Therapeutic potential of targeting mTOR in T-cell acute lymphoblastic leukemia (Review)

  • Authors:
    • Camilla Evangelisti
    • Cecilia Evangelisti
    • Francesca Chiarini
    • Annalisa Lonetti
    • Francesca Buontempo
    • Daniela Bressanin
    • Alessandra Cappellini
    • Ester Orsini
    • James A. McCubrey
    • Alberto M. Martelli
  • View Affiliations

  • Published online on: June 26, 2014     https://doi.org/10.3892/ijo.2014.2525
  • Pages: 909-918
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

T-cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous neoplastic disorder of immature hematopoietic precursors committed to the T-cell lineage. T-ALL comprises about 15% of pediatric and 25% of adult ALL cases. Even if the prognosis of T-ALL has improved especially in the childhood due to the use of new intensified treatment protocols, the outcome of relapsed patients who are resistant to conventional chemotherapeutic drugs or who relapse is still poor. For this reason, there is a need for novel and less toxic targeted therapies against signaling pathways aberrantly activated in T-ALL, such as the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR). Small molecules designed to target key components of this signaling axis have proven their efficacy both in vitro and in vivo in pre-clinical settings of T-ALL. In particular, different classes of mTOR inhibitors have been disclosed by pharmaceutical companies, and they are currently being tested in clinical trials for treating T-ALL patients. One of the most promising approaches for the treatment of T-ALL seems to be the combination of mTOR inhibitors with traditional chemotherapeutic agents. This could lead to a lower drug dosage that may circumvent the systemic side effects of chemotherapeutics. In this review, we focus on the different classes of mTOR inhibitors that will possibly have an impact on the therapeutic arsenal we have at our disposal against T-ALL.

1. Introduction

Acute lymphoblastic leukemia (ALL) is caused by the uncontrolled clonal proliferation of immature lymphoid cells which accumulate in the bone marrow (BM) and other body sites. The neoplastic lymphoblasts display an impaired differentiation program, are blocked at various maturation steps and are resistant to apoptotic stimuli and cell death. ALL accounts for approximately 20% of acute leukemias in the adult, however it is the most common malignant disease in the childhood (1). The clinical management of ALL is challenging, especially in the adults, even though current therapies can induce a complete remission in 65–90% of patients. Nevertheless, patients who are refractory to induction therapy or relapse after induction face a poor prognosis (2). ALL can be classified in two main subgroups, namely B-cell and T-cell ALL (B-ALL and T-ALL, respectively) (3).

T-ALL is an aggressive form of leukemia which arises in the thymus from T-cell progenitors expressing immature T-cell immunophenotypic markers (4,5). T-ALL accounts for 10–15% and 25% of pediatric and adult ALL, respectively. In the childhood, cure rate for T-ALL patients reaches 70–75%. In the adults, the cure rate remains low: 30–40% for adults below 60 years of age and 10% above this age (6,7). By immunophenotyping, it is possible to distinguish three subtypes of T-ALL, i.e., early, cortical and mature, which reflect different stages of healthy thymocyte differentiation. This classification is prognostically relevant, as early and mature T-ALLs have a poorer outcome than cortical T-ALL (8).

Recent findings have documented that T-ALL is an extremely heterogeneous disease, characterized by chromosomal rearrangements causing aberrant expression of transcription factors (Myb; TAL/SCL; HOX) (9,10), altered expression of oncogenes (10), somatic gene mutations (11,12), multiple signal transduction pathway impairment (1316) and microRNA dysregulation (1719).

Activating mutations in Notch-1, the master regulator of T-cell development, are found in more than 60% of T-ALL patients, independently of the subtype (20). All of these alterations impact on T-ALL cell proliferation, differentiation, survival and drug-resistance (21).

In general, leukemia pathogenesis, treatment resistance and relapse are thought to be caused by leukemic stem cells or leukemia initiating cells (LICs) (22). LICs are characterized by asymmetric cell division and self-renewal capacity, unlimited repopulating potential and production of partially differentiated cells. Whereas the bulk of leukemic cells rapidly proliferate, LICs are mainly quiescent (23). This feature is associated with chemoresistance, as conventional chemotherapy strategies mainly target rapidly dividing cells (24).

The phenotype of T-ALL LICs is still under discussion. Cox et al (25) reported that either CD34+/CD4 or CD34+/CD7 cells were capable of serial engraftment in NOD/SCID mice (25). Afterwards, the leukemia initiating potential in xenografts of the CD7+/CD1a subset of primary T-ALL samples was found to be superior to other subsets (26). The importance of CD34 as a marker of LIC activity in T-ALL patients has nevertheless been documented by independent groups (27,28). However, it has been shown that also CD34/CD7+ T-ALL cells displayed LIC proprieties, although at lower levels than CD34+ cells (28). The above-outlined discrepancies could well reflect differences among distinct molecular T-ALL subtypes. Nevertheless, these studies have disclosed the complexity of LICs in human T-ALL.

Among the deregulated signaling pathways that have been identified in T-ALL, the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling network has been reported to be active in a high percentage (75–80%) of patients, where it portends a poorer prognosis (29).

Over the last 10 years, mTOR has become an attractive therapeutic target in cancer patients, as several small molecule mTOR inhibitors have been developed and are being tested as monotherapy in clinical trials (3032). Moreover, the use of targeted drugs combined with traditional anticancer agents could increase treatment efficacy, by lowering the required dosage of chemotherapeutic drugs and limiting their systemic side effects (33). In this review, we will describe the potential of several strategies for mTOR inhibition to improve the outcome of T-ALL patients.

2. The PI3K/Akt/mTOR pathway

mTOR is a 289-kDa serine/threonine (Ser/Thr) kinase which belongs to the phosphoinositide kinase-related family of protein kinases (PIKK) (34). The PIKK family includes ataxia telangiectasia mutated (ATM), ataxia telangiectasia- and RAD3-related (ATR), human suppressor of morphogenesis in genitalia-1 (hSMG-1) and the catalytic subunit of DNA-dependent protein kinase (DNA-PK) (35).

mTOR collects input from several signal transduction networks, such as the PI3K/Akt, the Ras/Raf/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) and the AMP-activated protein kinase (AMPK) pathways, for regulating several physiological events. Indeed, mTOR is involved in cell cycle progression, cell survival, translation, metabolism, motility, autophagy and ageing (36). mTOR is the catalytic subunit of two distinct multi-protein complexes known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), both of which are characterized by their different partner proteins and their substrate specificity (36) (Fig. 1).

Figure 1

The PI3K/Akt/mTOR pathway. PI3K generates PtdIns(3,4,5)P3 from PtdIns(4,5)P2. PtdIns(3,4,5)P3 attracts to the plasma membrane PDK1 which phosphorylates Akt at Thr 308. Full Akt activation requires Ser 473 phosphorylation by mTORC2. Active Akt inhibits TSC2 activity through direct phosphorylation. TSC2 is a GTP-ase activating protein (GAP) that functions in association with TSC1 to inactivate the small G protein Rheb. Akt-driven TSC1/TSC2 complex inactivation allows Rheb to accumulate in a GTP-bound state. Rheb-GTP then upregulates mTORC1 activity. However, mTORC1 is controlled by Akt also through PRAS40 phosphorylation. The activation mechanisms of mTORC2 are not fully understood yet, but they require PI3K activity. Arrows indicate activating events, while perpendicular lines indicate inhibitory events. 4E-BP1, eukaryotic initiation factor 4E-binding protein 1; Deptor, DEP-domain-containing mTOR interacting protein; eIF4E, eukaryotic initiation factor 4E; FKBP38, FK-506 binding protein 38; mLST8, mammalian lethal-with-sec-thirteen 8; mTOR, mammalian target of rapamycin; mTORC1, mTOR complex 1; mTORC2, mTOR complex 2; PDK1, phosphoinositide-dependent kinase 1; PI3K, phosphoinositide 3-kinase; PKCα, protein kinase C α; PRAS40, proline-rich Akt substrate of 40-kDa; Protor, protein observed with Rictor; PtdIns(4,5)P2, phosphoinositide (4,5) bisphosphate; PtdIns(3,4,5)P3, phosphoinositide (3,4,5) trisphosphate; PTEN, phosphatase and tensin homolog deleted on chromosome ten; p70S6K, p70S6 kinase; Raptor, regulatory associated protein of mTOR; Rheb, Ras homolog enriched in brain; Rictor, rapamycin insensitive companion of mTOR; S6RP, S6 ribosomal protein; SGK, serum- and glucocorticoid-stimulated kinase; SIN1, stress-activated protein kinase-interacting protein 1; TSC1, tuberous sclerosis 1; TSC2, tuberous sclerosis 2.

