Tumor cells rewire metabolic pathways to adapt to their increased nutritional demands for energy, reducing equivalents, and cellular biosynthesis. Alternations in amino acid metabolism are 1 modality for satisfying those demands. Amino acids are not only components of proteins but also intermediate metabolites fueling multiple biosynthetic pathways. Amino acid–depletion therapies target amino acid uptake and catabolism using heterologous enzymes or recombinant or engineered human enzymes. Notably, such therapies have minimal effect on normal cells due to their lower demand for amino acids compared with tumor cells and their ability to synthesize the targeted amino acids under conditions of nutrient stress. Here, we review novel aspects of amino acid metabolism in hematologic malignancies and deprivation strategies, focusing on 4 key amino acids: arginine, asparagine, glutamine, and cysteine. We also present the roles of amino acid metabolism in the immunosuppressive tumor microenvironment and in drug resistance. This summary also offers an argument for the reclassification of amino acid–depleting enzymes as targeted therapeutic agents.

Amino acids perform critical metabolic functions. They are used for the synthesis of proteins, the formation of other low-molecular-weight compounds, and energy production by intermediate metabolites fueling other biosynthetic pathways. They have traditionally been classified as nutritionally essential (indispensable) or nonessential (dispensable) for animals and humans.1  Within the nutritionally essential amino acids, “essential” refers to those whose carbon skeletons cannot be synthesized de novo; “semiessential” refers to those that can be synthesized de novo but not in amounts necessary to maintain human health, necessitating dietary supplementation. Among 20 proteogenic amino acids, 9 are “essential” in humans by that definition: phenylalanine, valine (Val), threonine, tryptophan, methionine, leucine, isoleucine, lysine, and histidine (His). Six are “semiessential:” arginine (Arg), cysteine (Cys), glycine, glutamine (Gln), proline (Pro), and tyrosine. Nonessential amino acids are those that can be synthesized de novo in adequate amounts: aspartate (aspartic acid [Asp]), asparagine (Asn), glutamate (glutamic acid; Glu), alanine, and serine (Ser).2 

For the maintenance of hematopoietic stem cells (HSCs), the essential amino acid Val is indispensable, and depletion of Val decreases the number of native HSCs.3  On the other hand, the altered metabolic wiring of leukemic cells enables survival in the metabolically stressful leukemic microenvironment.4  That reprogrammed leukemic cell metabolome plays an important role in tumor growth and chemoresistance. Targeting such metabolic vulnerability, specifically the differences in amino acid dependency between HSCs and leukemia stem cells (LSCs), may unveil novel, targeted leukemia therapies with acceptable therapeutic windows.

The stress responses triggered by amino acid loss are observed in both LSCs and HSCs. Even normal homeostatic processes generate low-level stress and activation of the integrated stress response (ISR) pathway. Activation of the ISR results in eIF2α phosphorylation-dependent upregulation of activating transcription factor 4 (ATF4) that facilitates survival of both HSCs and LSCs.5  However, leukemia cells tend to require a greater supply of nutrients for growth and proliferation than normal cells,6,7  and the leukemia environment is characterized by lower levels of glucose, Gln, and oxygen compared with the normal hematopoietic microenvironment.8 

The process of rewiring metabolism to meet increased demands for energy, reducing equivalents, and cellular building blocks often results in cancer cells developing auxotrophy: the inability to synthesize sufficient quantities of amino acids due to dependence on the extracellular pool of amino acids. In contrast, normal cells exhibit lower demand for amino acids.9  That difference establishes a metabolic vulnerability in malignant cells, providing a rationale for amino acid–depletion therapy. Given that leukemia is a heterogeneous clonal disorder originating from LSCs,10  it is important to note that genomically heterogeneous LSC subpopulations propagate in the bone marrow (BM) microenvironment and likely exhibit different metabolic patterns and responses to amino acid–targeting therapies.11 

Fueled in part by advances in protein-engineering technology and improved understanding of the metabolic differences between cancer and normal cells, the full potential of amino acid–depletion therapy is starting to be realized9 ; for instance, l-asparaginase has produced excellent results in the treatment of acute lymphoblastic leukemia (ALL) and is a component of the standard of care.12 

This review describes novel aspects of amino acid metabolism in leukemia cells and deprivation strategies using heterologous or engineered enzymes that have been successfully used in the treatment of hematopoietic malignancies. We focus on 4 key amino acids, Arg, Asn, Gln, and Cys (Figure 1), along with the roles of amino acid metabolism in the immunosuppressive tumor microenvironment and in drug resistance. We also present the case for reclassifying amino acid–targeting therapies as targeted therapies against LSCs, which require specific amino acid–metabolic pathways for survival.

Figure 1.

Therapeutic targeting of amino acids in tumors and the tumor microenvironment. The most common amino acid–depletion strategies include depletion of Asn with asparaginase (ASNase), Arg with arginase (ARGase), or Arg deiminase (ADI), Glu with glutaminase (GLS) inhibitors, and Cys/cysteine (CSSC) with cysteinase. Stromal cells or adipocytes produce Asn and Glu, supporting tumor metabolism. ARGase secretion by tumor cells (which depletes extracellular Arg) and/or tumor consumption of Gln may deprive immune cells of these nutrients, and metabolic inhibitors of ARGase or GLNase reverse immune suppression. GSH, glutathione; NK, natural killer; ROS, reactive oxygen species; TCA, the citric acid.

Figure 1.

Therapeutic targeting of amino acids in tumors and the tumor microenvironment. The most common amino acid–depletion strategies include depletion of Asn with asparaginase (ASNase), Arg with arginase (ARGase), or Arg deiminase (ADI), Glu with glutaminase (GLS) inhibitors, and Cys/cysteine (CSSC) with cysteinase. Stromal cells or adipocytes produce Asn and Glu, supporting tumor metabolism. ARGase secretion by tumor cells (which depletes extracellular Arg) and/or tumor consumption of Gln may deprive immune cells of these nutrients, and metabolic inhibitors of ARGase or GLNase reverse immune suppression. GSH, glutathione; NK, natural killer; ROS, reactive oxygen species; TCA, the citric acid.

Close modal

Arg has been classified as a semiessential amino acid because healthy individuals can synthesize Arg from other amino acids, such as Gln, Glu, and Pro.13  In pathologic conditions, however, the human body becomes dependent on Arg intake.14  In addition, Arg serves as an intermediate or a substrate in many biological pathways, such as the urea cycle, TCA cycle, and thymotropic activities.15,16  Arg is also the major precursor for synthesis of cancer-associated compounds such as polyamines, nitric oxide (NO), and NO synthetase itself.16,,-19  Previous studies have shown that low NO concentrations promote cancer cell proliferation and angiogenesis, whereas high NO concentrations lead to DNA damage and cell death.20,,,-24 

As cancer cells require nutrients to sustain growth, exhausting endogenous sources of Arg is a potential therapeutic intervention. That strategy was successfully demonstrated to be effective in previous studies using polyethylene glycol–conjugated (PEGylated) forms of ADI (ADI-PEG20) or ARGase (PEG-ARGase), the 2 critical enzymes of the Arg metabolism/urea cycle.23,,-26 Figure 2 illustrates the Arg-metabolism cycle. Several studies have shown the beneficial effects of Arg degradation to citrulline by ADI in preventing proliferation of tumor cells such as melanoma, lung cancer, renal cell carcinoma, and hepatocellular carcinoma.27,28  In those experiments, researchers noted similarities among the malignancies, including deficiencies in OTC or arginosuccinate synthetase-1 (ASS1) (http://www.proteinatlas.org). Treatment with ADI depleted the endogenous Arg reservoirs of tumor cells because the cells failed to replace Arg by hydrolyzing citrulline.29 

Figure 2.

