Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Mar 1.
Published in final edited form as: Mol Genet Metab. 2014 Dec 5;114(3):397–402. doi: 10.1016/j.ymgme.2014.11.017

_targeting Novel Signaling Pathways for Resistant Acute Myeloid Leukemia

Kathleen M Sakamoto 1,*, Steven Grant 2, Diana Saleiro 3, John Crispino 3, Nobuko Hijiya 5, Francis Giles 3, Leonidas Platanias 3,4, Elizabeth A Eklund 3,4
PMCID: PMC4355162  NIHMSID: NIHMS647532  PMID: 25533111

Abstract

Acute myeloid leukemia (AML) is a hematologic malignancy that is the most common type of acute leukemia diagnosed in adults and the second most common type in children. The overall survival is poor and treatment is associated with significant complications and even death. In addition, a significant number of patients will not respond to therapy or relapse. In this review, several new signaling proteins aberrantly regulated in AML are described, including CREB, Triad1, Bcl-2 family members, Stat3, and mTOR/MEK. Identifying more effective and less toxic agents will provide novel approaches to treat AML.

Keywords: acute myeloid leukemia, signaling pathways, novel therapies, resistance

Introduction

Acute Myeloid Leukemia (AML) is a hematologic malignancy that originates in hematopoietic stem and myeloid progenitor cells (1-3). AML is the most common type of acute leukemia diagnosed in adults and the second most common leukemia in children. Despite intensive chemotherapy and stem cell transplantation, the overall survival is less than 30% for adults and 60% for children (3). In addition, the treatment for AML results in significant morbidity and late effects, including secondary malignancies or graft vs. host disease after stem cell transplant. The major challenge in finding a cure for AML is the development of resistant disease and subsequent relapse. Current advances in technology include genomic sequencing, epigenetic and phenotypic characterization, and identification of novel signaling pathways. Current efforts in improving AML therapy focus on inhibiting proteins that promote drug resistance and survival of AML initiating cells. Here, we review select signaling molecules and pathways including CREB, Triad1, Bcl-2 family members, Stat3, and mTOR/MEK that are aberrantly regulated in AML and potential _targets for therapy in patients with resistant disease.

CREB

Cyclic AMP Response Element Binding Protein (CREB) is a leucine zipper transcription factor that is critical for cell proliferation, survival and differentiation (4). CREB can act as an activator or repressor to regulate genes that control cell cycle, metabolism, and apoptosis in diverse cell types (5, 6). CREB-dependent signaling is important for gluconeogenesis, memory, neuronal plasticity, and hippocampal development. Upstream kinases that activate CREB include MAP kinase, RSK, adenylate cyclase, and CAMKIV (4, 7). A critical phosphorylation site in CREB identified in leukemia cells is serine 133, through MAP kinase/RSK activation (4-6). CREB phosphorylation leads to interaction with the histone acetyltransferase, CREB Binding Protein (CBP) and induction of specific CREB _target genes (5, 6).

CREB is overexpressed in acute leukemia cells from the majority of patients with both AML and acute lymphocytic leukemia (8, 9). The expression of CREB was identified primarily at the protein level but in certain AML samples, also at the mRNA level (10, 11). Expression of CREB protein in leukemia cells from AML patients was associated with decreased event-free survival and increased risk of relapse (5). Overexpression of CREB in AML cell lines resulted in increased cell proliferation and growth in the absence of cytokines (5). Lentiviral transduction of CREB in primary hematopoietic cells results in increased myeloid progenitors and colony formation (5). Transgenic mice expressing CREB in myeloid progenitors leads to a myeloproliferative syndrome after approximately 1 year, but not AML, suggesting additional cooperating oncogenes required for full transformation (5). Retroviral insertional mutagenesis led to the finding that the proto-oncogene Sox4 cooperates with CREB towards leukemogenesis (12). In addition, Sox4 and CREB are both highly expressed in AML cells from a significant number of patients with the disease (12).