mTORC1 is composed of the regulatory associated protein of mTOR (Raptor, a scaffolding protein), mammalian Lethal-with-Sec-Thirteen 8 (mLST8), proline-rich Akt substrate of 40-kDa (PRAS40), FK-506 binding protein 38 (FKBP38) and DEP-domain-containing mTOR interacting protein (Deptor). mTORC1 is sensitive to rapamycin and its derivatives (rapalogs) (37). Multiple exogenous stimuli regulate mTORC1 activity, including growth factors such as insulin and insulin-like growth factor-1 (IGF-1), stress signals, cellular energy status and amino acids (38).

mTORC1 activation is mainly regulated by PI3K/Akt signaling. Akt phosphorylates 200-kDa tuberous sclerosis 2 (TSC2 or hamartin). TSC2 is a GTPase-activating protein (GAP) that associates with TSC1 (tuberous sclerosis 1 or tuberin) for inactivating the small G protein Rheb (Ras homolog enriched in brain). Once TSC2 is phosphorylated by Akt, the GAP activity of the TSC1/TSC2 complex is repressed, allowing Rheb to accumulate in a GTP-bound state. As a consequence, Rheb-GTP upregulates the protein kinase activity of mTORC1 (39). Furthermore, Akt phosphorylates PRAS40 at Thr246. Phosphorylated PRAS40 dissociates from mTORC1 in response to growth factors, as well as glucose and nutrients, and thereby releases the inhibitory function of PRAS40 on mTORC1 (40).

mTORC1 positively regulates cell growth and proliferation by promoting many anabolic processes and by limiting catabolic processes such as autophagy (41) (Fig. 1). Regarding protein translation, mTORC1 phosphorylates components of the protein synthesis machinery, such as p70 S6 kinase (p70S6K) and eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1). In turn, p70S6K phosphorylates the 40S ribosomal protein S6 (S6RP), leading to active translation of mRNA involved in ribosome biogenesis (42), while 4E-BP1 interacts with the eukaryotic initiation factor 4E (eIF4E), which critically regulates cap-dependent mRNA translation (43). Once phosphorylated by mTORC1, 4E-BP1 releases eIF4E, which then associates with eIF4G to stimulate translation initiation (44,45).

In addition to its role in protein translation, activation of mTORC1 triggers metabolic changes that are critically important in carcinogenesis, such as mitochondrial biogenesis and oxidative metabolism, aerobic glycolysis and de novo lipogenesis (41). mTORC1 controls mitochondrial biogenesis and oxidative metabolism by regulating the interactions between the transcription factor yin-yang 1 (YY1) and the peroxisomal proliferator-activated receptor γ (PPARγ) coactivator 1 (PGC-1), thereby preventing the coactivation of YY1 (46).

As far as aerobic glycolysis is concerned, mTORC1 promotes it through induction of a transcriptional program affecting metabolic glycolytic gene targets of hypoxia-inducible factor 1α (HIF1α) (47,48).

Regarding lipid synthesis, mTORC1 activates the transcription factors sterol regulatory element binding protein 1 (SREBP1) and PPARγ which are necessary and sufficient for the differentiation of preadipocytes and lipid accumulation (41).

mTORC1 negatively regulates autophagy, a complex catabolic process that sustains cellular metabolism through recycling of cellular components during growth unfavorable conditions. Nevertheless, autophagy has also been associated with promoting cell survival during nutrient or hypoxic stress and may promote cancer cell survival (49). mTORC1 suppresses the kinase activity of unc-51-like kinase 1 (ULK1), thus preventing the ULK1/autophagy-related gene 13 (Atg13)/FIP200 complex formation (50) that plays an essential role at the early stages of autophagosome formation (51).

mTORC2 comprises rapamycin-insensitive companion of mTOR (Rictor), mLST8, stress-activated protein kinase-interacting protein 1 (SIN1), protein observed with Rictor (Protor), and Deptor, and is generally described as being insensitive to rapamycin/rapalogs. Nevertheless, it has been demonstrated that long-term rapamycin treatment leads to dissociation of mTORC2 with resulting inhibition of Akt feedback phosphorylation at Ser 473, in primary leukemic cells both in vitro and in vivo (52). mTORC2 is mainly activated by growth factors through PI3K/Akt, and controls several downstream AGC kinases such as Akt itself, serum- and glucocorticoid stimulated kinase (SGK) and protein kinases Cα (PKCα) (5355) (Fig. 1). Therefore, mTORC2 regulates cell proliferation, but it is also involved in the spatial control of cell growth via cytoskeleton regulation, through actin fibers, paxillin, RhoA, Rac1 and PKCα (56).

The regulation of PI3K/Akt/mTOR axis is extremely complex, and this is due mainly to the existence of multiple feedback loops and direct activation mechanisms that place mTOR both upstream and downstream of several oncogenic pathways. Importantly, these regulation loops are relevant in vivo and influence therapeutic responses based on mTOR inhibition, contributing to the drug-resistance that can occur in mTOR-targeted therapies using rapamycin or rapalogs (45). When Akt activates mTORC1, a negative feedback circuit antagonizes the formation of mTORC2 and reduces Akt activity (57). Moreover, when activated, mTORC1 phosphorylates p70S6K, which in turn inhibits insulin receptor substrate 1 (IRS-1) by phosphorylating it at multiple sites (Ser 307 and Ser 636/639), inducing its degradation and altering its localization, all of which ultimately dampen PI3K/Akt/mTORC1 activation (5861). mTORC1 is also capable of downregulating IRS-2 expression by enhancing its proteosomal degradation (62). Recent findings have also highlighted the existence of a rapamycin-sensitive, mTORC1/p70S6K-mediated phosphorylation of Rictor on Thr1135. This phosphorylative event exerts a negative regulatory effect on the mTORC2-dependent phosphorylation of Akt at Ser 473 in vivo (63).

PI3K/Akt/mTOR signaling is antagonized by phosphatases. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a potent repressor of this pathway that removes 3′-phosphate from phosphoinositide (3,4,5) trisphosphate [PtdIns(3,4,5)P3] to yield PtdIns (4,5) bisphosphate [PtdIns(4,5)P2] (64), thus counterbalancing the action of PI3K (Fig. 1). Loss of PTEN, due to inactivating mutations or silencing, has been reported in a wide range of sporadic human cancers, including leukemias, and it has been correlated to cellular proliferation, cancer susceptibility and tumor progression (65). PTEN plays an important role in T-ALL pathophysiology (see below).

The lipid phosphatases, Src homology domain-containing inositol phosphatase (SHIP) 1 and 2, remove 5′-phosphate from PtdIns(3,4,5)P3 to yield PtdIns(3,4)P2 (66), and play a fundamental role in the inhibition of proliferation and survival of hematopoietic cells (67). Mutations of SHIP1, that is predominantly expressed in hematopoietic cells, have been implicated in the development of different blood disorders, including T-ALL (68). Also protein phosphatases, such as protein phosphatase 2A (PP2A), impact on PI3K/Akt/mTOR signaling, as PP2A dephosphorylates Akt at Thr308 (69).

3. Disregulated mTOR activity and T-ALL development

It is established that PTEN deletion led to T-ALL development in mice (70) and that rapamycin treatment of preleukemic mice prevented LIC formation and halted T-ALL development (71). Both mTORC1 and mTORC2 have been implicated in T-ALL pathophysiology. Regarding mTORC1, it has been documented that loss of mTORC1 activity caused by Raptor deficiency, eradicated T-ALL in a murine model of disease, suggesting that mTORC1 played a key role in T-ALL LIC survival (72). However, rapamycin was not sufficient for T-ALL eradication. This could be due to the fact that rapamycin is an incomplete blocker of mTORC1 outputs (73). Therefore, dual PI3K/mTOR inhibitors or ATP-competitive mTORC1/mTORC2 inhibitors (see below) could be more effective agents against T-ALL, as they efficiently targeted rapamycin-resistant mTORC1 activity in T-ALL cells (7476).