Targeting Arg metabolism. Arg can be synthesized from other amino acids via urea cycle; however, tumor cells lacking the urea cycle enzyme arginosuccinate synthetase (ASS) rely on exogenous supply of Arg. The endogenous sources of Arg can be used as a potential therapeutic intervention in treating cancer. ADI catalyzes Arg degradation to citrulline and ammonium. ARGase generates ornithine as an intermediate, and ornithine carbomoyltransferase (OTC) catalyzes the conversion of ornithine to citrulline. Upon condensation of Asp and citrulline by the enzymatic activity of ASS, arginosuccinate intermediate is generated followed by the reformation of Arg.

Figure 2.

Targeting Arg metabolism. Arg can be synthesized from other amino acids via urea cycle; however, tumor cells lacking the urea cycle enzyme arginosuccinate synthetase (ASS) rely on exogenous supply of Arg. The endogenous sources of Arg can be used as a potential therapeutic intervention in treating cancer. ADI catalyzes Arg degradation to citrulline and ammonium. ARGase generates ornithine as an intermediate, and ornithine carbomoyltransferase (OTC) catalyzes the conversion of ornithine to citrulline. Upon condensation of Asp and citrulline by the enzymatic activity of ASS, arginosuccinate intermediate is generated followed by the reformation of Arg.

Close modal

One limitation in using ADI for cancer therapy is that ADI provokes an immune response in human cells because the enzyme is originally isolated from prokaryotes, specifically Mycoplasma arginini. Furthermore, the short half-life of the native drug (4 hours) places a restraining factor on its efficacy in cancer treatment.24,25  ADI-PEG20 was developed to diminish the drug immunogenicity and to extend the half-life of the enzyme to 6 days.25 

Several studies have tested the effect of ADI-PEG20 monotherapy in melanoma and advanced hepatocellular carcinoma; despite the transient drop of Arg levels observed, tumor progression was sustained and a neutralizing antibody was detected in the patients’ plasma.30,,-33  Additional resistance mechanisms that resulted from ADI-PEG20 treatment were autophagy and overexpression of the ASS1 protein. Notably, blocking autophagy with chloroquine in combination with ADI increased the rate of apoptosis in sarcoma cells in vitro, and treating sarcoma samples expressing low levels of ASS1 with ADI-PEG20 reduced cancer proliferation.34,35 

Many attempts have been made to combat the resistance that develops to ADI-PEG20 monotherapy. For instance, Beddowes et al proposed that ADI-PEG20 would act synergistically in combination with cisplatin and pemetrexed in thoracic cancer. Their results showed a sustainable drop in Arg level and a significant decrease in the production of neutralizing antibodies.36 

A recent study of the efficacy of ADI-PEG20 in combination with cytarabine in treating acute myeloid leukemia (AML) in mouse models and in vitro demonstrated that the combination significantly reduced AML cell percentage and plasma Arg level.37  ADI-PEG20–resistant AMLs showed higher expression of ASS1 and argininosuccinate lyase than ADI-PEG20–sensitive AMLs, whereas ADI-PEG20–sensitive AMLs showed greater Arg uptake from the extracellular milieu via high-affinity cationic amino acid transporter-1 (CAT1), consistent with the Arg-auxotrophic nature of ADI-PEG20–sensitive AMLs. These findings lend support to the suggestion that ADI-PEG20–resistant AMLs enhance the endogenous production of Arg through the ASS1 pathway.37 

The other Arg-depleting strategy is ARGase, a catabolic enzyme that converts Arg to ornithine in the urea cycle.38  Mammals express 2 isoforms of this enzyme, ARGase I and ARGase II, which have 60% similarity in their amino acid sequence. ARGase I is a cytosolic enzyme in the human liver and ARGase II is a mitochondrial enzyme in other tissues that is thought to play no role in the urea cycle.39,40  Like ADI, recombinant ARGase has been pegylated to extend its half-life from a few hours to 3 to 4 days. PEG-ARGase treatment diminished the proliferation of OTC- and ASS1-deficient cancer cells, unlike ADI-PEG20, which inhibited only tumors lacking ASS1 expression.38  Mussai et al41  demonstrated the antiproliferative effect of a pegylated human recombinant ARGase (BCT-100) on AML blasts. The mechanism involved the ARG-auxotrophic nature of AML blasts (which constitutively express the Arg transporters, CAT1, and CAT2B) that mediate extracellular Arg uptake. BCT-100 treatment led to rapid depletion of extracellular and intracellular Arg reservoirs, resulting in decreased proliferation of AML blasts.

Depleting Arg in tumor cells by using PEG-ARGase in combination with other drugs has been a focus of various studies; as 1 example, a recent study indicated the antitumor potential of PEG–ARGase I and 5-fluorouracil in hepatocellular carcinoma in vivo.38  Additionally, it has been reported that PEG–ARGase I in combination with cytarabine is an effective approach in treating AML and ALL both in vivo and in vitro.41,42  Despite the great efficacy of that combination, both studies failed to achieve durable responses and resulted in the development of resistance.41,42  PEG–ARGase I treatment decreased the expression of cyclin D3, a fundamental protein in T-cell ALL tumor proliferation, and consequently induced apoptosis in T-cell ALL cell lines isolated from mouse models.42  Furthermore, pegylated human recombinant ARGase impeded T-cell proliferation in ALL in vivo.42 

Asn is classified as a nonessential amino acid, meaning that it can be synthesized by human cells.43  In glycoprotein synthesis, the amide nitrogen of Asn residues in proteins provides a site of glycosylation to the carbohydrate chain to form the N-linkage.44  Additionally, Asn shows exchange-factor capacity to coordinate the uptake of extracellular Arg, His, and Ser, and it can modulate ammonia metabolism.44,45  The capacity of Asn to suppress apoptosis is evident by its ability to negatively modulate endoplasmic reticulum stress and translation-dependent apoptosis.46 

Asn starvation is a potential strategy to induce apoptosis in cancer cells that express a low level or are deficient in Asn synthetase (ASNS).47  Examples of the tumors lacking ASNS are leukemia and lymphoma, which depend on external sources of Asn.48,49  Expression of ASNS has been found to be positively correlated with poor prognosis in selected human tumors.46  Evidence of Asn auxotrophy was first discovered through the antitumor effect of asparaginase (ASNase) in guinea pig serum on lymphoma cells.50,-52  Later, ASNase was isolated from Escherichia coli to treat lymphoblastic leukemia.53 

ASNase monotherapy has been an effective approach to treat ALL and some cases of lymphomas, and the combination of vincristine and prednisone with ASNase has resulted in a remission rate of >90% in children with ALL.54,55  In breast cancer, a synergistic interaction between ASNase and doxorubicin in induction of apoptosis has been reported.56  ASNase and temozolomide triggered a synergistic antitumor effect in the growth of subcutaneous gliomas in vivo.57 

Although the majority of pediatric ALL patients achieve plasma levels of ASNase that deplete Asn effectively,58  some patients exhibit apparent pharmacokinetic/pharmacodynamic inadequacies.59,-61  Resistance to ASNase is currently thought to follow 1 of 4 patterns, as follows. First, normal and leukemic lymphoblasts may eliminate the ASNase enzyme through immune responses triggered by recognition of ASNase as a foreign protein.61  This results in the secretion of a neutralizing anti-ASNase antibody and blocks the drug activity.62,63  Second, leukemia blasts that rapidly upregulate ASNS protein expression may develop resistance to ASNase.64,65  Third, enhanced production of Gln synthetase and increased activity of Gln transporters (SLC1A5, SLC3A2, SLC38A2, SLC38A3, SLC38A4, SLC38A5) in cancer cells is believed to contribute to ASNase resistance, which can be resolved by inhibiting Gln synthetase.66  Fourth, BM stromal cells, an important element in the cancer microenvironment, release Asn when leukemia blasts are treated with ASNase.67  Previous studies showed that BM mesenchymal stem cells (MSCs) mediate resistance to most chemotherapy reagents. Because MSCs show sensitivity to cytoskeleton-targeting drugs,68  administration of vincristine prior to ASNase treatment has been proposed to impede the MSC-mediated resistance of ALL blasts.69 

It has been further reported that ASNase treatment depletes Gln, which can induce apoptosis because many cancer cells require large quantities of Gln to maintain TCA cycle anaplerosis and support cell survival.70  Interestingly, Asn supplementation was reported to be necessary and sufficient to suppress Gln withdrawal–induced apoptosis without restoring the levels of other nonessential amino acids or TCA-cycle intermediates.