Another interesting mechanism of CREB regulation is by microRNA34b (miR34b) (13, 14). In AML cells, the promoter of miR34b is highly methylated leading to increased AML cell proliferation. Mir34b is a negative regulator of CREB expression and binds directly to the 3’ UTR of CREB mRNA. Mir34b is upregulated in cells from patients with JMML and MPN/MDS, but downregulated in AML cells. These data suggest that CREB is critical for full transformation to AML (13).

Since CREB is overexpressed in leukemia cells from the majority of AML patients and is associated with a worse prognosis, CREB might be a potential _target for therapy. Knockdown of CREB using shRNA lentiviral constructs significantly inhibited the growth of AML cells (15, 16). This appears to be in part due to inhibition of expression of cell cycle genes, such as cyclins A and D. Furthermore, in vivo knockdown of CREB in an aggressive model of BCR/ABL-driven leukemia resulted in a statistically significant increase in median survival (16). Transduction of CREB shRNA in normal hematopoietic stem cells did not affect long-term engraftment, although there were transient effects on short-term engraftment and myelopoiesis (16). Therefore, CREB is critical for AML cells but is not required for normal hematopoietic stem cell activity. Taken together, these results suggest that CREB and CREB-dependent pathways are potential avenues for drug development for treatment of AML. Small molecule inhibitors of CREB are currently under development (17).

Triad1

The ARIH2 gene encodes Triad1; an E3 ubiquitin ligase that is expressed in bone marrow progenitor cells and increases in expression during granulopoiesis (18). Interaction of Triad1 with Ubc7 or Ubc13 (E2 ligases) results in K48 or K63 linked ubiquitin chains, respectively (19, 20). K48-linked chains lead to proteasomal or lysosomal degradation, and K63 only the latter. Consistent with this, Triad1 participates in lysosomal degradation of EGF-R and GH-R in epithelial cells, but may also facilitate proteasomal degradation of Mdm2 in multiple cell types (21, 22). Therefore, Triad1-inhibition results in recycling and sustained signaling by these growth factor receptors, and enhances degradation of Mdm2 substrates such as p53. Furthermore, Triad1 inhibits Mdm2 and Mdm2 degrades p53. Increased Triad1 stabilizes p53 by impairing Mdm2 activity, which is anti-oncogenic. Inhibition of Triad1 stabilizes Mdm2 resulting in increased p53 degradation, which would facilitate cell survival along with stabilization of growth factor receptors. Triad1 also associates with cullin proteins (23). The significance of this interaction is unknown, but suggests that Triad1 may broadly effect protein-ubiquitination by regulating cullin ligases.

A number of laboratories found that engineered overexpression of Triad1 in bone marrow progenitor cells decreases colony formation and impairs cytokine stimulated proliferation (18, 21, 24). The mechanism for this is unknown and no hematopoietic specific Triad1 substrates have been identified. Disruption of the ARIH2 gene in mice is embryonic lethal due to hepatocyte apoptosis at ~E16 (25). Despite this, E16 fetal liver-hematopoietic cells are normal in colony forming assays and repopulate hematopoiesis in irradiated wild type mice (25). However, recipients of Triad1−/− hematopoietic cells die rapidly of an inflammatory process that involves dendritic cell activation (25).

Clinical correlative studies suggest that Triad1 may function as a leukemia suppressor. For example, ARIH2 is located on chromosome 3p21 and deletion of this region is reported in AML and blast crisis CML (26-28). And, examination of publically available databases defines a specific decrease in Triad1 mRNA in AML with chromosomal translocations involving the MLL1- gene (11q23-AML), MYST4/CREBB gene translocation, and FLT3 internal tandem duplication (ITD) (29). The first two are associated with increased expression of a set of homeodomain transcription factors that includes HoxB4, B4, A7-11 and Meis1 (30, 31). And, incidence of ITD-FLT3 mutation is increased in Hox-overexpressing-AML (29). Additionally, a set of twins were reported both of whom had the same MLL1-gene translocation, but leukemia only developed in the twin who also had a deletion of 3p21 (28).

