway for nonoxidative disposal of the byproducts of glycolysis. Lactic acid is a significant byproduct of cancer cell metabolism and is often exported from the cell to remove excess carbon and maintain cellular NADPH stores (4). Inhibition of MCT1-based lactate export through treatment with α-cyano-4-OH-cinnamate (CHC) or lonidamine causes the tumor microenvironment to decrease in pH, with significant levels of cellular death occurring as the pH reaches 6.5, the typical microenvironmental tumor pH (64–66). CHC treatment has been shown to effectively slow the tumor growth rate in murine models of lung adenocarcinoma and colorectal adenocarcinoma xenografts, whereas Lewis lung carcinoma tumor-bearing mice treated with CHC showed greatly increased sensitivity to a single 6 Gy dose of radiation (65, 66). Mild side effects have been observed in patients after lonidamine treatment, with approximately half displaying myalgia; similarly, animal models indicate that the negative side effects of lonidamine are less likely to occur when it is given orally rather than intravenously. The LDH-A isoform has been shown to be upregulated in response to the overexpression of oncogenes such as HER2 and MYCN (64, 67). Targeting the conversion of pyruvate to lactate has also been shown to be a productive anticancer strategy. Glycolytic cell types have shown susceptibility to FX11-based LDH-A inhibition, exemplified by inhibition of tumor growth formation in P198 pancreatic xenografts, p493 β-lymphoma xenografts, and pancreatic LZ10.7 cells. Inhibition of LDH-A via siRNA or FX11 treatment results in increased mitochondrial function, evidenced by higher oxygen consumption and ROS generation, as well as an increase in necrosis and cellular death. When FX11 was combined with FK866, a nicotinamide phosphoribosyltransferase inhibitor, tumors underwent regression, whereas FK866 or FX11 alone merely slowed tumor growth (68, 69). Although solubility may require further derivatization of FX11, animals undergoing FX11 treatment did not lose weight or exhibit any symptoms of toxicity (68, 69). In addition, we have shown that the inhibition of LDH-A, either through siRNA or oxamate treatment, can overcome resistance to both paclitaxel and trastuzumab (37, 70).

Citric acid cycle

Metformin is a complex I inhibitor for the mitochondrial respiratory chain that is best known for its use as a treatment of diabetes. A study of the incidence rate of cancer within diabetics linked metformin use to lower incidence rates of cancer and better survival for diabetic patients who developed pancreatic cancer (71–74). In addition to the initial studies linking metformin to better clinical outcomes for diabetic patients with pancreatic cancer, newer work has investigated the ability of metformin to address therapeutic resistance in a variety of cancer types.

Cisplatin-resistant ovarian cancer cell lines showed a synergistic response to treatment with metformin and phenethyl isothiocyanate (PEITC), resulting in slower cancer cell growth and induction of cell death for ovarian cancer cells (75). Similarly, pancreatic cancer ductal adenocarcinoma cell lines (SUIT-2 and MIAPaCa-2) exhibiting varying degrees of gemcitabine resistance have been used to test the efficacy of gemcitabine treatment in combination with metformin or metformin and R1507 [a humanized anti–insulin-like growth factor-IR (IGF-IR) monoclonal antibody]. The combination of gemcitabine with metformin or metformin and R1507 induced greater inhibition of cellular proliferation and more apoptosis than solo treatment, whereas gemcitabine treatment alone increased the expression of genes indicative of chemoresistance, such as survivin and XIAP (76). Metformin has also been tested with respect to trastuzumab-resistant HER2-amplified breast cancers (77). Selectively targeting more chemoresistant cell populations, such as cancer stem cells (CSC), may provide a strategy to resensitize a tumor to chemotherapeutic compounds. Metformin has been reported to preferentially target CSC-like cells and is significantly more effective in CD44+/CD24/low JIMT-1 cells, a cell line derived from a pleural metastasis of a patient resistant to trastuzumab, compared with non-the CD44+/CD24/low JIMT-1 cell population, in vitro (77). Similarly, xenografts of JIMT1 were resistant to trastuzumab treatment, as no reduction in tumor growth was observed over a 7-week course of treatment. However, when treated with metformin alone or trastuzumab in combination with metformin, tumor volumes were reduced by 2- or 4-fold respectively, indicating that metformin treatment can help overcome trastuzumab resistance (77).

Pyruvate dehydrogenase kinase (PDK) phosphorylates and inhibits pyruvate dehydrogenase, preventing the formation and entry of acetyl-CoA into the citric acid cycle. Increased expression or activity of PDK isoforms may play a role in the metabolic shift away from mitochondrial oxidation and contributes to drug resistance, as siRNA-targeting PDK restores mitochondrial oxidation in cancerous, but not normal, cells (78, 79). The small-molecule dichloroacetate (DCA) also inhibits PDK-triggering apoptosis in cancerous cells but requires pharmacologically prohibitive concentrations to be effective, likely due to the decrease in expression of the cellular transporter responsible for DCA uptake in cancer cells (78, 80). Combination therapy with other agents, such as omeprazole or temozolomide, has shown to be more effective than solo treatments (81, 82). This shows that targeting components related to the citric acid cycle can be an effective strategy in the design of new therapeutics.