Gln depletion by the glutaminase (GLNase) activity of ASNase has been associated clinically with toxicities such as acute pancreatitis, thrombotic disorders, and immunosuppressive effects71,,-74  (Figures 3 and 4). One approach that has been suggested to solve this issue is to isolate or engineer Glu with GLS-free ASNase.75,76  Nevertheless, ASNase purified from bacteria triggers hypersensitivity or anaphylaxis.76,77 E coli expresses 2 isoforms of ASNase: ASNase I and ASNase II. The former is a cytosolic protein that has a low affinity to Asn, and the latter is a periplasmic enzyme that is commonly used in cancer therapy because of its high affinity to Asn.78  In a comparison of PEG–E coli ASNase and Erwinia chrysanthemi ([Erw] now Dickeya dadantii) ASNase (ERWase), E coli ASNase exhibited lower immunogenicity than Erw ASNase, whereas Erw ASNase resulted in less toxicity and is a preferred treatment of patients who are hypersensitive to E coli ASNase.79,-81 

Figure 3.

Therapeutic targeting of Asn and Gln. Asn is catalyzed by ASNS, the primary mediator of resistance to ASNase. ASNase hydrolyzes ASN into Asp and ammonia. Asn starvation by ASNase is a potential strategy of apoptosis induction in leukemia and lymphoma cells lacking ASNS and depend on the exogenous supply of Asn. The secondary activity present in ASNase is an GLS activity, which drives hydrolysis of Gln to Glu and ammonia. AMP, adenosine monophosphate; ATP, adenosine triphosphate; PPi, proton pump inhibitor.

Figure 3.

Therapeutic targeting of Asn and Gln. Asn is catalyzed by ASNS, the primary mediator of resistance to ASNase. ASNase hydrolyzes ASN into Asp and ammonia. Asn starvation by ASNase is a potential strategy of apoptosis induction in leukemia and lymphoma cells lacking ASNS and depend on the exogenous supply of Asn. The secondary activity present in ASNase is an GLS activity, which drives hydrolysis of Gln to Glu and ammonia. AMP, adenosine monophosphate; ATP, adenosine triphosphate; PPi, proton pump inhibitor.

Close modal
Figure 4.

Gln metabolism in AML in tumor microenvironment. (A) Tumor cells avidly consume Gln to fuel biosynthesis and proliferation. GLS converts Gln to Glu, which is a necessary step for Gln to enter the TCA cycle. BCAT1, a cytosolic aminotransferase for BCAAs (leucine, isoleucine, and Val), is a critical enzyme that initiates the catabolism of BCAAs and results in the production of Glu. (B) Consumption of Gln by tumor cells deprives immune cells of this critical nutrient. GLS inhibitor blocks Gln consumption by tumor cells and elevates Glu in the tumor microenvironment supporting immune cell function.

Figure 4.

Gln metabolism in AML in tumor microenvironment. (A) Tumor cells avidly consume Gln to fuel biosynthesis and proliferation. GLS converts Gln to Glu, which is a necessary step for Gln to enter the TCA cycle. BCAT1, a cytosolic aminotransferase for BCAAs (leucine, isoleucine, and Val), is a critical enzyme that initiates the catabolism of BCAAs and results in the production of Glu. (B) Consumption of Gln by tumor cells deprives immune cells of this critical nutrient. GLS inhibitor blocks Gln consumption by tumor cells and elevates Glu in the tumor microenvironment supporting immune cell function.

Close modal

Recently, the ASNase and GLNase activities of 4 different isoforms of ASNase were investigated: E coli ASNase, ERWase, and native and mutant isoforms of Helicobacter pylori ASNase (HpA), the native wild-type (wt) HpA and the mutant developed mutant (dm) HpA.82  All 4 ASNase isoforms had similar ASNase activities, but ERWase exhibited the highest and dm HpA showed the lowest GLS activities. Of the 4, ERWase exhibited the highest cytotoxic activity on ALL samples, indicating an important role for the GLS activity of ASNase in the drug’s overall anticancer activity. Asn-dependent ALL cells showed no difference in sensitivity to any of the 4 tested isoforms.82 

Notably, a direct correlation between obesity and the incidence of relapse in children with ALL has been reported. Parmentier et al82  and Ehsanipour et al83  reported the counteractive effects of BM adipocytes on leukemia cells after ASNase treatment. They demonstrated that the Gln-synthetase production by BM adipocytes was upregulated upon exposure to chemotherapy, although there was no change in plasma levels of Gln or Asn. Similarly, in xenograft models, adipocytes inhibited the cytotoxic activity of ASNase by releasing Gln into the leukemia microenvironment. Both native and pegylated forms of ASNase prolonged survival in nonobese mice with ALL; however, pegylated ASNase failed to prolong survival in their obese counterparts. Furthermore, although adipocytes secrete both Asn and Gln, Asn did not protect ALL cells from the cytotoxic effects of ASNase, and blocking Gln synthetase in adipocytes enhanced ASNase activity.83 

Given the pivotal role of Gln in tumor cell survival, direct targeting of this pathway has been pursued through pharmacological GLS inhibitors. In preclinical studies, GLS inhibitor CB-839 decreased mitochondrial oxidative phosphorylation and inhibited AML cell growth.84  CB-839 further impaired antioxidant GSH production, which resulted in accumulation of mitochondrial ROS (mitoROS) followed by apoptosis induction in AML cells without cytotoxic effects on normal hematopoietic progenitors.85,86  The phase 1 clinical trial of CB-839 in relapsed/refractory AML demonstrated excellent tolerance but lack of objective responses when used as a single agent.87  A recent preliminary report of the ongoing combination trial of CB-839 and 5-azacitidine demonstrated rapid reduction of BM blasts in high-risk myelodysplastic syndrome patients.88  In preclinical studies with AML cell lines and patient samples harboring isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) mutations, CB-839 decreased production of oncometabolite 2-hydroxyglutarate, inducing differentiation,89  and the synergy between GLS inhibition and B-cell lymphoma 2 (BCL-2) inhibitor venetoclax has been reported.85  Gallipoli et al86  demonstrated the role of Gln on the metabolic adaptations supporting both mitochondrial function and cellular redox metabolism as resistance mechanisms of FLT3 internal tandem duplication (FLT3-ITD)–mutated AML to direct FLT3 kinase inhibitors. FLT3 inhibitors quizartinib has been shown to inhibit glucose uptake and glycolysis, rendering cells dependent on Gln metabolism; in turn, the combination of CB-839 with AC220 synergistically depleted GSH, induced mitoROS, and caused apoptotic cell death in both in vitro and in vivo mouse models.86,90  These studies indicate that GLS inhibition in combination with other targeted agents including BCL-2 and FLT3 tyrosine kinase inhibitors may offer therapeutic strategies targeting metabolism and survival in AML. Currently, the combination of CB839 and hypomethylating agent azacitidine is ongoing in high-risk myelodysplastic syndrome patients.88 

Recently, the oncogenic function of branched-chain aminotransferase 1 (BCAT1), a cytosolic aminotransferase for branched-chain amino acids (BCAAs; leucine, isoleucine, and Val), was described as a critical enzyme for α-ketoglutarate (αKG) homeostasis related to leukemia metabolism.91,92  BCAT1 transfers α-amino groups from BCAAs to αKG, which results in the production of Glu (Figure 4). BCAT1 has been shown to be enriched and activated in LSCs in human AML and blast-crisis chronic myeloid leukemia, linking BCAA metabolism to epigenomic and posttranslational hypoxia-inducible factor 1α (HIF1α) regulation via αKG-dependent dioxygenases.93  Raffel et al demonstrated that knockdown of BCAT1 caused accumulation of αKG and HIF1α protein degradation, which translated into a growth-and-survival defect and abrogated leukemia-initiating potential. In turn, overexpression of BCAT1 decreased intracellular αKG levels and caused DNA hypermethylation through altered TET activity, similar to the phenotype exhibited by leukemia cells harboring mutant IDH.93  These findings suggest that αKG is a naturally occurring metabolite with tumor-suppressive potential, and suggest the BCAA-BCAT1-αKG pathway as a therapeutic target to compromise LSC function.

