An important mediator of cytokine signaling implicated in regulation of hematopoiesis is the PI3K/protein kinase B (PKB/c-Akt) signaling module. Constitutive activation of this signaling module has been observed in a large group of leukemias. Because activation of this signaling pathway has been demonstrated to be sufficient to induce hematologic malignancies and is thought to correlate with poor prognosis and enhanced drug resistance, it is considered to be a promising target for therapy. A high number of pharmacologic inhibitors directed against either individual or multiple components of this pathway have already been developed to improve therapy. In this review, the safety and efficacy of both single and dual-specificity inhibitors will be discussed as well as the potential of combination therapy with either inhibitors directed against other signal transduction molecules or classic chemotherapy.

The PI3K family consists of 3 distinct subclasses of which, to date, only the class I isoforms have been implicated in regulation of hematopoiesis. Three distinct catalytic class IA isoforms have been identified: p110α, p110β, and p110δ.1  These isoforms are predominantly activated by protein tyrosine kinases and form heterodimers with a group a regulatory adapter molecules, including p85α, p85β, p50α, p55α, and p55γ.1  In addition, a single-class 1B isoform termed p110γ has been described, which can be specifically activated by G-protein coupled receptors and associates with a p101 regulatory molecule.1  The most important substrate for these class I PI3Ks is phosphatidylinositol 4,5 bisphosphate [PI(4,5)P2] which can be phosphorylated at the D3 position of the inositol ring on extracellular stimulation, resulting in the formation of phosphatidylinositol 3,4,5 trisphosphate [PI(3,4,5)P3]1  (Figure 1). The activity of PI3K can be inhibited by phosphate and tensin homologue (PTEN), a ubiquitously expressed tumor suppressor protein that can dephosphorylate PIP3, resulting in the formation of PI(4,5)P2.1,2  Similarly, SH2-containing inositol-5′-phosphatase 1 (SHIP1), a protein predominantly expressed in hematopoietic cells, can hydrolyze PIP3 to generate PI(3,4)P2.1,2  Although both PTEN and SHIP1 act on the main product of PI3K, PI(3,4,5)P3, the generated products PI(3,4)P2 and PI(4,5)P2 both act as discrete second messengers activating distinct downstream events.2  Recently, pleckstrin homology (PH) domain leucine-rich repeat protein phosphatase (PHLPP) was found to terminate PKB signaling by directly dephosphorylating and inactivating PKB.3  However, a role for PHLPP in regulation of hematopoiesis remains to be determined. Inositol 1,3,4,5-tetrakiphosphate [Ins(1,3,4,5)P4], which is generated from inositol 1,4,5-triphosphate [Ins(1,4,5)P3] by inositol triphosphate 3-kinase B (InsP3KB), is considered to be a fourth negative regulator of the PI3K/PKB signaling module. The activity of downstream effectors of PI3K, including PKB, can be abrogated because of binding of PI(3,4,5)P3-specific pleckstrin homology (PH) domains to Ins(1,3,4,5)P4.4  Since the identification of PI3K, it has become evident that it plays an essential role in proliferation, survival, and differentiation of many different cell types. Correct regulation of the activity of PI3K has been demonstrated to be essential for maintenance of hematopoietic stem cells using PTEN- and SHIP1-deficient mice that displayed constitutive activation of PI3K activity. In those mice, an initial expansion of HSCs could be observed that was followed by a depletion of long-term repopulating HSCs.5  The role of class I PI3K isoforms in lineage development has been investigated in more detail. Combined deletion of p85α, p55α, and p50α has, for example, resulted in a complete block in B lymphocyte development.6  Similarly, introduction of a mutated, catalytically inactive p110δ (p110δD910A) in the normal p110δ locus also resulted in a block in early B lymphocyte development, whereas T-cell development was unaffected.7  These results indicate that PI3K activity is essential for normal B lymphocyte development. In addition, conditional deletion of either PTEN or SHIP1 in adult HSCs, resulting in activation of the PI3K pathway, not only reduced the level of B-lymphocytes but also enhanced the level of myeloid cells.8,9  Furthermore, enhanced levels of megakaryocyte progenitors have been observed in SHIP1-deficient mice.10  In time, both PTEN- and SHIP1-deficient mice developed a myeloproliferative disorder that progressed to leukemia.8,9  In PTEN heterozygote (+/−) SHIP null (−/−) mice, a more severe myeloproliferative phenotype, displayed by reduced erythrocyte and platelet numbers and enhanced white blood cell counts including elevated levels of neutrophils and monocytes in the peripheral blood, could be observed.11  Recently, a shorter SHIP1 isoform (s-SHIP1), which is transcribed from an internal promoter in the SHIP1 gene, has also been implicated in positive regulation of lymphocyte development during hematopoiesis.12  Pharmacologic inhibition of PI3K activity in human umbilical cord blood-derived CD34+ HSCs and progenitor cells revealed that inhibition of the activity of PI3K is sufficient to completely abrogate both proliferation and differentiation of eosinophil and neutrophil progenitors eventually leading to cell death.13  Finally, mice deficient for InsP3KB, resulting in an induction of PKB activity, display higher levels of granulocyte-macrophage progenitors and mature neutrophils in the bone marrow14  and dramatically reduced levels of mature CD4+ and CD8+ T lymphocytes.15  Together, these studies suggest that correct temporal regulation of PI3K activity is critical for both HSC maintenance and regulation of lineage development.

Figure 1

Schematic representation of the PI3K/PKB signaling module. Activation of PI3K by receptor stimulation results in the production of PtdIns(3,4,5)P3 at the plasma membrane. PKB subsequently translocates to the plasma membrane where it is phosphorylated by PDK1 and the mTORC2 complex. On phosphorylation, PKB is released into the cytoplasm where it can both inhibitory phosphorylate multiple substrates, including FoxO transcription factors and GSK-3, and induce the activity of other substrates, such as mTOR as part of the mTORC1 complex. Negative regulators of the PI3K/PKB signaling module include PTEN, SHIP1, Ins(1,3,4,5)P4, and PHLPP1/2.

Figure 1

Schematic representation of the PI3K/PKB signaling module. Activation of PI3K by receptor stimulation results in the production of PtdIns(3,4,5)P3 at the plasma membrane. PKB subsequently translocates to the plasma membrane where it is phosphorylated by PDK1 and the mTORC2 complex. On phosphorylation, PKB is released into the cytoplasm where it can both inhibitory phosphorylate multiple substrates, including FoxO transcription factors and GSK-3, and induce the activity of other substrates, such as mTOR as part of the mTORC1 complex. Negative regulators of the PI3K/PKB signaling module include PTEN, SHIP1, Ins(1,3,4,5)P4, and PHLPP1/2.

Close modal

An important mediator of PI3K signaling is protein kinase B (PKB/c-akt). Whereas PKB is cytosolic in unstimulated cells, PI3K-mediated activation of PKB requires translocation of PKB to the membrane. PI(3,4,5)P3, which is generated by PI3K, serves as an anchor for PH domain–containing proteins in the membrane, including PKB.16  Activation of PKB requires phosphorylation on both Thr308 in the activation loop, by phosphoinositide-dependent kinase 1 (PDK1) and Ser473, within the carboxyl-terminal hydrophobic motif, by the MTORC2 complex that consists of multiple proteins, including mTOR, Rictor, mLST8, DEPTOR, Sin1, and PROTOR17  (Figure 1).

Three highly homologous PKB isoforms have been described to be expressed in mammalian cells: PKBα, PKBβ, and PKBγ. PKB has been demonstrated to play an important role in regulation of cell survival and proliferation in a large variety of cell types.18  Analysis of HSCs derived from PKBα/PKBβ double-knockout mice revealed that PKB plays an important role in maintenance of long-term repopulating HSCs. These PKBα/PKBβ double-deficient HSCs were found to persist in the G0 phase of the cell cycle, suggesting that the functional defects observed in these mice with regard to long-term hematopoiesis were caused by enhanced quiescence.19  In contrast, loss of only one of the isoforms only minimally affected HSCs.19  In addition, ectopic expression of constitutively active PKB in mouse HSCs conversely resulted in transient expansion and increased cycling of HSCs, followed by apoptosis and expansion of immature progenitors in BM and spleen, which was also associated with impaired engraftment.20  In addition to HSC maintenance, PKB also plays a critical role in the regulation of cell fate decisions during hematopoietic lineage development. High PKB activity in human umbilical cord blood-derived hematopoietic progenitors was, for example, found to promote neutrophil and monocyte differentiation and to inhibit B lymphocyte development, whereas reduction of PKB activity has been demonstrated to be required for optimal eosinophil maturation.13  In addition, PKB plays an important role in regulation of proliferation and survival, but not maturation, of human dendritic cell (DC) progenitors.21  The development of a myeloproliferative disease, characterized by extramedullary hematopoiesis in liver and spleen, and lymphoblastic thymic T-cell lymphoma in the majority of mice transplanted with mouse bone marrow cells ectopically expressing constitutively active PKB further demonstrates the importance for correct regulation of PKB activity in HSCs and progenitor cells.20  In addition, recent findings suggest that constitutive activation of PKBβ is sufficient to accelerate MYC-induced T-cell acute lymphoblastic leukemia (T-ALL) in zebrafish.22  Furthermore, analysis of mice deficient for both PKBα and PKBβ, but not single knockout mice, revealed that the generation of marginal zone and B1 B cells and the survival of mature follicular B cells also depend on correct regulation of PKB activity.23 

Multiple PKB substrates have been identified, including members of the FoxO subfamily of forkhead transcription factors FoxO1, FoxO3, and FoxO4, the serine/threonine kinase glycogen synthase kinase-3 (GSK-3), and the serine/threonine kinase mammalian target of rapamycin (mTOR) as part of the MTORC1 complex, which also includes the regulatory associated protein of mTOR (Raptor; Figure 1).18  FoxO transcription factors, which are inhibitory phosphorylated by PKB,18  are known to play an important role in regulation of proliferation and survival of various cell types. In addition, these transcription factors play an important role in HSC maintenance, which has been demonstrated by conditional deletion of FoxO1, FoxO3, and FoxO4 in the adult hematopoietic system.24  An initial expansion of HSCs has been observed in those mice, which correlated with an HSC-specific up-regulation of cyclin D2 and down-regulation of cyclin G2, p130/Rb, p27, and p21.24  After this initial expansion, a reduction in HSC numbers was observed, resulting in a defective long-term repopulating capacity.24  Similarly, competitive repopulation experiments revealed that deletion of FoxO3 alone is also sufficient to impair long-term reconstitution.25  In addition, in aging mice deficient for FoxO3, the frequency of HSCs was increased compared with wild-type littermate controls.25  Both mouse models have also been used to examine the role of FoxO transcription factors in lineage development. Conditional deletion of FoxO1, FoxO3, and FoxO4 resulted in enhanced levels of myeloid cells and decreased numbers of peripheral blood lymphocytes under normal conditions. In time, these mice developed leukocytosis characterized by a relative neutrophilia and lymphopenia.24  In contrast, in FoxO3-deficient mice, neutrophilia only developed during aging and under myelosuppressive stress conditions.26  Although deletion of FoxO3 alone was not sufficient to induce neutrophilia, ectopic expression of a constitutively active, nonphosphorylatable, FoxO3 mutant in mouse hematopoietic progenitors did result in a decrease in the formation of both myeloid and erythroid colonies,27  suggesting that FoxO3 does play an important role in lineage development. Modulation of the activity of either PKB or FoxO transcription factors has been observed to alter the level of reactive oxygen species (ROS). Although ROS levels are reduced in PKBα/β-deficient mice,19  increased levels have been observed in mice deficient for FoxO.25  Increasing the ROS levels in PKBα/β-deficient mice was sufficient to rescue differentiation defects, but not impair long-term hematopoiesis.19  Reducing the ROS levels in FoxO-deficient mice with N-acetyl-L-cysteine, an antioxidative agent, was sufficient to abrogate the enhanced levels of proliferation and apoptosis in FoxO-deficient HSCs and to restore the reduced colony-forming ability of these cells.24  These studies demonstrate that correct regulation of ROS by FoxO transcription factors is essential for normal hematopoiesis.