Fatty acid synthesis

ATP citrate lyase (ACLY) catalyzes the ATP-dependant conversion of citrate to acetyl-CoA and oxaloacetate, a key step in fatty acid biosynthesis. As expected, treatment with the ACLY inhibitor SB-204990 reduced cellular acetyl-CoA concentrations (83). In A549 xenografts, ACLY inhibition is cytostatic and induces differentiation, whereas for pancreatic xenografts SB-204990 treatment resulted in significant growth inhibition. ACLY inhibition results in more severe effects for more glycolytic cells, as ACLY inhibition hinders glucose-dependant lipid synthesis, and this is expected to make the effects of compounds that inhibit ACLY favor tumor cells over nonglycolytic and vegetative cells, as the latter are less dependant on ACLY activity (83). SB-204990 is orally bioavailable and shows no toxicity in animal models, but may have low in vivo stability (83).

Synthesis of the fatty acids used for lipogenesis requires precursors derived from the citric acid cycle, such as acetyl-CoA or malonyl-CoA. These precursors are then used by fatty acid synthase (FASN) enzyme complex to produce components necessary for lipogenesis, such as palmitate. FASN is upregulated in many cancers and correlates with poor patient outcomes, such as metastasis. FASN knockdown xenografts had smaller, slower-growing tumors with lower tumorigenic potential and less metastasis relative to FASN-expressing xenografts (84). FASN inhibitors, such as G28UCM, were able to effectively reduce the size of established xenografts by 20% to 90%. This decrease in size was accompanied by increased apoptotic cell death and decreased HER2 phosphorylation (85). Most FASN inhibitors (e.g., cerulenin, C75, EGCG) are unstable or impotent, or have significant side effects (anorexia and weight loss) in vivo, yet G28UCM-treated animals displayed no anorexia or weight loss, whereas G28UCM retained the ability to inhibit FASN activity and reduce tumor size (85). In vitro, G28UCM showed synergistic interactions with trastuzumab, lapatinib, erlotinib, and gefitinib; whereas trastuzumab (AU565TR) or lapatinib (AU565LR)–resistant cells retained FASN expression and sensitivity—providing an alternative method for overcoming drug resistance (85).

Earlier we have provided examples of compounds that can effectively resensitize cancer cells to therapeutics to which they had shown resistance. The focus of this article is on metabolic inhibitors that target either signaling transduction networks or key metabolic enzymes that are commonly dysregulated in cancer (Fig. 1). Although both strategies can be effective, directly targeting signal transduction networks often results in compensation through other closely related pathways, whereas the mechanisms allowing a cell to compensate for loss or inhibition of a rate-limiting or pathway-initiating enzymes in the essential metabolic pathways are more limited (86).

Figure 1.

A wide range of metabolic components supports the cancerous phenotype. Glucose and glutamine are metabolized into critical intermediates necessary for cellular growth, such as amino acids, nucleotides, and fatty acids. The metabolic inhibitors discussed in this review are shown in red near their target of inhibition. Disruption of these metabolic networks can reduce drug resistance and render cancer cells more susceptible to current therapeutic regimens.

Figure 1.

A wide range of metabolic components supports the cancerous phenotype. Glucose and glutamine are metabolized into critical intermediates necessary for cellular growth, such as amino acids, nucleotides, and fatty acids. The metabolic inhibitors discussed in this review are shown in red near their target of inhibition. Disruption of these metabolic networks can reduce drug resistance and render cancer cells more susceptible to current therapeutic regimens.

Close modal

Metabolic pathway components, such as those involved in glucose usage and amino acid biosynthesis, are likely to be good targets. Inhibition of PFKFB3 represses phosphofructokinase 1 (PFK1) by reducing fructose 2,6 bisphosphate (F2,6BP) synthesis. This leads to a decreased glycolytic characteristics evidenced by lower glucose uptake and less production of lactate, ATP, NAD+, and NADH (87). 3-(3-Pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO) is a small-molecule inhibitor of PFKFB3 that has shown selectivity for transformed cells in vitro and suppresses growth of Lewis lung carcinoma, MDA-MB-231 breast adenocarcinoma, and HL-60 promyelocytic leukemia xenografts (87). Given the observed effects on tumor growth in a variety of cancers as well as the metabolic changes 3PO treatment induces, PFK1 inhibition may produce chemosensitization in a similar manner to what happens when other glycolytic enzymes are inhibited.