Cys is a semiessential amino acid. Previous studies highlighted the elevated intracellular levels of ROS in cancer cells, a result of genetic and microenvironment alterations and high metabolism rate.94  Those high levels of ROS contribute directly to oxidative stress. To counterbalance high levels of ROS, tumor cells enhance the production of reducing equivalents in part through maintenance of the reduced form of GSH.94,95  Because Cys is 1 of the building blocks of GSH, elevated production of GSH may exhaust endogenous sources of Cys.96  Subsequently, to survive and proliferate, cancer cells accelerate the uptake of extracellular cystine (CSSC; Cys disulfide) via the CSSC/Glu antiporter (xCT transporter).96,97 

Cysteinase, which decreases Cys levels in serum and depletes extracellular Cys, is a potential cancer therapeutic tool.98  To address pharmacokinetic limitations including short half-life, Cramer et al98  engineered cysteinase conjugated to PEG. An in vivo study in cynomolgus monkeys showed that the engineered cysteinase consumed both CSSC and Cys with a high kinetic rate and impeded survival of HMVP2 prostate cancer cells. Cysteinase treatment caused an increase in intracellular ROS accumulation and increased autophagy by elevating 5′ adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and decreasing mammalian target of rapamycin (mTOR) phosphorylation at Ser 2448.98,-100  In a murine model, intraperitoneal administration of cysteinase decreased. CSSC and Cys levels in serum were almost completely eliminated, but the levels recovered to preinjection concentrations after 4 days and 2 days, respectively. No side effects such as weight loss or organ anomalies were observed in either model. Cysteinase treatment significantly inhibited proliferation of the DU145 (P < .0001 at 100 mg/kg) and PC3 (P < .0001 at 50 and 100 mg/kg) prostate cancer cell lines in xenograft murine models.98 

Zhang et al96  demonstrated that, whereas primary chronic lymphoid leukemia (CLL) cells exhibited a limited capacity to transport CSSC into the cells due to low expression of xCT, the cells effectively imported Cys produced by BM stromal cells, which expressed high levels of xCT, imported CSSC, converted it to Cys, and exported Cys into the BM microenvironment. The authors also highlighted the reliance of CLL cells on extracellular Cys for synthesizing GSH, which enhanced CLL cell survival and protected CLL cells from drug-induced cytotoxicity.96  That stroma-leukemia interaction represents 1 of the key amino acid–metabolic pathways for effectively targeting residual leukemia cells in the BM microenvironment (Figure 1).

Very recently, it has been reported that LSCs are auxotrophic to Cys to sustain energy metabolism and that targeting this axis with a cysteine-degrading enzyme may represent an effective therapeutic strategy.101  Cys depletion led to impaired GSH synthesis, decreased glutathionylation of succinate dehydrogenase A (a key component of electron transport chain complex II), and decreased oxidative phosphorylation, and, consequently, decreased production of adenosine triphosphate, leading to LSC death.101 

To mimic the malignant BM microenvironment, Carmer et al98  cocultured splenocytes from mice bearing the aggressive TLC1-Tg:p53−/− CLL, which develop disease resembling human CLL, with BM stromal cells. They observed that cysteinase effectively decreased the viability of those splenocytes. Furthermore, TLC1-Tg:p53−/− CLL-bearing mice treated with cysteinase monotherapy showed a significantly higher survival rate (median, 7 months) than the untreated mice (median, 3.5 months; P < .0001). Cysteinase as monotherapy or combined with fludarabine, tested on CLL with 17p deletions (which is more aggressive and resistant to standard of drugs than CLL without these deletions),102  were both shown to be effective in killing the CLL cells in coculture with stromal cells.98 

Studies of cysteine have prompted the need for attention to the importance of media usage in the context of in vitro metabolomics studies. Cantor et al103  demonstrated that traditional standard culture media and mouse plasma poorly reflect the metabolite composition of human plasma. On the other hand, culture media with polar metabolite concentrations comparable to those of human plasma yielded physiologically relevant alterations of the cellular redox state, glucose utilization, and overall metabolome.103  Further physiological relevance with respect to discovery of unforeseen metabolic wiring and novel metabolite-drug interactions can be achieved through the incorporation of coculture with stromal cells to properly model the hematological microenvironment.

The tumor microenvironment mediates resistance to amino acid–depletion therapies. Recent insights into the immunological component of the tumor microenvironment highlight the functional interplay between tumor cells, amino acids, and immune cells.104 

In probing the mechanism by which patients with AML manifest lymphopenia and pancytopenia, Mussai et al showed that AML blasts and myeloid-derived suppressor cells (MDSCs) exhibit similar capacity to block the proliferation of T cells and human hematopoietic progenitor cells.105  They further demonstrated that MDSCs express ARGase I, and AML blasts secrete ARGase II, both of which induce the specific M-2 phenotype in the surrounding monocytes to suppress immunity. Those findings are consistent with the clinical symptoms of lymphopenia and pancytopenia in AML patients and confirm the immunosuppressive activity of AML blasts in blood and BM.105  (Figure 2) Furthermore, Steggerda et al highlighted the suppressive mechanism of granulocytes, myeloid cells that reside in the peripheral blood and aggregate to the site of infection or injury. Once activated, granulocytes release ARGase I from the cytosol to the extracellular microenvironment. ARGase I consumes the Arg required for immune cell proliferation by converting it to ornithine, thereby impeding proliferation of T cells and natural killer cells. The authors found that a potent and selective ARGase I inhibitor, CB-1158, inhibits Arg depletion in culture medium. Notably, the drug restored proliferation of MDSCs and granulocytes purified from patients with lung or head and neck cancer.106 

GLS inhibitors, which block Gln consumption, and ARGase inhibitors, which block Arg consumption, increase availability of their respective target nutrients for immune cells (Figure 4).107  Ongoing clinical trials are exploring these inhibitors as monotherapy and in combination with anti-PD1 blockade.108  The results summarized here, however, provide a cautionary note for the development of therapies that deplete Arg because Arg can both positively modulate AML growth and negatively modulate an immunosuppressive microenvironment. The question of how AML cells tolerate Arg depletion mediated by self-produced ARGase II in their microenvironment remains unknown. It is possible that AML cells maintain a delicate balance between reserved intracellular Arg and secreted Arg and/or activate alternative metabolic pathways to survive when Arg is depleted. To mitigate negative impact on the AML microenvironment and effectively eradicate AML cells, adjunct approaches such as combinations of immune therapies and amino acid–targeted therapies should be evaluated.

Hematological malignancies frequently develop dependency on specific amino acids for their survival, and insufficient production of these amino acids creates a metabolic vulnerability and therapeutic opportunity.109  Amino acid–targeted enzymes such as ASNase, the first enzyme approved by the US Food and Drug Administration for the treatment of ALL, have shown promising results.12  For 5 decades, ASNase has been classified as a chemotherapy, but as highlighted in this review, ASNase and other amino acid–targeting enzymes fulfill the major requirements of targeted therapies: targeting a specific molecule that is upregulated or specific to cancer cells over normal cells. Therefore, we encourage the oncology community to reclassify ASNase and similar amino acid–targeted enzymes as targeted therapies.