Functional studies of bone marrow progenitor cells also support a role for Triad1 as a leukemia suppressor. HoxA10 activates ARIH2 gene transcription during myelopoiesis in a tyrosine phosphorylation dependent manner (24). HoxA10 is a substrate for Shp2 protein tyrosine phosphatase, and HoxA10-induced Triad1 expression is blocked by constitutive activation of Shp2 (24,32-35). Engineered overexpression of HoxA10 in bone marrow progenitor cells increases cytokine induced proliferation in vitro, and results in a myeloproliferative neoplasm with neutrophilia in vivo (35-38). However, effects of HoxA10-overexpression are paradoxically antagonized by the increase in Triad1 expression that is observed in these cells (24). This suggests the possibility that Triad1 antagonizes generally pro-proliferative effects of Hox proteins via ubiquitin-mediated degradation of proteins that are involved in hematopoietic stem and progenitor cell expansion.

The myeloproliferative neoplasm that develops in mice with HoxA10-overexpressing bone marrow progresses to AML with time (35, 36). This latency suggests that additional mutations are required for induction of AML. Such mutations may include events that silence leukemia suppressors, such as Triad1. Perhaps in support of this hypothesis, AML develops rapidly in mice transplanted with bone marrow that is co-overexpressing HoxA10 plus a constitutively active form of Shp2 (35). Both Shp2 activation and increased HOX expression are associated with FLT3 internal tandem duplication in human AML (19, 39, 40).

ITD-FLT3 mutation and increased HOX expression are associated with drug resistance and poor outcomes in AML Defining relevant substrates for Triad1 in leukemic progenitor cells may suggest novel molecular therapeutic _targets to address this problem.

_targeting MLL leukemias with BH3-mimetics and survival signaling inhibitors

Rearrangements of the mixed lineage leukemia (MLL) gene on chromosome 11q23 occurs in up to 10% of acute myelogenous leukemia (AML), and is generally associated with a relatively unfavorable prognosis (41). Consequently, this AML sub-type may represent a prototype of intrinsically resistant AML. The MLL rearrangement is associated with multiple translocation partners, most frequently AF9 in AML (42), which result in a variety of genetic and epigenetic aberrations culminating in increased cell survival (43). Over the last decade, rational and highly specific therapeutic strategies _targeting MLL leukemias have focused on disrupting MLL fusion partners and enzymes implicated in leukemogenesis. These _targets have included menin, the histone methyltransferase DOT1L, the transcriptional elongation factor p-TEFb, and bromodomain proteins (e.g. BRD4), among others (42).

Resistance to therapy of AML in general, and MLL leukemias in particular, may also stem from aberrant expression of Bcl-2 family proteins (44). This consideration has prompted interest in BH3-mimetics, such as the Bcl-2/Bcl-xL antagonist ABT-737 and the Bcl-2-selective antagonist ABT-199 as therapeutic candidates in AML (45, 46). BH3-mimetics mimic the actions of BH3-only pro-apoptotic proteins such as Bim and effectively neutralize the pro-survival functions of anti-apoptotic multi-domain Bcl-2 family members. Past efforts have highlighted the value of BH3-profiling in predicting whether a particular tumor type may or may not be susceptible to such BH3-mimetics (47). Interestingly, MLL leukemias have very recently been shown to be extremely sensitive to ABT-199, suggesting that these cells are highly dependent upon Bcl-2 functions for their survival (48).