Recent work also shows that for some forms of breast cancer, a key component of the serine biosynthesis pathway, phosphoglycerate dehydrogenase (PHGDH), is often amplified in cancer resulting in increased activity of the serine biosynthesis pathway (88). Knockdown of PHGDH in breast cancer cell lines exhibiting increased PHGDH expression reduces α-ketoglutarate (α-KG) levels, inhibits proliferation, and induces cellular death (88). Because increased PHGDH is observed in approximately 70% of ER breast cancers, it is reasonable to expect that treatments disrupting PHGDH function and enzymes related to serine biosynthesis will present an attractive strategy for overcoming resistance to chemotherapeutics (88, 89). Similarly, cellular stress pathways have been shown to be upregulated in cancerous cells (13). New research linking cellular stress to metabolism indicates that components within the ROS and heat shock response pathways might also be targets capable of producing highly selective synergistic responses with current chemotherapeutic agents (13, 14, 67, 90, 91).

The link between therapeutic sensitivity and dysregulated cancer cell metabolism is quickly being dissected in a variety of studies about a diverse array of cancers and therapeutic agents. It is becoming clear that this may provide a strategy for surmounting the drug resistance that endangers so many patients with cancer. We have described a variety of metabolic aspects ranging from resource uptake to breakdown or biosynthesis that can contribute to or be targeted to overcome therapeutic resistance to an assortment of agents. Although no unifying mechanism is apparent to show how inhibition of metabolic pathways can result in resensitization to chemotherapeutics, common themes do occur. When cancer cell metabolism is disrupted, the presumed resource shortfalls are often not fatal. Many of the metabolic inhibitors or gene knockdowns produced inhibitory effects on the cellular growth rate but do not induce cellular death. Rapid cellular growth seems to have a protective function, possibly due to the fact that rapidly dividing cells have access to sufficient resources to enact energy-intensive therapeutic resistance pathways on demand (15, 16). The extra resources available may allow for the synthesis and operation of drug efflux pumps and antiapoptotic genes such as surviving, and inhibition of the dysregulated metabolism often reduces ATP and other metabolic levels which may prevent efficient activation of resistance pathways (15, 76). Genomic instability frequently observed in cancers also ensures that a population of growing cells is more likely to have the genetic diversity to adapt to the selective pressures induced by chemotherapeutic treatments (92–95). By curtailing or preventing continued cellular growth, metabolic disruption might limit the ability of the tumor to adapt to the chemotherapeutic regimen. Finally, in many cases the disruption caused by inhibiting glycolytic metabolism results in increased cellular stress (often in the form of higher levels of ROS) or decreased protective response (such as NADPH levels), which may predispose cells toward apoptosis or limit the ability of the tumor to address further chemotherapeutic insult (14, 20, 68, 69). Many survival pathways have both cytoprotective and cytotoxic functions, and the increase in cellular stress induced by metabolic inhibition may precondition the stress response pathways toward apoptotic outcomes (14). As evidenced by the diverse array of cancers discussed earlier, this method of attacking therapeutic resistance in cancer is demonstratively effective in a wide variety of cancer types and merits continued investigation.

No potential conflicts of interest were disclosed.

Conception and design: E.B. Butler, Y. Zhao, M. Tan

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): E.B. Butler, M. Tan

Writing, review, and/or revision of the manuscript: E.B. Butler, C. Muñoz-Pinedo, J. Lu, M. Tan

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Zhao, M. Tan

Study supervision: M. Tan

This study was supported by Fondo de Investigaciones Sanitarias-ISCIII from Spain (PI10/0104 and RTICC RD06/0020 to C. Muñoz-Pinedo); NIH grants RO1CA137021 (J. Lu) and RO1CA149646 (M. Tan); The Vincent F. Kilborn, Jr. Cancer Research Foundation (M. Tan).