Recent findings have demonstrated that correct regulation of the activity of GSK-3, which is inhibitory phosphorylated by PKB,18  is also essential for HSC maintenance. A reduction in long-term, but not short-term, repopulating HSCs has, for example, been observed in GSK3-deficient mice.28  In addition, disruption of GSK-3 activity in mice with a pharmacologic inhibitor or shRNAs has been shown to transiently induce expansion of both HSCs and progenitor cells followed by exhaustion of long-term repopulation HSCs.28,29  An acceleration of neutrophil and megakaryocyte recovery could be observed after transplantation of mice treated with a GSK-3 inhibitor, suggesting a role for GSK-3 in lineage development.29  Indeed, ex vivo experiments with human hematopoietic progenitors revealed that GSK-3 can enhance eosinophil differentiation and inhibit neutrophil development at least in part via regulation of C/EBPα, a key regulator of hematopoiesis.13 

A third, important mediator of PI3K/PKB signaling is mTOR.18  In contrast to GSK-3 and FoxO transcription factors, which are inhibitory phosphorylated by PKB, the activity of mTOR is positively regulated by PKB. Inhibition of the GTPase activating protein Tuberous sclerosis protein 2 (TSC2)/TSC1 complex by PKB results in accumulation of GTP-bound Rheb and subsequent activation of mTOR.30  In addition, because rapamycin was sufficient to revert the HSC phenotype of mice in which GSK-3 was depleted, it is probable that mTOR is an important effector of GSK-3.28  Conditional deletion of TSC1 in mice, resulting in activation of mTOR, has been demonstrated to enhance the percentage of cycling HSCs and to reduce the self-renewal capacity of HSCs in serial transplantation assays.31  In addition, treatment of PTEN-deficient mice with rapamycin appears to be sufficient to revert the HSC phenotype in those mice, indicating that mTORC1 is an important mediator of PI3K in regulation of HSC proliferation.32  In addition, deletion of TSC1 has been demonstrated to induce ROS levels in HSCs. In vivo treatment of TSC1-deficient mice with an ROS antagonist restored HSC numbers and function,31  suggesting that TSC1, similar to FoxO transcription factors, regulates HSC numbers at least in part via ROS. Although mice deficient of TSC1 also display reduced granulocyte and lymphocyte numbers, this is probably because of reduced progenitor proliferation. In contrast to PKB, which regulates both proliferation and differentiation of myeloid progenitors,13  mTOR primarily regulates expansion of hematopoietic progenitors, as was demonstrated for, for example, granulocyte progenitors33  and progenitors for interstitial DCs and Langerhans cells.21  Although the molecular mechanisms underlying mTOR-mediated regulation of lineage development are largely unknown, it has recently been demonstrated that mTOR decreases the ratio of wild-type C/EBPα (C/EBPαp42) and truncated C/EBPαp30, which is generated by alternative translation initiation, resulting in high levels of the smaller p30 C/EBPα isoform,34  which inhibits trans-activation of C/EBPα target genes in a dominant-negative manner and binds to the promoters of a unique set of target genes to suppress their transcription.

Together, these studies show that FoxO transcription factors GSK-3 and mTOR are all important mediators of PI3K in terms of HSC maintenance and lineage development.

The studies described in the first three sections of this review clearly demonstrate that deregulation of the PI3K/PKB signaling module dramatically affects hematopoiesis resulting in the development of hematologic malignancies. To investigate whether, in patients, the development of leukemia can be caused by aberrant regulation of this signaling module, research has focused on identifying mutations in PI3K and upstream and downstream regulators of this molecule (Figure 2). Constitutive activation of class I PI3K isoforms has been observed in a high percentage of patients with acute35,36  and chronic leukemia.37  In contrast to the expression of p110α, β, and γ, which is only up-regulated in leukemic blasts of some patients, p110δ expression appears to be consistently up-regulated in cells from patients with either acute myeloid leukemia (AML) or acute promyelocytic leukemia.36,38  Activating mutations in the kinase domain (H1047R) and helical domain (E545A) of p110α have been detected in a wide variety of human solid tumors.39  The latter has also been detected in acute leukemia, albeit in a very low percentage (1 of 88).39  In addition, another activating helical domain mutation (E545G) has recently been detected in a chronic myeloid leukemia (CML) cell line.40  Furthermore, both activating mutations in p110α (PIK3CA) and in-frame insertions/deletions in the PI3K regulatory subunit p85α (PIK3R1) have been observed in a low number of pediatric T-ALL patients (both 2 of 44).41  In a small group of myelodysplastic syndrome (MDS) patients, however, the above described mutations could not be detected.42 

Figure 2

Schematic representation of the known mutations in the PI3K/PKB signaling module in leukemia. Aberrant regulation of the PI3K/PKB signaling module has been found in a large group of patients with leukemia. However, only few mutations are known to directly affect hematopoiesis. Mutations indentified in leukemic cells are indicated above the linear (unscaled) representations of PI3K, SHIP, PTEN, and PKB. Grey arrows/lines indicate activating mutations; black arrows, inactivating mutations/deletions. *Mutations found in AML and ALL cell lines. **Mutation found in CML cell line. #Mutations inducing leukemia in mouse model. BD indicates binding domain; C2, C2 domain (putative membrane binding domain); SH2, Src homology 2 region (tyrosine-phosphorylated residue binding region); BHD, Bcr homology domain; SH3, Src homology 3 region (proline-rich protein-protein interaction region); and BR, binding region.

Figure 2

Schematic representation of the known mutations in the PI3K/PKB signaling module in leukemia. Aberrant regulation of the PI3K/PKB signaling module has been found in a large group of patients with leukemia. However, only few mutations are known to directly affect hematopoiesis. Mutations indentified in leukemic cells are indicated above the linear (unscaled) representations of PI3K, SHIP, PTEN, and PKB. Grey arrows/lines indicate activating mutations; black arrows, inactivating mutations/deletions. *Mutations found in AML and ALL cell lines. **Mutation found in CML cell line. #Mutations inducing leukemia in mouse model. BD indicates binding domain; C2, C2 domain (putative membrane binding domain); SH2, Src homology 2 region (tyrosine-phosphorylated residue binding region); BHD, Bcr homology domain; SH3, Src homology 3 region (proline-rich protein-protein interaction region); and BR, binding region.

Close modal

Importantly, ectopic expression of mutated p110α has been demonstrated to be sufficient to induce leukemia in a mouse transplantation model.43  Alternatively, constitutive activation of PI3K may also be caused by either aberrant expression or activation of PTEN. Reduced expression of PTEN has, for example, been observed in different types of leukemia. Although analysis of both myeloid leukemic cell lines and primary AML blasts indicates that PTEN mutations are rare in AML,44,45  both homozygous and heterozygous deletion of PTEN as well as nonsynonymous sequence alterations in exon 7 have been detected in approximately 15% and 25% of T-ALL patients, respectively.41  In addition, analysis of primary T-ALL cells revealed that, as a result of mutation-induced alternative splicing, full-length SHIP1 expression is often low or undetectable.46  In addition, an inactivating mutation in the phosphatase domain of SHIP1 has also been detected in primary AML blasts.47 

Although constitutive activation of PKB, resulting in enhanced survival signals,48-50  has been demonstrated in a significant fraction of AML51,52  and chronic lymphoid leukemia (CLL) patients,49,53,54  until recently, no PKB mutations were found in patients with leukemia. However, an activating mutation in the pleckstrin homology domain of PKB (E17K) has recently been detected in various solid tumors55  and one pediatric T-ALL patient.41  In contrast, this mutation could not be detected in a several cohorts of CLL,56,57  CML,58  and AML patients,57,59  suggesting that this particular mutation is very rare. However, transplantation of mice with bone marrow cells ectopically expressing this E17K mutation was sufficient to induce leukemia.55  Although mutations in mTOR have thus far not been described in hematologic malignancies, a recent study has identified several mTOR mutations in solid tumors.60  Interestingly, 2 single amino acid mutations, S2215Y and R2505P, identified in large intestine adenocarcinoma and renal cell carcinoma, respectively, appear to induce constitutive activation of mTOR, even under nutrient starvation conditions.60 

In addition to mutations in the PI3K signaling module itself, mutations in cytokine receptors, including constitutive activation of FMS-like tyrosine kinase 3 (FLT3) by internal tandem duplication (Flt3-ITD)61  and mutation in c-Kit43,62  have been described to affect PI3K signaling in hematologic malignancies. In addition to these tyrosine kinase receptors, the activity of the PI3K/PKB pathway can also be enhanced by several fusion proteins, including Bcr-Abl,63  which can be detected in virtually all patients with CML and in a subset of patients with ALL.64  The mechanisms underlying Bcr-Abl–mediated activation of the PI3K/PKB signaling pathway have been investigated extensively. Coimmunoprecipitation experiments revealed that Bcr-Abl associates with Shc,65  which subsequently binds to the p85α subunit of PI3K66  resulting in activation. In addition, expression of Bcr-Abl has been shown to reduce the levels of PHLPP1 and PHLPP2, which are negative regulators of PKB phosphorylation,67  and to up-regulate the level of p110γ.68  Bcr-Abl has also been shown to indirectly induce the activity of the PI3K/PKB signaling module by, for example, increasing the level of Nox-4–generated ROS resulting in inhibition of PP1a, a negative regulator of PI3K/PKB.69 

Other potential regulators of PI3K often mutated or aberrantly expressed in leukemia include Ras,1  Evi1,70  PP2A,52  and casein kinase 2 (CK2).35  Inhibition of CK2 has been shown to reduce the activity of the PI3K/PKB signaling module by dephosphorylation and activation of PTEN, resulting in induction of apoptosis in primary CLL cells in vitro.71,72  Interestingly, in a subset of MDS patients, it was demonstrated that deletion of glutathione S-transferase θ can result in the generation of a DNA sequence homologous to mTOR, thereby inducing the activity of downstream effectors of mTOR in a PI3K/PKB-independent manner. Inhibition of mTOR in these cells was sufficient to induce apoptosis.73,74 

It is evident that the activity of the PI3K/PKB signaling module is regulated by various extracellular factors, suggesting that deregulation of the production of those factors by components of the microenvironment could also result in aberrant hematopoiesis. Several studies have revealed that the microenvironment of CLL patients constitutively activates PI3K in leukemic cells, induces proliferation and survival of CLL cells, and contributes to the resistance of leukemic cells to cytotoxic agents.75-80  Inhibition of the PI3K/PKB signaling pathway was shown to be sufficient to induce apoptosis in CLL cells.75,76  In addition, the migratory capacity of primary CLL cells has been demonstrated to be enhanced by CX3CL1 in an autocrine manner.78  Constitutive release of soluble CX3CL1 by CLL cells results in a PKB-dependent up-regulation of CXCR4, a chemokine receptor that is important for CLL cell migration.78 

Although the studies described in this section clearly demonstrate that constitutive activation of the PI3K/PKB signaling module can result in the development of leukemia, enhanced FoxO activity has also been observed in 40% of the AML patients.81  Similarly, enhanced FoxO activity could also be observed in MLL-AF9–induced AML in a mouse model.81  Activation of PKB or deletion of FoxO1/3/4 was sufficient to reduce leukemic cell growth, suggesting that inactivation of the PI3K/PKB signaling module contributes to the development of MLL-AF9–induced leukemia.81  Together, these studies demonstrate that, in addition to constitutive activation of the PI3K/PKB signaling pathway, inactivation of this module can also result in the development of leukemia.

Because the PI3K/PKB signaling module is aberrantly regulated in many patients with leukemia, the question was raised whether the level of PI3K/PKB activation in leukemic blasts could be used as a prognostic marker. Mouse transplantation studies with blasts from pediatric de novo B-ALL patients revealed that a rapid induction of leukemia correlates with enhanced mTOR activity in the leukemic blasts.82  In addition, comparison of pediatric T-ALL patients revealed that the survival rate of patients positively correlates with the level of PTEN.83  Similar observations were made in a different cohort of pediatric T-ALL patients, in which PTEN deletions correlated with early treatment failure in T-ALL.41  Furthermore, constitutive activation of the PI3K pathway, as measured by enhanced FoxO3 expression84  or phosphorylation,85  enhanced levels of phosphorylated, and therefore inactive, PTEN86  or enhanced levels of phosphorylated PKB52,87  is also considered to be an independent adverse prognostic factor in AML patients. Similarly, in MDS, activation of the PI3K/PKB pathway has only been detected in high-risk MDS, but not in low-risk MDS patients,88,89  suggesting that high PKB activity may also be an adverse prognostic factor for MDS.

In contrast, Tamburini et al suggest that enhanced PKB phosphorylation positively correlates with the survival of AML patients.90  Although the short-term survival rate (within 12 months) appeared to be slightly lower in the group displaying high PKB phosphorylation compared with the group with low levels of phosphorylated PKB, both the long-term survival and relapse-free survival were significantly enhanced.90  Except for this last study, all other studies suggest that enhanced PI3K/PKB activity correlates with reduced survival rate in both ALL and AML patients.

