<<

Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 https://doi.org/10.1007/s12288-019-01107-8

REVIEW ARTICLE

The Metabolic Profiles in Hematological Malignancies

1 1 2 Tao Liu • Xing-Chun Peng • Bin Li

Received: 13 September 2018 / Accepted: 25 February 2019 / Published online: 23 April 2019 Ó Indian Society of Hematology and Blood Transfusion 2019

Abstract Leukemia is one of the most aggressive hema- Abbreviations tological malignancies. Leukemia stem cells account for AML Acute myeloid leukemia the poor prognosis and relapse of the disease. Decades of CML Chronic myeloid leukemia investigations have been performed to figure out how to T-ALL T-cell lymphoblastic leukemia eradicate the leukemia stem cells. It has also been known B-ALL B-cell lymphoblastic leukemia that cancer cells especially solid cancer cells use energy CLL Chronic lymphocytic leukemia differently than most of the cell types. The same thing MM Multiple myeloma happens to leukemia. Since there are metabolic differences MDS Myelodysplastic syndrome between the hematopoietic stem cells and their immediate LSCs Leukemia stem cells descendants, we aim at manipulating the energy sources OXPHOS Oxidative phosphorylation with which that could have an effect on leukemia stem FAO Fatty acid oxidation cells while sparing the normal blood cells. In this review PPP Pentose phosphate pathway we summarize the metabolic characteristics of distinct Glut1 Glucose transporter 1 leukemias such as acute myeloid leukemia, chronic mye- Ara-C Arabinofuranosyl cytidine loid leukemia, T cell lymphoblastic leukemia, B-cell 2-DG 2-Deoxy-D-glucose lymphoblastic leukemia, chronic lymphocytic leukemia RTKs Receptor tyrosine and other leukemia associated hematological malignancies ROS Reactive oxygen species such as multiple myeloma and myelodysplastic syndrome. CSCs Cancer stem cells A better understanding of the metabolic profiles in distinct PPARc Proliferator-activated receptor gamma ligands leukemias might provide novel perspectives and shed light CPT1 Carnitine O-palmitoyltransferase I on novel metabolic targeting strategies towards the clinical GAT Gonadal adipose tissue treatment of leukemias. GA IDH1/2 Isocitrate 1 and 2 Keywords Hematological malignancies Á Leukemia stem PHGDH Phosphoglycerate dehydrogenas cells Á Metabolism I-ASP l- GS ASNS Asparagine synthetase & Bin Li PRC2 Polycomb repressive complex 2 [email protected]

1 Department of Pathology, People’s Hospital of Longhua, Shenzhen 518131, People’s Republic of China 2 Department of Pathology, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai Clinical Center, CAS, Huaihai Road 966, Shanghai City 200031, Shanghai, People’s Republic of China 123 626 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634

Background discuss the metabolic profiles of four major leukemias as well as other hematological malignancies. In particular, we Leukemia is a kind of hematological malignancy that often underscore potential metabolic vulnerabilities for each type initiates in the bone marrow and leads to abnormal and of leukemia. immature white blood cells which are also named blasts or leukemia cells. In a contemporary view, LSCs is respon- sible for the initiation and development of the disease and Glucose Metabolism can explain most of the poor prognosis in clinical trials. Numerous studies have shed light on eliminating LSCs The well established Warburg effect has demonstrated that based on their distinct genetic and epigenetic characteris- solid cancer cells mainly use glycolysis to meet their tics compared with the normal blood stem cells [1, 2]. In energy requirement [10]. The Warburg effect had long recent decades, awareness that the metabolic phenotype of been believed to be an adaptation to low-oxygen environ- leukemia cells is heterogeneous and distinct from that of ment in cancer cells. Since most of the raw materials and their normal counterparts—is increasing. How the leuke- energy required for cell proliferation are from glycolysis, mia cells survive and propagate in adverse environment cancer cells need to activate glycolysis pathways although such as low glucose, hypoxia and oxidative stress remains they mostly reside in a condition that has enough oxygen. poorly understood [3, 4]. Targeting the glucose metabolism of leukemia cells has Glucose, free fatty acids, glutamine, amino acids are already becoming an emerging trend for leukemia basic and important materials for the catabolism pathways treatment. that support the growth and survival of the cancer cells. These catabolites are either assimilated from the blood Role in AML circulation or synthesized within cancer cells. Glucose is the among the most available nutrients in blood and a There are still some controversies concerning whether metabolic substrate generally utilized by cancer cells [5]. It AML cells use glycolytic metabolism or OXPHOS meta- is well known that anaerobic glycolysis and aerobic bolism although the glucose metabolism in AML has been mediated oxidative phosphorylation (OXPHOS) are the implicated in many studies. An interesting study by Wang two major pathways in glucose metabolism. Thus, various et al. had demonstrated that AML cells are so unique in the drugs that interfere with glycolysis and OXPHOS are being way they sense the glucose availability and utilization investigated and designed as anti-cancer agents. Fatty acids while normal cells do not get disrupted because they have are also essential substrates for catabolic pathways in other glucose metabolism in place in case of an emergency. cancers. The pentose phosphate pathway (PPP) mediated They suggested that by precise control of the level of glucose catabolism is known to be an important initiating glycolysis would be a novel therapeutical strategy for point for the production of NADPH and maintenance of a treating AML without affecting the normal function of the redox balance in normal cells [6]. However, fatty acid HSCs [11]. Besides, Saito et al. indicated that AMPK oxidation (FAO) take the place of PPP in cancer cells and deletion substantially delays leukemogenesis and eradi- contributes substantially to these processes under meta- cates LSCs in MLL-AF9 induced AML by downregulating bolic stress due to the low glucose levels [7]. Contrary to the expression of glucose transporter 1 (Glut1), enhancing the FAO catabolism, lipids and steroids are de novo syn- oxidative stress and inducing DNA damage. This study thesized to generate the new phospholipid bilayers, which powerfully supports that the Warburg effect does exist in are essential for the fast proliferation and division of the leukemia [12]. Moreover, Chen et al. [13] showed that cancer cells [8]. Glutamine is another indispensable nutri- elevated glycolysis decrease the vulnerability to anti-leu- ent for cancer cell growth since the amido nitrogen is an kemia agent arabinofuranosyl cytidine (Ara-C) while essential substrate for hexosamine and nucleotide synthesis inhibition of glycolysis suppresses AML cell growth and [9], and is also getting involved in TCA cycle to produce enhances cytotoxicity of Ara-C in killing leukemic cells. energy for cancer cells. Other critical metabolic processes Also, Larrue et al. demonstrated that the glycolytic inhi- include asparagine metabolism, folate metabolism and bitor 2-deoxy-D-glucose (2-DG) exhibited great anti-leu- oxidative stress metabolism. Like solid cancer cells, leu- kemia activity through modulating the expression of kemia cells are not metabolically homogenous and distinct receptor tyrosine kinases (RTKs) such as FLT-ITD and types of leukemia cells utilize special metabolic raw KIT. Modulating glycolysis by using 2-DG provides a material preferentially. Thus, targeting the metabolic dif- therapeutic approach in some subset of AML harboring ferences between leukemia cells and normal blood cells mutated RTKs [14]. One of the traditional ways of treating provides novel anti-leukemia strategy. In this review, we AML is by chemotherapy but sometimes it is not quite effective because of chemoresistance. A recent research by 123 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 627