An important role for mTORC2 in T-ALL development is suggested by the findings of another group (77). It was documented that deletion of the mTORC2 component, Rictor, prevented leukemogenesis and hematopoietic stem cell (HSC) depletion after PTEN deletion in adult mice. These observations implicated an important role for mTORC2 activation in these processes. However, Rictor deletion (and hence mTORC2 function inactivation) had little effect on the physiology of healthy (i.e., non-PTEN-deleted) HSCs. Moreover, PTEN deletion from neonatal HSCs did not activate PI3K/Akt signaling or promote HSC proliferation/depletion or leukemogenesis. Therefore, it was concluded that PTEN is required in adult, but not neonatal, HSCs for inhibiting mTORC2 signaling downstream of PI3K/Akt (77). These findings could explain why B-ALL, where PTEN deletions are very uncommon (78), is a disease of the early childhood with a peak incidence at 2–5 years of age (79), whereas pediatric T-ALL, in which PTEN deletion/inactivation is quite frequently observed (80), displays an older mean age of presentation (approximately 9–10 years) (81).

4. Causes for PI3K/Akt/mTOR pathway activation in T-ALL

PI3K/Akt/mTOR pathway aberrant activation is a common feature in T-ALL, being detectable in 70–85% of the patients (82) and is associated with a poorer outcome (80,83).

Mutations in PI3K, Akt and PTEN have been described in T-ALL patients. Collectively, they were found in about 50% of 44 T-ALL samples (84). However, while PI3K or Akt mutations are extremely rare (two and one case, respectively, in the above mentioned study), PTEN mutations occur more frequently in both adult and pediatric T-ALL (85,86). In adults, PTEN mutations have been identified in 10% of patients in a study in which 90 T-ALL cases were analyzed (87), whereas in children, PTEN was found mutated in 52 out of 301 (17.3%) patients (85). However, some PTEN mutations affected exon 7, and were predicted to truncate the C2 domain without disrupting the lipid phosphatase domain of PTEN (84). Therefore, these mutations should not impact on PI3K/Akt/mTOR signaling, even though this has never been documented.

Moreover, PTEN could be either deleted (84) or repressed due to several mechanisms. In T-ALLs displaying Notch-1 activation (50–60% of cases), PTEN could be repressed through the hairy enhancer of Split-1 (HES-1), a downstream target of Notch-1 signaling (88). Another Notch-1 target gene which negatively impacts on PTEN expression is c-Myc (89,90). Overexpression of miR-19 has also been documented in T-ALL patients and resulted in lower expression of several genes controlling the PI3K/Akt/mTOR cascade, including PTEN (91).

However, in most pediatric T-ALL clinical samples, PTEN is expressed, but displays elevated phosphorylation at the C-terminal Ser/Thr cluster, due to phosphorylation by casein kinase 2 (CK2), and/or oxidation by reactive oxygen species (ROS). Phosphorylation and/or oxidation resulted in PTEN stabilization and functional inactivation, with ensuing overactivation of PI3K/Akt/mTOR signaling (92). Decreased activity of PP2A on Thr308 p-Akt could also account for PI3K/Akt/mTOR upregulation in PTEN-null T-ALL cells (93).

IGF-1/IGF-1R signaling plays an important role in the activation of the PI3K/Akt/mTOR cascade in T-ALL cells. Indeed, pharmacologic inhibition or genetic deletion of IGF-1R negatively affected T-ALL cell proliferation and survival (94). Interestingly, IGF-1R is a Notch-1 target gene and Notch-1 was required to maintain IGF-1R expression at high levels in T-ALL cells. Furthermore, a moderate decrease in IGF1-R signaling compromised T-ALL LIC activity (94).

Cytokines produced by the thymic/BM microenvironment, including interleukin (IL)-4 (95) and IL-7 (96), could be involved in upregulation of PI3K/Akt/mTOR signaling in T-ALL. An important source for IL-7 could be represented by thymic epithelial cells (97). In this connection, it has been recently reported that ROS, produced through IL-7 signaling, are critical for activating PI3K/Akt/mTOR which in turn mediates proliferation and survival of T-ALL cells (29). However, in T-ALL patients, increased signaling downstream of the IL-7 receptor α chain (IL-7Rα) could also be a consequence of gain-of-function IL-7Rα mutations, which were detected in about 9% of pediatric T-ALL patients (98).

CXC chemokine ligand 12 (CXCL12), referred to as SDF-1α (stromal cell-derived factor 1α), the ligand for the CXC chemokine receptor 4 (CXCR4), is another cytokine with the potential for activating PI3K/Akt/mTOR signaling (99). CXCL12 is produced by BM stromal cells in T-ALL patients (100) and it has been recently demonstrated to be involved in PI3K/Akt/mTOR activation and drug-resistance in T-ALL cells (101).

5. mTOR inhibitors

We will summarize the three main classes of mTOR inhibitors that have been tested in pre-clinical models and/or entered clinical trials for treatment of T-ALL: rapamycin/rapalogs that are allosteric mTORC1 inhibitors; dual PI3K/mTOR inhibitors that target both PI3K and mTORC1/mTORC2; ATP-competitive, ‘active-site’ mTORC1/mTORC2 inhibitors that target the catalytic site of mTOR (Fig. 2).

Rapamycin/rapalogs

Rapamycin (sirolimus), a natural compound discovered from the bacterium Streptomyces hygroscopicus in the Easter Island more than 30 years ago, is an allosteric mTORC1 inhibitor that at first interacts with the intracellular protein, FK506 binding protein 12 (FKBP12) (102). The rapamycin/FKBP12 complex results in the dissociation of Raptor from mTORC1 and loss of contact between mTORC1 and its substrates (103). Therefore, rapamycin does not directly target the mTOR catalytic site and does not affect mTORC2 activity, except in some cell types after prolonged exposure (52). Since no other cellular protein has been identified as rapamycin targets and a cofactor (i.e., FKBP12) is required, rapamycin is a very selective mTORC1 inhibitor. Rapamycin is now FDA-approved as an immunosuppressive agent in solid organ transplantation.

Rapamycin derivatives (rapalogs) display an improved bioavailability when compared to rapamycin, and include CCI-779 (temsirolimus), RAD001 (everolimus) and AP23573 (ridaforolimus). The orally available RAD001 is more efficacious than rapamycin, as it has a higher affinity to FKBP12 (104).

Rapamycin has been tested in vitro in pre-clinical models of T-ALL, where it induced apoptosis and/or cell cycle arrest and synergized with chemotherapeutic drugs (doxorubicin, idarubicin, dexamethasone) (105107). Interestingly, rapamycin synergized with the glycolysis inhibitor, 3-BrOP in T-ALL cell lines, where the combined treatment induced apoptosis (108). It was concluded that when ATP is depleted by glycolysis inhibition, blocking mTORC1 may further limit nutrient uptake, which resulted in additional cytotoxicity.

CCI-779 was able to block in vitro IL-7-induced proliferation, survival and cell cycle progression of primary T-ALL cells, and synergized with both doxorubicin and dexamethasone (109).

The Pediatric Preclinical Testing Program (PPTP) evaluated rapamycin against T-ALL cell lines and xenografts. Rapamycin induced regression in the two T-cell ALL xenografts studied during PPTP (110).

However, the efficacy of rapamycin/rapalogs as broad-based monotherapies for acute leukemia treatment has not been as promising as initially expected. Indeed, several mechanisms emerged as barriers to anti-leukemic activity of this class of mTORC1 inhibitors which could explain the mostly disappointing results of clinical trials (111,112).

The rapamycin/rapalog modest effects on leukemic cells, could be due to several reasons. Firstly, these drugs have only a poor pro-apoptotic activity, being mainly cytostatic. Secondly, they do not target all the mTORC1 outputs. In particular, phosphorylation of 4E-BP1 is usually resistant to rapamycin/rapalogs (73,113,114). This is a very critical issue, as 4E-BP1 controls the cap-dependent translation of mRNAs coding for critical factors which regulates cell survival and proliferation in cancer cells. These include c-Myc, cyclin-dependent kinase-2 (CDK-2), cyclin D1, signal activator and transducer of transcription-3 (STAT-3), B-cell lymphoma 2 (Bcl-2), Bcl-xL, survivin, myeloid cell leukemia-1 (Mcl-1), ornithine decarboxylase (45,82). Thirdly, upregulation of PIM protein kinase activity has been shown to contribute to resistance to rapamycin (115). Indeed, PIM1 protein kinase phosphorylates PRAS40 at the same amino acidic residue (Thr246) as Akt and, by doing so, activates mTORC1 (116). Moreover, PIM2 protein kinase phosphorylated 4E-BP1 at Ser 65 residue and this phosphorylative event was documented to be essential for oncogenic protein translation independent of mTORC1 activity in acute myelogenous leukemia cells (113). Interestingly, a small molecule inhibitor of PIM protein kinases (SMI-4a) was cytotoxic to T-ALL cell lines through the induction of a G1 phase cell cycle arrest, and apoptosis (117). SMI-4a treatment reduced mTORC1, but not mTORC2, activity. However, it upregulated MEK/ERK signaling, possibly due to mTORC1/p70S6K inhibition (117).