1.
Warburg
O
. 
On respiratory impairment in cancer cells
.
Science
1956
;
124
:
269
70
.
2.
Koppenol
WH
,
Bounds
PL
,
Dang
CV
. 
Otto Warburg's contributions to current concepts of cancer metabolism
.
Nat Rev Cancer
2011
;
11
:
325
37
.
3.
Kilburn
DG
,
Lilly
MD
,
Webb
FC
. 
The energetics of mammalian cell growth
.
J Cell Sci
1969
;
4
:
645
54
.
4.
Vander Heiden
MG
,
Cantley
LC
,
Thompson
CB
. 
Understanding the Warburg effect: the metabolic requirements of cell proliferation
.
Science
2009
;
324
:
1029
33
.
5.
Bollig-Fischer
A
,
Dziubinski
M
,
Boyer
A
,
Haddad
R
,
Giroux
CN
,
Ethier
SP
. 
HER-2 signaling, acquisition of growth factor independence, and regulation of biological networks associated with cell transformation
.
Cancer Res
2010
;
70
:
7862
73
.
6.
Bauer
DE
,
Harris
MH
,
Plas
DR
,
Lum
JJ
,
Hammerman
PS
,
Rathmell
JC
, et al
Cytokine stimulation of aerobic glycolysis in hematopoietic cells exceeds proliferative demand
.
FASEB J
2004
;
18
:
1303
5
.
7.
Fantin
VR
,
St-Pierre
J
,
Leder
P
. 
Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance
.
Cancer Cell
2006
;
9
:
425
34
.
8.
Levine
AJ
,
Puzio-Kuter
AM
. 
The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes
.
Science
2010
;
330
:
1340
4
.
9.
Matoba
S
,
Kang
JG
,
Patino
WD
,
Wragg
A
,
Boehm
M
,
Gavrilova
O
, et al
p53 regulates mitochondrial respiration
.
Science
2006
;
312
:
1650
3
.
10.
DeBerardinis
RJ
,
Mancuso
A
,
Daikhin
E
,
Nissim
I
,
Yudkoff
M
,
Wehrli
S
, et al
Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis
.
Proc Natl Acad Sci U S A
2007
;
104
:
19345
50
.
11.
Wise
DR
,
Thompson
CB
. 
Glutamine addiction: a new therapeutic target in cancer
.
Trends Biochem Sci
2010
;
35
:
427
33
.
12.
Vander Heiden
MG
,
Locasale
JW
,
Swanson
KD
,
Sharfi
H
,
Heffron
GJ
,
Amador-Noguez
D
, et al
Evidence for an alternative glycolytic pathway in rapidly proliferating cells
.
Science
2010
;
329
:
1492
9
.
13.
Riggs
JW
,
Barrilleaux
B
,
Varlakhanova
N
,
Bush
K
,
Chan
V
,
Knoepfler
P
. 
Induced pluripotency and oncogenic transformation are related processes
.
Stem Cells Dev
2013
;
22
:
37
50
.
14.
Raj
L
,
Ide
T
,
Gurkar
AU
,
Foley
M
,
Schenone
M
,
Li
X
, et al
Selective killing of cancer cells by a small molecule targeting the stress response to ROS
.
Nature
2011
;
475
:
231
4
.
15.
Fanciulli
M
,
Bruno
T
,
Giovannelli
A
,
Gentile
FP
,
Di Padova
M
,
Rubiu
O
, et al
Energy metabolism of human LoVo colon carcinoma cells: correlation to drug resistance and influence of lonidamine
.
Clin Cancer Res
2000
;
6
:
1590
7
.
16.
Zhou
Y
,
Tozzi
F
,
Chen
J
,
Fan
F
,
Xia
L
,
Wang
J
, et al
Intracellular ATP levels are a pivotal determinant of chemoresistance in colon cancer cells
.
Cancer Res
2012
;
72
:
304
14
.
17.
Longley
DB
,
Johnston
PG
. 
Molecular mechanisms of drug resistance
.
J Pathol
2005
;
205
:
275
92
.
18.
McDonald
JT
,
Kim
K
,
Norris
AJ
,
Vlashi
E
,
Phillips
TM
,
Lagadec
C
, et al
Ionizing radiation activates the Nrf2 antioxidant response
.
Cancer Res
2010
;
70
:
8886
95
.
19.
Coriat
R
,
Nicco
C
,
Chereau
C
,
Mir
O
,
Alexandre
J
,
Ropert
S
, et al
Sorafenib-induced hepatocellular carcinoma cell death depends on reactive oxygen species production in vitro and in vivo
.
Mol Cancer Ther
2012
;
11
:
2284
93
.
20.
Tamada
M
,
Nagano
O
,
Tateyama
S
,
Ohmura
M
,
Yae
T
,
Ishimoto
T
, et al
Modulation of glucose metabolism by CD44 contributes to antioxidant status and drug resistance in cancer cells
.
Cancer Res
2012
;
72
:
1438
48
.
21.
Tome
ME
,
Frye
JB
,
Coyle
DL
,
Jacobson
EL
,
Samulitis
BK
,
Dvorak
K
, et al
Lymphoma cells with increased anti-oxidant defenses acquire chemoresistance
.
Exp Ther Med
2012
;
3
:
845
52
.
22.