Song et al. revealed that increased glycolysis and decreased metabolic regulators (mTORC1 and AMPK) could target OXPHOS may result in the drug resistance of AML cells. the CLL lymphocytes by reducing lactate production, thus Targeting anaerobic glycolysis metabolism is an effective decreasing aerobic glycolysis [19]. strategy for manipulating chemoresistance in AML [15]. Despite the numerous evidence proofing that AML mainly Role in CML uses glycolysis as their glucose utilization pathways, which is consistant with the solid cancer cells, an intriguing Glycolytic dependencies is also reported in CML. For investigation by Lagadinou et al. reported that OXPHOS is instance, Gottschalk et al. showed that imatinib (STI571), the unique fuel source for LSCs of AML. They discovered which is a classic inhibitor of the BCR-ABL oncoprotein, that LSCs in AML reside in a low levels of reactive oxygen induces cell apoptosis in human leukemia BCR-ABL species (termed ‘‘ROS-low’’) condition and exhibit a positive cells by altering the glucose metabolism from slower rate of energy metabolism compared with the nor- glycolysis to mitochondrial metabolism [20]. mal blood cells. Interestingly, they found that the anti- To sum up, leukemia cells may not always depend on apoptotic molecule BCL-2 is aberrantly overexpressed in anaerobic respiration as their energy source. Moderated ROS-low LSCs. Targeting the BCL-2 dependent OXPHOS aerobic glycolysis, along with alternative metabolic path- could selectively kill LSCs while spare the normal blood ways like OXPHOS, may also be necessary to promote the stem cells [16] (Table 1). growth and survival of leukemia cells especially when they are in an adverse environment in the long run. The Role in ALL and CLL heterogeneity of glucose metabolism in distinct leukemias remains to be studied and more evidence is needed to get a Intriguingly, T-ALL leukemia cells might also prefer to comprehensive map of leukemic glucose metabolism for reprogram glycolysis to OXPHOS to survive under the treatment in the future (Fig. 1). metabolic stress. For example, a study by Kishton et al. [17] revealed that AMPK is essential to maintain mito- Lipogenesis Metabolism chondrial metabolism which alleviates metabolic stress and prevents T-ALL cells from apoptosis. Moreover, Liu et al. The process of lipogenesis, also named lipid synthesis, implied that specifically targeting Glut1 impaired the gly- plays an important role in the pathogenesis of solid cancer. colysis and anabolic metabolism of B-ALL cells harboring Several are getting involved in the lipid synthesis BCR-ABL fusion [18]. CLL leukemic cells also pathways, including , acetyl-CoA car- tend to use glycolysis. Synergistically using a tyrosine boxylase, ATP citrate and choline [21–25]. kinase inhibitor dasatinib and inhibiting two master Choline is the raw material in regenerating the membrane phospholipid phosphatidylcholine of cancer cells. The expression and activity of are increased in Table 1 Select agents targeting metabolism that are approved, or are in trials, for the treatment of cancer, focusing on targets discussed in various solid human cancers because of growth factor this review stimulation and activation of Ras signaling pathway [23]. Many studies support that lipid synthesis favors leukemo- Drug Target genesis (Table 2). 2-DG RTK such as FLT-ITD and KIT Imatinib (STI571) BCR-ABL oncoprotein Role in AML Inhibitor ST1326 CPT1a Avocatin B Mitochondrial A recent finding by Fraineau et al. [29] revealed that ABT-737 BCL-2 mesenchymal stromal cells (MSCs) derived from bone NXT629 PPAR-a marrow niche create a favorable microenvironment that ABT-199 BCL-2 sustains the initiation and progression of AML. This dis- BPTES Glutaminase covery also indicates that novel therapeutic treatment could I-ASP Circulating asparagine target the leukemia niche which favors more for the sur- Methotrexate (MTX) Dihydrofolate (DHFR) vival of cancer cells than the normal blood cells. However, SU9516 (DHFR) there are evidence showing that adipogenesis was inhibited mTOR inhibitors (MTIs) mTOR in some subtype of AML. For example, Kim et al. sug- 3-DeazaneplanocinA (DZNep) Histone gested that adipogenic associated transcription factors are AZD1208 Pan-Pim kinase negatively regulated in the promyelocytic leukemia (PML). This contributes to fat accumulation by blocking the dif- ferentiation of preadipocytes into adipocytes [27]. In 123 628 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634