The molecular mechanisms underlying this reduced prognosis are, thus far, incompletely understood. However, a reduced apoptotic response has been observed in AML blasts displaying enhanced PI3K/PKB activation.91  In addition, PI3K has been demonstrated to induce expression of the multidrug resistance-associated protein 1, a member of the ATP-binding cassette membrane transporters that functions as a drug efflux pump,92  suggesting that high PI3K activity induces drug resistance. The observation that high levels of multidrug resistance-associated protein 1 correlate with enhanced drug resistance in AML cells and poor prognosis supports that hypothesis.93 

Because aberrant regulation of the PI3K signaling module has frequently been observed in leukemic cells, PI3K and its downstream effectors are considered to be promising targets for therapy (Figure 3; Table 1). The functionality of LY294002 and wortmannin, 2 well-known PI3K inhibitors that prevent ATP to bind to and activate PI3K, has for example been investigated extensively. Preclinical experiments indicate that both LY294002 and wortmannin induce apoptosis in leukemic cells and rescue drug sensitivity. However, it has also been demonstrated that both inhibitors are detrimental for normal cells.13,94  In addition, both inhibitors exhibit little specificity within the PI3K family and inhibit the activity of other kinases, including CK2 and smMLCK, respectively.95  Recently, several novel PI3K inhibitors, including S14161,96  the p110α-selective inhibitor AS702630,97  the p110β-selective inhibitor TGX-115,36  and the p110δ inhibitors IC8711436,38  and CAL-10180,98-100  have been discovered that also appear to affect proliferation and survival of leukemic blasts. It has, for example, been demonstrated that IC87114 reduces proliferation and survival of both AML blasts38  and acute promyelocytic leukemia cells36  without affecting the proliferation of normal hematopoietic progenitors.38  For CAL-101, a dual mechanism of action has been revealed in patients with CLL. CAL-101 both decreases the survival of CLL cells directly and abrogates cellular interactions between CLL cells components of the tissue microenvironment.99,100  Phase 1 or 2 clinical trials (NCT00710528 and NCT01090414) have therefore been initiated to examine the efficacy of CAL-101 as a therapeutic agent in CLL.

Figure 3

Schematic representation of the PI3K/PKB signaling module and the available inhibitors inhibiting this pathway. Several inhibitors have been developed to inhibit aberrant regulation of the PI3K/PKB signaling module in leukemia. These inhibitors target single or multiple proteins in the pathway. Black boxes represent groups of similar inhibitors; and black arrows, the specific target of the inhibitors.

Figure 3

Schematic representation of the PI3K/PKB signaling module and the available inhibitors inhibiting this pathway. Several inhibitors have been developed to inhibit aberrant regulation of the PI3K/PKB signaling module in leukemia. These inhibitors target single or multiple proteins in the pathway. Black boxes represent groups of similar inhibitors; and black arrows, the specific target of the inhibitors.

Close modal
Table 1

Inhibitors of PI3K/PKB signaling pathway

TargetCompoundEffect
Clinical trials (phase)LeukemiaReference(s)
In vitroIn vivo
PI3K Wortmannin −   147,148  
 LY294002 −   37,148,150  
 AS702630 −   105  
 S14161   96  
p110β TGX-115 −   36,105  
p110δ IC87114 −   36,38,105,145  
AMG 319 − − NCT01300026 (1) CLL and ALL  
CAL-101 NCT01090414 (1) CLL and AML 80,98,99  
NCT00710528 (1) CLL and AML 80,98,99  
PDK1 UCN-01 −   122  
PKB Perifosine − NCT00391560 (2) RRL 101,103  
− − NCT00873457 (2) CLL  
PIA −   104  
A-443654 −   53  
AiX −   107  
Triciribine (API-2) − NCT00363454 (1)  106  
AKT-I-1/2 −   52,53  
GSK690693 − NCT00493818 (1)  151  
MK2206 − − NCT01231919 (1) RRL  
− − NCT01253447 (2) AML  
SR13668 − − NCT00896207 (1)   
GSK2141795 − − NCT00920257 (1)   
GSK21110183 − − NCT00881946 (1/2) RRL  
mTOR Rapamycin NCT00795886 (2) ALL 112,152,153  
NCT00068302 (1) ALL and AML 112,152,153  
(2) CML 154,155  
RAD001 NCT00636922 (1) (1/2) AML AML and MDS 138  
(2) B-CLL 109  
(2) CLL 116  
CCI-779 NCT00290472 (2) CLL  
NCT00084916 (2) RRL  
NCT00084474 (2) CLL  
(2) ALL 156  
AP23573 − − (2) AML 113  
  NCT00086125 (2) RRL  
PP242   117,118,157  
OSI-027 −   118,120  
AZD-8050 −   118  
PI3K/mTOR PI-103   76,126,127,158  
NVP-BEZ235   123,125  
PI3K/PDK1 BAG956   121  
PKB/PDK1/Flt3 KP372-1 −   128  
TargetCompoundEffect
Clinical trials (phase)LeukemiaReference(s)
In vitroIn vivo
PI3K Wortmannin −   147,148  
 LY294002 −   37,148,150  
 AS702630 −   105  
 S14161   96  
p110β TGX-115 −   36,105  
p110δ IC87114 −   36,38,105,145  
AMG 319 − − NCT01300026 (1) CLL and ALL  
CAL-101 NCT01090414 (1) CLL and AML 80,98,99  
NCT00710528 (1) CLL and AML 80,98,99  
PDK1 UCN-01 −   122  
PKB Perifosine − NCT00391560 (2) RRL 101,103  
− − NCT00873457 (2) CLL  
PIA −   104  
A-443654 −   53  
AiX −   107  
Triciribine (API-2) − NCT00363454 (1)  106  
AKT-I-1/2 −   52,53  
GSK690693 − NCT00493818 (1)  151  
MK2206 − − NCT01231919 (1) RRL  
− − NCT01253447 (2) AML  
SR13668 − − NCT00896207 (1)   
GSK2141795 − − NCT00920257 (1)   
GSK21110183 − − NCT00881946 (1/2) RRL  
mTOR Rapamycin NCT00795886 (2) ALL 112,152,153  
NCT00068302 (1) ALL and AML 112,152,153  
(2) CML 154,155  
RAD001 NCT00636922 (1) (1/2) AML AML and MDS 138  
(2) B-CLL 109  
(2) CLL 116  
CCI-779 NCT00290472 (2) CLL  
NCT00084916 (2) RRL  
NCT00084474 (2) CLL  
(2) ALL 156  
AP23573 − − (2) AML 113  
  NCT00086125 (2) RRL  
PP242   117,118,157  
OSI-027 −   118,120  
AZD-8050 −   118  
PI3K/mTOR PI-103   76,126,127,158  
NVP-BEZ235   123,125  
PI3K/PDK1 BAG956   121  
PKB/PDK1/Flt3 KP372-1 −   128  

RRL indicates relapsed and refractory leukemia.

Research has also focused on the development of pharmacologic compounds that inhibit PKB. One such compound is perifosine, a synthetic alkylphosphocholine with oral bioavailability that inhibits PKB phosphorylation by competitive interaction with its PH domain and promotes degradation of PKB, mTOR, Raptor, Rictor, p70S6K, and 4E-BP1.101  It has been demonstrated that this compound induces apoptosis in multidrug-resistant human T-ALL cells and primary AML cells102,103  but does not affect normal CD34+ hematopoietic progenitor cells.103  The efficacy of perifosine in treatment of different types of leukemia is currently examined in several phase 2 clinical trials (NCT00391560 and NCT00873457). Phosphatidylinositol ether lipid analogs inhibit PKB activity in a similar manner compared with perifosine. Treatment of HL60 cells with phosphatidylinositol ether lipid analogs resulted in inhibition of proliferation and sensitization to chemotherapeutic agents in concentrations that did not affect proliferation of normal hematopoietic progenitors.104  Another specific PKB inhibitor (AKT-I-1/2 inhibitor) has been demonstrated to efficiently reduce colony formation in high-risk AML samples52  and induces apoptosis in primary CLL cells.105  The PKB inhibitor triciribine (API-2), a purine analog, has been demonstrated to interact with the PH domain of PKB and can thus prevent association of PKB with PI(3,4,5)P3. In T-ALL cell lines, API-2 has been demonstrated to induce cell cycle arrest and apoptosis.106  The safety of this inhibitor is currently examined in a phase 1 clinical trial in patients with advanced hematologic malignancies (#NCT00363454). Recently, a novel specific PKB inhibitor termed AiX has been demonstrated to preferentially induce apoptosis in CLL cells with a normal immunoglobulin status that correlates with poor clinical outcome.107 

Rapamycin and its analogs RAD001 (everolimus), CCI-779 (temsirolimus), AP23573 (deforolimus), and RAD001108,109  inhibit the mTORC1 complex by association with FKBP-12, which abrogates the association of Raptor with mTOR.110  The efficacy of these compounds as therapeutic drugs has been examined in various preclinical and clinical studies for a wide range of malignancies. Although both rapamycin and its analog CCI-779 exhibit strong anti–tumor capacities in vitro,82,111,112  only a partial response was observed in clinical trials with rapamycin112  or its analog AP23573 in hematologic malignancies.113  The observed induction of PKB activity in AML blasts treated with rapamycin or AP23573 may explain the limited therapeutic effects of these compounds.114  Furthermore, experiments with PTEN-deficient mice revealed that withdrawal of rapamycin results in a rapid reinduction of leukemia and death in the majority of mice, which is probably because of failure to eliminate the leukemic stem cell population.115  In a subset of CLL patients, RAD-001 displays a modest antitumor activity.116  In addition, treatment of CLL patients with RAD-001 results in mobilization of malignant cells from the protective nodal masses into the peripheral blood.116  As an alternative, ATP-competitive mTOR inhibitors have recently been generated that inhibit the activity of both mTORC1 and mTORC2.117  Compared with rapamycin, the mTORC 1/2 inhibitor PP242 more efficiently reduced the development of leukemia in mice transplanted with primary ALL blasts or preleukemic thymocytes overexpressing PKB,117  while inducing less adverse effects on proliferation and function of normal lymphocytes.117,118  Similarly, OSI-027, another mTORC1/2 inhibitor,118  has been demonstrated to exhibit anti–leukemic effects in both Ph+ ALL and CML cells,119  and, compared with rapamycin, to efficiently suppress proliferation of AML cell lines.120 

Although the single-specificity inhibitors do affect the survival of leukemic cells, their effect in patients appears to be modest. The efficacy of combination therapy using multiple inhibitors directed against different intermediates of the PI3K signaling module is therefore also under investigation. Combined inhibition of mTOR and p110δ with RAD001 and IC87114, respectively, has for example been demonstrated to synergistically reduce proliferation of AML blasts.114  Similarly, combining the PI3K/PDK1 inhibitor BAG956 with RAD001 also resulted in a synergistic reduction in tumor volume in a mouse model transplanted with BCR-ABL–expressing cells.121  Finally, the efficacy of a novel combination regimen, including both perifosine and UCN-01 (NCT00301938), a PDK1 inhibitor that is known to induce apoptosis in AML cells in vitro,122  is currently examined in a phase 1 trial.

To further optimize inhibition of the PI3K signaling module, dual-specificity inhibitors have also been generated (Table 1). Recently, NVP-BEZ235, an orally bioavailable imidazoquinoline derivative that inhibits the activity of both PI3K and mTOR by binding to their ATP-binding pocket, has been identified.123  This compound significantly reduced proliferation and survival in both primary T-ALL124  and AML cells125  as well as leukemic cell lines124,125  without affecting the clonogenic capacity of normal hematopoietic progenitors.125  Another potent dual inhibitor for both class I PI3K isoforms and mTORC1 is PI-103, a synthetic small molecule of the pyridofuropyrimidine class. PI-103 has been demonstrated to reduce proliferation and survival of cells from T-ALL,126  AML,127  and CLL patients76  and appears to exhibit a stronger anti–leukemic activity compared with both rapamycin126  and the combination of RAD001 and IC87114.127  Importantly, although PI-103 reduces proliferation of normal hematopoietic progenitors, survival is not affected.127  A dual PI3K/PDK1 inhibitor called BAG956 has also recently been described. Although this compound can inhibit proliferation of BCR-ABL and FLT3-ITD–expressing cells, its anti–leukemic capacity is reduced compared with RAD001.121  In addition to these 3 dual-specificity inhibitors, KP372-1, a multiple kinase inhibitor capable of inhibiting PKB, PDK1, and FLT3, has been generated.128  It has been demonstrated that KP372-1 can induce apoptosis in primary AML cells and leukemic cell lines128  without affecting the survival of normal hematopoietic progenitors.128 

Leukemogenesis involves aberrant regulation of various signal transduction pathways, including, but not limited to, the PI3K signaling module. Simultaneous targeting of multiple aberrantly regulated signal transduction pathways is therefore considered to be a promising therapeutic strategy (Tables 2 and 3). Histone deacetylase inhibitors have, for example, emerged as a promising class of anti–tumor agents. Although, in mouse models, the histone deacetylase inhibitor MS-275 only partially inhibited leukemic cells, combined administration of MS-275 and RAD001 potentiated the effect of both inhibitors individually both in vitro andin vivo.129  Synergistic negative effects on proliferation and survival of leukemic cell lines have also been observed after coadministration of histone deacetylase inhibitors and perisofine.130  Although proteosome inhibitors are considered to be a new class of therapeutic agents, treatment of pediatric and adult B-ALL patients with such an inhibitor (bortezomib) was not sufficient to induce a robust anti–tumor response.131  Experiments in leukemic cell lines and primary cells from B-ALL patients revealed that, whereas MG132, a proteosome inhibitor, and RAD001 alone only modestly reduce cell viability, combined inhibition of proteosomes and mTOR significantly enhanced cell death.132  In addition to proteosome inhibitors, the efficacy of specific tyrosine kinase inhibitors, including inhibitors of Flt3, Abl, and c-Kit, has been investigated in preclinical and clinical models. Combined inhibition of tyrosine kinases and the PI3K/PKB pathway resulted in a synergistically enhanced anti–leukemia effect in ALL121  and AML cells121  compared with the individual inhibitors. Phase 1 or 2 clinical trials have therefore been initiated to investigate the synergistic effects of combined inhibition of PI3K/PKB and Flt3 (NCT00819546) or c-Kit (NCT00762632). The occurrence of clinical resistance to STI-571 is a major problem for CML patients. Although the most common mechanisms of induction of resistance are mutations or alterations in the Bcr-Abl gene,133  a STI-571–induced compensatory PKB/mTor activation has also been described as a potential mechanism for the persistence of Bcr-Abl-positive cells in STI-571–treated patients.134-136  A combination of pharmacologic compounds inhibiting the PI3K/PKB pathway and STI-571 has been shown to be effective in cells from STI-571–resistant CML patients.119,121,137-142  Several phase 1 or 2 clinical trials have therefore been initiated to determine the effectiveness of such a combination therapy in STI-571–resistant CML patients (NCT01188889, NCT00093639, and NCT00101088).