Liu
Y
,
Cao
Y
,
Zhang
W
,
Bergmeier
S
,
Qian
Y
,
Akbar
H
, et al
A small molecule inhibitor of glucose transporter 1 down-regulates glycolysis, induces cell cycle arrest, and inhibits cancer cell growth in vitro and in vivo
.
Mol Cancer Ther
2012
;
11
:
1672
82
.
23.
Chan
DA
,
Sutphin
PD
,
Nguyen
P
,
Turcotte
S
,
Lai
EW
,
Banh
A
, et al
Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality
.
Sci Transl Med
2011
;
3
:
94ra70
.
24.
McBrayer
SK
,
Cheng
JC
,
Singhal
S
,
Krett
NL
,
Rosen
ST
,
Shanmugam
M
. 
Multiple myeloma exhibits novel dependence on GLUT4, GLUT8, and GLUT11: implications for glucose transporter-directed therapy
.
Blood
2012
;
119
:
4686
97
.
25.
Cao
X
,
Fang
L
,
Gibbs
S
,
Huang
Y
,
Dai
Z
,
Wen
P
, et al
Glucose uptake inhibitor sensitizes cancer cells to daunorubicin and overcomes drug resistance in hypoxia
.
Cancer Chemother Pharmacol
2007
;
59
:
495
505
.
26.
Le Calve
B
,
Rynkowski
M
,
Le Mercier
M
,
Bruyere
C
,
Lonez
C
,
Gras
T
, et al
Long-term in vitro treatment of human glioblastoma cells with temozolomide increases resistance in vivo through up-regulation of GLUT transporter and aldo-keto reductase enzyme AKR1C expression
.
Neoplasia
2010
;
12
:
727
39
.
27.
Katt
WP
,
Ramachandran
S
,
Erickson
JW
,
Cerione
RA
. 
Dibenzophenanthridines as inhibitors of glutaminase C and cancer cell proliferation
.
Mol Cancer Ther
2012
;
11
:
1269
78
.
28.
Liu
W
,
Le
A
,
Hancock
C
,
Lane
AN
,
Dang
CV
,
Fan
TW
, et al
Reprogramming of proline and glutamine metabolism contributes to the proliferative and metabolic responses regulated by oncogenic transcription factor c-MYC
.
Proc Natl Acad Sci U S A
2012
;
109
:
8983
8
.
29.
Yuneva
MO
,
Fan
TW
,
Allen
TD
,
Higashi
RM
,
Ferraris
DV
,
Tsukamoto
T
, et al
The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type
.
Cell Metab
2012
;
15
:
157
70
.
30.
Seltzer
MJ
,
Bennett
BD
,
Joshi
AD
,
Gao
P
,
Thomas
AG
,
Ferraris
DV
, et al
Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1
.
Cancer Res
2010
;
70
:
8981
7
.
31.
Fuchs
BC
,
Finger
RE
,
Onan
MC
,
Bode
BP
. 
ASCT2 silencing regulates mammalian target-of-rapamycin growth and survival signaling in human hepatoma cells
.
Am J Physiol Cell Physiol
2007
;
293
:
C55
63
.
32.
Todorova
VK
,
Kaufmann
Y
,
Luo
S
,
Klimberg
VS
. 
Tamoxifen and raloxifene suppress the proliferation of estrogen receptor-negative cells through inhibition of glutamine uptake
.
Cancer Chemother Pharmacol
2011
;
67
:
285
91
.
33.
Karunakaran
S
,
Ramachandran
S
,
Coothankandaswamy
V
,
Elangovan
S
,
Babu
E
,
Periyasamy-Thandavan
S
, et al
SLC6A14 (ATB0,+) protein, a highly concentrative and broad specific amino acid transporter, is a novel and effective drug target for treatment of estrogen receptor-positive breast cancer
.
J Biol Chem
2011
;
286
:
31830
8
.
34.
Ramirez-Peinado
S
,
Alcazar-Limones
F
,
Lagares-Tena
L
,
El Mjiyad
N
,
Caro-Maldonado
A
,
Tirado
OM
, et al
2-Deoxyglucose induces Noxa-dependent apoptosis in alveolar rhabdomyosarcoma
.
Cancer Res
2011
;
71
:
6796
806
.
35.
Liu
H
,
Hu
YP
,
Savaraj
N
,
Priebe
W
,
Lampidis
TJ
. 
Hypersensitization of tumor cells to glycolytic inhibitors
.
Biochemistry
2001
;
40
:
5542
7
.
36.
Maschek
G
,
Savaraj
N
,
Priebe
W
,
Braunschweiger
P
,
Hamilton
K
,
Tidmarsh
GF
, et al
2-Deoxy-d-glucose increases the efficacy of adriamycin and paclitaxel in human osteosarcoma and non–small cell lung cancers in vivo
.
Cancer Res
2004
;
64
:
31
4
.
37.
Zhao
Y
,
Liu
H
,
Liu
Z
,
Ding
Y
,
Ledoux
SP
,
Wilson
GL
, et al
Overcoming trastuzumab resistance in breast cancer by targeting dysregulated glucose metabolism
.
Cancer Res
2011
;
71
:
4585
97
.
38.
Meynet
O
,
Beneteau
M
,
Jacquin