In this connection, the disappointing performances of rapamycin/rapalogs have been also ascribed to the feedback loops that lead to re-activation of either PI3K/Akt and/or MEK/ERK signaling upon mTORC1 inhibition (118121). However, it should be pointed out that the existence of these feedback loops has never been documented in T-ALL cells treated with rapamycin/rapalogs.

In agreement with pre-clinical studies, clinical trials with rapalogs combined with chemotherapy have provided more encouraging clinical results (122,123). Phase I/II clinical trials are ongoing in which CCI-779 is being tested in combination with intensive re-induction therapy (dexamethasone, mitoxantrone, vincristine and PEG-asparaginase) in children with relapsed T-ALL (ClinicalTrials.gov: NCT01403415). Also RAD001 has entered phase I/II clinical trials for T-ALL, in combination with standard chemotherapy regimens (ClinicalTrials.gov: NCT00968253; NCT01523977; NCT01403415).

Dual PI3K/mTOR inhibitors

PI3K and mTOR belong to the PIKK family of kinases, and share high sequence homology in their catalytic domains. Dual PI3K/mTOR inhibitors are ATP-competitive inhibitors that target the active sites of both the holoenzymes. The first compound of this class to be disclosed was the morpholinoquinazoline derivative, PI-103 (124). Dual PI3K/mTOR inhibitors downregulate signaling both upstream and downstream of Akt, thus avoiding the issue of Akt re-activation which follows mTORC1 inhibition. These compounds are more powerful apoptotic inducers than rapamycin/rapalogs and inhibit rapamycin-resistant mTORC1 outputs (125,126). They also target mTORC2 activity (127). PI-103 was cytotoxic to T-ALL cell lines and patient samples, where it inhibited 4E-BP1 phosphorylation, as well as oncogenic protein translation, more efficiently than rapamycin (74,75). Interestingly, Shepherd and coworkers have documented that PI-103 treatment of T-ALL cell lines with activating Notch-1 mutations, caused a compensatory upregulation of Notch-1 signaling, as demonstrated by increased levels of c-Myc (128). PI-103 and a γ-secretase inhibitor (compound E, which targets Notch-1 signaling) synergized in inducing T-ALL cell death, thus providing a rational basis for the use of drug combinations that target both the signaling networks (128). Although PI-103 displayed low toxicity and was well tolerated in mouse xenografts (124), it did not enter clinical trials, mainly because of its rapid in vivo metabolism (129).

NVP-BEZ235 is an orally bioavailable imidazoquinoline dual PI3K/mTOR inhibitor (130) that has entered a phase I clinical trial for relapsed/refractory ALL patients (ClinicalTrials.gov:NCT01756118). NVP-BEZ235 inhibited the proliferation and induced apoptosis in T-ALL cell lines and primary lymphoblasts (114). The drug synergized with several chemotherapeutic agents (cyclophosphamide, Ara-C, dexamethasone) currently used for treating T-ALL patients (114,131). In this connection, it is very important to emphasize that NVP-BEZ235 also inhibited DNA-PK and ATM/ATR kinases, that are key players of DNA damage response (DDR) (132). Chemotherapeutic drugs, such as Ara-C and doxorubicin, induce DDR and activate ATR (17,133). Therefore, abrogation of DNA repair by NVP-BEZ235 could potentiate the effects of traditional chemotherapeutic drugs.

NVP-BGT226 is another dual PI3K/mTOR inhibitor which has been tested in vitro against T-ALL cell lines and primary lymphoblasts (134). NVP-BGT226 was more powerful in inducing apoptosis than NVP-BEZ235. Nevertheless, a phase I clinical study of NVP-BTG226 in patients with advanced solid tumors, revealed that the drug displayed only a limited anti-neoplastic activity and inconsistent target inhibition. This was probably due to the fact that efficacious plasma concentrations were not achieved at the maximum safety dose (135).

The main limit of dual PI3K/mTOR inhibitors is that these drugs, by inhibiting PIKK family of kinases, could also result in more toxic side effects than rapamycin/rapalogs (136), even if they seem to be well tolerated when administered orally (137,138).

ATP-competitive mTORC1/mTORC2 inhibitors

Due to the limited success of rapalogs in the treatment of leukemia, a new generation of mTOR inhibitors, which target the ATP-binding site of mTOR and inhibit the catalytic activity of both mTORC1 and mTORC2, were developed. Acting on both mTOR complexes, these compounds display stronger effects on cell growth, proliferation and survival than rapalogs, and they offer a more efficient alternative to rapalogs in the treatment of T-ALL. Their use also minimize the re-activation feedback loops of Akt seen with rapamycin/rapalogs (139). This class of inhibitors displayed, in pre-clinical evaluations, more potent anti-leukemic effects when compared with rapamycin/rapalogs. In particular, they strongly suppressed both mTORC1-dependent phosphorylation of p70S6K and 4E-BP1 (140,141) and mTORC2-dependent phosphorylation of Akt at Ser 473, without affecting PI3K (136,142).

PP-242 was one of the first mTORC1/mTORC2 ATP- competitive inhibitors to be disclosed (143). PP-242 displayed cytotoxic activity against T-ALL and was a potent repressor of cap-dependent mRNA translation in T-ALL cells, most likely via inhibition of the rapamycin-resistant phosphorylation of 4E-BP1 (75). PP-242 has not been developed into the clinic, however its derivative, MLN0128 (formerly INK128), has entered phase I/II clinical trials for cancer patients, including hematological malignancies (e.g., ClinicalTrials.gov: NCT01058707; NCT01351350). MLN0128 displayed potent anti-leukemic activity in pre-clinical models of B-ALL (144).

Other mTORC1/mTORC2 ATP-competitive inhibitors which have been successfully tested in vitro against T-ALL cells include AZD-8055 and OSI-027 (75,76). Both of these drugs are being evaluated in clinical trials for individuals with lymphomas (ClinicalTrials.gov: NCT01194193; NCT00698243).

6. Conclusion

mTOR is activated in most T-ALL cell lines and primary samples, due to several mechanisms, which include PTEN gene deletion/suppression or PTEN protein phosphorylation/oxidation. mTOR activation confers a poorer prognosis to T-ALL patients. Both mTORC1 and mTORC2 play an important role in the pathophysiology of T-ALL, as they are involved in the proliferation/survival of T-ALL LICs.

Three main classes of mTOR inhibitors have been tested both in vitro and in vivo in pre-clinical settings of T-ALL: allosteric mTORC1 inhibitors (rapamycin/rapalogs), dual PI3K/mTOR inhibitors and ATP-competitive mTORC1/mTORC2 inhibitors. Some of these are now being tested, alone or in combination with chemotherapeutic drugs, in T-ALL patients. Therefore, in the future also T-ALL could be added to the growing list of disorders where mTOR inhibition is beneficial to patient outcome.

7. Perspectives

A growing body of evidence has documented that mTOR is a key node of the PI3K/Akt/mTOR signaling pathway, which is by far one of the most commonly upregulated signal transduction cascades in human cancer (32). The literature reviewed in this article suggests that there is a strong rationale for targeting mTOR in T-ALL, including the fact that both mTORC1 and mTORC2 are important for T-ALL LIC survival (72). These findings suggest that mTOR inhibition, by targeting LICs, has the potential for eradicating T-ALL.

Could it be possible to specifically target mTOR signaling in T-ALL LICs, without affecting the functions of healthy HSCs? Indeed, evidence suggests that mTOR is important for the biology of normal HSCs (145). However, preliminary findings have indicated that there are subtle differences in how HSCs and LICs utilize the same signaling pathways. This has been demonstrated in murine LICs treated with rapamycin (146), where the drug did not affect HSCs, while it was cytotoxic to LICs. Some of the side effects of rapamycin/rapalogs (anemia, leukopenia, thrombocytopenia) seem to indicate that this class of drugs does indeed affect normal hematopoiesis. However, these side effects are usually quite mild (147). The side effects of dual PI3K/mTOR inhibitors and of ATP-competitive mTORC1/mTORC2 inhibitors on healthy HSCs are at present not well known, although the only hematological toxicity which emerged from a phase I study of BGT-226 was anemia (148).

A major challenge in the clinical use of mTOR inhibitors remains the identification of patients who will likely respond to the treatment. For example, it has been recently documented that B-lymphoma cell lines which did not express 4E-BP1, were resistant to ATP-competitive mTORC1/mTORC2 inhibitors (149).