While the use of such BH3-mimetics in combination with standard chemotherapy to overcome resistance in MLL or other forms of AML represents a logical strategy, an alternative approach involves disrupting survival signaling pathways, including the MAPK and PI3K/AKT/mTOR pathways, with the goal of further enhancing BH3-mimetic anti-leukemic potency. For example, activation of the MEK1/2/ERK1/2 pathway regulates the abundance of the pro-apoptotic molecule Bim by phosphorylating it and promoting its proteasomal degradation (49). Indeed, MEK1/2 inhibitors have been shown to interact synergistically with ABT-737 in AML cells, in part by diminishing the expression of the anti-apoptotic protein Mcl-1 (50). Similarly, activity of the PI3K/AKT/mTOR pathway is known to be important for AML cell survival (51) as well as maintenance of expression of Mcl-1 through a GSK-dependent pathway (52). Notably, PI3K inhibitors have recently been reported to increase the anti-leukemic activity of ABT-737 in diverse AML cell types, including those exhibiting MLL rearrangements (e.g. MV4-11) (53). Given the intrinsic sensitivity of MLL leukemias to BH3-mimetics, such dual _targeting may be highly appropriate in this setting. Collectively, these findings raise the possibility that in addition to _targeting proteins directly implicated in MLL-related leukemogenesis (e.g. menin, DOT1L etc.), a strategy combining clinically relevant BH3-mimetics with PI3K or MEK1/2 inhibitors could prove to be particularly effective against this intrinsically resistant form of AML.

_targeting MAPK and mTOR pathways in AML

Recent studies have shown that AML relapse is associated with the gain of additional mutations and clonal evolution, due to the cytotoxic chemotherapy that patients receive and survival of preleukemic clones (54-57). Thus, the need for novel and effective therapies for AML remains a high clinical priority. This is particularly true for selective _targeted therapeutic approaches that can be combined with chemotherapy.

Several mutations lead to constitutive activation of signals that promote expansion and survival of the leukemic clones (58, 59). Among them, the mitogen-activated protein kinase (MAPK) and mammalian _target of rapamycin (mTOR) pathways play prominent roles (60-63). MAPK pathways include at least four major signaling cascades: extracellular signal-related kinases (ERK) 1 and 2, c-JUN N-terminal kinase (JNK) 1, 2, and 3, p38 MAPK, and ERK5 (63). Extensive studies from many groups have shown that molecular or pharmacological _targeting of MAPK signaling cascades, alone or in combination with other drugs, results in enhanced anti-leukemic responses in AML (64, 65). Work from our laboratory has shown that the downstream effector of MAPK pathways, Mnk kinases, may be attractive _targets for the treatment of AML (66, 67). Specifically molecular or pharmacological _targeting of Mnk kinases promotes anti-leukemic responses in pre-clinical AML models (66, 67).

mTOR can form two distinct multiprotein complexes: mTOR complex 1 (mTORC1) and 2 (mTORC2) (60, 61). Although the complexes include mTOR as their central common element, they have different purposes and functions (60, 61). Activation of mTORC1 complexes controls mRNA translation of oncogenic proteins, cell cycle progression, autophagy, and cellular growth and metabolism (60, 61), while mTORC2 complexes regulate cellular metabolism, and promotes malignant cell survival (60, 61). Inhibition of mTORC1 pathways using rapamycin or other rapalogs alone or in combination with other anti-leukemic agents, including chemotherapeutic regimens, have shown potent anti-leukemic properties in vitro and in vivo (reviewed in 68). Recent studies have also raised the possibility that catalytic mTOR inhibitors alone or in combination with other chemotherapeutic agents may induce more potent anti-leukemic effects than single- or combined-rapamycin treatment (69-71). This is important as such agents inhibit both mTORC1 and mTORC2 complexes and, potentially, other mTOR complexes that may exist but have not been yet identified. Other studies have also described potent anti-leukemic properties of dual PI3K-mTORC1/2 inhibitors, such as NVP-BGT226 (72) and NVP-BEZ235 (73), in AML.