Antimetabolites that interrupt amino acid synthesis have also been developed and are undergoing clinical trials as cancer therapeutics.110,-112  However, the lack of antimetabolite specificity often leads to undesirable side effects. Moreover, cancer cells can develop resistance due to upregulation of endogenous enzymes, collateral pathways, and the microenvironment when used as single agents.81  Therefore, drug combinations are being studied extensively to optimize amino acid–targeted enzyme therapy. In fact, combination of the BCL-2 inhibitor venetoclax with the hypomethylating agent azacitidine acts to inhibit amino acid uptake and catabolism, and the combination has been shown to be effective against LSCs, which depend on amino acid metabolism for survival.109  These findings provide a molecular mechanism for targeting LSCs and indicate that clinically relevant eradication of LSCs can be achieved with drugs that target LSC metabolic vulnerabilities.109  On the other hand, amino acid–modulating therapies that enhance the supply of amino acids to tumor-infiltrating lymphocytes may function as adjunct approaches in immune therapies.

The authors thank Sanaz Ghotbaldini for assistance with manuscript preparation, and Kathryn Hale, Department of Scientific Publications, The University of Texas Anderson Cancer Center, for manuscript review.

This work was supported in part by the National Institutes of Health, National Cancer Institute (R01 CA206210-01) (M.K.).

Contribution: All authors have contributed to the preparation of the manuscript and reviewed/approved it in its final form.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Marina Konopleva, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Unit 428, 1515 Holcombe Blvd, Houston, TX 77030; e-mail: mkonople@mdanderson.org.