Table 2

Combination regimens with single specificity inhibitors

TargetCompoundCombination regimensEffects
Clinical trials (phase)LeukemiaReference(s)
In vitroIn vivo
PI3K Wortmannin ATRA (DA) −   159  
STI-571 (Bcr-Abl TKI) −   141  
LY294002 Apigenin (CK2 I) −   160  
ATRA (DA) −   159  
STI-571 (Bcr-Abl TKI) −   141  
Arsenic trioxide −   161  
p110δ IC87114 VP16 (CT) −   145  
CAL-101 Lenalidomide −   98  
CT and mAb CD20 − − NCT01088048 (1) CLL  
Rituximab (mAb CD20) − − NCT01203930 (2) CLL  
PDK1 UCN-01 Ara-C (CT) − (2) AML 146  
Cytarabine (CT) − − NCT00004263 (1) AML and MDS  
Fludarabine (CT) − − NCT00019838 (1) RRL  
OSU-03012 STI-571 (Bcr-Abl TKI) −   140  
PP2A Forskolin Idarubicin/Ara-C −   162  
PKB Perifosine UCN-01 − − NCT00301938 (1) RRL  
HDAC I −   130  
TRAIL (AI) −   163  
Etoposide (CT −   103  
PIA CT −   104  
Triciribine Cytarabine (CT) −   106  
MK2206 Rituximab (mAb CD20) − − NCT01369849 (1/2) CLL  
Arsenic trioxide −   161  
mTOR Rapamycin UCN-01 −   122  
3-BrOP (glycolysis I) −   164  
STI-571 (Bcr-Abl TKI) −   139  
Notch I −   165  
Erlotinib (EGFR-TKI) −   166  
Curcumin −   167  
Dexamethasone −   168,169  
Etoposide (CT)   144  
Decatibine (CT) − − NCT00861874 (1) AML  
Aracytin (CT) − − NCT00235560 (2) AML  
Methotrexate (CT) NCT01162551 (2) ALL and CLL 111  
Anthracyclin (CT) −   170  
Daunorubicin (CT) −   171  
CT − NCT00776373 (1/2) ALL and CML 152  
− NCT01184898 (1/2) AML 152  
− NCT00780104 (1/2) AML and CML 152  
RAD001 IC87114 −   114  
BAG956   121  
Bortezomib (PI) −   132  
MS-275 (HDAC I)   129  
PKC412 (Flt3 TKI) − − NCT00819546 (1) AML and MDS  
Imatinib (Bcr-Abl TKI) − NCT00093639 (1/2) CML 138  
− NCT01188889 (1/2) CML 138  
Nilotinib (c-Kit-TKI) − − NCT00762632 (1/2) AML  
Alemtuzumab (mAb CD52) − − NCT00935792 (1/2) CLL  
ATRA (DA)   172  
Ara-C (CT) −   132,149  
Vincristine (CT)   173  
CT − − NCT00544999 (1) AML  
− − NCT01154439 (1) AML  
CCI-779 Methotrexate (CT)   111  
Clofarabine (CT) − − NCT00775593 (2) AML  
 STI-571 (Bcr-Abl TKI) − − NCT00101088 (1) CML  
PP242 Vincristine (CT) −   118  
OSI-027 STI-571 (Bcr-Abl TKI) −   119  
GILZ STI-571 (Bcr-Abl TKI) −   137  
TargetCompoundCombination regimensEffects
Clinical trials (phase)LeukemiaReference(s)
In vitroIn vivo
PI3K Wortmannin ATRA (DA) −   159  
STI-571 (Bcr-Abl TKI) −   141  
LY294002 Apigenin (CK2 I) −   160  
ATRA (DA) −   159  
STI-571 (Bcr-Abl TKI) −   141  
Arsenic trioxide −   161  
p110δ IC87114 VP16 (CT) −   145  
CAL-101 Lenalidomide −   98  
CT and mAb CD20 − − NCT01088048 (1) CLL  
Rituximab (mAb CD20) − − NCT01203930 (2) CLL  
PDK1 UCN-01 Ara-C (CT) − (2) AML 146  
Cytarabine (CT) − − NCT00004263 (1) AML and MDS  
Fludarabine (CT) − − NCT00019838 (1) RRL  
OSU-03012 STI-571 (Bcr-Abl TKI) −   140  
PP2A Forskolin Idarubicin/Ara-C −   162  
PKB Perifosine UCN-01 − − NCT00301938 (1) RRL  
HDAC I −   130  
TRAIL (AI) −   163  
Etoposide (CT −   103  
PIA CT −   104  
Triciribine Cytarabine (CT) −   106  
MK2206 Rituximab (mAb CD20) − − NCT01369849 (1/2) CLL  
Arsenic trioxide −   161  
mTOR Rapamycin UCN-01 −   122  
3-BrOP (glycolysis I) −   164  
STI-571 (Bcr-Abl TKI) −   139  
Notch I −   165  
Erlotinib (EGFR-TKI) −   166  
Curcumin −   167  
Dexamethasone −   168,169  
Etoposide (CT)   144  
Decatibine (CT) − − NCT00861874 (1) AML  
Aracytin (CT) − − NCT00235560 (2) AML  
Methotrexate (CT) NCT01162551 (2) ALL and CLL 111  
Anthracyclin (CT) −   170  
Daunorubicin (CT) −   171  
CT − NCT00776373 (1/2) ALL and CML 152  
− NCT01184898 (1/2) AML 152  
− NCT00780104 (1/2) AML and CML 152  
RAD001 IC87114 −   114  
BAG956   121  
Bortezomib (PI) −   132  
MS-275 (HDAC I)   129  
PKC412 (Flt3 TKI) − − NCT00819546 (1) AML and MDS  
Imatinib (Bcr-Abl TKI) − NCT00093639 (1/2) CML 138  
− NCT01188889 (1/2) CML 138  
Nilotinib (c-Kit-TKI) − − NCT00762632 (1/2) AML  
Alemtuzumab (mAb CD52) − − NCT00935792 (1/2) CLL  
ATRA (DA)   172  
Ara-C (CT) −   132,149  
Vincristine (CT)   173  
CT − − NCT00544999 (1) AML  
− − NCT01154439 (1) AML  
CCI-779 Methotrexate (CT)   111  
Clofarabine (CT) − − NCT00775593 (2) AML  
 STI-571 (Bcr-Abl TKI) − − NCT00101088 (1) CML  
PP242 Vincristine (CT) −   118  
OSI-027 STI-571 (Bcr-Abl TKI) −   119  
GILZ STI-571 (Bcr-Abl TKI) −   137  

DA indicates differentiating agents; TKI, tyrosine kinase inhibitor; I, inhibitor; CT, chemotherapy; AI, apoptosis inducer; RRL, relapsed and refractory leukemia; and PI, proteasome inhibitor.

Table 3

Combination regimens with dual specificity inhibitors

TargetCompoundCombination regimensEffects
Clinical trials (phase)References
In vitroIn vivo
PI3K/mTOR PI-103 Nutlin-3 (MDM2-I) −  158  
STI-571 (Bcr-Abl-TKI) −  142  
Arsenic disulfide −  174  
Vincristine (CT) −  126  
Fludarabine (CT) −  76  
PI3K/mTOR NVP-BEZ235 CT −  124  
PI3K/PDK1 BAG956 STI-571 (Bcr-Abl-TKI)  121  
Rapamycin/RAD-001 −  121  
PKC412 (Flt3 TKI)  121  
TargetCompoundCombination regimensEffects
Clinical trials (phase)References
In vitroIn vivo
PI3K/mTOR PI-103 Nutlin-3 (MDM2-I) −  158  
STI-571 (Bcr-Abl-TKI) −  142  
Arsenic disulfide −  174  
Vincristine (CT) −  126  
Fludarabine (CT) −  76  
PI3K/mTOR NVP-BEZ235 CT −  124  
PI3K/PDK1 BAG956 STI-571 (Bcr-Abl-TKI)  121  
Rapamycin/RAD-001 −  121  
PKC412 (Flt3 TKI)  121  

I indicates inhibitor; TKI, tyrosine kinase inhibitor; and CT, chemotherapy.

Chemotherapy has been shown to be effective in a subset of patients. However, incomplete remission and the development of a refractory disease have been observed in many patients with acute leukemia.143  To optimize treatment, chemotherapy could potentially be combined with specific pharmacologic inhibitors (Table 2). Coadministration of mTOR inhibitors with different types of chemotherapeutic drugs, including etoposide, Ara-C, cytarabine, and dexamethasone, has, for example, been demonstrated to induce synergistic anti–leukemia effects in cells from AML patients144  and ALL patients.111,132  Several phase 1 or 2 clinical trials have therefore been initiated to investigate and optimize the synergistic effect of mTOR inhibitors and chemotherapeutic drugs in patients (NCT00544999, NCT01184898, NCT00780104, NCT01162551 and NCT00776373, NCT00861874, NCT00235560, and NCT01154439) In addition, synergistic effects have been observed in AML cells after coadministration of chemotherapeutic agents with IC87114,145  UCN-01,146  or triciribine.106  Similar results were obtained in T-ALL cells when combining the dual-specificity inhibitors PI-103 and NVP-BEZ235 with chemotherapy.124,126  Finally, inhibition of the activity of p110δ with CAL-101 was shown to be sufficient to abrogate lenalidomide-induced activation of primary CCL cells.98  In addition to the clinical trials focusing on CAL-101 alone, the efficacy of a combination regimen with both CAL-101 and lenalidomide is therefore also under investigation in phase 1 or 2 clinical trials (NCT01203930 and NCT01088048).

Future perspectives

During the last 2 decades, it has become clear that the PI3K/PKB signal transduction pathway plays an important role in both normal and malignant hematopoiesis. As discussed in “Derequlated P13K/PKB signaling in malignant hematopoiesis,” aberrant regulation of this signaling module has been observed in a large group of acute leukemias. Although mutations in PI3K, PKB, or the upstream regulators PTEN and SHIP1 have been detected in cells from patients with leukemia, these mutations appear to be rare. These mutations can therefore not account for the large incidence of constitutive activation of PI3K in patients with leukemia. In contrast, several oncogenic fusion proteins and mutated tyrosine kinase receptors have been demonstrated to induce hematologic malignancies by constitutive activation of the PI3K/PKB signaling module. Because PI3K is frequently activated in leukemia and activation of this molecule is thought to correlate with poor prognosis and drug resistance, it is considered to be a promising target for therapy. A high number of pharmacologic inhibitors directed against both individual and multiple components of this pathway have already been developed to improve therapy. Although the single-specificity inhibitors do affect the survival of leukemic cells, their effect in patients appears to be modest. This can be explained by both inhibitor-induced abrogation of negative feedback loops and alternative mechanisms of activation of downstream effectors of PI3K/PKB. Although further research is required to examine the safety and efficacy of the dual-specificity inhibitors, these compounds appear to possess more promising anti–leukemic activities compared with single-specificity inhibitors. In addition, to further optimize therapeutic regimens, research has focused on coadministration of inhibitors of the PI3K signaling module with either classic chemotherapy or inhibitors directed against other signal transduction molecules. The in vitro studies and mouse transplantation experiments described herein strongly suggest that both strategies could indeed be used to improve current therapeutic regimens in specific patient groups.