MA
,
Pradelli
LA
,
Cornille
A
,
Carles
M
, et al
Glycolysis inhibition targets Mcl-1 to restore sensitivity of lymphoma cells to ABT-737-induced apoptosis
.
Leukemia
2012
;
26
:
1145
7
.
39.
Nakano
A
,
Tsuji
D
,
Miki
H
,
Cui
Q
,
El Sayed
SM
,
Ikegame
A
, et al
Glycolysis inhibition inactivates ABC transporters to restore drug sensitivity in malignant cells
.
PLoS ONE
2011
;
6
:
e27222
.
40.
Christofk
HR
,
Vander Heiden
MG
,
Wu
N
,
Asara
JM
,
Cantley
LC
. 
Pyruvate kinase M2 is a phosphotyrosine-binding protein
.
Nature
2008
;
452
:
181
6
.
41.
Keri
G
,
Erchegyi
J
,
Horvath
A
,
Mezo
I
,
Idei
M
,
Vantus
T
, et al
A tumor-selective somatostatin analog (TT-232) with strong in vitro and in vivo antitumor activity
.
Proc Natl Acad Sci U S A
1996
;
93
:
12513
8
.
42.
Tejeda
M
,
Gaal
D
,
Barna
K
,
Csuka
O
,
Keri
G
. 
The antitumor activity of the somatostatin structural derivative (TT-232) on different human tumor xenografts
.
Anticancer Res
2003
;
23
:
4061
6
.
43.
Tejeda
M
,
Gaal
D
,
Csuka
O
,
Keri
G
. 
Growth inhibitory effect of the somatostatin structural derivative (TT-232) on leukemia models
.
Anticancer Res
2005
;
25
:
325
30
.
44.
Tejeda
M
,
Gaal
D
,
Hullan
L
,
Hegymegi-Barakonyi
B
,
Keri
G
. 
Evaluation of the antitumor efficacy of the somatostatin structural derivative TT-232 on different tumor models
.
Anticancer Res
2006
;
26
:
3477
83
.
45.
Tejeda
M
,
Gaal
D
,
Hullan
L
,
Schwab
R
,
Szokoloczi
O
,
Keri
G
. 
Antitumor activity of the somatostatin structural derivative (TT-232), against mouse and human melanoma tumor models
.
Anticancer Res
2007
;
27
:
4015
9
.
46.
Walsh
MJ
,
Brimacombe
KR
,
Veith
H
,
Bougie
JM
,
Daniel
T
,
Leister
W
, et al
2-Oxo-N-aryl-1,2,3,4-tetrahydroquinoline-6-sulfonamides as activators of the tumor cell specific M2 isoform of pyruvate kinase
.
Bioorg Med Chem Lett
2011
;
21
:
6322
7
.
47.
Wang
JB
,
Erickson
JW
,
Fuji
R
,
Ramachandran
S
,
Gao
P
,
Dinavahi
R
, et al
Targeting mitochondrial glutaminase activity inhibits oncogenic transformation
.
Cancer Cell
2010
;
18
:
207
19
.
48.
Duran
RV
,
Oppliger
W
,
Robitaille
AM
,
Heiserich
L
,
Skendaj
R
,
Gottlieb
E
, et al
Glutaminolysis activates Rag-mTORC1 signaling
.
Mol Cell
2012
;
47
:
349
58
.
49.
Kamata
S
,
Kishimoto
T
,
Kobayashi
S
,
Miyazaki
M
,
Ishikura
H
. 
Possible involvement of persistent activity of the mammalian target of rapamycin pathway in the cisplatin resistance of AFP-producing gastric cancer cells
.
Cancer Biol Ther
2007
;
6
:
1036
43
.
50.
Chiarini
F
,
Grimaldi
C
,
Ricci
F
,
Tazzari
PL
,
Evangelisti
C
,
Ognibene
A
, et al
Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 against T-cell acute lymphoblastic leukemia
.
Cancer Res
2010
;
70
:
8097
107
.
51.
Busaidy
NL
,
Farooki
A
,
Dowlati
A
,
Perentesis
JP
,
Dancey
JE
,
Doyle
LA
, et al
Management of metabolic effects associated with anticancer agents targeting the PI3K-Akt-mTOR pathway
.
J Clin Oncol
2012
;
30
:
2919
28
.
52.
Vilar
E
,
Perez-Garcia
J
,
Tabernero
J
. 
Pushing the envelope in the mTOR pathway: the second generation of inhibitors
.
Mol Cancer Ther
2011
;
10
:
395
403
.
53.
Zoncu
R
,
Efeyan
A
,
Sabatini
DM
. 
mTOR: from growth signal integration to cancer, diabetes and ageing
.
Nat Rev Mol Cell Biol
2011
;
12
:
21
35
.
54.
van Veelen
W
,
Korsse
SE
,
van de Laar
L
,
Peppelenbosch
MP
. 
The long and winding road to rational treatment of cancer associated with LKB1/AMPK/TSC/mTORC1 signaling
.
Oncogene
2011
;
30
:
2289
303
.
55.
Martelli
AM
,
Evangelisti
C
,
Chappell
W
,
Abrams
SL
,
Basecke
J
,
Stivala
F
, et al
Targeting the translational apparatus to improve leukemia therapy: roles of the PI3K/PTEN/Akt/mTOR pathway
.
Leukemia