Fig. 1 Basic metabolic pathways in leukemia cells

addition, Yasugi et al. [28] showed that peroxisome pro- discovery about the glycolysis in cancer cells can be liferator-activated receptor gamma ligands (PPARc) pro- explained as a result of the preferential oxidation of fatty mote the lipogenesis as well as myeloid differentiation in acids in the mitochondrial of cancer cells. In fact, targeting human NB4 cells. This finding seemed to indicate the FAO or other anaplerotic pathways that compensate FAO association of the AML differentiation with lipogenesis but has been demonstrated to provide novel therapeutic it did not clarify a definite cause and effect relationship strategies in treating distinct leukemias [30]. between them. It remains to be explored whether lipoge- nesis contributes to the myeloid differentiation or vice Role in AML and CML versa. Carnitine O-palmitoyltransferase I (CPT1) is a critical rate- Role in Other Hematological Malignancies limiting enzyme, which catalyzes the transfer of the acyl group of a long-chain fatty acyl-CoA from coenzyme A to A study by Medina et al. demonstrate that adiponectin, l-carnitine in FAO. Inhibition of CPT1 has been implicated cytokine generated by adipocytes could significantly sup- its anti-leukemia effect in vivo and intro. For example, press the survival of MM cells by activating PKA/AMPK Ricciard et al. [31] demonstrated that a novel CPT1a pathway and inactivating the ACC1 activity, thus allevi- inhibitor ST1326 exhibits high anti-leukemia activity by ating lipogenesis [26]. These results not only clarify the inducing cell cycle arrest, mitochondrial damage, and cell positive association between obesity and the trend to apoptosis in several leukemia cell lines including AML, develop into MM but also provide a therapeutic strategy by ALL and CLL, with the best efficacy for AML cells. targeting the lipogenesis of MM cancer cells. Despite the catalytic enzymes involved in FAO in cyto- To summarize, whether lipogenesis promote the devel- plasm, the cell surface fatty acid transporters are also novel opment of leukemia or not is still elusive. Leukemic therapeutic targets for AML. Recently, Ye et al. showed dependency on lipogenesis may be determined by the that fatty acid transporter CD36 positive LSCs, which were genetic and epigenetic characteristics of distinct leukemias. substantially enriched in gonadal adipose tissue (GAT), have unique metabolic properties and could evade from chemotherapy by the GAT niche. Targeting the CPT1 FAO Metabolism mediated FAO pathway upsteam of CD36 provides a unique strategy of selective targeting the quiescent LSCs The fatty acid oxidation (FAO) is considered to be the both in human AML and CML treatment [32]. Since free reverse lipid metabolism in terms of lipogenesis. It is fatty acids are not able to cross the mitochondrial mem- noteworthy that FAO has been associated with chemore- branes without help, carnitine-acylcarnitine translocase, sistance and mitochondrial uncoupling. Warburg’s also named carnitine transporter, is responsible for

123 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 629

Table 2 Metabolism dependency in distinct hematological malignancies Role in ALL and CLL References Messmer et al. revealed that one antagonist of PPARa called Glucose metabolism NXT629 efficiently eliminates the CLL cells by suppressing Glycolysis dependency in AML [11–15] peroxisome proliferator-activated receptor (PPAR)-a, OXPHOS dependency in AML [16] which is a main transcriptional modulator of FAO. This OXPHOS dependency in T-ALL [17] finding strongly supported that the survival and growth of Glycolysis dependency in CML [18, 20] CLL cells also depend on the fatty acid metabolism [36]. In Glycolysis dependency in CLL [19] addition, Hermanova et al. suggested that pharmacological Lipogenesis metabolism inhibition of FAO greatly sensitizes the ALL cells to L-ASP, Adipogenesis independency in AML [27, 28] which is a key agent in treating T-ALL. Lipogenesis dependency in MM [26] To sum up, targeting the leukemia’s ‘‘fatty tooth’’ has FAO metabolism exerted a great potential and efficacy in treating distinct FAO dependency in AML [31–35] kinds of leukemias. Targeting FAO as well as other FAO dependency in CML [37] metabolic pathways simultaneously, with moderate inhi- FAO dependency in CLL [36] bition by combination therapy, provide a great value in Glutaminolysis metabolism leukemia treatment. Glutaminolysis dependency in AML [38–40] Glutaminolysis dependency in CML [43–45] Glutaminolysis dependency in T-ALL [41, 42] Glutaminolysis Metabolism Aspartate metabolism Aspartate dependency in AML [48] Glutamine is the most abundant amino acid in amino acid Aspartate dependency in T-ALL [46, 47, 49] metabolism. Glutaminolysis, a process in which glutamine Aspartate independency in B-ALL [50] is discomposed to glutamate, aspartate, CO2, pyruvate, lactate, alanine and citric acid, is an extra energy resource Folate metabolism in cancer cells particularly when glycolysis is relatively Folate dependency in AML [51] low due to the accumulation of a dimeric form of PKM2. Folate dependency in T-ALL [52–54] The rate-limiting step in intracellular glutamine catabolism Folate independency in T-ALL [55] is catalyzed by glutaminase (GA) which converts glu- Oxidative stress metabolism tamine to glutamate. The genes that encode GA are called ROS dependency in AML [58, 59] GLS1 and GLS2. They are alternatively spliced and dis- ROS dependency in T-ALL [60] tinct GA isoforms are generated. ROS dependency in CML [61] Role in AML and CML