Inhibition of aberrantly regulated intracellular signal transduction pathways provides an important means to improve therapeutic regimens for patients with leukemia. Because the PI3K/PKB signaling pathway appears to be highly deregulated in a large number of patients with leukemia, this pathway is considered to be a promising target for therapy. Compared with targeting individual components of the PI3K/PKB signaling module alone, either abrogating this pathway at multiple levels using dual-specificity inhibitors or combining pathway specific inhibitors with classic regimens appears to be a more effective therapeutic strategy for patients with leukemia. Further research is therefore warranted to examine the safety and efficacy of these regimens in leukemia patients.

R.P. was supported by KiKa (Children Cancer Free grant).

The authors apologize that, because of space restrictions, some references to original papers may not have been included in this review.

Contribution: R.P. and M.B. wrote the manuscript.

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

Correspondence: Miranda Buitenhuis, Department of Hematology, Erasmus MC, Dr Molewaterplein 50, Faculty Building Office H-Ee1330F, 3015 GE Rotterdam, The Netherlands; e-mail: m.buitenhuis@erasmusmc.nl.

1
Vanhaesebroeck
 
B
Guillermet-Guibert
 
J
Graupera
 
M
Bilanges
 
B
The emerging mechanisms of isoform-specific PI3K signalling.
Nat Rev Mol Cell Biol
2010
, vol. 
11
 
5
(pg. 
329
-
341
)
2
Buitenhuis
 
M
Coffer
 
PJ
The role of the PI3K-PKB signaling module in regulation of hematopoiesis.
Cell Cycle
2009
, vol. 
8
 
4
(pg. 
560
-
566
)
3
Brognard
 
J
Sierecki
 
E
Gao
 
T
Newton
 
AC
PHLPP and a second isoform, PHLPP2, differentially attenuate the amplitude of Akt signaling by regulating distinct Akt isoforms.
Mol Cell
2007
, vol. 
25
 
6
(pg. 
917
-
931
)
4
Jia
 
Y
Subramanian
 
KK
Erneux
 
C
, et al. 
Inositol 1,3,4,5-tetrakisphosphate negatively regulates phosphatidylinositol-3,4,5-trisphosphate signaling in neutrophils.
Immunity
2007
, vol. 
27
 
3
(pg. 
453
-
467
)
5
Helgason
 
CD
Antonchuk
 
J
Bodner
 
C
Humphries
 
RK
Homeostasis and regeneration of the hematopoietic stem cell pool are altered in SHIP-deficient mice.
Blood
2003
, vol. 
102
 
10
(pg. 
3541
-
3547
)
6
Fruman
 
DA
Mauvais-Jarvis
 
F
Pollard
 
DA
, et al. 
Hypoglycaemia, liver necrosis and perinatal death in mice lacking all isoforms of phosphoinositide 3-kinase p85 alpha.
Nat Genet
2000
, vol. 
26
 
3
(pg. 
379
-
382
)
7
Okkenhaug
 
K
Bilancio
 
A
Farjot
 
G
, et al. 
Impaired B and T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice.
Science
2002
, vol. 
297
 
5583
(pg. 
1031
-
1034
)
8
Zhang
 
J
Grindley
 
JC
Yin
 
T
, et al. 
PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention.
Nature
2006
, vol. 
441
 
7092
(pg. 
518
-
522
)
9
Helgason
 
CD
Damen
 
JE
Rosten
 
P
, et al. 
Targeted disruption of SHIP leads to hemopoietic perturbations, lung pathology, and a shortened life span.
Genes Dev
1998
, vol. 
12
 
11
(pg. 
1610
-
1620
)
10
Perez
 
LE
Desponts
 
C
Parquet
 
N
Kerr
 
WG
SH2-inositol phosphatase 1 negatively influences early megakaryocyte progenitors.
PLoS One
2008
, vol. 
3
 
10
pg. 
e3565
 
11
Moody
 
JL
Xu
 
L
Helgason
 
CD
Jirik
 
FR
Anemia, thrombocytopenia, leukocytosis, extramedullary hematopoiesis, and impaired progenitor function in Pten+/−SHIP−/− mice: a novel model of myelodysplasia.
Blood
2004
, vol. 
103
 
12
(pg. 
4503
-
4510
)
12
Nguyen
 
NY
Maxwell
 
MJ
Ooms
 
LM
, et al. 
An ENU-induced mouse mutant of SHIP1 reveals a critical role of the stem cell isoform for suppression of macrophage activation.
Blood
2011
, vol. 
117
 
20
(pg. 
5362
-
5371
)
13
Buitenhuis
 
M
Verhagen
 
LP
van Deutekom
 
HW
, et al. 
Protein kinase B (c-akt) regulates hematopoietic lineage choice decisions during myelopoiesis.
Blood
2008
, vol. 
111
 
1
(pg. 
112
-
121
)
14
Jia
 
Y
Loison
 
F
Hattori
 
H
, et al. 
Inositol trisphosphate 3-kinase B (InsP3KB) as a physiological modulator of myelopoiesis.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
12
(pg. 
4739
-
4744
)
15
Pouillon
 
V
Hascakova-Bartova
 
R
Pajak
 
B
, et al. 
Inositol 1,3,4,5-tetrakisphosphate is essential for T lymphocyte development.
Nat Immunol
2003
, vol. 
4
 
11
(pg. 
1136
-
1143
)
16
Burgering
 
BM
Coffer
 
PJ
Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction.
Nature
1995
, vol. 
376
 
6541
(pg. 
599
-
602
)
17
Oh
 
WJ
Jacinto
 
E
mTOR complex 2 signaling and functions.
Cell Cycle
2011
, vol. 
10
 
14
(pg. 
2305
-
2316
)
18
Manning
 
BD
Cantley
 
LC
AKT/PKB signaling: navigating downstream.
Cell
2007
, vol. 
129
 
7
(pg. 
1261
-
1274
)
19
Juntilla
 
MM
Patil
 
VD
Calamito
 
M
Joshi
 
RP
Birnbaum
 
MJ
Koretzky
 
GA
AKT1 and AKT2 maintain hematopoietic stem cell function by regulating reactive oxygen species.
Blood
2010
, vol. 
115
 
20
(pg. 
4030
-
4038
)
20
Kharas
 
MG
Okabe
 
R
Ganis
 
JJ
, et al. 
Constitutively active AKT depletes hematopoietic stem cells and induces leukemia in mice.
Blood
2010
, vol. 
115
 
7
(pg. 
1406
-
1415
)
21
van de Laar
 
L
Buitenhuis
 
M
Wensveen
 
FM
Janssen
 
HL
Coffer
 
PJ
Woltman
 
AM
Human CD34-derived myeloid dendritic cell development requires intact phosphatidylinositol 3-kinase-protein kinase B-mammalian target of rapamycin signaling.
J Immunol
2010
, vol. 
184
 
12
(pg. 
6600
-
6611
)
22
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
2011
, vol. 
208
 
8
(pg. 
1595
-
1603
)
23
Calamito
 
M
Juntilla
 
MM
Thomas
 
M
, et al. 
Akt1 and Akt2 promote peripheral B-cell maturation and survival.
Blood
2010
, vol. 
115
 
20
(pg. 
4043
-
4050
)
24
Tothova
 
Z
Kollipara
 
R
Huntly
 
BJ
, et al. 
FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress.
Cell
2007
, vol. 
128
 
2
(pg. 
325
-
339
)
25
Miyamoto
 
K
Araki
 
KY
Naka
 
K
, et al. 
Foxo3a is essential for maintenance of the hematopoietic stem cell pool.
Cell Stem Cell
2007
, vol. 
1
 
1
(pg. 
101
-
112
)
26
Miyamoto
 
K
Miyamoto
 
T
Kato
 
R
Yoshimura
 
A
Motoyama
 
N
Suda
 
T
FoxO3a regulates hematopoietic homeostasis through a negative feedback pathway in conditions of stress or aging.
Blood
2008
, vol. 
112
 
12
(pg. 
4485
-
4493
)
27
Engström
 
M
Karlsson
 
R
Jonsson
 
JI
Inactivation of the forkhead transcription factor FoxO3 is essential for PKB-mediated survival of hematopoietic progenitor cells by kit ligand.
Exp Hematol
2003
, vol. 
31
 
4
(pg. 
316
-
323
)
28
Huang
 
J
Zhang
 
Y
Bersenev
 
A
, et al. 
Pivotal role for glycogen synthase kinase-3 in hematopoietic stem cell homeostasis in mice.
J Clin Invest
2009
, vol. 
119
 
12
(pg. 
3519
-
3529
)
29
Trowbridge
 
JJ
Xenocostas
 
A
Moon
 
RT
Bhatia
 
M
Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation.
Nat Med
2006
, vol. 
12
 
1
(pg. 
89
-
98
)
30
Sengupta
 
S
Peterson
 
TR
Sabatini
 
DM
Regulation of the mTOR complex 1 pathway by nutrients, growth factors, and stress.
Mol Cell
2010
, vol. 
40
 
2
(pg. 
310
-
322
)
31
Chen
 
C
Liu
 
Y
Liu
 
R
Ikenoue
 
T
Guan
 
KL
Zheng
 
P
TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species.
J Exp Med
2008
, vol. 
205
 
10
(pg. 
2397
-
2408
)
32
Yilmaz
 
OH
Valdez
 
R
Theisen
 
BK
, et al. 
Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells.
Nature
2006
, vol. 
441
 
7092
(pg. 
475
-
482
)
33
Geest
 
CR
Zwartkruis
 
FJ
Vellenga
 
E
Coffer
 
PJ
Buitenhuis
 
M
Mammalian target of rapamycin activity is required for expansion of CD34+ hematopoietic progenitor cells.
Haematologica
2009
, vol. 
94
 
7
(pg. 
901
-
910
)
34
Fu
 
CT
Zhu
 
KY
Mi
 
JQ
, et al. 
An evolutionarily conserved PTEN-C/EBPalpha-CTNNA1 axis controls myeloid development and transformation.
Blood
2010
, vol. 
115
 
23
(pg. 
4715
-
4724
)
35
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
2008
, vol. 
118
 
11
(pg. 
3762
-
3774
)
36
Billottet
 
C
Banerjee
 
L
Vanhaesebroeck
 
B
Khwaja
 
A
Inhibition of class I phosphoinositide 3-kinase activity impairs proliferation and triggers apoptosis in acute promyelocytic leukemia without affecting atra-induced differentiation.
Cancer Res
2009
, vol. 
69
 
3
(pg. 
1027
-
1036
)
37
Ringshausen
 
I
Schneller
 
F
Bogner
 
C
, et al. 
Constitutively activated phosphatidylinositol-3 kinase (PI-3K) is involved in the defect of apoptosis in B-CLL: association with protein kinase Cdelta.
Blood
2002
, vol. 
100
 
10
(pg. 
3741
-
3748
)
38
Sujobert
 
P
Bardet
 
V
Cornillet-Lefebvre
 
P
, et al. 
Essential role for the p110delta isoform in phosphoinositide 3-kinase activation and cell proliferation in acute myeloid leukemia.
Blood
2005
, vol. 
106
 
3
(pg. 
1063
-
1066
)
39
Lee
 
JW
Soung
 
YH
Kim
 
SY
, et al. 
PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas.
Oncogene
2005
, vol. 
24
 
8
(pg. 
1477
-
1480
)
40
Quentmeier
 
H
Eberth
 
S
Romani
 
J
Zaborski
 
M
Drexler
 
HG
BCR-ABL1-independent PI3Kinase activation causing imatinib-resistance.
J Hematol Oncol
2011
, vol. 
4
 pg. 
6
 
41
Gutierrez
 
A
Sanda
 
T
Grebliunaite
 
R
, et al. 
High frequency of PTEN, PI3K, and AKT abnormalities in T-cell acute lymphoblastic leukemia.
Blood
2009
, vol. 
114
 
3
(pg. 
647
-
650
)
42
Machado-Neto
 
JA
Traina
 
F
Lazarini
 
M
, et al. 
Screening for hotspot mutations in PI3K, JAK2, FLT3 and NPM1 in patients with myelodysplastic syndromes.
Clinics (Sao Paulo)
2011
, vol. 
66
 
5
(pg. 
793
-
799
)
43
Horn
 
S
Bergholz
 
U
Jucker
 
M
, et al. 
Mutations in the catalytic subunit of class IA PI3K confer leukemogenic potential to hematopoietic cells.
Oncogene
2008
, vol. 
27
 