One approach that may result in more potent anti-leukemic effects includes co-_targeting negative feedback regulatory cascades that may be activated during AML treatment of cells with chemotherapy, or selective _targeted therapy (67, 68, 75). The combination of MAPK signaling inhibitors and mTORC1/2 inhibitors is such an example (76-78). In fact, the combination of the catalytic mTOR inhibitor AZD8055 with the MEK inhibitor selumetinib was shown to exert synergistic proapoptotic effects in AML cells (78). Altogether, there is accumulating evidence supporting the development of future clinical trials combining such agents for the treatment of AML.

_targeting STAT3 in myeloid malignancies

Activation of JAK/STAT signaling is a hallmark of the myeloproliferative neoplasms (MPNs). Mutations in JAK2, MPL, and CALR, which account for the vast majority of MPN cases, all result in increased STAT activation and subsequent cytokine hypersensitivity or independence (79, 80). The identification of the V617F activating allele of JAK2 in primary myelofibrosis (PMF) led to rapid clinical development of JAK inhibitors, with the hope that such drugs would be analogous to imatinib in CML. Unfortunately, despite providing important clinical benefits and a survival advantage (81, 82), the only FDA approved JAK inhibitor, ruxolitinib, does not cure PMF (83). In addition, recent studies have found that myelofibrosis cells develop JAK inhibitor resistance (84) as well as have an intrinsic resistance (85), which in both cases, allows for continued STAT3/5 phosphorylation.

Activating JAK2 mutations are less commonly seen in other myeloid malignancies, with one large study reporting an incidence of 3% in de novo AML (86). Despite this low incidence of JAK mutations, an intriguing report demonstrated that STAT3 activation is common in de novo AML (87). Moreover, several studies have revealed that other oncogenic events can drive enhanced STAT phosphorylation. For example, the t(8;21) fusion has been shown to enhance the JAK/STAT pathway (88). FLT3 ITD also leads to enhanced JAK/STAT signaling (89). The knowledge that the JAK/STAT pathway is activated in many cases of AML, and data from pre-clinical studies that have shown an anti-tumor effect for JAK inhibitors (for example, see ref 88), have led to initiation of clinical trials of JAK inhibitors in AML. However, in a Phase 2 study, ruxolitinib only showed modest anti-leukemia activity as a single agent, with 3 significant responses in a study of 18 post-MPN AML patients (90).

Although there has been a focus on the upstream kinase JAK2 as a _target for therapy, many studies suggest that directly _targeting STAT3 may be effective. Indeed, several types of STAT inhibitors are under development as cancer therapies (91). For example, the small molecule STAT3 inhibitor C188-9 induced apoptosis in multiple AML cell lines and primary cells (92). A different STAT3 inhibitor, OPB-31121, inhibited STAT3 and STAT5 phosphorylation and had strong anti-growth effect on human leukemia cells, including those with FLT3-ITD or JAK2 V617F (93). _targeting STAT3 may also be key in treatment of chemotherapy resistant AML. A recent study revealed that feedback activation of STAT3 in cancer cells facilitated drug resistance in tumors with activation of a variety of kinases, including EGFR, MET and KRAS (94). Thus, combining STAT3 inhibitor with other _targeted therapies or conventional chemotherapy may be effective in combating refractory and relapsed AML.

In conclusion, although advances have been made in the treatment of certain types of cancer due to development of _targeted therapies, patients with AML continue to receive intensive chemotherapy and stem cell transplantation. These approaches result in significant toxicities and long-term complications. The primary challenge in finding a cure for AML, is the development of resistance following chemotherapy. The identification and characterization of signaling molecules discussed in this review provide potentially novel _targets for drug development to treat AML patients in the future.

Research Highlights.

Standard treatment for AML is associated with significant toxicities and late effects.

Characterization of signaling pathways aberrantly regulated in AML cells could lead to identification of novel _targets for therapy.

CREB is a transcription factor that is overexpressed in AML cells and is required for proliferation and survival, suggesting that inhibiting this protein is a novel approach to treat AML.