Additional work is therefore required to identify and confirm predictive biomarkers of constitutive/acquired resistance and sensitivity to each drug in large scale clinical trials using homogeneous patient populations (32). Future studies could also benefit from a more thorough analysis of the entire PI3K/Akt/mTOR pathway and of its cross-talk with other signal transduction networks aberrantly activated in T-ALL, including the Notch-1 pathway (85,86). All of these studies could provide the rationale for developing personalized pharmacological treatments, based on mTOR inhibitors, with or without chemotherapeutics or other targeted agents, aimed at T-ALL eradication.

Acknowledgements

This study was supported by a grant from MIUR FIRB 2010 (RBAP10447J_003) to A.M.M.

References

1 

Farhi DC and Rosenthal NS: Acute lymphoblastic leukemia. Clin Lab Med. 20:17–28. 2000.

2 

Mullighan CG: Molecular genetics of B-precursor acute lymphoblastic leukemia. J Clin Invest. 122:3407–3415. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Brown C: The genomics revolution: relevance in healthcare today and tomorrow. J R Coll Physicians Edinb. 42:248–250. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Zhao WL: Targeted therapy in T-cell malignancies: dysregulation of the cellular signaling pathways. Leukemia. 24:13–21. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Kox C, Zimmermann M, Stanulla M, et al: The favorable effect of activating NOTCH1 receptor mutations on long-term outcome in T-ALL patients treated on the ALL-BFM 2000 protocol can be separated from FBXW7 loss of function. Leukemia. 24:2005–2013. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Pui CH, Robison LL and Look AT: Acute lymphoblastic leukaemia. Lancet. 371:1030–1043. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Koch U and Radtke F: Notch in T-ALL: new players in a complex disease. Trends Immunol. 32:434–442. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Hoelzer D and Gokbuget N: T-cell lymphoblastic lymphoma and T-cell acute lymphoblastic leukemia: a separate entity? Clin Lymphoma Myeloma. 9:S214–S221. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Alharbi RA, Pettengell R, Pandha HS and Morgan R: The role of HOX genes in normal hematopoiesis and acute leukemia. Leukemia. 27:1000–1008. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Iacobucci I, Papayannidis C, Lonetti A, Ferrari A, Baccarani M and Martinelli G: Cytogenetic and molecular predictors of outcome in acute lymphocytic leukemia: recent developments. Curr Hematol Malig Rep. 7:133–143. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Bains T, Heinrich MC, Loriaux MM, et al: Newly described activating JAK3 mutations in T-cell acute lymphoblastic leukemia. Leukemia. 26:2144–2146. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Jenkinson S, Koo K, Mansour MR, et al: Impact of NOTCH1/FBXW7 mutations on outcome in pediatric T-cell acute lymphoblastic leukemia patients treated on the MRC UKALL 2003 trial. Leukemia. 27:41–47. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Blackburn JS, Liu S, Raiser DM, et al: Notch signaling expands a pre-malignant pool of T-cell acute lymphoblastic leukemia clones without affecting leukemia-propagating cell frequency. Leukemia. 26:2069–2078. 2012. View Article : Google Scholar

14 

Lhermitte L, Ben Abdelali R, Villarese P, et al: Receptor kinase profiles identify a rationale for multitarget kinase inhibition in immature T-ALL. Leukemia. 27:305–314. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Cialfi S, Palermo R, Manca S, et al: Glucocorticoid sensitivity of T-cell lymphoblastic leukemia/lymphoma is associated with glucocorticoid receptor-mediated inhibition of Notch1 expression. Leukemia. 27:485–488. 2013. View Article : Google Scholar

16 

Malyukova A, Brown S, Papa R, et al: FBXW7 regulates glucocorticoid response in T-cell acute lymphoblastic leukaemia by targeting the glucocorticoid receptor for degradation. Leukemia. 27:1053–1062. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Correia NC, Durinck K, Leite AP, et al: Novel TAL1 targets beyond protein-coding genes: identification of TAL1-regulated microRNAs in T-cell acute lymphoblastic leukemia. Leukemia. 27:1603–1606. 2013. View Article : Google Scholar

18 

Lv M, Zhang X, Jia H, et al: An oncogenic role of miR-142-3p in human T-cell acute lymphoblastic leukemia (T-ALL) by targeting glucocorticoid receptor-a and cAMP/PKA pathways. Leukemia. 26:769–777. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Schotte D, Pieters R and Den Boer ML: MicroRNAs in acute leukemia: from biological players to clinical contributors. Leukemia. 26:1–12. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Tosello V and Ferrando AA: The NOTCH signaling pathway: role in the pathogenesis of T-cell acute lymphoblastic leukemia and implication for therapy. Ther Adv Hematol. 4:199–210. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Van Vlierberghe P and Ferrando A: The molecular basis of T cell acute lymphoblastic leukemia. J Clin Invest. 122:3398–3406. 2012.PubMed/NCBI

22 

Buss EC and Ho AD: Leukemia stem cells. Int J Cancer. 129:2328–2336. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Clevers H: The cancer stem cell: premises, promises and challenges. Nat Med. 17:313–319. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Kreso A and Dick JE: Evolution of the cancer stem cell model. Cell Stem Cell. 14:275–291. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Cox CV, Martin HM, Kearns PR, Virgo P, Evely RS and Blair A: Characterization of a progenitor cell population in childhood T-cell acute lymphoblastic leukemia. Blood. 109:674–682. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Chiu PP, Jiang H and Dick JE: Leukemia-initiating cells in human T-lymphoblastic leukemia exhibit glucocorticoid resistance. Blood. 116:5268–5279. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Ma W, Gutierrez A, Goff DJ, et al: NOTCH1 signaling promotes human T-cell acute lymphoblastic leukemia initiating cell regeneration in supportive niches. PloS One. 7:e397252012. View Article : Google Scholar : PubMed/NCBI

28 

Gerby B, Clappier E, Armstrong F, et al: Expression of CD34 and CD7 on human T-cell acute lymphoblastic leukemia discriminates functionally heterogeneous cell populations. Leukemia. 25:1249–1258. 2011. View Article : Google Scholar

29 

Silva A, Girio A, Cebola I, Santos CI, Antunes F and Barata JT: Intracellular reactive oxygen species are essential for PI3K/Akt/mTOR-dependent IL-7-mediated viability of T-cell acute lymphoblastic leukemia cells. Leukemia. 25:960–967. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Benjamin D, Colombi M, Moroni C and Hall MN: Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov. 10:868–880. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Lv X, Ma X and Hu Y: Furthering the design and the discovery of small molecule ATP-competitive mTOR inhibitors as an effective cancer treatment. Expert Opin Drug Discov. 8:991–1012. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Dienstmann R, Rodon J, Serra V and Tabernero J: Picking the point of inhibition: a comparative review of PI3K/AKT/mTOR pathway inhibitors. Mol Cancer Ther. 13:1021–1031. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Steelman LS, Franklin RA, Abrams SL, et al: Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy. Leukemia. 25:1080–1094. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Memmott RM and Dennis PA: Akt-dependent and -independent mechanisms of mTOR regulation in cancer. Cell Signal. 21:656–664. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Finlay MR and Griffin RJ: Modulation of DNA repair by pharmacological inhibitors of the PIKK protein kinase family. Bioorg Med Chem Lett. 22:5352–5359. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Laplante M and Sabatini DM: mTOR signaling in growth control and disease. Cell. 149:274–293. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Zoncu R, Efeyan A and Sabatini DM: mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 12:21–35. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Fingar DC and Blenis J: Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene. 23:3151–3171. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Inoki K, Li Y, Zhu T, Wu J and Guan KL: TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 4:648–657. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Volkers M and Sussman M: mTOR/PRAS40 interaction: hypertrophy or proliferation. Cell Cycle. 12:3579–3580. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Laplante M and Sabatini DM: mTOR signaling at a glance. J Cell Sci. 122:3589–3594. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Browne GJ and Proud CG: A novel mTOR-regulated phosphorylation site in elongation f actor 2 kinase modulates the activity of the kinase and its binding to calmodulin. Mol Cell Biol. 24:2986–2997. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Ma XM and Blenis J: Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol. 10:307–318. 2009. View Article : Google Scholar : PubMed/NCBI

44 

McCubrey JA, Steelman LS, Chappell WH, et al: Mutations and deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascades which alter therapy response. Oncotarget. 3:954–987. 2012.PubMed/NCBI

45 

Martelli AM, Evangelisti C, Chappell W, et al: Targeting the translational apparatus to improve leukemia therapy: roles of the PI3K/PTEN/Akt/mTOR pathway. Leukemia. 25:1064–1079. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Cunningham JT, Rodgers JT, Arlow DH, Vazquez F, Mootha VK and Puigserver P: mTOR controls mitochondrial oxidative function through a YY1-PGC-1a transcriptional complex. Nature. 450:736–740. 2007. View Article : Google Scholar