29
(pg. 
4096
-
4106
)
44
Liu
 
TC
Lin
 
PM
Chang
 
JG
Lee
 
JP
Chen
 
TP
Lin
 
SF
Mutation analysis of PTEN/MMAC1 in acute myeloid leukemia.
Am J Hematol
2000
, vol. 
63
 
4
(pg. 
170
-
175
)
45
Aggerholm
 
A
Gronbaek
 
K
Guldberg
 
P
Hokland
 
P
Mutational analysis of the tumour suppressor gene MMAC1/PTEN in malignant myeloid disorders.
Eur J Haematol
2000
, vol. 
65
 
2
(pg. 
109
-
113
)
46
Lo
 
TC
Barnhill
 
LM
Kim
 
Y
Nakae
 
EA
Yu
 
AL
Diccianni
 
MB
Inactivation of SHIP1 in T-cell acute lymphoblastic leukemia due to mutation and extensive alternative splicing.
Leuk Res
2009
, vol. 
33
 
11
(pg. 
1562
-
1566
)
47
Luo
 
JM
Yoshida
 
H
Komura
 
S
, et al. 
Possible dominant-negative mutation of the SHIP gene in acute myeloid leukemia.
Leukemia
2003
, vol. 
17
 
1
(pg. 
1
-
8
)
48
Longo
 
PG
Laurenti
 
L
Gobessi
 
S
Sica
 
S
Leone
 
G
Efremov
 
DG
The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells.
Blood
2008
, vol. 
111
 
2
(pg. 
846
-
855
)
49
Schade
 
AE
Powers
 
JJ
Wlodarski
 
MW
Maciejewski
 
JP
Phosphatidylinositol-3-phosphate kinase pathway activation protects leukemic large granular lymphocytes from undergoing homeostatic apoptosis.
Blood
2006
, vol. 
107
 
12
(pg. 
4834
-
4840
)
50
Cuní
 
S
Perez-Aciego
 
P
Perez-Chacon
 
G
, et al. 
A sustained activation of PI3K/NF-kappaB pathway is critical for the survival of chronic lymphocytic leukemia B cells.
Leukemia
2004
, vol. 
18
 
8
(pg. 
1391
-
1400
)
51
Min
 
YH
Eom
 
JI
Cheong
 
JW
, et al. 
Constitutive phosphorylation of Akt/PKB protein in acute myeloid leukemia: its significance as a prognostic variable.
Leukemia
2003
, vol. 
17
 
5
(pg. 
995
-
997
)
52
Gallay
 
N
Dos Santos
 
C
Cuzin
 
L
, et al. 
The level of AKT phosphorylation on threonine 308 but not on serine 473 is associated with high-risk cytogenetics and predicts poor overall survival in acute myeloid leukaemia.
Leukemia
2009
, vol. 
23
 
6
(pg. 
1029
-
1038
)
53
Zhuang
 
J
Hawkins
 
SF
Glenn
 
MA
, et al. 
Akt is activated in chronic lymphocytic leukemia cells and delivers a pro-survival signal: the therapeutic potential of Akt inhibition.
Haematologica
2010
, vol. 
95
 
1
(pg. 
110
-
118
)
54
Schade
 
AE
Wlodarski
 
MW
Maciejewski
 
JP
Pathophysiology defined by altered signal transduction pathways: the role of JAK-STAT and PI3K signaling in leukemic large granular lymphocytes.
Cell Cycle
2006
, vol. 
5
 
22
(pg. 
2571
-
2574
)
55
Carpten
 
JD
Faber
 
AL
Horn
 
C
, et al. 
A transforming mutation in the pleckstrin homology domain of AKT1 in cancer.
Nature
2007
, vol. 
448
 
7152
(pg. 
439
-
444
)
56
Mahmoud
 
IS
Sughayer
 
MA
Mohammad
 
HA
Awidi
 
AS
EL-Khateeb
 
MS
Ismail
 
SI
The transforming mutation E17K/AKT1 is not a major event in B-cell-derived lymphoid leukaemias.
Br J Cancer
2008
, vol. 
99
 
3
(pg. 
488
-
490
)
57
Zenz
 
T
Dohner
 
K
Denzel
 
T
Dohner
 
H
Stilgenbauer
 
S
Bullinger
 
L
Chronic lymphocytic leukaemia and acute myeloid leukaemia are not associated with AKT1 pleckstrin homology domain (E17K) mutations.
Br J Haematol
2008
, vol. 
141
 
5
(pg. 
742
-
743
)
58
He
 
Y
Zheng
 
J
Hu
 
Y
, et al. 
Chronic myeloid leukemia and BCR/ABL signal pathways are not associated with AKT1 pleckstrin homology domain (E17K) mutations.
Eur J Haematol
2010
, vol. 
84
 
1
(pg. 
87
-
88
)
59
Tibes
 
R
Kornblau
 
SM
Qiu
 
Y
, et al. 
PI3K/AKT pathway activation in acute myeloid leukaemias is not associated with AKT1 pleckstrin homology domain mutation.
Br J Haematol
2008
, vol. 
140
 
3
(pg. 
344
-
347
)
60
Sato
 
T
Nakashima
 
A
Guo
 
L
Coffman
 
K
Tamanoi
 
F
Single amino-acid changes that confer constitutive activation of mTOR are discovered in human cancer.
Oncogene
2010
, vol. 
29
 
18
(pg. 
2746
-
2752
)
61
Brandts
 
CH
Sargin
 
B
Rode
 
M
, et al. 
Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation.
Cancer Res
2005
, vol. 
65
 
21
(pg. 
9643
-
9650
)
62
Hashimoto
 
K
Matsumura
 
I
Tsujimura
 
T
, et al. 
Necessity of tyrosine 719 and phosphatidylinositol 3′-kinase-mediated signal pathway in constitutive activation and oncogenic potential of c-kit receptor tyrosine kinase with the Asp814Val mutation.
Blood
2003
, vol. 
101
 
3
(pg. 
1094
-
1102
)
63
Skorski
 
T
Bellacosa
 
A
Nieborowska-Skorska
 
M
, et al. 
Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway.
EMBO J
1997
, vol. 
16
 
20
(pg. 
6151
-
6161
)
64
Clark
 
SS
McLaughlin
 
J
Timmons
 
M
, et al. 
Expression of a distinctive BCR-ABL oncogene in Ph1-positive acute lymphocytic leukemia (ALL).
Science
1988
, vol. 
239
 
4841
(pg. 
775
-
777
)
65
Harrison-Findik
 
D
Susa
 
M
Varticovski
 
L
Association of phosphatidylinositol 3-kinase with SHC in chronic myelogeneous leukemia cells.
Oncogene
1995
, vol. 
10
 
7
(pg. 
1385
-
1391
)
66
Ren
 
SY
Xue
 
F
Feng
 
J
Skorski
 
T
Intrinsic regulation of the interactions between the SH3 domain of p85 subunit of phosphatidylinositol-3 kinase and the protein network of BCR/ABL oncogenic tyrosine kinase.
Exp Hematol
2005
, vol. 
33
 
10
(pg. 
1222
-
1228
)
67
Hirano
 
I
Nakamura
 
S
Yokota
 
D
, et al. 
Depletion of Pleckstrin homology domain leucine-rich repeat protein phosphatases 1 and 2 by Bcr-Abl promotes chronic myelogenous leukemia cell proliferation through continuous phosphorylation of Akt isoforms.
J Biol Chem
2009
, vol. 
284
 
33
(pg. 
22155
-
22165
)
68
Hickey
 
FB
Cotter
 
TG
BCR-ABL regulates phosphatidylinositol 3-kinase-p110gamma transcription and activation and is required for proliferation and drug resistance.
J Biol Chem
2006
, vol. 
281
 
5
(pg. 
2441
-
2450
)
69
Naughton
 
R
Quiney
 
C
Turner
 
SD
Cotter
 
TG
Bcr-Abl-mediated redox regulation of the PI3K/AKT pathway.
Leukemia
2009
, vol. 
23
 
8
(pg. 
1432
-
1440
)
70
Yoshimi
 
A
Goyama
 
S
Watanabe-Okochi
 
N
, et al. 
Evi1 represses PTEN expression by interacting with polycomb complexes and activates PI3K/AKT/mTOR signaling.
Blood
2011
, vol. 
117
 
13
(pg. 
3617
-
3628
)
71
Martins
 
LR
Lucio
 
P
Silva
 
MC
Gameiro
 
P
Silva
 
MG
Barata
 
JT
On CK2 regulation of chronic lymphocytic leukemia cell viability.
Mol Cell Biochem
2011
, vol. 
356
 
1
(pg. 
51
-
55
)
72
Martins
 
LR
Lucio
 
P
Silva
 
MC
, et al. 
Targeting CK2 overexpression and hyperactivation as a novel therapeutic tool in chronic lymphocytic leukemia.
Blood
2010
, vol. 
116
 
15
(pg. 
2724
-
2731
)
73
Maeda
 
Y
Yamaguchi
 
T
Ueda
 
S
, et al. 
Mutant type glutathione S-transferase theta 1 gene homologue to mTOR in myelodysplastic syndrome: possible clinical application of rapamycin.
Leuk Lymphoma
2003
, vol. 
44
 
7
(pg. 
1179
-
1185
)
74
Maeda
 
Y
Yamaguchi
 
T
Sasakawa
 
A
, et al. 
Relationship between expression of mutant type glutathione S-transferase theta-1 gene and reactivity of rapamycin in myelodysplastic syndrome.
Hematology
2009
, vol. 
14
 
5
(pg. 
266
-
270
)
75
Shehata
 
M
Schnabl
 
S
Demirtas
 
D
, et al. 
Reconstitution of PTEN activity by CK2 inhibitors and interference with the PI3-K/Akt cascade counteract the antiapoptotic effect of human stromal cells in chronic lymphocytic leukemia.
Blood
2010
, vol. 
116
 
14
(pg. 
2513
-
2521
)
76
Niedermeier
 
M
Hennessy
 
BT
Knight
 
ZA
, et al. 
Isoform-selective phosphoinositide 3′-kinase inhibitors inhibit CXCR4 signaling and overcome stromal cell-mediated drug resistance in chronic lymphocytic leukemia: a novel therapeutic approach.
Blood
2009
, vol. 
113
 
22
(pg. 
5549
-
5557
)
77
Buchner
 
M
Baer
 
C
Prinz
 
G
, et al. 
Spleen tyrosine kinase inhibition prevents chemokine- and integrin-mediated stromal protective effects in chronic lymphocytic leukemia.
Blood
2010
, vol. 
115
 
22
(pg. 
4497
-
4506
)
78
Ferretti
 
E
Bertolotto
 
M
Deaglio
 
S
, et al. 
A novel role of the CX3CR1/CX3CL1 system in the cross-talk between chronic lymphocytic leukemia cells and tumor microenvironment.
Leukemia
2011
, vol. 
25
 
8
(pg. 
1268
-
1277
)
79
Ding
 
W
Nowakowski
 
GS
Knox
 
TR
, et al. 
Bi-directional activation between mesenchymal stem cells and CLL B-cells: implication for CLL disease progression.
Br J Haematol
2009
, vol. 
147
 
4
(pg. 
471
-
483
)
80
Lannutti
 
BJ
Meadows
 
SA
Herman
 
SE
, et al. 
CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability.
Blood
2011
, vol. 
117
 
2
(pg. 
591
-
594
)
81
Sykes
 
SM
Lane
 
SW
Bullinger
 
L
, et al. 
AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias.
Cell
2011
, vol. 
146
 
5
(pg. 
697
-
708
)
82
Meyer
 
LH
Eckhoff
 
SM
Queudeville
 
M
, et al. 
Early relapse in ALL is identified by time to leukemia in NOD/SCID mice and is characterized by a gene signature involving survival pathways.
Cancer Cell
2011
, vol. 
19
 
2
(pg. 
206
-
217
)
83
Jotta
 
PY
Ganazza
 
MA
Silva
 
A
, et al. 
Negative prognostic impact of PTEN mutation in pediatric T-cell acute lymphoblastic leukemia.
Leukemia
2010
, vol. 
24
 
1
(pg. 
239
-
242
)
84
Santamaría
 
CM
Chillon
 
MC
Garcia-Sanz
 
R
, et al. 
High FOXO3a expression is associated with a poorer prognosis in AML with normal cytogenetics.
Leuk Res
2009
, vol. 
33
 
12
(pg. 
1706
-
1709
)
85
Kornblau
 
SM
Singh
 
N
Qiu
 
Y
Chen
 
W
Zhang
 
N
Coombes
 
KR
Highly phosphorylated FOXO3A is an adverse prognostic factor in acute myeloid leukemia.
Clin Cancer Res
2010
, vol. 
16
 
6
(pg. 
1865
-
1874
)
86
Cheong
 
JW
Eom
 
JI
Maeng
 
HY
, et al. 
Phosphatase and tensin homologue phosphorylation in the C-terminal regulatory domain is frequently observed in acute myeloid leukaemia and associated with poor clinical outcome.
Br J Haematol
2003
, vol. 
122
 