Glutaminolysis plays an important role in maintaining the transporting both carnitine-fatty acid complexes and free growth and survival of AML cells as is evidenced by the carnitine across the inner mitochondrial membrane. A indication that glutaminolysis inhibition through CB-839 study by Wu et al. revealed that targeting the carnitine concomitant with using the BCL-2 inhibitor ABT-199 transporter 2 (CT2, SLC22A16) which is overexpressed in induced GLS1 inhibition contributes to the leukemic cell AML shows the great efficacy in eradicating AML cells cycle arrest and apoptosis without affecting their normal [33]. Since FAO is carrying out in mitochondrial, a study human CD34? progenitors [38]. Moreover, an in vitro by Lee et al. [34] revealed that avocatin B could inhibit the assay by Goto et al. [39] suggested that glutamine depri- mitochondrial function, which provides a novel strategy for vation reduces the intracellular content and selectively eradicating leukemia cells while sparing their increases reactive oxygen species (ROS) most significantly normal counterparts. Moreover, Velez et al. [35] implied in four AML cell lines especially in HL-60. The incidence that metformin and phenformin inhibit the FAO and sen- of mutations in 1 and 2 (IDH1/2) sitize leukemia cells to the BCL-2 inhibitor ABT-737 is frequently found in AML. These mutations contribute to induced cell-intrinsic apoptosis. Interestingly, Shinohara the glutamine dependency as the main source for a-ke- et al. [37] showed that by synergistically using the anti- toglutarate which could be converted to the oncoprotein cancer fatty acid derivative AIC-47 and imatinib could 2-hydroxyglutarate in cancer cells. Targeting the glutamine strengthen the attack on CML cells by inhibiting CPT1c dependency of the leukemia cells would be mostly bene- expression and FAO metabolism. ficial to a specific subset of AML with IDH mutations. For 123 630 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 example, Emadi et al. [40] demonstrate that a small Role in AML molecule of glutaminase inhibitor called Bis-2-(5-pheny- lacetamido-1,2,4-thiadiazol-2-yl) ethyl sulfide BPTES, I-ASP shows great efficacy in treating AML disease. For exclusively suppresses the survival of human primary example, Parmentier et al. [47] showed that asparaginase AML cells expressing mutated IDH compared with those activity of I-ASP alone may not be enough for the cyto- expressing the normal IDH. The impact of glycosylation of toxicity to the leukemia cells in T-ALL, and that glutam- ASCT2 which was ever underestimated in metabolic inase activity may be essential for its full anti-leukemia reprogramming could be a therapeutic target in leukemia activity. Moreover, Willems et al. [48] reveal that l-ASP treatment. For example, Polet et al. [43] showed that the increases the expression of the glutamine synthase (GS) defect in glycosylation of glutamine transporters could and knockdown of GS contributes to l-ASP induced result in glycolytic inhibition in AML as well as in CML. autophagic process in AML cells. Drug resistance resulting from the metabolic reprogram- ming accounts for the relapse of leukemia treatment. For Role in ALL and CLL instance, Sta¨ubert et al. [44] demonstrated that resistant leukemia cells alter their metabolism characterized by I-ASP has also been demonstrated to be a efficacious reduced dependency on glutamine metabolism at the cost in treating T-ALL and natural killer (NK) of a higher requirement for glucose and enhanced FAO cell lymphoma by inhibiting the protein synthesis, inducing with decreased pantothenic acid uptake ability. Moreover, the cell cycle arrest and enhancing the ROS level to the Polet et al. implied that phosphoglycerate dehydrogenas mitochondrial permeabilization and subsequent cell apop- (PHGDH) is upregulated after glutamine deprivation. tosis in cancer cells [46]. However, an study done by Chan Silencing PHGDH in vitro and the use of diet without et al. showed that the glutaminase activity of L-ASP is not serine in vivo suppresses AML. Besides, the glutamine always indispensable for anticancer effect in solid tumors. dependency could also be observed in BCR-ABL positive They implied that the L-ASP’s glutaminase activity is CML. Sontakke et al. indicated that the expression of essential for inhibiting the asparagine synthetase (ASNS) glutamine importer SLC1A5 is increased in BCR-ABL positive cells but not for ASNS negative cells. The thera- transduced human cord blood CD341 cells and these peutic significance of this observation is that glutaminase cancer cells are more susceptible to the glutaminase inhi- negative variants of L-ASP instead of wild-type L-ASP bitor BPTES, which implies that the glutamine metabolism should be explored for better treatment of ASNS negative is elevated in CML to maintain OXPHOS level [45]. cancers [49]. Although different L-ASP based chemother- apy are highly efficient, disease relapse could happen fre- Roles in ALL quently. For example, Chien et al. [50] reported that cleavage of anti-apoptotic molecule BCL-2A1 contribute Glutaminolysis has been also highly implicated to be a to the sensitivity of malignant natural killer (NK) cell lines critical process in other forms of leukemia such as T-ALL. and B-ALL cells. For example, Herranz et al. [41] revealed that glu- In summary, l-ASP associated therapeutics towards taminolysis is an important pathway for mutated NOTCH1 distinct types of leukemia provide one-carbon metabolism induced T-ALL growth and a critical determinant of sen- targeting strategy in leukemia treatment. sitivity in anti-NOTCH1 leukemia treatment in vivo. ASCT2, also named SLC1A5, which is known as a major glutamine transporter, has been reported to participate in Folate Metabolism the growth of solid cancer cells [42]. To sum up, targeting glutaminolysis have shown a great Folate metabolism is considered one of another important efficacy in treating AML as well as ALL. More research is one-carbon metabolism besides the aspartate metabolism. needed to determine whether distinct types of leukemia Antifolates are the first effective chemotherapy drugs cells glutamine metabolism as a complemental source. which suppress the growth and survival of cancer cells by inhibiting DNA and RNA synthesis.