1.
Rose
WC
,
Oesterling
MJ
,
Womack
M
.
Comparative growth on diets containing ten and 19 amino acids, with further observations upon the role of glutamic and aspartic acids
.
J Biol Chem
.
1948
;
176
(
2
):
753
-
762
.
2.
Fernandes
HS
,
Silva Teixeira
CS
,
Fernandes
PA
,
Ramos
MJ
,
Cerqueira
NM
.
Amino acid deprivation using enzymes as a targeted therapy for cancer and viral infections
.
Expert Opin Ther Pat
.
2017
;
27
(
3
):
283
-
297
.
3.
Taya
Y
,
Ota
Y
,
Wilkinson
AC
, et al
.
Depleting dietary valine permits nonmyeloablative mouse hematopoietic stem cell transplantation
.
Science
.
2016
;
354
(
6316
):
1152
-
1155
.
4.
Eales
KL
,
Hollinshead
KE
,
Tennant
DA
.
Hypoxia and metabolic adaptation of cancer cells
.
Oncogenesis
.
2016
;
5
:
e190
.
5.
van Galen
P
,
Mbong
N
,
Kreso
A
, et al
.
Integrated stress response activity marks stem cells in normal hematopoiesis and leukemia
.
Cell Reports
.
2018
;
25
(
5
):
1109
-
1117.e5
.
6.
Ward
PS
,
Thompson
CB
.
Metabolic reprogramming: a cancer hallmark even Warburg did not anticipate
.
Cancer Cell
.
2012
;
21
(
3
):
297
-
308
.
7.
Zhu
J
,
Thompson
CB
.
Metabolic regulation of cell growth and proliferation
.
Nat Rev Mol Cell Biol
.
2019
;
20
(
7
):
436
-
450
.
8.
Cantor
JR
,
Sabatini
DM
.
Cancer cell metabolism: one hallmark, many faces
.
Cancer Discov
.
2012
;
2
(
10
):
881
-
898
.
9.
Cantor
JR
,
Panayiotou
V
,
Agnello
G
,
Georgiou
G
,
Stone
EM
.
Engineering reduced-immunogenicity enzymes for amino acid depletion therapy in cancer
.
Methods Enzymol
.
2012
;
502
:
291
-
319
.
10.
Estey
E
,
Döhner
H
.
Acute myeloid leukaemia
.
Lancet
.
2006
;
368
(
9550
):
1894
-
1907
.
11.
Ye
H
,
Adane
B
,
Khan
N
, et al
.
Leukemic stem cells evade chemotherapy by metabolic adaptation to an adipose tissue niche
.
Cell Stem Cell
.
2016
;
19
(
1
):
23
-
37
.
12.
Dinndorf
PA
,
Gootenberg
J
,
Cohen
MH
,
Keegan
P
,
Pazdur
R
.
FDA drug approval summary: pegaspargase (oncaspar) for the first-line treatment of children with acute lymphoblastic leukemia (ALL)
.
Oncologist
.
2007
;
12
(
8
):
991
-
998
.
13.
Morris
SM
Jr
.
Arginine: beyond protein
.
Am J Clin Nutr
.
2006
;
83
(
2
):
508S
-
512S
.
14.
Reeds
PJ
.
Dispensable and indispensable amino acids for humans
.
J Nutr
.
2000
;
130
(
7
):
1835S
-
1840S
.
15.
Barbul
A
,
Rettura
G
,
Levenson
SM
,
Seifter
E
.
Wound healing and thymotropic effects of arginine: a pituitary mechanism of action
.
Am J Clin Nutr
.
1983
;
37
(
5
):
786
-
794
.
16.
Patil
MD
,
Bhaumik
J
,
Babykutty
S
,
Banerjee
UC
,
Fukumura
D
.
Arginine dependence of tumor cells: targeting a chink in cancer’s armor
.
Oncogene
.
2016
;
35
(
38
):
4957
-
4972
.
17.
Yoshiki
N
,
Kubota
T
,
Aso
T
.
Expression and localization of heme oxygenase in human placental villi
.
Biochem Biophys Res Commun
.
2000
;
276
(
3
):
1136
-
1142
.
18.
Pegg
AE
.
Mammalian polyamine metabolism and function
.
IUBMB Life
.
2009
;
61
(
9
):
880
-
894
.
19.
Morris
SM
Jr
.
Enzymes of arginine metabolism
.
J Nutr
.
2004
;
134
(
suppl 10
):
2743S
-
2747S
,
discussion 2765S-2767S
.
20.
Lind
DS
.
Arginine and cancer
.
J Nutr
.
2004
;
134
(
suppl 10
):
2837S
-
2841S
,
discussion 2853S
.
21.
Pervin
S
,
Singh
R
,
Hernandez
E
,
Wu
G
,
Chaudhuri
G
.
Nitric oxide in physiologic concentrations targets the translational machinery to increase the proliferation of human breast cancer cells: involvement of mammalian target of rapamycin/eIF4E pathway
.
Cancer Res
.
2007
;
67
(
1
):
289
-
299
.
22.
Choudhari
SK
,
Chaudhary
M
,
Bagde
S
,
Gadbail
AR
,
Joshi
V
.
Nitric oxide and cancer: a review
.
World J Surg Oncol
.
2013
;
11
:
118
.
23.
Phoa
N
,
Epe
B
.
Influence of nitric oxide on the generation and repair of oxidative DNA damage in mammalian cells
.
Carcinogenesis
.
2002
;
23
(
3
):
469
-
475
.
24.
Zhang
L
,
Liu
M
,
Jamil
S
,
Han
R
,
Xu
G
,
Ni
Y
.
PEGylation and pharmacological characterization of a potential anti-tumor drug, an engineered arginine deiminase originated from Pseudomonas plecoglossicida
.
Cancer Lett
.
2015
;
357
(
1
):
346
-
354
.
25.
Ensor
CM
,
Holtsberg
FW
,
Bomalaski
JS
,
Clark
MA
.
Pegylated arginine deiminase (ADI-SS PEG20,000 mw) inhibits human melanomas and hepatocellular carcinomas in vitro and in vivo
.
Cancer Res
.
2002
;
62
(
19
):
5443
-
5450
.
26.
Cheng
PN
,
Leung
YC
,
Lo
WH
,
Tsui
SM
,
Lam
KC
.
Remission of hepatocellular carcinoma with arginine depletion induced by systemic release of endogenous hepatic arginase due to transhepatic arterial embolisation, augmented by high-dose insulin: arginase as a potential drug candidate for hepatocellular carcinoma
.
Cancer Lett
.
2005
;
224
(
1
):
67
-
80
.
27.
Feun
L
,
You
M
,
Wu
CJ
, et al
.
Arginine deprivation as a targeted therapy for cancer
.
Curr Pharm Des
.
2008
;
14
(
11
):
1049
-
1057
.
28.
Liu
J
,
Ma
J
,
Wu
Z
, et al
.
Arginine deiminase augments the chemosensitivity of argininosuccinate synthetase-deficient pancreatic cancer cells to gemcitabine via inhibition of NF-κB signaling
.
BMC Cancer
.
2014
;
14
:
686
.
29.
Feun
L
,
Savaraj
N
.
Pegylated arginine deiminase: a novel anticancer enzyme agent
.
Expert Opin Investig Drugs
.
2006
;
15
(
7
):
815
-
822
.
30.
Ott
PA
,
Carvajal
RD
,
Pandit-Taskar
N
, et al
.
Phase I/II study of pegylated arginine deiminase (ADI-PEG 20) in patients with advanced melanoma
.
Invest New Drugs
.
2013
;
31
(
2
):
425
-
434
.
31.
Ascierto
PA
,
Scala
S
,
Castello
G
, et al
.
Pegylated arginine deiminase treatment of patients with metastatic melanoma: results from phase I and II studies
.
J Clin Oncol
.
2005
;
23
(
30
):
7660
-
7668
.
32.
Glazer
ES
,
Piccirillo
M
,
Albino
V
, et al
.
Phase II study of pegylated arginine deiminase for nonresectable and metastatic hepatocellular carcinoma
.
J Clin Oncol
.
2010
;
28
(
13
):
2220
-
2226
.
33.
Yang
TS
,
Lu
SN
,
Chao
Y
, et al
.
A randomised phase II study of pegylated arginine deiminase (ADI-PEG 20) in Asian advanced hepatocellular carcinoma patients
.
Br J Cancer
.
2010
;
103
(
7
):
954
-
960
.
34.
Bean
GR
,
Kremer
JC
,
Prudner
BC
, et al
.
A metabolic synthetic lethal strategy with arginine deprivation and chloroquine leads to cell death in ASS1-deficient sarcomas
.
Cell Death Dis
.
2016
;
7
(
10
):
e2406
.
35.
Tsai
WB
,
Aiba
I
,
Long
Y
, et al
.
Activation of Ras/PI3K/ERK pathway induces c-Myc stabilization to upregulate argininosuccinate synthetase, leading to arginine deiminase resistance in melanoma cells
.
Cancer Res
.
2012
;
72
(
10
):
2622
-
2633
.
36.
Beddowes
E
,
Spicer
J
,
Chan
PY
, et al
.
Phase 1 dose-escalation study of pegylated arginine deiminase, cisplatin, and pemetrexed in patients with argininosuccinate synthetase 1-deficient thoracic cancers
.
J Clin Oncol
.
2017
;
35
(
16
):
1778
-
1785
.
37.
Miraki-Moud
F
,
Ghazaly
E
,
Ariza-McNaughton
L
, et al
.
Arginine deprivation using pegylated arginine deiminase has activity against primary acute myeloid leukemia cells in vivo
.
Blood
.
2015
;
125
(
26
):
4060
-
4068
.
38.
Cheng
PN
,
Lam
TL
,
Lam
WM
, et al
.
Pegylated recombinant human arginase (rhArg-peg5,000mw) inhibits the in vitro and in vivo proliferation of human hepatocellular carcinoma through arginine depletion
.
Cancer Res
.
2007
;
67
(
1
):
309
-
317
.
39.
Krebs
HA
,
Hems
R
,
Lund
P
.