3
(pg. 
454
-
456
)
87
Kornblau
 
SM
Womble
 
M
Qiu
 
YH
, et al. 
Simultaneous activation of multiple signal transduction pathways confers poor prognosis in acute myelogenous leukemia.
Blood
2006
, vol. 
108
 
7
(pg. 
2358
-
2365
)
88
Follo
 
MY
Mongiorgi
 
S
Bosi
 
C
, et al. 
The Akt/mammalian target of rapamycin signal transduction pathway is activated in high-risk myelodysplastic syndromes and influences cell survival and proliferation.
Cancer Res
2007
, vol. 
67
 
9
(pg. 
4287
-
4294
)
89
Nyåkern
 
M
Tazzari
 
PL
Finelli
 
C
, et al. 
Frequent elevation of Akt kinase phosphorylation in blood marrow and peripheral blood mononuclear cells from high-risk myelodysplastic syndrome patients.
Leukemia
2006
, vol. 
20
 
2
(pg. 
230
-
238
)
90
Tamburini
 
J
Elie
 
C
Bardet
 
V
, et al. 
Constitutive phosphoinositide 3-kinase/Akt activation represents a favorable prognostic factor in de novo acute myelogenous leukemia patients.
Blood
2007
, vol. 
110
 
3
(pg. 
1025
-
1028
)
91
Rosen
 
DB
Putta
 
S
Covey
 
T
, et al. 
Distinct patterns of DNA damage response and apoptosis correlate with Jak/Stat and PI3kinase response profiles in human acute myelogenous leukemia.
PLoS One
2010
, vol. 
5
 
8
pg. 
e12405
 
92
Tazzari
 
PL
Cappellini
 
A
Ricci
 
F
, et al. 
Multidrug resistance-associated protein 1 expression is under the control of the phosphoinositide 3 kinase/Akt signal transduction network in human acute myelogenous leukemia blasts.
Leukemia
2007
, vol. 
21
 
3
(pg. 
427
-
438
)
93
Mahadevan
 
D
List
 
AF
Targeting the multidrug resistance-1 transporter in AML: molecular regulation and therapeutic strategies.
Blood
2004
, vol. 
104
 
7
(pg. 
1940
-
1951
)
94
Gunther
 
R
Abbas
 
HK
Mirocha
 
CJ
Acute pathological effects on rats of orally administered wortmannin-containing preparations and purified wortmannin from Fusarium oxysporum.
Food Chem Toxicol
1989
, vol. 
27
 
3
(pg. 
173
-
179
)
95
Davies
 
SP
Reddy
 
H
Caivano
 
M
Cohen
 
P
Specificity and mechanism of action of some commonly used protein kinase inhibitors.
Biochem J
2000
, vol. 
351
 
1
(pg. 
95
-
105
)
96
Mao
 
X
Cao
 
B
Wood
 
TE
, et al. 
A small-molecule inhibitor of D-cyclin transactivation displays preclinical efficacy in myeloma and leukemia via phosphoinositide 3-kinase pathway.
Blood
2011
, vol. 
117
 
6
(pg. 
1986
-
1997
)
97
de Frias
 
M
Iglesias-Serret
 
D
Cosialls
 
AM
, et al. 
Isoform-selective phosphoinositide 3-kinase inhibitors induce apoptosis in chronic lymphocytic leukaemia cells.
Br J Haematol
2010
, vol. 
150
 
1
(pg. 
108
-
111
)
98
Herman
 
SE
Lapalombella
 
R
Gordon
 
AL
, et al. 
The role of phosphatidylinositol 3-kinase-delta in the immunomodulatory effects of lenalidomide in chronic lymphocytic leukemia.
Blood
2011
, vol. 
117
 
16
(pg. 
4323
-
4327
)
99
Hoellenriegel
 
J
Meadows
 
SA
Sivina
 
M
, et al. 
The phosphoinositide 3′-kinase delta inhibitor, CAL-101, inhibits B-cell receptor signaling and chemokine networks in chronic lymphocytic leukemia.
Blood
2011
, vol. 
118
 
13
(pg. 
3603
-
3612
)
100
Herman
 
SE
Gordon
 
AL
Wagner
 
AJ
, et al. 
Phosphatidylinositol 3-kinase-delta inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals.
Blood
2010
, vol. 
116
 
12
(pg. 
2078
-
2088
)
101
Fu
 
L
Kim
 
YA
Wang
 
X
, et al. 
Perifosine inhibits mammalian target of rapamycin signaling through facilitating degradation of major components in the mTOR axis and induces autophagy.
Cancer Res
2009
, vol. 
69
 
23
(pg. 
8967
-
8976
)
102
Chiarini
 
F
Del Sole
 
M
Mongiorgi
 
S
, et al. 
The novel Akt inhibitor, perifosine, induces caspase-dependent apoptosis and downregulates P-glycoprotein expression in multidrug-resistant human T-acute leukemia cells by a JNK-dependent mechanism.
Leukemia
2008
, vol. 
22
 
6
(pg. 
1106
-
1116
)
103
Papa
 
V
Tazzari
 
PL
Chiarini
 
F
, et al. 
Proapoptotic activity and chemosensitizing effect of the novel Akt inhibitor perifosine in acute myelogenous leukemia cells.
Leukemia
2008
, vol. 
22
 
1
(pg. 
147
-
160
)
104
Tabellini
 
G
Tazzari
 
PL
Bortul
 
R
, et al. 
Novel 2′-substituted, 3′-deoxy-phosphatidyl-myoinositol analogues reduce drug resistance in human leukaemia cell lines with an activated phosphoinositide 3-kinase/Akt pathway.
Br J Haematol
2004
, vol. 
126
 
4
(pg. 
574
-
582
)
105
de Frias
 
M
Iglesias-Serret
 
D
Cosialls
 
AM
, et al. 
Akt inhibitors induce apoptosis in chronic lymphocytic leukemia cells.
Haematologica
2009
, vol. 
94
 
12
(pg. 
1698
-
1707
)
106
Evangelisti
 
C
Ricci
 
F
Tazzari
 
P
, et al. 
Preclinical testing of the Akt inhibitor triciribine in T-cell acute lymphoblastic leukemia.
J Cell Physiol
2011
, vol. 
226
 
3
(pg. 
822
-
831
)
107
Hofbauer
 
SW
Pinon
 
JD
Brachtl
 
G
, et al. 
Modifying akt signaling in B-cell chronic lymphocytic leukemia cells.
Cancer Res
2010
, vol. 
70
 
18
(pg. 
7336
-
7344
)
108
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
2006
, vol. 
12
 
17
(pg. 
5165
-
5173
)
109
Decker
 
T
Sandherr
 
M
Goetze
 
K
Oelsner
 
M
Ringshausen
 
I
Peschel
 
C
A pilot trial of the mTOR (mammalian target of rapamycin) inhibitor RAD001 in patients with advanced B-CLL.
Ann Hematol
2009
, vol. 
88
 
3
(pg. 
221
-
227
)
110
Choi
 
J
Chen
 
J
Schreiber
 
SL
Clardy
 
J
Structure of the FKBP12-rapamycin complex interacting with the binding domain of human FRAP.
Science
1996
, vol. 
273
 
5272
(pg. 
239
-
242
)
111
Teachey
 
DT
Sheen
 
C
Hall
 
J
, et al. 
mTOR inhibitors are synergistic with methotrexate: an effective combination to treat acute lymphoblastic leukemia.
Blood
2008
, vol. 
112
 
5
(pg. 
2020
-
2023
)
112
Récher
 
C
Beyne-Rauzy
 
O
Demur
 
C
, et al. 
Antileukemic activity of rapamycin in acute myeloid leukemia.
Blood
2005
, vol. 
105
 
6
(pg. 
2527
-
2534
)
113
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
2008
, vol. 
14
 
9
(pg. 
2756
-
2762
)
114
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
2008
, vol. 
111
 
1
(pg. 
379
-
382
)
115
Guo
 
W
Schubbert
 
S
Chen
 
JY
, et al. 
Suppression of leukemia development caused by PTEN loss.
Proc Natl Acad Sci U S A
2011
, vol. 
108
 
4
(pg. 
1409
-
1414
)
116
Zent
 
CS
LaPlant
 
BR
Johnston
 
PB
, et al. 
The treatment of recurrent/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) with everolimus results in clinical responses and mobilization of CLL cells into the circulation.
Cancer
2010
, vol. 
116
 
9
(pg. 
2201
-
2207
)
117
Janes
 
MR
Limon
 
JJ
So
 
L
, et al. 
Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor.
Nat Med
2010
, vol. 
16
 
2
(pg. 
205
-
213
)
118
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
2011
, vol. 
25
 
5
(pg. 
781
-
791
)
119
Carayol
 
N
Vakana
 
E
Sassano
 
A
, et al. 
Critical roles for mTORC2- and rapamycin-insensitive mTORC1-complexes in growth and survival of BCR-ABL-expressing leukemic cells.
Proc Natl Acad Sci U S A
2010
, vol. 
107
 
28
(pg. 
12469
-
12474
)
120
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
2011
, vol. 
17
 
13
(pg. 
4378
-
4388
)
121
Weisberg
 
E
Banerji
 
L
Wright
 
RD
, et al. 
Potentiation of antileukemic therapies by the dual PI3K/PDK-1 inhibitor, BAG956: effects on BCR-ABL- and mutant FLT3-expressing cells.
Blood
2008
, vol. 
111
 
7
(pg. 
3723
-
3734
)
122
Hahn
 
M
Li
 
W
Yu
 
C
Rahmani
 
M
Dent
 
P
Grant
 
S
Rapamycin and UCN-01 synergistically induce apoptosis in human leukemia cells through a process that is regulated by the Raf-1/MEK/ERK, Akt, and JNK signal transduction pathways.
Mol Cancer Ther
2005
, vol. 
4
 
3
(pg. 
457
-
470
)
123
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
2008
, vol. 
7
 
7
(pg. 
1851
-
1863
)
124
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
2010
, vol. 
70
 
20
(pg. 
8097
-
8107
)
125
Chapuis
 
N
Tamburini
 
J
Green
 
AS
, et al. 
Dual inhibition of PI3K and mTORC1/2 signaling by NVP-BEZ235 as a new therapeutic strategy for acute myeloid leukemia.
Clin Cancer Res
2010
, vol. 
16
 
22
(pg. 
5424
-
5435
)
126
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
2009
, vol. 
69
 
8
(pg. 
3520
-
3528
)
127
Park
 
S
Chapuis
 
N
Bardet
 
V
, et al. 
PI-103, a dual inhibitor of Class IA phosphatidylinositide 3-kinase and mTOR, has antileukemic activity in AML.
Leukemia
2008
, vol. 
22
 
9
(pg. 
1698
-
1706
)
128
Zeng
 
Z
Samudio
 
IJ
Zhang
 
W
, et al. 
Simultaneous inhibition of PDK1/AKT and Fms-like tyrosine kinase 3 signaling by a small-molecule KP372-1 induces mitochondrial dysfunction and apoptosis in acute myelogenous leukemia.
Cancer Res
2006
, vol. 
66
 
7
(pg. 
3737
-
3746
)
129
Nishioka
 
C
Ikezoe
 
T
Yang
 
J
Koeffler
 
HP
Yokoyama
 
A
Blockade of mTOR signaling potentiates the ability of histone deacetylase inhibitor to induce growth arrest and differentiation of acute myelogenous leukemia cells.
Leukemia
2008
, vol. 
22
 
12
(pg. 
2159
-
2168
)
130
Rahmani
 
M
Reese
 
E
Dai
 
Y
, et al. 
Coadministration of histone deacetylase inhibitors and perifosine synergistically induces apoptosis in human leukemia cells through Akt and ERK1/2 inactivation and the generation of ceramide and reactive oxygen species.
Cancer Res
2005
, vol. 
65
 
6
(pg. 
2422
-
2432
)
131
Horton
 
TM
Pati
 
D
Plon
 
SE
, et al. 
A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children's Oncology Group study.
Clin Cancer Res
2007
, vol. 
13
 
5
(pg. 
1516
-
1522
)
132
Saunders
 
P
Cisterne
 
A
Weiss
 
J
Bradstock
 
KF
Bendall
 
LJ
The mammalian target of rapamycin inhibitor RAD001 (everolimus) synergizes with chemotherapeutic agents, ionizing radiation and proteasome inhibitors in pre-B acute lymphocytic leukemia.
Haematologica
2011
, vol. 
96
 
1
(pg. 
69
-
77
)
133
Gorre
 
ME
Mohammed
 
M
Ellwood
 
K
, et al. 
Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification.
Science
2001
, vol. 
293
 