Aspartate Metabolism Role in AML l-Asparaginase (I-ASP, erwinia l-asparaginase), which A study by Lynn et al. discovered that the expression of converts L-asparagine to ammonia and , is an folate receptor b (FRb) is elevated when treating AML anticancer agent exhibiting both asparaginase and glu- cells with all-trans retinoic acid (ATRA), which improves taminase activity. immune therapy by using CAR-T cell therapy. This 123 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 631 observation provides novel therapeutic treatment by tar- contribute to cell apoptosis by inducing increased DNA geting the folate metabolism [51]. damage and enhanced ROS level in AML with FLT3-ITD translocation. In addition, Liu et al. revealed that bone Role in ALL marrow niche induces oxidative adaptation and protects leukemia cells from apoptosis in ALL, thus providing cues In addition, Methotrexate (MTX) is well known for its role that targeting redox balance by suppressing antioxidant in treating various hematological malignancies by inhibit- generation and anti-apoptosis pathways could overcome ing dihydrofolate reductase (DHFR). For example, drug resistance [60]. Moreover, the therapeutic approach Uchiyama et al. [52] demonstrated that a cyclin-dependent by oxidative stress enhancement to kill leukemia cells kinase inhibitor called SU9516, which downregulates the could be also applied to CML. For example, a recent study expression of both DHFR mRNA and protein, leads to by Singh et al. [61] suggested that high endogenous ROS enhanced sensitivity to MTX in human T-cell leukemia level is able to weaken the nitric oxide (NO) generation in Jurkat cell line. However, the polymorphisms of DHFR are neutrophils (PMN) of CML. To briefly conclude, targeting concerned with worse ALL outcome, which is associated the oxidative condition represents a general strategy in with higher DHFR expression [53]. Combined effects treating distinct types of leukemias. should be carefully examined on other genes involved in folate metabolism. Drug combination shows very high efficacy as it is evidenced by Teachey et al. [54] showed Conclusions that mTOR inhibitors (MTIs) substantially increase the sensitivity of ALL cells to MTX by downregulation of both Though sharing many similarities, leukemia is genetically DHFR and cell cycle promotor cyclin D1. Drug resistance and epigenetically different from solid tumor. Besides, the also gives rise to major obstacles in applying MTX to treat emerging trend of using metabolic strategies to treat leu- T-ALL [55]. Thus, individual and personalized treatment kemia is more or less enlightened by the metabolic treat- based on molecular diagnosis should be provided to the ment in solid tumor. Since leukemia originates from a patients who are possibly resistant to MTX. In general, small subset of cell population called LSCs, it is critical to although most of the current investigations of folate figure out the metabolic differences between LSCs and metabolism focus on T-ALL treatment, future directions HSCs and further develop strategies by specifically tar- would be extended to other forms of hematological geting LSCs. Similar to their normal counterparts, LSCs malignancies to advance the understanding of folate are orchestrated in a high hierarchy which is from stem metabolism in leukemia. cells, progenitor cells to more differentiated cell lineages. The metabolic profiles might depend on diverse cell lin- eages that determine the LSC origin in distinct leukemias. Oxidative Stress Metabolism Therefore it is not difficult to understand why different type of leukemia would prefer different ways of utilizing energy Role in AML and CML resources. Moreover, microenvironment could also exert an influence on the leukemia cells and is constantly reshaping Oxidative stress and OXPHOS are interconnected: their ‘‘metabolic behavior’’. Due to the complicated niche OXPHOS occurs in mitochondrial, induces oxidative stress where the leukemia cells reside in, it is essential to dig into [56], which in turn reduces OXPHOS flux [57]. Reactive the composition of the niche for each type of leukemia, oxygen species (ROS) is generated during oxidative stress thus figure out more clearly how the niche changes the and plays an important role in regulating cell growth, ‘‘metabolic behavior’’ in distinct leukemia cells. In other division, differentiation and apoptosis. A moderate level of words, it is critical to take the context-dependent deter- is essential to maintain the survival of the cancer cells. minants into consideration in attempts to study metabo- However, the LSCs might be vulnerable to high ROS lisms in diverse leukemias. To summarize, a better condition, and upregulation of ROS level might even understanding of the connection between the cell origin induce the differentiation of leukemia cells. For example, and their metabolic status, and the function of each leu- Zhou et al. showed that 3-Deazaneplanocin A (DZNep), a kemia niche would better map the metabolic profiles in histone methyltransferase inhibitor, downregulates poly- distinct hematological malignancies. comb-repressive complex 2 (PRC2) and induces cell With regards to specific targeting strategies based on the apoptosis by enhancing the oxidative stress in primary metabolic profiles in distinct hematological malignancies, AML samples [58]. Recently, a study from Doshi et al. [59] high throughout sequencing and metabolomics would show indicated that by jointed using pan-Pim kinase inhibitor their power in discovering more potential targets. For AZD1208 and 2 inhibitors significantly example, by comparing different metabolic profiles in the 123 632 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 normal