Accumulation of amino acids by the perfused rat liver in the presence of ethanol
.
Biochem J
.
1973
;
134
(
3
):
697
-
705
.
40.
Morris
SM
Jr
,
Bhamidipati
D
,
Kepka-Lenhart
D
.
Human type II arginase: sequence analysis and tissue-specific expression
.
Gene
.
1997
;
193
(
2
):
157
-
161
.
41.
Mussai
F
,
Egan
S
,
Higginbotham-Jones
J
, et al
.
Arginine dependence of acute myeloid leukemia blast proliferation: a novel therapeutic target
.
Blood
.
2015
;
125
(
15
):
2386
-
2396
.
42.
Hernandez
CP
,
Morrow
K
,
Lopez-Barcons
LA
, et al
.
Pegylated arginase I: a potential therapeutic approach in T-ALL
.
Blood
.
2010
;
115
(
25
):
5214
-
5221
.
43.
Reubi
JC
,
Toggenburger
G
,
Cuénod
M
.
Asparagine as precursor for transmitter aspartate in corticostriatal fibres
.
J Neurochem
.
1980
;
35
(
4
):
1015
-
1017
.
44.
Richards
NG
,
Kilberg
MS
.
Asparagine synthetase chemotherapy
.
Annu Rev Biochem
.
2006
;
75
:
629
-
654
.
45.
Krall
AS
,
Xu
S
,
Graeber
TG
,
Braas
D
,
Christofk
HR
.
Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor
.
Nat Commun
.
2016
;
7
:
11457
.
46.
Zhang
J
,
Fan
J
,
Venneti
S
, et al
.
Asparagine plays a critical role in regulating cellular adaptation to glutamine depletion
.
Mol Cell
.
2014
;
56
(
2
):
205
-
218
.
47.
Story
MD
,
Voehringer
DW
,
Stephens
LC
,
Meyn
RE
.
L-asparaginase kills lymphoma cells by apoptosis
.
Cancer Chemother Pharmacol
.
1993
;
32
(
2
):
129
-
133
.
48.
Cooney
DA
,
Capizzi
RL
,
Handschumacher
RE
.
Evaluation of L-asparagine metabolism in animals and man
.
Cancer Res
.
1970
;
30
(
4
):
929
-
935
.
49.
Prager
MD
,
Bachynsky
N
.
Asparagine synthetase in normal and malignant tissues: correlation with tumor sensitivity to asparaginase
.
Arch Biochem Biophys
.
1968
;
127
(
1
):
645
-
654
.
50.
Kidd
JG
.
Regression of transplanted lymphomas induced in vivo by means of normal guinea pig serum. II. Studies on the nature of the active serum constituent: histological mechanism of the regression: tests for effects of guinea pig serum on lymphoma cells in vitro: discussion
.
J Exp Med
.
1953
;
98
(
6
):
583
-
606
.
51.
Broome
JD
.
Evidence that the L-asparaginase of guinea pig serum is responsible for its antilymphoma effects. II. Lymphoma 6C3HED cells cultured in a medium devoid of L-asparagine lose their susceptibility to the effects of guinea pig serum in vivo
.
J Exp Med
.
1963
;
118
:
121
-
148
.
52.
McCredie
KB
,
Ho
DH
,
Freireich
EJ
.
L-asparaginase for the treatment of cancer
.
CA Cancer J Clin
.
1973
;
23
(
4
):
220
-
227
.
53.
Miller
HK
,
Salser
JS
,
Balis
ME
.
Amino acid levels following L-asparagine amidohydrolase (EC.3.5.1.1) therapy
.
Cancer Res
.
1969
;
29
(
1
):
183
-
187
.
54.
Beard
ME
,
Crowther
D
,
Galton
DA
, et al
.
L-asparaginase in treatment of acute leukaemia and lymphosarcoma
.
BMJ
.
1970
;
1
(
5690
):
191
-
195
.
55.
Ortega
JA
,
Nesbit
ME
Jr
,
Donaldson
MH
, et al
.
L-Asparaginase, vincristine, and prednisone for induction of first remission in acute lymphocytic leukemia
.
Cancer Res
.
1977
;
37
(
2
):
535
-
540
.
56.
Abakumova
OI
,
Podobed
OV
,
Karalkin
PA
,
Kondakova
LI
,
Sokolov
NN
.
Antitumor activity of L-asparaginase from Erwinia carotovora from against different leukemic and solid tumours cell lines [in Russian]
.
Biomed Khim
.
2013
;
59
(
5
):
498
-
513
.
57.
Panosyan
EH
,
Wang
Y
,
Xia
P
, et al
.
Asparagine depletion potentiates the cytotoxic effect of chemotherapy against brain tumors
.
Mol Cancer Res
.
2014
;
12
(
5
):
694
-
702
.
58.
Appel
IM
,
Kazemier
KM
,
Boos
J
, et al
.
Pharmacokinetic, pharmacodynamic and intracellular effects of PEG-asparaginase in newly diagnosed childhood acute lymphoblastic leukemia: results from a single agent window study
.
Leukemia
.
2008
;
22
(
9
):
1665
-
1679
.
59.
Zalewska-Szewczyk
B
,
Andrzejewski
W
,
Młynarski
W
,
Jedrychowska-Dańska
K
,
Witas
H
,
Bodalski
J
.
The anti-asparagines antibodies correlate with L-asparagines activity and may affect clinical outcome of childhood acute lymphoblastic leukemia
.
Leuk Lymphoma
.
2007
;
48
(
5
):
931
-
936
.
60.
Avramis
VI
,
Panosyan
EH
.
Pharmacokinetic/pharmacodynamic relationships of asparaginase formulations: the past, the present and recommendations for the future
.
Clin Pharmacokinet
.
2005
;
44
(
4
):
367
-
393
.
61.
Patel
N
,
Krishnan
S
,
Offman
MN
, et al
.
A dyad of lymphoblastic lysosomal cysteine proteases degrades the antileukemic drug L-asparaginase
.
J Clin Invest
.
2009
;
119
(
7
):
1964
-
1973
.
62.
Avramis
VI
,
Sencer
S
,
Periclou
AP
, et al
.
A randomized comparison of native Escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a Children’s Cancer Group study
.
Blood
.
2002
;
99
(
6
):
1986
-
1994
.
63.
Peterson
RG
,
Handschumacher
RE
,
Mitchell
MS
.
Immunological responses to L-asparaginase
.
J Clin Invest
.
1971
;
50
(
5
):
1080
-
1090
.
64.
Aslanian
AM
,
Fletcher
BS
,
Kilberg
MS
.
Asparagine synthetase expression alone is sufficient to induce l-asparaginase resistance in MOLT-4 human leukaemia cells
.
Biochem J
.
2001
;
357
(
pt 1
):
321
-
328
.
65.
Haskell
CM
,
Canellos
GP
.
l-asparaginase resistance in human leukemia--asparagine synthetase
.
Biochem Pharmacol
.
1969
;
18
(
10
):
2578
-
2580
.
66.
Chien
WW
,
Le Beux
C
,
Rachinel
N
, et al
.
Differential mechanisms of asparaginase resistance in B-type acute lymphoblastic leukemia and malignant natural killer cell lines
.
Sci Rep
.
2015
;
5
:
8068
.
67.
Iwamoto
S
,
Mihara
K
,
Downing
JR
,
Pui
CH
,
Campana
D
.
Mesenchymal cells regulate the response of acute lymphoblastic leukemia cells to asparaginase
.
J Clin Invest
.
2007
;
117
(
4
):
1049
-
1057
.
68.
Li
J
,
Law
HK
,
Lau
YL
,
Chan
GC
.
Differential damage and recovery of human mesenchymal stem cells after exposure to chemotherapeutic agents
.
Br J Haematol
.
2004
;
127
(
3
):
326
-
334
.
69.
Fung
KL
,
Liang
RH
,
Chan
GC
.
Vincristine but not imatinib could suppress mesenchymal niche’s support to lymphoid leukemic cells
.
Leuk Lymphoma
.
2010
;
51
(
3
):
515
-
522
.
70.
DeBerardinis
RJ
,
Mancuso
A
,
Daikhin
E
, 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 USA
.
2007
;
104
(
49
):
19345
-
19350
.
71.
Samudio
I
,
Konopleva
M
.
Asparaginase unveils glutamine-addicted AML
.
Blood
.
2013
;
122
(
20
):
3398
-
3400
.
72.
Avramis
VI
.
Asparaginases: biochemical pharmacology and modes of drug resistance
.
Anticancer Res
.
2012
;
32
(
7
):
2423
-
2437
.
73.
Lessner
HE
,
Valenstein
S
,
Kaplan
R
,
DeSimone
P
,
Yunis
A
.
Phase II study of L-asparaginase in the treatment of pancreatic carcinoma
.
Cancer Treat Rep
.
1980
;
64
(
12
):
1359
-
1361
.
74.
Chan
WK
,
Lorenzi
PL
,
Anishkin
A
, et al
.
The glutaminase activity of L-asparaginase is not required for anticancer activity against ASNS-negative cells
.
Blood
.
2014
;
123
(
23
):
3596
-
3606
.
75.
Kumar
S
,
Venkata Dasu
V
,
Pakshirajan
K
.
Purification and characterization of glutaminase-free L-asparaginase from Pectobacterium carotovorum MTCC 1428
.
Bioresour Technol
.
2011
;
102
(
2
):
2077
-
2082
.
76.
Offman
MN
,
Krol
M
,
Patel
N
, et al
.
Rational engineering of L-asparaginase reveals importance of dual activity for cancer cell toxicity
.
Blood
.
2011
;
117
(
5
):
1614
-
1621
.
77.
Zalewska-Szewczyk
B
,
Andrzejewski
W
,
Bodalski
J
.