47 

Majumder PK, Febbo PG, Bikoff R, et al: mTOR inhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation of apoptotic and HIF-1-dependent pathways. Nat Med. 10:594–601. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Yecies JL and Manning BD: Transcriptional control of cellular metabolism by mTOR signaling. Cancer Res. 71:2815–2820. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Levine B and Kroemer G: Autophagy in the pathogenesis of disease. Cell. 132:27–42. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Hosokawa N, Hara T, Kaizuka T, et al: Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol Biol Cell. 20:1981–1991. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Mizushima N: The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol. 22:132–139. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Zeng Z, Sarbassov dos D, Samudio IJ, et al: Rapamycin derivatives reduce mTORC2 signaling and inhibit AKT activation in AML. Blood. 109:3509–3512. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Sarbassov DD, Guertin DA, Ali SM and Sabatini DM: Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 307:1098–1101. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Garcia-Martinez JM and Alessi DR: mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1). Biochem J. 416:375–385. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Ikenoue T, Inoki K, Yang Q, Zhou X and Guan KL: Essential function of TORC2 in PKC and Akt turn motif phosphorylation, maturation and signalling. EMBO J. 27:1919–1931. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Oh WJ and Jacinto E: mTOR complex 2 signaling and functions. Cell Cycle. 10:2305–2316. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Tamburini J, Green AS, Chapuis N, et al: Targeting translation in acute myeloid leukemia: a new paradigm for therapy? Cell Cycle. 8:3893–3899. 2009. View Article : Google Scholar : PubMed/NCBI

58 

Shah OJ, Wang Z and Hunter T: Inappropriate activation of the TSC/Rheb/mTOR/S6K cassette induces IRS1/2 depletion, insulin resistance, and cell survival deficiencies. Curr Biol. 14:1650–1656. 2004. View Article : Google Scholar : PubMed/NCBI

59 

Bhaskar PT and Hay N: The two TORCs and Akt. Dev Cell. 12:487–502. 2007. View Article : Google Scholar : PubMed/NCBI

60 

Lang SA, Hackl C, Moser C, et al: Implication of RICTOR in the mTOR inhibitor-mediated induction of insulin-like growth factor-I receptor (IGF-IR) and human epidermal growth factor receptor-2 (Her2) expression in gastrointestinal cancer cells. Biochim Biophys Acta. 1803:435–442. 2010. View Article : Google Scholar

61 

Xu X, Sarikas A, Dias-Santagata DC, et al: The CUL7 E3 ubiquitin ligase targets insulin receptor substrate 1 for ubiquitin-dependent degradation. Mol Cell. 30:403–414. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Sriburi R, Jackowski S, Mori K and Brewer JW: XBP1: a link between the unfolded protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J Cell Biol. 167:35–41. 2004. View Article : Google Scholar : PubMed/NCBI

63 

Boulbes D, Chen CH, Shaikenov T, et al: Rictor phosphorylation on the Thr-1135 site does not require mammalian target of rapamycin complex 2. Mol Cancer Res. 8:896–906. 2010. View Article : Google Scholar : PubMed/NCBI

64 

Maehama T and Dixon JE: The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J Biol Chem. 273:13375–13378. 1998. View Article : Google Scholar : PubMed/NCBI

65 

Sansal I and Sellers WR: The biology and clinical relevance of the PTEN tumor suppressor pathway. J Clin Oncol. 22:2954–2963. 2004. View Article : Google Scholar

66 

Kalesnikoff J, Sly LM, Hughes MR, et al: The role of SHIP in cytokine-induced signaling. Rev Physiol Biochem Pharmacol. 149:87–103. 2003. View Article : Google Scholar : PubMed/NCBI

67 

Liu Q, Sasaki T, Kozieradzki I, et al: SHIP is a negative regulator of growth factor receptor-mediated PKB/Akt activation and myeloid cell survival. Genes Dev. 13:786–791. 1999. View Article : Google Scholar : PubMed/NCBI

68 

Bunney TD and Katan M: Phosphoinositide signalling in cancer: beyond PI3K and PTEN. Nat Rev Cancer. 10:342–352. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Seshacharyulu P, Pandey P, Datta K and Batra SK: Phosphatase: PP2A structural importance, regulation and its aberrant expression in cancer. Cancer Lett. 335:9–18. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Guo W, Lasky JL, Chang CJ, et al: Multi-genetic events collaboratively contribute to Pten-null leukaemia stem-cell formation. Nature. 453:529–533. 2008. View Article : Google Scholar

71 

Guo W, Schubbert S, Chen JY, et al: Suppression of leukemia development caused by PTEN loss. Proc Natl Acad Sci USA. 108:1409–1414. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Hoshii T, Kasada A, Hatakeyama T, et al: Loss of mTOR complex 1 induces developmental blockage in early T-lymphopoiesis and eradicates T-cell acute lymphoblastic leukemia cells. Proc Natl Acad Sci USA. 111:3805–3810. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Kang SA, Pacold ME, Cervantes CL, et al: mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin. Science. 341:12365662013. View Article : Google Scholar : PubMed/NCBI

74 

Chiarini F, Fala F, Tazzari PL, et al: Dual inhibition of class IA phosphatidylinositol 3-kinase and mammalian target of rapamycin as a new therapeutic option for T-cell acute lymphoblastic leukemia. Cancer Res. 69:3520–3528. 2009. View Article : Google Scholar : PubMed/NCBI

75 

Evangelisti C, Ricci F, Tazzari P, et al: Targeted inhibition of mTORC1 and mTORC2 by active-site mTOR inhibitors has cytotoxic effects in T-cell acute lymphoblastic leukemia. Leukemia. 25:781–791. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Bressanin D, Evangelisti C, Ricci F, et al: Harnessing the PI3K/Akt/mTOR pathway in T-cell acute lymphoblastic leukemia: eliminating activity by targeting at different levels. Oncotarget. 3:811–823. 2012.PubMed/NCBI

77 

Magee JA, Ikenoue T, Nakada D, Lee JY, Guan KL and Morrison SJ: Temporal changes in PTEN and mTORC2 regulation of hematopoietic stem cell self-renewal and leukemia suppression. Cell Stem Cell. 11:415–428. 2012. View Article : Google Scholar : PubMed/NCBI

78 

Mullighan CG: Genomic profiling of B-progenitor acute lymphoblastic leukemia. Best Pract Res Clin Haematol. 24:489–503. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Inaba H, Greaves M and Mullighan CG: Acute lymphoblastic leukaemia. Lancet. 381:1943–1955. 2013. View Article : Google Scholar

80 

Jotta PY, Ganazza MA, Silva A, et al: Negative prognostic impact of PTEN mutation in pediatric T-cell acute lymphoblastic leukemia. Leukemia. 24:239–242. 2010. View Article : Google Scholar : PubMed/NCBI

81 

Karrman K, Forestier E, Heyman M, et al: Clinical and cytogenetic features of a population-based consecutive series of 285 pediatric T-cell acute lymphoblastic leukemias: rare T-cell receptor gene rearrangements are associated with poor outcome. Genes Chromosomes Cancer. 48:795–805. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Martelli AM, Chiarini F, Evangelisti C, et al: Two hits are better than one: targeting both phosphatidylinositol 3-kinase and mammalian target of rapamycin as a therapeutic strategy for acute leukemia treatment. Oncotarget. 3:371–394. 2012.PubMed/NCBI

83 

Nemes K, Sebestyen A, Mark A, et al: Mammalian target of rapamycin (mTOR) activity dependent phospho-protein expression in childhood acute lymphoblastic leukemia (ALL). PLoS One. 8:e593352013. View Article : Google Scholar : PubMed/NCBI

84 

Gutierrez A, Sanda T, Grebliunaite R, et al: High frequency of PTEN, PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia. Blood. 114:647–650. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Bandapalli OR, Zimmermann M, Kox C, et al: NOTCH1 activation clinically antagonizes the unfavorable effect of PTEN inactivation in BFM-treated children with precursor T-cell acute lymphoblastic leukemia. Haematologica. 98:928–936. 2013. View Article : Google Scholar

86 

Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, et al: Toward a NOTCH1/FBXW7/RAS/PTEN-based oncogenetic risk classification of adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult Acute Lymphoblastic Leukemia study. J Clin Oncol. 31:4333–4342. 2013. View Article : Google Scholar