2011
;
25
:
1064
79
.
56.
Willems
L
,
Tamburini
J
,
Chapuis
N
,
Lacombe
C
,
Mayeux
P
,
Bouscary
D
. 
PI3K and mTOR signaling pathways in cancer: new data on targeted therapies
.
Curr Oncol Rep
2012
;
14
:
129
38
.
57.
Sabatini
DM
. 
mTOR and cancer: insights into a complex relationship
.
Nat Rev Cancer
2006
;
6
:
729
34
.
58.
Mendoza
MC
,
Er
EE
,
Blenis
J
. 
The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation
.
Trends Biochem Sci
2011
;
36
:
320
8
.
59.
Dituri
F
,
Mazzocca
A
,
Giannelli
G
,
Antonaci
S
. 
PI3K functions in cancer progression, anticancer immunity and immune evasion by tumors
.
Clin Dev Immunol
2011
;
2011
:
947858
.
60.
Polak
R
,
Buitenhuis
M
. 
The PI3K/PKB signaling module as key regulator of hematopoiesis: implications for therapeutic strategies in leukemia
.
Blood
2012
;
119
:
911
23
.
61.
Aksamitiene
E
,
Kiyatkin
A
,
Kholodenko
BN
. 
Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: a fine balance
.
Biochem Soc Trans
2012
;
40
:
139
46
.
62.
Davies
MA
. 
The role of the PI3K-AKT pathway in melanoma
.
Cancer J
2012
;
18
:
142
7
.
63.
Sheppard
K
,
Kinross
KM
,
Solomon
B
,
Pearson
RB
,
Phillips
WA
. 
Targeting PI3 kinase/AKT/mTOR signaling in cancer
.
Crit Rev Oncog
2012
;
17
:
69
95
.
64.
Fang
J
,
Quinones
QJ
,
Holman
TL
,
Morowitz
MJ
,
Wang
Q
,
Zhao
H
, et al
The H+-linked monocarboxylate transporter (MCT1/SLC16A1): a potential therapeutic target for high-risk neuroblastoma
.
Mol Pharmacol
2006
;
70
:
2108
15
.
65.
Sonveaux
P
,
Copetti
T
,
De Saedeleer
CJ
,
Vegran
F
,
Verrax
J
,
Kennedy
KM
, et al
Targeting the lactate transporter MCT1 in endothelial cells inhibits lactate-induced HIF-1 activation and tumor angiogenesis
.
PLoS ONE
2012
;
7
:
e33418
.
66.
Sonveaux
P
,
Vegran
F
,
Schroeder
T
,
Wergin
MC
,
Verrax
J
,
Rabbani
ZN
, et al
Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice
.
J Clin Invest
2008
;
118
:
3930
42
.
67.
Zhao
YH
,
Zhou
M
,
Liu
H
,
Ding
Y
,
Khong
HT
,
Yu
D
, et al
Upregulation of lactate dehydrogenase A by ErbB2 through heat shock factor 1 promotes breast cancer cell glycolysis and growth
.
Oncogene
2009
;
28
:
3689
701
.
68.
Hasmann
M
,
Schemainda
I
. 
FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis
.
Cancer Res
2003
;
63
:
7436
42
.
69.
Le
A
,
Cooper
CR
,
Gouw
AM
,
Dinavahi
R
,
Maitra
A
,
Deck
LM
, et al
Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression
.
Proc Natl Acad Sci U S A
2010
;
107
:
2037
42
.
70.
Zhou
M
,
Zhao
Y
,
Ding
Y
,
Liu
H
,
Liu
Z
,
Fodstad
O
, et al
Warburg effect in chemosensitivity: targeting lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol
.
Mol Cancer
2010
;
9
:
33
.
71.
Li
D
,
Yeung
SC
,
Hassan
MM
,
Konopleva
M
,
Abbruzzese
JL
. 
Antidiabetic therapies affect risk of pancreatic cancer
.
Gastroenterology
2009
;
137
:
482
8
.
72.
Evans
JM
,
Donnelly
LA
,
Emslie-Smith
AM
,
Alessi
DR
,
Morris
AD
. 
Metformin and reduced risk of cancer in diabetic patients
.
BMJ
2005
;
330
:
1304
5
.
73.
Libby
G
,
Donnelly
LA
,
Donnan
PT
,
Alessi
DR
,
Morris
AD
,
Evans
JM
. 
New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes
.
Diabetes Care
2009
;
32
:
1620
5
.
74.
Sadeghi
N
,
Abbruzzese
JL
,
Yeung
SC
,
Hassan
M
,
Li
D
. 
Metformin use is associated with better survival of diabetic patients with pancreatic cancer
.
Clin Cancer Res
2012
;
18
:
2905
12
.
75.
Chan
DK
,
Miskimins
WK
. 
Metformin and phenethyl isothiocyanate combined treatment in vitro is cytotoxic to ovarian cancer cultures
.
J Ovarian Res
2012
;
5
:
19
.
76.
Kawanami
T
,
Takiguchi
S
,
Ikeda
N
,
Funakoshi
A
. 
A humanized anti-IGF-1R monoclonal antibody (R1507) and/or metformin enhance gemcitabine-induced apoptosis in pancreatic cancer cells