Triad1 is an E3 ubiquitin ligase and leukemia suppressor that is decreased in AML with MLL1 gene rearrangements, MYST4/CREBB gene translocations, and FLT3 internal tandem duplications.

BH3-mimetics in combination with chemotherapy overcomes resistance in AML cells.

Downstream effectors of MAPK pathways, Mnk kinases, may be attractive _targets for treatment of AML.

Stat3 activation is common in newly diagnosed AML and small molecules to inhibit STAT3 induce apoptosis in AML cells.

Acknowledgements

K.M.S. is supported by the NIH (R01 HL75826, R01 GM087305), Leukemia and Lymphoma Society of America Screen to Lead Program, Hyundai Hope on Wheels, Stanford SPARK/Child Health Research Institute, S.G. is supported by the NIH (CA142509, CA167708) and Leukemia and Lymphoma Society of America (#6472-15). L.C.P. is supported by grants from the NIH (R01 CA77816, CA155566, CA161196 and CA121192) and Department of Veterans Affairs (I01CX000916). J.C. is supported by the Samuel Waxman Cancer Research Foundation and NIH (R01 CA101774). E.A.E. is supported by NIH (R01 CA174205, DK098812) and VA Merit Review.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Gojo I, Karp JE. New Strategies in Acute Myelogenous Leukemia: Leukemogenesis and Personalized Medicine. Clin Cancer Res. 2014 doi: 10.1158/1078-0432.CCR-14-0900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Estey EH. Acute myeloid leukemia: 2014 Update on risk-stratification and management. Am J Hematol. 2014;89(11):1063–81. doi: 10.1002/ajh.23834. [DOI] [PubMed] [Google Scholar]
  • 3.Perry AM, Attar EC. New Insights in AML Biology From Genomic Analysis. Semin Hematol. 2014;51(4):282–97. doi: 10.1053/j.seminhematol.2014.08.005. [DOI] [PubMed] [Google Scholar]
  • 4.Shaywitz AJ, Greenberg ME. CREB: a stimulus-induced transcription factor activated by a diverse array of extracellular signals. Annu Rev Biochem. 1999;68:821–61. doi: 10.1146/annurev.biochem.68.1.821. [DOI] [PubMed] [Google Scholar]
  • 5.Shankar DB, Cheng JC, Kinjo K, Federman N, Moore TB, Gill A, Rao NP, Landaw EM, Sakamoto KM. The role of CREB as a proto-oncogene in hematopoiesis and in acute myeloid leukemia. Cancer Cell. 2005;7(4):351–62. doi: 10.1016/j.ccr.2005.02.018. [DOI] [PubMed] [Google Scholar]
  • 6.Shankar DB, Cheng JC, Sakamoto KM. Role of cyclic AMP response element binding protein in human leukemias. Cancer. 2005;104(9):1819–24. doi: 10.1002/cncr.21401. [DOI] [PubMed] [Google Scholar]
  • 7.Ma H, Groth RD, Cohen SM, Emery JF, Li B, Hoedt E, Zhang G, Neubert TA, Tsien RW. gammaCaMKII Shuttles Ca(2+)/CaM to the Nucleus to Trigger CREB Phosphorylation and Gene Expression. Cell. 2014;159(2):281–94. doi: 10.1016/j.cell.2014.09.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cho EC, Mitton B, Sakamoto KM. CREB and leukemogenesis. Crit Rev Oncog. 2011;16(1-2):37–46. doi: 10.1615/critrevoncog.v16.i1-2.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Esparza SD, Chang J, Shankar DB, Zhang B, Nelson SF, Sakamoto KM. CREB regulates Meis1 expression in normal and malignant hematopoietic cells. Leukemia. 2008;22(3):665–7. doi: 10.1038/sj.leu.2404933. [DOI] [PubMed] [Google Scholar]