human CD34? cord blood samples and the primary 4. Schulze A, Harris AL (2012) How cancer metabolism is tuned for AML patient samples, one can do an analytic selection to proliferation and vulnerable to disruption. Nature 491(7424):364–373 target the most potential molecule or molecule combina- 5. Boroughs LK, DeBerardinis RJ (2015) Metabolic pathways pro- tion. Drug inhibition of the metabolic signaling pathways moting cancer cell survival and growth. Nat Cell Biol and molecules has been implicated in clinical treatment of 17(4):351–359 leukemias but it is also noteworthy that the normal blood 6. Kruiswijk F, Labuschagne CF, Vousden KH (2015) p53 in sur- vival, death and metabolic health: a lifeguard with a licence to cells should be spared when designing small inhibitors for kill. Nat Rev Mol Cell Biol 16(7):393–405 the potential targets. And sometimes combinatorial use of 7. Jeon SM, Chandel NS, Hay N (2012) AMPK regulates NADPH drugs would potentiate the efficiency in eliminating the homeostasis to promote tumour cell survival during energy stress. leukemia cells and even the LSCs. Moreover, drug depri- Nature 485(7400):661–665 8. Martinez-Outschoorn UE, Peiris-Pages M, Pestell RG, Sotgia F, vation contributes to metabolic compensation, which is Lisanti MP (2017) Cancer metabolism: a therapeutic perspective. recognized as a metabolic adaption and reprogramming. Nat Rev Clin Oncol 14(2):113 Combined modulation of related metabolic signaling 9. DeBerardinis RJ, Cheng T (2010) Q’s next: the diverse functions pathways is indispensable because leukemia cells would of glutamine in metabolism, cell biology and cancer. Oncogene 29(3):313–324 bypass or utilize other pathways in order to better prolif- 10. Kumazaki M, Shinohara H, Taniguchi K, Takai T, Kuranaga Y, erate and survive. Finally, figuring out novel functional Sugito N et al (2016) Perturbation of the Warburg effect increases sites of metabolic targets would be helpful of targeting the sensitivity of cancer cells to TRAIL-induced cell death. Exp genetic mutation induced drug resistance. Finally mapping Cell Res 347(1):133–142 11. Wang Y-H, Israelsen William J, Lee D, Yu Vionnie WC, Jeanson a whole and comprehensive metabolic profiles of distinct Nathaniel T, Clish Clary B et al (2014) Cell-state-specific hematological malignancies is the first and most critical metabolic dependency in hematopoiesis and leukemogenesis. step to address all of the issues that are constantly bewil- Cell 158(6):1309–1323 dering us. 12. Saito Y, Chapple RH, Lin A, Kitano A, Nakada D (2015) AMPK protects leukemia-initiating cells in myeloid leukemias from metabolic stress in the bone marrow. Cell Stem Cell Acknowledgements This study was supported by grants from Project 17(5):585–596 of Shanghai Municipal Health Bureau (Surface Program, 13. Chen WL, Wang JH, Zhao AH, Xu X, Wang YH, Chen TL et al SHXH201402). (2014) A distinct glucose metabolism signature of acute myeloid leukemia with prognostic value. Blood 124(10):1645–1654 Authors’ Contribution All authors read and approved the final 14. Larrue C, Saland E, Vergez F, Serhan N, Delabesse E, Mansat-De manuscript. BL designed the manuscript and TL and XP wrote the Mas V et al (2015) Antileukemic activity of 2-deoxy-D-glucose review. through inhibition of N-linked glycosylation in acute myeloid leukemia with FLT3-ITD or c-KIT mutations. Mol Cancer Ther Compliance with Ethical Standards 14(10):2364–2373 15. Song K, Li M, Xu X, Xuan LI, Huang G, Liu Q (2016) Resistance Conflict of interest The authors declare that they have no conflict of to chemotherapy is associated with altered glucose metabolism in interest. acute myeloid leukemia. Oncol Lett 12(1):334–342 16. Lagadinou ED, Sach A, Callahan K, Rossi RM, Neering SJ, Ethics approval and consent to participate All authors are com- Minhajuddin M et al (2013) BCL-2 inhibition targets oxidative pliant with ethical standards. phosphorylation and selectively eradicates quiescent human leu- kemia stem cells. Cell Stem Cell 12(3):329–341 Consent for publication All authors approve the manuscript for 17. Kishton RJ, Barnes CE, Nichols AG, Cohen S, Gerriets VA, publication. Siska PJ et al (2016) AMPK is essential to balance glycolysis and mitochondrial metabolism to control T-all cell stress and survival. Availability of data and materials Data and materials related to this Cell Metab 23(4):649–662 work are available upon request. 18. Liu T, Kishton RJ, Macintyre AN, Gerriets VA, Xiang H, Liu X et al (2014) Glucose transporter 1-mediated glucose uptake is limiting for B-cell acute lymphoblastic leukemia anabolic meta- References bolism and resistance to apoptosis. Cell Death Dis 5(10):e1470 19. Martinez Marignac VL, Smith S, Toban N, Bazile M, Aloyz R (2013) Resistance to dasatinib in primary chronic lymphocytic 1. Ito K, Bernardi R, Morotti A, Matsuoka S, Saglio G, Ikeda Y leukemia lymphocytes involves AMPK-mediated energetic re- (2008) PML targeting eradicates quiescent leukaemia-initiating programming. Oncotarget 4(12):2550–2566 cells. Nature 453(7198):1072–1078 20. Gottschalk S, Anderson N, Hainz C, Eckhardt SG, Serkova NJ 2. Holtz M, Forman SJ, Bhatia R (2007) Growth factor stimulation (2004) Imatinib (STI571)-mediated changes in glucose metabo- reduces residual quiescent chronic myelogenous leukemia pro- lism in human leukemia BCR-ABL-positive cells. Clin Cancer genitors remaining after imatinib treatment. Can Res Res 10(19):6661–6668 67(3):1113–1120 21. Bauer DE, Hatzivassiliou G, Zhao F, Andreadis C, Thompson CB 3. Cairns RA, Harris IS, Mak TW (2011) Regulation of cancer cell (2005) ATP citrate lyase is an important component of cell metabolism. Nat Rev Cancer 11(2):85–95 growth and transformation. Oncogene 24(41):6314–6322