.
Oncol Rep
2012
;
27
:
867
72
.
77.
Cufi
S
,
Corominas-Faja
B
,
Vazquez-Martin
A
,
Oliveras-Ferraros
C
,
Dorca
J
,
Bosch-Barrera
J
, et al
Metformin-induced preferential killing of breast cancer initiating CD44+CD24-/low cells is sufficient to overcome primary resistance to trastuzumab in HER2+ human breast cancer xenografts
.
Oncotarget
2012
;
3
:
395
8
.
78.
Bonnet
S
,
Archer
SL
,
Allalunis-Turner
J
,
Haromy
A
,
Beaulieu
C
,
Thompson
R
, et al
A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth
.
Cancer Cell
2007
;
11
:
37
51
.
79.
Lu
CW
,
Lin
SC
,
Chien
CW
,
Lin
SC
,
Lee
CT
,
Lin
BW
, et al
Overexpression of pyruvate dehydrogenase kinase 3 increases drug resistance and early recurrence in colon cancer
.
Am J Pathol
2011
;
179
:
1405
14
.
80.
Babu
E
,
Ramachandran
S
,
CoothanKandaswamy
V
,
Elangovan
S
,
Prasad
PD
,
Ganapathy
V
, et al
Role of SLC5A8, a plasma membrane transporter and a tumor suppressor, in the antitumor activity of dichloroacetate
.
Oncogene
2011
;
30
:
4026
37
.
81.
Heshe
D
,
Hoogestraat
S
,
Brauckmann
C
,
Karst
U
,
Boos
J
,
Lanvers-Kaminsky
C
. 
Dichloroacetate metabolically targeted therapy defeats cytotoxicity of standard anticancer drugs
.
Cancer Chemother Pharmacol
2011
;
67
:
647
55
.
82.
Ishiguro
T
,
Ishiguro
M
,
Ishiguro
R
,
Iwai
S
. 
Cotreatment with dichloroacetate and omeprazole exhibits a synergistic antiproliferative effect on malignant tumors
.
Oncol Lett
2012
;
3
:
726
8
.
83.
Hatzivassiliou
G
,
Zhao
F
,
Bauer
DE
,
Andreadis
C
,
Shaw
AN
,
Dhanak
D
, et al
ATP citrate lyase inhibition can suppress tumor cell growth
.
Cancer Cell
2005
;
8
:
311
21
.
84.
Zaytseva
YY
,
Rychahou
PG
,
Gulhati
P
,
Elliott
VA
,
Mustain
WC
,
O'Connor
K
, et al
Inhibition of fatty acid synthase attenuates CD44-associated signaling and reduces metastasis in colorectal cancer
.
Cancer Res
2012
;
72
:
1504
17
.
85.
Puig
T
,
Aguilar
H
,
Cufi
S
,
Oliveras
G
,
Turrado
C
,
Ortega-Gutierrez
S
, et al
A novel inhibitor of fatty acid synthase shows activity against HER2+ breast cancer xenografts and is active in anti-HER2 drug-resistant cell lines
.
Breast Cancer Res
2011
;
13
:
R131
.
86.
McCormick
F
. 
Mutant onco-proteins as drug targets: successes, failures, and future prospects
.
Curr Opin Genet Dev
2011
;
21
:
29
33
.
87.
Clem
B
,
Telang
S
,
Clem
A
,
Yalcin
A
,
Meier
J
,
Simmons
A
, et al
Small-molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glycolytic flux and tumor growth
.
Mol Cancer Ther
2008
;
7
:
110
20
.
88.
Possemato
R
,
Marks
KM
,
Shaul
YD
,
Pacold
ME
,
Kim
D
,
Birsoy
K
, et al
Functional genomics reveal that the serine synthesis pathway is essential in breast cancer
.
Nature
2011
;
476
:
346
50
.
89.
Davis
ME
,
Zuckerman
JE
,
Choi
CH
,
Seligson
D
,
Tolcher
A
,
Alabi
CA
, et al
Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles
.
Nature
2010
;
464
:
1067
70
.
90.
Dai
C
,
Whitesell
L
,
Rogers
AB
,
Lindquist
S
. 
Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis
.
Cell
2007
;
130
:
1005
18
.
91.
Solimini
NL
,
Luo
J
,
Elledge
SJ
. 
Non-oncogene addiction and the stress phenotype of cancer cells
.
Cell
2007
;
130
:
986
8
.
92.
Greaves
M
,
Maley
CC
. 
Clonal evolution in cancer
.
Nature
2012
;
481
:
306
13
.
93.
Chen
KG
,
Sikic
BI
. 
Molecular pathways: regulation and therapeutic implications of multidrug resistance
.
Clin Cancer Res
2012
;
18
:
1863
9
.
94.
Hodgkinson
A
,
Chen
Y
,
Eyre-Walker
A
. 
The large-scale distribution of somatic mutations in cancer genomes
.
Hum Mutat
2012
;
33
:
136
43
.
95.
Hodgkinson
A
,
Eyre-Walker
A
. 
Variation in the mutation rate across mammalian genomes
.
Nat Rev Genet
2011
;
12
:
756
66
.