5531
(pg. 
876
-
880
)
134
Burchert
 
A
Wang
 
Y
Cai
 
D
, et al. 
Compensatory PI3-kinase/Akt/mTor activation regulates imatinib resistance development.
Leukemia
2005
, vol. 
19
 
10
(pg. 
1774
-
1782
)
135
Okabe
 
S
Tauchi
 
T
Ohyashiki
 
K
Characteristics of dasatinib- and imatinib-resistant chronic myelogenous leukemia cells.
Clin Cancer Res
2008
, vol. 
14
 
19
(pg. 
6181
-
6186
)
136
Hui
 
RC
Gomes
 
AR
Constantinidou
 
D
, et al. 
The forkhead transcription factor FOXO3a increases phosphoinositide-3 kinase/Akt activity in drug-resistant leukemic cells through induction of PIK3CA expression.
Mol Cell Biol
2008
, vol. 
28
 
19
(pg. 
5886
-
5898
)
137
Joha
 
S
Nugues
 
AL
Hetuin
 
D
, et al. 
GILZ inhibits the mTORC2/AKT pathway in BCR-ABL(+) cells. [published online adead of print, August 1, 2011].
Oncogene
 
138
Mancini
 
M
Corradi
 
V
Petta
 
S
Martinelli
 
G
Barbieri
 
E
Santucci
 
MA
mTOR inhibitor RAD001 (Everolimus) enhances the effects of imatinib in chronic myeloid leukemia by raising the nuclear expression of c-ABL protein.
Leuk Res
2010
, vol. 
34
 
5
(pg. 
641
-
648
)
139
Parmar
 
S
Smith
 
J
Sassano
 
A
, et al. 
Differential regulation of the p70 S6 kinase pathway by interferon alpha (IFNalpha) and imatinib mesylate (STI571) in chronic myelogenous leukemia cells.
Blood
2005
, vol. 
106
 
7
(pg. 
2436
-
2443
)
140
Tseng
 
PH
Lin
 
HP
Zhu
 
J
, et al. 
Synergistic interactions between imatinib mesylate and the novel phosphoinositide-dependent kinase-1 inhibitor OSU-03012 in overcoming imatinib mesylate resistance.
Blood
2005
, vol. 
105
 
10
(pg. 
4021
-
4027
)
141
Klejman
 
A
Rushen
 
L
Morrione
 
A
Slupianek
 
A
Skorski
 
T
Phosphatidylinositol-3 kinase inhibitors enhance the anti–leukemia effect of STI571.
Oncogene
2002
, vol. 
21
 
38
(pg. 
5868
-
5876
)
142
Kharas
 
MG
Janes
 
MR
Scarfone
 
VM
, et al. 
Ablation of PI3K blocks BCR-ABL leukemogenesis in mice, and a dual PI3K/mTOR inhibitor prevents expansion of human BCR-ABL+ leukemia cells.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3038
-
3050
)
143
Burnett
 
A
Wetzler
 
M
Lowenberg
 
B
Therapeutic advances in acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
5
(pg. 
487
-
494
)
144
Xu
 
Q
Thompson
 
JE
Carroll
 
M
mTOR regulates cell survival after etoposide treatment in primary AML cells.
Blood
2005
, vol. 
106
 
13
(pg. 
4261
-
4268
)
145
Billottet
 
C
Grandage
 
VL
Gale
 
RE
, et al. 
A selective inhibitor of the p110delta isoform of PI 3-kinase inhibits AML cell proliferation and survival and increases the cytotoxic effects of VP16.
Oncogene
2006
, vol. 
25
 
50
(pg. 
6648
-
6659
)
146
Sampath
 
D
Cortes
 
J
Estrov
 
Z
, et al. 
Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial.
Blood
2006
, vol. 
107
 
6
(pg. 
2517
-
2524
)
147
Wymann
 
MP
Bulgarelli-Leva
 
G
Zvelebil
 
MJ
, et al. 
Wortmannin inactivates phosphoinositide 3-kinase by covalent modification of Lys-802, a residue involved in the phosphate transfer reaction.
Mol Cell Biol
1996
, vol. 
16
 
4
(pg. 
1722
-
1733
)
148
Marley
 
SB
Lewis
 
JL
Schneider
 
H
Rudd
 
CE
Gordon
 
MY
Phosphatidylinositol-3 kinase inhibitors reproduce the selective antiproliferative effects of imatinib on chronic myeloid leukaemia progenitor cells.
Br J Haematol
2004
, vol. 
125
 
4
(pg. 
500
-
511
)
149
Xu
 
Q
Simpson
 
SE
Scialla
 
TJ
Bagg
 
A
Carroll
 
M
Survival of acute myeloid leukemia cells requires PI3 kinase activation.
Blood
2003
, vol. 
102
 
3
(pg. 
972
-
980
)
150
Zhao
 
S
Konopleva
 
M
Cabreira-Hansen
 
M
, et al. 
Inhibition of phosphatidylinositol 3-kinase dephosphorylates BAD and promotes apoptosis in myeloid leukemias.
Leukemia
2004
, vol. 
18
 
2
(pg. 
267
-
275
)
151
Levy
 
DS
Kahana
 
JA
Kumar
 
R
AKT inhibitor, GSK690693, induces growth inhibition and apoptosis in acute lymphoblastic leukemia cell lines.
Blood
2009
, vol. 
113
 
8
(pg. 
1723
-
1729
)
152
Aleskog
 
A
Norberg
 
M
Nygren
 
P
, et al. 
Rapamycin shows anticancer activity in primary chronic lymphocytic leukemia cells in vitro, as single agent and in drug combination.
Leuk Lymphoma
2008
, vol. 
49
 
12
(pg. 
2333
-
2343
)
153
Hirase
 
C
Maeda
 
Y
Takai
 
S
Kanamaru
 
A
Hypersensitivity of Ph-positive lymphoid cell lines to rapamycin: possible clinical application of mTOR inhibitor.
Leuk Res
2009
, vol. 
33
 
3
(pg. 
450
-
459
)
154
Sillaber
 
C
Mayerhofer
 
M
Bohm
 
A
, et al. 
Evaluation of antileukaemic effects of rapamycin in patients with imatinib-resistant chronic myeloid leukaemia.
Eur J Clin Invest
2008
, vol. 
38
 
1
(pg. 
43
-
52
)
155
Mayerhofer
 
M
Aichberger
 
KJ
Florian
 
S
, et al. 
Identification of mTOR as a novel bifunctional target in chronic myeloid leukemia: dissection of growth-inhibitory and VEGF-suppressive effects of rapamycin in leukemic cells.
FASEB J
2005
, vol. 
19
 
8
(pg. 
960
-
962
)
156
Teachey
 
DT
Obzut
 
DA
Cooperman
 
J
, et al. 
The mTOR inhibitor CCI-779 induces apoptosis and inhibits growth in preclinical models of primary adult human ALL.
Blood
2006
, vol. 
107
 
3
(pg. 
1149
-
1155
)
157
Hsieh
 
AC
Costa
 
M
Zollo
 
O
, et al. 
Genetic dissection of the oncogenic mTOR pathway reveals druggable addiction to translational control via 4EBP-eIF4E.
Cancer Cell
2010
, vol. 
17
 
3
(pg. 
249
-
261
)
158
Kojima
 
K
Shimanuki
 
M
Shikami
 
M
, et al. 
The dual PI3 kinase/mTOR inhibitor PI-103 prevents p53 induction by Mdm2 inhibition but enhances p53-mediated mitochondrial apoptosis in p53 wild-type AML.
Leukemia
2008
, vol. 
22
 
9
(pg. 
1728
-
1736
)
159
Neri
 
LM
Borgatti
 
P
Tazzari
 
PL
, et al. 
The phosphoinositide 3-kinase/AKT1 pathway involvement in drug and all-trans-retinoic acid resistance of leukemia cells.
Mol Cancer Res
2003
, vol. 
1
 
3
(pg. 
234
-
246
)
160
Cheong
 
JW
Min
 
YH
Eom
 
JI
Kim
 
SJ
Jeung
 
HK
Kim
 
JS
Inhibition of CK2α and PI3K/Akt synergistically induces apoptosis of CD34+CD38− leukaemia cells while sparing haematopoietic stem cells.
Anticancer Res
2010
, vol. 
30
 
11
(pg. 
4625
-
4634
)
161
Redondo-Muñoz
 
J
Escobar-Diaz
 
E
Hernandez Del Cerro
 
M
, et al. 
Induction of B-chronic lymphocytic leukemia cell apoptosis by arsenic trioxide involves suppression of the phosphoinositide 3-kinase/Akt survival pathway via c-jun-NH2 terminal kinase activation and PTEN upregulation.
Clin Cancer Res
2010
, vol. 
16
 
17
(pg. 
4382
-
4391
)
162
Cristóbal
 
I
Garcia-Orti
 
L
Cirauqui
 
C
Alonso
 
MM
Calasanz
 
MJ
Odero
 
MD
PP2A impaired activity is a common event in acute myeloid leukemia and its activation by forskolin has a potent anti–leukemic effect.
Leukemia
2011
, vol. 
25
 
4
(pg. 
606
-
614
)
163
Tazzari
 
PL
Tabellini
 
G
Ricci
 
F
, et al. 
Synergistic proapoptotic activity of recombinant TRAIL plus the Akt inhibitor Perifosine in acute myelogenous leukemia cells.
Cancer Res
2008
, vol. 
68
 
22
(pg. 
9394
-
9403
)
164
Akers
 
LJ
Fang
 
W
Levy
 
AG
Franklin
 
AR
Huang
 
P
Zweidler-McKay
 
PA
Targeting glycolysis in leukemia: A novel inhibitor 3-BrOP in combination with rapamycin.
Leuk Res
2011
, vol. 
35
 
6
(pg. 
814
-
820
)
165
Chan
 
SM
Weng
 
AP
Tibshirani
 
R
Aster
 
JC
Utz
 
PJ
Notch signals positively regulate activity of the mTOR pathway in T-cell acute lymphoblastic leukemia.
Blood
2007
, vol. 
110
 
1
(pg. 
278
-
286
)
166
Boehrer
 
S
Galluzzi
 
L
Lainey
 
E
, et al. 
Erlotinib antagonizes constitutive activation of SRC family kinases and mTOR in acute myeloid leukemia.
Cell Cycle
2011
, vol. 
10
 
18
(pg. 
3168
-
3175
)
167
Hayun
 
R
Okun
 
E
Berrebi
 
A
, et al. 
Rapamycin and curcumin induce apoptosis in primary resting B chronic lymphocytic leukemia cells.
Leuk Lymphoma
2009
, vol. 
50
 
4
(pg. 
625
-
632
)
168
Gu
 
L
Zhou
 
C
Liu
 
H
, et al. 
Rapamycin sensitizes T-ALL cells to dexamethasone-induced apoptosis.
J Exp Clin Cancer Res
2010
, vol. 
29
 pg. 
150
 
169
Bonapace
 
L
Bornhauser
 
BC
Schmitz
 
M
, et al. 
Induction of autophagy-dependent necroptosis is required for childhood acute lymphoblastic leukemia cells to overcome glucocorticoid resistance.
J Clin Invest
2010
, vol. 
120
 
4
(pg. 
1310
-
1323
)
170
Avellino
 
R
Romano
 
S
Parasole
 
R
, et al. 
Rapamycin stimulates apoptosis of childhood acute lymphoblastic leukemia cells.
Blood
2005
, vol. 
106
 
4
(pg. 
1400
-
1406
)
171
Yang
 
X
Lin
 
J
Gong
 
Y
, et al. 
Antileukaemia effect of rapamycin alone or in combination with daunorubicin on Ph+ acute lymphoblastic leukaemia cell line. [published online ahead of print, September 4, 2011].
Hematol Oncol
 
172
Nishioka
 
C
Ikezoe
 
T
Yang
 
J
Gery
 
S
Koeffler
 
HP
Yokoyama
 
A
Inhibition of mammalian target of rapamycin signaling potentiates the effects of all-trans retinoic acid to induce growth arrest and differentiation of human acute myelogenous leukemia cells.
Int J Cancer
2009
, vol. 
125
 
7
(pg. 
1710
-
1720
)
173
Crazzolara
 
R
Cisterne
 
A
Thien
 
M
, et al. 
Potentiating effects of RAD001 (Everolimus) on vincristine therapy in childhood acute lymphoblastic leukemia.
Blood
2009
, vol. 
113
 
14
(pg. 
3297
-
3306
)
174
Hong
 
Z
Xiao
 
M
Yang
 
Y
, et al. 
Arsenic disulfide synergizes with the phosphoinositide 3-kinase inhibitor PI-103 to eradicate acute myeloid leukemia stem cells by inducing differentiation.
Carcinogenesis
2011
, vol. 
32
 
10
(pg. 
1550
-
1558
)
Sign in via your Institution