123 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634 633

22. Chajes V, Cambot M, Moreau K, Lenoir GM, Joulin V (2006) 39. Goto M, Miwa H, Shikami M, Tsunekawa-Imai N, Suganuma K, Acetyl-CoA carboxylase alpha is essential to breast cancer cell Mizuno S et al (2014) Importance of glutamine metabolism in survival. Can Res 66(10):5287–5294 leukemia cells by energy production through TCA cycle and by 23. Clem BF, Clem AL, Yalcin A, Goswami U, Arumugam S, Telang redox homeostasis. Cancer Investig 32(6):241–247 S et al (2011) A novel small molecule antagonist of choline 40. Emadi A, Jun SA, Tsukamoto T, Fathi AT, Minden MD, Dang kinase-alpha that simultaneously suppresses MAPK and PI3K/ CV (2014) Inhibition of glutaminase selectively suppresses the AKT signaling. Oncogene 30(30):3370–3380 growth of primary acute myeloid leukemia cells with IDH 24. Flavin R, Peluso S, Nguyen PL, Loda M (2010) Fatty acid syn- mutations. Exp Hematol 42(4):247–251 thase as a potential therapeutic target in cancer. Future Oncol 41. Herranz D, Ambesi-Impiombato A, Sudderth J, Sanchez-Martin 6(4):551–562 M, Belver L, Tosello V et al (2015) Metabolic reprogramming 25. Mulvihill MM, Nomura DK (2013) Therapeutic potential of induces resistance to anti-NOTCH1 therapies in T cell acute monoacylglycerol inhibitors. Life Sci 92(8–9):492–497 lymphoblastic leukemia. Nat Med 21(10):1182–1189 26. Medina EA, Oberheu K, Polusani SR, Ortega V, Velagaleti GV, 42. van Geldermalsen M, Wang Q, Nagarajah R, Marshall AD, Oyajobi BO (2014) PKA/AMPK signaling in relation to adipo- Thoeng A, Gao D et al (2016) ASCT2/SLC1A5 controls glu- nectin’s antiproliferative effect on multiple myeloma cells. Leu- tamine uptake and tumour growth in triple-negative basal-like kemia 28(10):2080–2089 breast cancer. Oncogene 35(24):3201–3208 27. Kim MK, Yang S, Lee KH, Um JH, Liu M, Kang H et al (2011) 43. Polet F, Martherus R, Corbet C, Pinto A, Feron O (2016) Inhi- Promyelocytic leukemia inhibits adipogenesis, and loss of bition of glucose metabolism prevents glycosylation of the glu- promyelocytic leukemia results in fat accumulation in mice. Am J tamine transporter ASCT2 and promotes compensatory LAT1 Physiol Endocrinol Metab 301(6):E1130–E1142 upregulation in leukemia cells. Oncotarget 7(29):46371–46383 28. Yasugi E, Horiuchi A, Uemura I, Okuma E, Nakatsu M, Saeki K 44. Sontakke P, Koczula KM, Jaques J, Wierenga AT, Brouwers-Vos et al (2006) Peroxisome proliferator-activated receptor gamma AZ, Pruis M et al (2016) Hypoxia-like signatures induced by ligands stimulate myeloid differentiation and lipogenensis in BCR-ABL potentially alter the glutamine uptake for maintaining human leukemia NB4 cells. Dev Growth Differ 48(3):177–188 oxidative phosphorylation. PLoS ONE 11(4):e0153226 29. Le Y, Fraineau S, Chandran P, Sabloff M, Brand M, Lavoie JR 45. Polet F, Corbet C, Pinto A, Rubio LI, Martherus R, Bol V et al et al (2016) Adipogenic mesenchymal stromal cells from bone (2016) Reducing the serine availability complements the inhibi- marrow and their hematopoietic supportive role: towards under- tion of the glutamine metabolism to block leukemia cell growth. standing the permissive marrow microenvironment in acute Oncotarget 7(2):1765–1776 myeloid leukemia. Stem Cell Rev 12(2):235–244 46. Alachkar H, Fulton N, Sanford B, Malnassy G, Mutonga M, 30. Samudio I, Fiegl M, Andreeff M (2009) Mitochondrial uncou- Larson RA et al (2017) Expression and polymorphism (rs4880) of pling and the Warburg effect: molecular basis for the repro- mitochondrial superoxide (SOD2) and asparaginase gramming of cancer cell metabolism. Can Res 69(6):2163–2166 induced hepatotoxicity in adult patients with acute lymphoblastic 31. Ricciardi MR, Mirabilii S, Allegretti M, Licchetta R, Calarco A, leukemia. Pharmacogenomics J 17(3):274–279 Torrisi MR et al (2015) Targeting the leukemia cell metabolism 47. Parmentier JH, Maggi M, Tarasco E, Scotti C, Avramis VI, by the CPT1a inhibition: functional preclinical effects in leuke- Mittelman SD (2015) Glutaminase activity determines cytotoxi- mias. Blood 126(16):1925–1929 city of L-asparaginases on most leukemia cell lines. Leuk Res 32. Ye H, Adane B, Khan N, Sullivan T, Minhajuddin M, Gasparetto 39(7):757–762 M et al (2016) Leukemic stem cells evade chemotherapy by 48. Willems L, Jacque N, Jacquel A, Neveux N, Maciel TT, Lambert metabolic adaptation to an adipose tissue niche. Cell Stem Cell M et al (2013) Inhibiting glutamine uptake represents an attrac- 19(1):23–37 tive new strategy for treating acute myeloid leukemia. Blood 33. Wu Y, Hurren R, MacLean N, Gronda M, Jitkova Y, Sukhai MA 122(20):3521–3532 et al (2015) Carnitine transporter CT2 (SLC22A16) is over-ex- 49. Chan WK, Lorenzi PL, Anishkin A, Purwaha P, Rogers DM, pressed in acute myeloid leukemia (AML) and target knockdown Sukharev S et al (2014) The glutaminase activity of L-asparagi- reduces growth and viability of AML cells. Apoptosis Int J nase is not required for anticancer activity against ASNS-negative Program Cell Death 20(8):1099–1108 cells. Blood 123(23):3596–3606 34. Lee EA, Angka L, Rota SG, Hanlon T, Mitchell A, Hurren R et al 50. Chien WW, Le Beux C, Rachinel N, Julien M, Lacroix CE, Allas (2015) Targeting mitochondria with avocatin B induces selective S et al (2015) Differential mechanisms of asparaginase resistance leukemia cell death. Can Res 75(12):2478–2488 in B-type acute lymphoblastic leukemia and malignant natural 35. Velez J, Pan R, Lee JT, Enciso L, Suarez M, Duque JE et al killer cell lines. Sci Rep 5:8068 (2016) Biguanides sensitize leukemia cells to ABT-737-induced 51. Lynn RC, Poussin M, Kalota A, Feng Y, Low PS, Dimitrov DS apoptosis by inhibiting mitochondrial electron transport. Onco- et al (2015) Targeting of folate receptor beta on acute myeloid target 7(32):51435–51449 leukemia blasts with chimeric antigen receptor-expressing T 36. Messmer D, Lorrain K, Stebbins K, Bravo Y, Stock N, Cabrera G cells. Blood 125(22):3466–3476 et al (2015) A selective novel peroxisome proliferator-activated 52. Uchiyama H, Sowa Y, Wakada M, Yogosawa M, Nakanishi R, receptor (PPAR)-alpha antagonist induces apoptosis and inhibits Horinaka M et al (2010) Cyclin-dependent kinase inhibitor proliferation of CLL cells in vitro and in vivo. Mol Med SU9516 enhances sensitivity to methotrexate in human T-cell 21:410–419 leukemia Jurkat cells. Cancer Sci 101(3):728–734 37. Shinohara H, Kumazaki M, Minami Y, Ito Y, Sugito N, Kuranaga 53. Dulucq S, St-Onge G, Gagne V, Ansari M, Sinnett D, Labuda D Y et al (2016) Perturbation of energy metabolism by fatty-acid et al (2008) DNA variants in the dihydrofolate reductase gene and derivative AIC-47 and imatinib in BCR-ABL-harboring leukemic outcome in childhood ALL. Blood 111(7):3692–3700 cells. Cancer Lett 371(1):1–11 54. Teachey DT, Sheen C, Hall J, Ryan T, Brown VI, Fish J et al 38. Jacque N, Ronchetti AM, Larrue C, Meunier G, Birsen R, (2008) mTOR inhibitors are synergistic with methotrexate: an Willems L et al (2015) Targeting glutaminolysis has antileukemic effective combination to treat acute lymphoblastic leukemia. activity in acute myeloid leukemia and synergizes with BCL-2 Blood 112(5):2020–2023 inhibition. Blood 126(11):1346–1356 55. Wojtuszkiewicz A, Peters GJ, van Woerden NL, Dubbelman B, Escherich G, Schmiegelow K et al (2015) Methotrexate