87 

Grossmann V, Haferlach C, Weissmann S, et al: The molecular profile of adult T-cell acute lymphoblastic leukemia: mutations in RUNX1 and DNMT3A are associated with poor prognosis in T-ALL. Genes Chromosomes Cancer. 52:410–422. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Palomero T, Sulis ML, Cortina M, et al: Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nat Med. 13:1203–1210. 2007. View Article : Google Scholar : PubMed/NCBI

89 

Palomero T, Lim WK, Odom DT, et al: NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth. Proc Natl Acad Sci USA. 103:18261–18266. 2006. View Article : Google Scholar : PubMed/NCBI

90 

Gutierrez A, Grebliunaite R, Feng H, et al: Pten mediates Myc oncogene dependence in a conditional zebrafish model of T cell acute lymphoblastic leukemia. J Exp Med. 208:1595–1603. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Mavrakis KJ, Wolfe AL, Oricchio E, et al: Genome-wide RNA-mediated interference screen identifies miR-19 targets in Notch-induced T-cell acute lymphoblastic leukaemia. Nat Cell Biol. 12:372–379. 2010. View Article : Google Scholar : PubMed/NCBI

92 

Silva A, Yunes JA, Cardoso BA, et al: PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability. J Clin Invest. 118:3762–3774. 2008. View Article : Google Scholar : PubMed/NCBI

93 

Hales EC, Orr SM, Larson Gedman A, Taub JW and Matherly LH: Notch1 receptor regulates AKT protein activation loop (Thr308) dephosphorylation through modulation of the PP2A phosphatase in phosphatase and tensin homolog (PTEN)-null T-cell acute lymphoblastic leukemia cells. J Biol Chem. 288:22836–22848. 2013. View Article : Google Scholar

94 

Medyouf H, Gusscott S, Wang H, et al: High-level IGF1R expression is required for leukemia-initiating cell activity in T-ALL and is supported by Notch signaling. J Exp Med. 208:1809–1822. 2011. View Article : Google Scholar : PubMed/NCBI

95 

Cardoso BA, Martins LR, Santos CI, et al: Interleukin-4 stimulates proliferation and growth of T-cell acute lymphoblastic leukemia cells by activating mTOR signaling. Leukemia. 23:206–208. 2009. View Article : Google Scholar : PubMed/NCBI

96 

Barata JT, Silva A, Brandao JG, Nadler LM, Cardoso AA and Boussiotis VA: Activation of PI3K is indispensable for interleukin 7-mediated viability, proliferation, glucose use, and growth of T cell acute lymphoblastic leukemia cells. J Exp Med. 200:659–669. 2004. View Article : Google Scholar

97 

Scupoli MT, Vinante F, Krampera M, et al: Thymic epithelial cells promote survival of human T-cell acute lymphoblastic leukemia blasts: the role of interleukin-7. Haematologica. 88:1229–1237. 2003.PubMed/NCBI

98 

Zenatti PP, Ribeiro D, Li W, et al: Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia. Nat Genet. 43:932–939. 2011. View Article : Google Scholar

99 

Wong D and Korz W: Translating an antagonist of chemokine receptor CXCR4: from bench to bedside. Clin Cancer Res. 14:7975–7980. 2008. View Article : Google Scholar

100 

Scupoli MT, Donadelli M, Cioffi F, et al: Bone marrow stromal cells and the upregulation of interleukin-8 production in human T-cell acute lymphoblastic leukemia through the CXCL12/CXCR4 axis and the NF-κB and JNK/AP-1 pathways. Haematologica. 93:524–532. 2008.PubMed/NCBI

101 

Pillozzi S, Masselli M, De Lorenzo E, et al: Chemotherapy resistance in acute lymphoblastic leukemia requires hERG1 channels and is overcome by hERG1 blockers. Blood. 117:902–914. 2011. View Article : Google Scholar

102 

Heitman J, Movva NR and Hall MN: Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science. 253:905–909. 1991. View Article : Google Scholar

103 

Zhou H, Luo Y and Huang S: Updates of mTOR inhibitors. Anticancer Agents Med Chem. 10:571–581. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Schuler W, Sedrani R, Cottens S, et al: SDZ RAD, a new rapamycin derivative: pharmacological properties in vitro and in vivo. Transplantation. 64:36–42. 1997. View Article : Google Scholar : PubMed/NCBI

105 

Avellino R, Romano S, Parasole R, et al: Rapamycin stimulates apoptosis of childhood acute lymphoblastic leukemia cells. Blood. 106:1400–1406. 2005. View Article : Google Scholar : PubMed/NCBI

106 

Chan SM, Weng AP, Tibshirani R, Aster JC and Utz PJ: Notch signals positively regulate activity of the mTOR pathway in T-cell acute lymphoblastic leukemia. Blood. 110:278–286. 2007. View Article : Google Scholar : PubMed/NCBI

107 

Wu KN, Zhao YM, He Y, et al: Rapamycin interacts synergistically with idarubicin to induce T-leukemia cell apoptosis in vitro and in a mesenchymal stem cell simulated drug-resistant microenvironment via Akt/mammalian target of rapamycin and extracellular signal-related kinase signaling pathways. Leuk Lymphoma. 55:668–676. 2014.

108 

Akers LJ, Fang W, Levy AG, Franklin AR, Huang P and Zweidler-McKay PA: Targeting glycolysis in leukemia: a novel inhibitor 3-BrOP in combination with rapamycin. Leukemia Res. 35:814–820. 2011. View Article : Google Scholar : PubMed/NCBI

109 

Batista A, Barata JT, Raderschall E, et al: Targeting of active mTOR inhibits primary leukemia T cells and synergizes with cytotoxic drugs and signaling inhibitors. Exp Hematol. 39:457–472. e4532011. View Article : Google Scholar : PubMed/NCBI

110 

Houghton PJ, Morton CL, Kolb EA, et al: Initial testing (stage 1) of the mTOR inhibitor rapamycin by the pediatric preclinical testing program. Pediatr Blood Cancer. 50:799–805. 2008. View Article : Google Scholar : PubMed/NCBI

111 

Yee KW, Zeng Z, Konopleva M, et al: Phase I/II study of the mammalian target of rapamycin inhibitor everolimus (RAD001) in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res. 12:5165–5173. 2006. View Article : Google Scholar : PubMed/NCBI

112 

Rizzieri DA, Feldman E, Dipersio JF, et al: A phase 2 clinical trial of deforolimus (AP23573, MK-8669), a novel mammalian target of rapamycin inhibitor, in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res. 14:2756–2762. 2008. View Article : Google Scholar

113 

Tamburini J, Green AS, Bardet V, et al: Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia. Blood. 114:1618–1627. 2009. View Article : Google Scholar : PubMed/NCBI

114 

Chiarini F, Grimaldi C, Ricci F, et al: Activity of the novel dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 against T-cell acute lymphoblastic leukemia. Cancer Res. 70:8097–8107. 2010. View Article : Google Scholar : PubMed/NCBI

115 

Fox CJ, Hammerman PS and Thompson CB: The Pim kinases control rapamycin-resistant T cell survival and activation. J Exp Med. 201:259–266. 2005. View Article : Google Scholar

116 

Zhang F, Beharry ZM, Harris TE, et al: PIM1 protein kinase regulates PRAS40 phosphorylation and mTOR activity in FDCP1 cells. Cancer Biol Ther. 8:846–853. 2009. View Article : Google Scholar : PubMed/NCBI

117 

Lin YW, Beharry ZM, Hill EG, et al: A small molecule inhibitor of Pim protein kinases blocks the growth of precursor T-cell lymphoblastic leukemia/lymphoma. Blood. 115:824–833. 2010. View Article : Google Scholar

118 

Tamburini J, Chapuis N, Bardet V, et al: Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulin-like growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways. Blood. 111:379–382. 2008. View Article : Google Scholar

119 

Carracedo A, Ma L, Teruya-Feldstein J, et al: Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J Clin Invest. 118:3065–3074. 2008.PubMed/NCBI

120 

Efeyan A and Sabatini DM: mTOR and cancer: many loops in one pathway. Curr Opin Cell Biol. 22:169–176. 2010. View Article : Google Scholar : PubMed/NCBI

121 

Bertacchini J, Guida M, Accordi B, et al: Feedbacks and adaptive capabilities of the PI3K/Akt/mTOR axis in acute myeloid leukemia revealed by pathway selective inhibition and phosphoproteome analysis. Leukemia. Apr 4–2014.(E-pub ahead of print).