Development of anti-asparaginase antibodies in childhood acute lymphoblastic leukemia
.
Pediatr Blood Cancer
.
2004
;
43
(
5
):
600
-
602
.
78.
Swain
AL
,
Jaskólski
M
,
Housset
D
,
Rao
JK
,
Wlodawer
A
.
Crystal structure of Escherichia coli L-asparaginase, an enzyme used in cancer therapy
.
Proc Natl Acad Sci USA
.
1993
;
90
(
4
):
1474
-
1478
.
79.
Keating
MJ
,
Holmes
R
,
Lerner
S
,
Ho
DH
.
L-asparaginase and PEG asparaginase–past, present, and future
.
Leuk Lymphoma
.
1993
;
10
(
suppl 1
):
153
-
157
.
80.
Salzer
W
,
Seibel
N
,
Smith
M
.
Erwinia asparaginase in pediatric acute lymphoblastic leukemia
.
Expert Opin Biol Ther
.
2012
;
12
(
10
):
1407
-
1414
.
81.
Kurtzberg
J
,
Asselin
B
,
Bernstein
M
,
Buchanan
GR
,
Pollock
BH
,
Camitta
BM
.
Polyethylene Glycol-conjugated L-asparaginase versus native L-asparaginase in combination with standard agents for children with acute lymphoblastic leukemia in second bone marrow relapse: a Children’s Oncology Group Study (POG 8866)
.
J Pediatr Hematol Oncol
.
2011
;
33
(
8
):
610
-
616
.
82.
Parmentier
JH
,
Maggi
M
,
Tarasco
E
,
Scotti
C
,
Avramis
VI
,
Mittelman
SD
.
Glutaminase activity determines cytotoxicity of L-asparaginases on most leukemia cell lines
.
Leuk Res
.
2015
;
39
(
7
):
757
-
762
.
83.
Ehsanipour
EA
,
Sheng
X
,
Behan
JW
, et al
.
Adipocytes cause leukemia cell resistance to L-asparaginase via release of glutamine
.
Cancer Res
.
2013
;
73
(
10
):
2998
-
3006
.
84.
Gregory
MA
,
Nemkov
T
,
Park
HJ
, et al
.
Targeting glutamine metabolism and redox state for leukemia therapy
.
Clin Cancer Res
.
2019
;
25
(
13
):
4079
-
4090
.
85.
Jacque
N
,
Ronchetti
AM
,
Larrue
C
, et al
.
Targeting glutaminolysis has antileukemic activity in acute myeloid leukemia and synergizes with BCL-2 inhibition
.
Blood
.
2015
;
126
(
11
):
1346
-
1356
.
86.
Gallipoli
P
,
Giotopoulos
G
,
Tzelepis
K
, et al
.
Glutaminolysis is a metabolic dependency in FLT3ITD acute myeloid leukemia unmasked by FLT3 tyrosine kinase inhibition
.
Blood
.
2018
;
131
(
15
):
1639
-
1653
.
87.
Wang
ES
,
Frankfurt
O
,
Orford
KW
, et al
.
Phase 1 study of CB-839, a first-in-class, orally administered small molecule inhibitor of glutaminase in patients with relapsed/refractory leukemia [abstract]
.
Blood
.
2015
;
126
(
23
). Abstract
2566
.
88.
Guerra
V
,
Dinardo
CD
,
Konopleva
M
, et al
.
Interim results from a phase Ib/II clinical study of the glutaminase inhibitor telaglenastat (CB-839) in combination with azacitidine in patients with advanced myelodysplastic syndrome (MDS) [abstract]
.
J Clin Oncol
.
2019
;
37
(
suppl 15
). Abstract
7037
.
89.
Matre
P
,
Velez
J
,
Jacamo
R
, et al
.
Inhibiting glutaminase in acute myeloid leukemia: metabolic dependency of selected AML subtypes
.
Oncotarget
.
2016
;
7
(
48
):
79722
-
79735
.
90.
Gregory
MA
,
Nemkov
T
,
Reisz
JA
, et al
.
Glutaminase inhibition improves FLT3 inhibitor therapy for acute myeloid leukemia
.
Exp Hematol
.
2018
;
58
:
52
-
58
.
91.
Medyouf
H
,
Mossner
M
,
Jann
JC
, et al
.
Myelodysplastic cells in patients reprogram mesenchymal stromal cells to establish a transplantable stem cell niche disease unit
.
Cell Stem Cell
.
2014
;
14
(
6
):
824
-
837
.
92.
Hattori
A
,
Tsunoda
M
,
Konuma
T
, et al
.
Cancer progression by reprogrammed BCAA metabolism in myeloid leukaemia
.
Nature
.
2017
;
545
(
7655
):
500
-
504
.
93.
Raffel
S
,
Falcone
M
,
Kneisel
N
, et al
.
BCAT1 restricts αKG levels in AML stem cells leading to IDHmut-like DNA hypermethylation [published correction appears in Nature. 2018;560(7718):E28]
.
Nature
.
2017
;
551
(
7680
):
384
-
388
.
94.
Trachootham
D
,
Alexandre
J
,
Huang
P
.
Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?
Nat Rev Drug Discov
.
2009
;
8
(
7
):
579
-
591
.
95.
Harris
IS
,
Treloar
AE
,
Inoue
S
, et al
.
Glutathione and thioredoxin antioxidant pathways synergize to drive cancer initiation and progression [published correction appears in Cancer Cell. 2015;27(2):314]
.
Cancer Cell
.
2015
;
27
(
2
):
211
-
222
.
96.
Zhang
W
,
Trachootham
D
,
Liu
J
, et al
.
Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia
.
Nat Cell Biol
.
2012
;
14
(
3
):
276
-
286
.
97.
Timmerman
LA
,
Holton
T
,
Yuneva
M
, et al
.
Glutamine sensitivity analysis identifies the xCT antiporter as a common triple-negative breast tumor therapeutic target
.
Cancer Cell
.
2013
;
24
(
4
):
450
-
465
.
98.
Cramer
SL
,
Saha
A
,
Liu
J
, et al
.
Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth
.
Nat Med
.
2017
;
23
(
1
):
120
-
127
.
99.
Ghislat
G
,
Patron
M
,
Rizzuto
R
,
Knecht
E
.
Withdrawal of essential amino acids increases autophagy by a pathway involving Ca2+/calmodulin-dependent kinase kinase-β (CaMKK-β)
.
J Biol Chem
.
2012
;
287
(
46
):
38625
-
38636
.
100.
Mungai
PT
,
Waypa
GB
,
Jairaman
A
, et al
.
Hypoxia triggers AMPK activation through reactive oxygen species-mediated activation of calcium release-activated calcium channels
.
Mol Cell Biol
.
2011
;
31
(
17
):
3531
-
3545
.
101.
Jones
CL
,
Stevens
BM
,
D’Alessandro
A
, et al
.
Cysteine depletion targets leukemia stem cells through inhibition of electron transport complex II
.
Blood
.
2019
;
134
(
4
):
389
-
394
.
102.
Liu
J
,
Chen
G
,
Feng
L
, et al
.
Loss of p53 and altered miR15-a/16-1 MCL-1 pathway in CLL: insights from TCL1-Tg:p53(-/-) mouse model and primary human leukemia cells
.
Leukemia
.
2014
;
28
(
1
):
118
-
128
.
103.
Cantor
JR
,
Abu-Remaileh
M
,
Kanarek
N
, et al
.
Physiologic medium rewires cellular metabolism and reveals uric acid as an endogenous inhibitor of UMP synthase
.
Cell
.
2017
;
169
(
2
):
258
-
272.e17
.
104.
Lemos
H
,
Huang
L
,
Prendergast
GC
,
Mellor
AL
.
Immune control by amino acid catabolism during tumorigenesis and therapy
.
Nat Rev Cancer
.
2019
;
19
(
3
):
162
-
175
.
105.
Mussai
F
,
De Santo
C
,
Abu-Dayyeh
I
, et al
.
Acute myeloid leukemia creates an arginase-dependent immunosuppressive microenvironment
.
Blood
.
2013
;
122
(
5
):
749
-
758
.
106.
Steggerda
SM
,
Bennett
MK
,
Chen
J
, et al
.
Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment
.
J Immunother Cancer
.
2017
;
5
(
1
):
101
.
107.
Fung
MKL
,
Chan
GC
.
Drug-induced amino acid deprivation as strategy for cancer therapy
.
J Hematol Oncol
.
2017
;
10
(
1
):
144
.
108.
Mussai
F
,
Wheat
R
,
Sarrou
E
, et al
.
Targeting the arginine metabolic brake enhances immunotherapy for leukaemia [published online ahead of print 28 November 2018]
.
Int J Cancer
.
doi:10.1002/ijc.32028
.
109.
Jones
CL
,
Stevens
BM
,
D’Alessandro
A
, et al
.
Inhibition of amino acid metabolism selectively targets human leukemia stem cells [published correction appears in Cancer Cell. 2019;35(2):333-335]
.
Cancer Cell
.
2018
;
34
(
5
):
724
-
740.e4
.
110.
Ertel
IJ
,
Nesbit
ME
,
Hammond
D
,
Weiner
J
,
Sather
H
.
Effective dose of L-asparaginase for induction of remission in previously treated children with acute lymphocytic leukemia: a report from Childrens Cancer Study Group
.
Cancer Res
.
1979
;
39
(
10
):
3893
-
3896
.
111.
Farber
S
,
Diamond
LK
.
Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid
.
N Engl J Med
.
1948
;
238
(
23
):
787
-
793
.
112.
Vander Heiden
MG
.
Targeting cancer metabolism: a therapeutic window opens
.
Nat Rev Drug Discov
.
2011
;
10
(
9
):
671
-
684
.
Sign in via your Institution