123 634 Indian J Hematol Blood Transfus (Oct-Dec 2019) 35(4):625–634

resistance in relation to treatment outcome in childhood acute increased DNA damage and oxidative stress. Oncotarget lymphoblastic leukemia. J Hematol Oncol 8:61 7(30):48280–48295 56. Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, 60. Liu J, Masurekar A, Johnson S, Chakraborty S, Griffiths J, Smith Lopez M et al (2010) Mitochondrial metabolism and ROS gen- D et al (2015) Stromal cell-mediated mitochondrial redox adap- eration are essential for Kras-mediated tumorigenicity. Proc Natl tation regulates drug resistance in childhood acute lymphoblastic Acad Sci USA 107(19):8788–8793 leukemia. Oncotarget 6(40):43048–43064 57. Sena LA, Chandel NS (2012) Physiological roles of mitochon- 61. Singh AK, Awasthi D, Dubey M, Nagarkoti S, Kumar A, drial reactive oxygen species. Mol Cell 48(2):158–167 Chandra T et al (2016) High oxidative stress adversely affects 58. Zhou J, Bi C, Cheong LL, Mahara S, Liu SC, Tay KG et al (2011) NFkappaB mediated induction of inducible The histone methyltransferase inhibitor, DZNep, up-regulates in human neutrophils: implications in chronic myeloid leukemia. TXNIP, increases ROS production, and targets leukemia cells in Nitric Oxide 58:28–41 AML. Blood 118(10):2830–2839 59. Doshi KA, Trotta R, Natarajan K, Rassool FV, Tron AE, Huszar Publisher’s Note Springer Nature remains neutral with regard to D et al (2016) Pim kinase inhibition sensitizes FLT3-ITD acute jurisdictional claims in published maps and institutional affiliations. myeloid leukemia cells to topoisomerase 2 inhibitors through

123