122 

Park S, Chapuis N, Saint Marcoux F, et al: A phase Ib GOELAMS study of the mTOR inhibitor RAD001 in association with chemotherapy for AML patients in first relapse. Leukemia. 27:1479–1486. 2013. View Article : Google Scholar : PubMed/NCBI

123 

Daver N, Kantarjian H, Thomas D, et al: A phase I/II study of hyper-CVAD plus everolimus in patients with relapsed/refractory acute lymphoblastic leukemia. In: 55th ASH Annual Meeting; Blood. 122. abs. 3916. 2013

124 

Fan QW, Knight ZA, Goldenberg DD, et al: A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell. 9:341–349. 2006. View Article : Google Scholar : PubMed/NCBI

125 

Cho DC, Cohen MB, Panka DJ, et al: The efficacy of the novel dual PI3-kinase/mTOR inhibitor NVP-BEZ235 compared with rapamycin in renal cell carcinoma. Clin Cancer Res. 16:3628–3638. 2010. View Article : Google Scholar : PubMed/NCBI

126 

Karar J, Cerniglia GJ, Lindsten T, Koumenis C and Maity A: Dual PI3K/mTOR inhibitor NVP-BEZ235 suppresses hypoxia-inducible factor (HIF)-1α expression by blocking protein translation and increases cell death under hypoxia. Cancer Biol Ther. 13:1102–1111. 2012.PubMed/NCBI

127 

Schenone S, Brullo C, Musumeci F, Radi M and Botta M: ATP-competitive inhibitors of mTOR: an update. Curr Med Chem. 18:2995–3014. 2011. View Article : Google Scholar : PubMed/NCBI

128 

Shepherd C, Banerjee L, Cheung CW, et al: PI3K/mTOR inhibition upregulates NOTCH-MYC signalling leading to an impaired cytotoxic response. Leukemia. 27:650–660. 2013. View Article : Google Scholar : PubMed/NCBI

129 

Raynaud FI, Eccles S, Clarke PA, et al: Pharmacologic characterization of a potent inhibitor of class I phosphatidylinositide 3-kinases. Cancer Res. 67:5840–5850. 2007. View Article : Google Scholar : PubMed/NCBI

130 

Maira SM, Stauffer F, Brueggen J, et al: Identification and characterization of NVP-BEZ235, a new orally available dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor with potent in vivo antitumor activity. Mol Cancer Ther. 7:1851–1863. 2008. View Article : Google Scholar : PubMed/NCBI

131 

Schult C, Dahlhaus M, Glass A, et al: The dual kinase inhibitor NVP-BEZ235 in combination with cytotoxic drugs exerts anti-proliferative activity towards acute lymphoblastic leukemia cells. Anticancer Res. 32:463–474. 2012.PubMed/NCBI

132 

Shortt J, Martin BP, Newbold A, et al: Combined inhibition of PI3K-related DNA damage response kinases and mTORC1 induces apoptosis in MYC-driven B-cell lymphomas. Blood. 121:2964–2974. 2013. View Article : Google Scholar : PubMed/NCBI

133 

Woods D and Turchi JJ: Chemotherapy induced DNA damage response: convergence of drugs and pathways. Cancer Biol Ther. 14:379–389. 2013. View Article : Google Scholar : PubMed/NCBI

134 

Kampa-Schittenhelm KM, Heinrich MC, Akmut F, et al: Cell cycle-dependent activity of the novel dual PI3K-mTORC1/2 inhibitor NVP-BGT226 in acute leukemia. Mol Cancer. 12:462013. View Article : Google Scholar : PubMed/NCBI

135 

Soria JC, Cortes J, Massard C, et al: Phase I safety, pharmacokinetic and pharmacodynamic trial of BMS-599626 (AC480), an oral pan-HER receptor tyrosine kinase inhibitor, in patients with advanced solid tumors. Ann Oncol. 23:463–471. 2012. View Article : Google Scholar : PubMed/NCBI

136 

Janes MR, Limon JJ, So L, et al: Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor. Nat Med. 16:205–213. 2010. View Article : Google Scholar : PubMed/NCBI

137 

Garcia-Echeverria C and Sellers WR: Drug discovery approaches targeting the PI3K/Akt pathway in cancer. Oncogene. 27:5511–5526. 2008. View Article : Google Scholar : PubMed/NCBI

138 

Garcia-Echeverria C: Allosteric and ATP-competitive kinase inhibitors of mTOR for cancer treatment. Bioorg Med Chem Lett. 20:4308–4312. 2010. View Article : Google Scholar : PubMed/NCBI

139 

Gentzler RD, Altman JK and Platanias LC: An overview of the mTOR pathway as a target in cancer therapy. Expert Opin Ther Targets. 16:481–489. 2012. View Article : Google Scholar : PubMed/NCBI

140 

Altman JK, Sassano A, Kaur S, et al: Dual mTORC2/mTORC1 targeting results in potent suppressive effects on acute myeloid leukemia (AML) progenitors. Clin Cancer Res. 17:4378–4388. 2011. View Article : Google Scholar : PubMed/NCBI

141 

Willems L, Chapuis N, Puissant A, et al: The dual mTORC1 and mTORC2 inhibitor AZD8055 has anti-tumor activity in acute myeloid leukemia. Leukemia. 26:1195–1202. 2012. View Article : Google Scholar : PubMed/NCBI

142 

Gupta M, Hendrickson AE, Yun SS, et al: Dual mTORC1/mTORC2 inhibition diminishes Akt activation and induces Puma-dependent apoptosis in lymphoid malignancies. Blood. 119:476–487. 2012. View Article : Google Scholar : PubMed/NCBI

143 

Feldman ME, Apsel B, Uotila A, et al: Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 7:e382009. View Article : Google Scholar : PubMed/NCBI

144 

Janes MR, Vu C, Mallya S, et al: Efficacy of the investigational mTOR kinase inhibitor MLN0128/INK128 in models of B-cell acute lymphoblastic leukemia. Leukemia. 27:586–594. 2013. View Article : Google Scholar : PubMed/NCBI

145 

Peng C, Chen Y, Li D and Li S: Role of Pten in leukemia stem cells. Oncotarget. 1:156–160. 2010.PubMed/NCBI

146 

Yilmaz OH, Valdez R, Theisen BK, et al: Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature. 441:475–482. 2006. View Article : Google Scholar : PubMed/NCBI

147 

Kaplan B, Qazi Y and Wellen JR: Strategies for the management of adverse events associated with mTOR inhibitors. Transplant Rev. Mar 12–2014.(Epub ahead of print).

148 

Markman B, Tabernero J, Krop I, et al: Phase I safety, pharmacokinetic, and pharmacodynamic study of the oral phosphatidylinositol-3-kinase and mTOR inhibitor BGT226 in patients with advanced solid tumors. Ann Oncol. 23:2399–2408. 2012. View Article : Google Scholar : PubMed/NCBI

149 

Mallya S, Fitch BA, Lee JS, So L, Janes MR and Fruman DA: Resistance to mTOR kinase inhibitors in lymphoma cells lacking 4EBP1. PloS One. 9:e888652014. View Article : Google Scholar

Related Articles

Journal Cover

September-2014
Volume 45 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Evangelisti C, Evangelisti C, Chiarini F, Lonetti A, Buontempo F, Bressanin D, Cappellini A, Orsini E, McCubrey JA, Martelli AM, Martelli AM, et al: Therapeutic potential of targeting mTOR in T-cell acute lymphoblastic leukemia (Review). Int J Oncol 45: 909-918, 2014
APA
Evangelisti, C., Evangelisti, C., Chiarini, F., Lonetti, A., Buontempo, F., Bressanin, D. ... Martelli, A.M. (2014). Therapeutic potential of targeting mTOR in T-cell acute lymphoblastic leukemia (Review). International Journal of Oncology, 45, 909-918. https://doi.org/10.3892/ijo.2014.2525
MLA
Evangelisti, C., Evangelisti, C., Chiarini, F., Lonetti, A., Buontempo, F., Bressanin, D., Cappellini, A., Orsini, E., McCubrey, J. A., Martelli, A. M."Therapeutic potential of targeting mTOR in T-cell acute lymphoblastic leukemia (Review)". International Journal of Oncology 45.3 (2014): 909-918.
Chicago
Evangelisti, C., Evangelisti, C., Chiarini, F., Lonetti, A., Buontempo, F., Bressanin, D., Cappellini, A., Orsini, E., McCubrey, J. A., Martelli, A. M."Therapeutic potential of targeting mTOR in T-cell acute lymphoblastic leukemia (Review)". International Journal of Oncology 45, no. 3 (2014): 909-918. https://doi.org/10.3892/ijo.2014.2525