  • 10.Mora-Garcia P, Cheng J, Crans-Vargas HN, Countouriotis A, Shankar D, Sakamoto KM. Transcriptional regulators and myelopoiesis: the role of serum response factor and CREB as _targets of cytokine signaling. Stem Cells. 2003;21(2):123–30. doi: 10.1634/stemcells.21-2-123. [DOI] [PubMed] [Google Scholar]
  • 11.Crans-Vargas HN, Landaw EM, Bhatia S, Sandusky G, Moore TB, Sakamoto KM. Expression of cyclic adenosine monophosphate response-element binding protein in acute leukemia. Blood. 2002;99(7):2617–9. doi: 10.1182/blood.v99.7.2617. [DOI] [PubMed] [Google Scholar]
  • 12.Sandoval S, Kraus C, Cho EC, Cho M, Bies J, Manara E, Accordi B, Landaw EM, Wolff L, Pigazzi M, et al. Sox4 cooperates with CREB in myeloid transformation. Blood. 2012;120(1):155–65. doi: 10.1182/blood-2011-05-357418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pigazzi M, Manara E, Bresolin S, Tregnago C, Beghin A, Baron E, Giarin E, Cho EC, Masetti R, Rao DS, et al. MicroRNA-34b promoter hypermethylation induces CREB overexpression and contributes to myeloid transformation. Haematologica. 2013;98(4):602–10. doi: 10.3324/haematol.2012.070664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Pigazzi M, Manara E, Baron E, Basso G. miR-34b _targets cyclic AMP-responsive element binding protein in acute myeloid leukemia. Cancer Res. 2009;69(6):2471–8. doi: 10.1158/0008-5472.CAN-08-3404. [DOI] [PubMed] [Google Scholar]
  • 15.Cheng JC, Esparza S, Sandoval S, Shankar D, Fu C, Sakamoto KM. Potential role of CREB as a prognostic marker in acute myeloid leukemia. Future Oncol. 2007;3(4):475–80. doi: 10.2217/14796694.3.4.475. [DOI] [PubMed] [Google Scholar]
  • 16.Cheng JC, Kinjo K, Judelson DR, Chang J, Wu WS, Schmid I, Shankar DB, Kasahara N, Stripecke R, Bhatia R, et al. CREB is a critical regulator of normal hematopoiesis and leukemogenesis. Blood. 2008;111(3):1182–92. doi: 10.1182/blood-2007-04-083600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.iao X, Li BX, Mitton B, Ikeda A, Sakamoto KM. _targeting CREB for cancer therapy: friend or foe. Curr Cancer Drug _targets. 2010;10(4):384–91. doi: 10.2174/156800910791208535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Marteijn JA, van Emst L, Erpelinck-Verschueren CA, Nikoloski G, Menke A, de Witte T, Löwenberg B, Jansen JH, van der Reijden BA. The E3 ubiquitin-protein ligase Triad1 inhibits clonogenic growth of primary myeloid progenitor cells. Blood. 2005;106(13):4114–23. doi: 10.1182/blood-2005-04-1450. [DOI] [PubMed] [Google Scholar]
  • 19.Marteijn JA, van der Meer LT, Smit JJ, Noordermeer SM, Wissink W, Jansen P, Swarts HG, Hibbert RG, de Witte T, Sixma TK, Jansen JH, van der Reijden BA. The ubiquitin ligase Triad1 inhibits myelopoiesis through UbcH7 and Ubc13 interacting domains. Leukemia. 2009;23(8):1480–9. doi: 10.1038/leu.2009.57. [DOI] [PubMed] [Google Scholar]
  • 20.Pietschmann K, Buchwald M, Müller S, Knauer SK, Kögl M, Heinzel T, Krämer OH. Differential regulation of PML-RARα stability by the ubiquitin ligases SIAH1/SIAH2 and TRIAD1. Int J Biochem Cell Biol. 2012;44(1):132–8. doi: 10.1016/j.biocel.2011.10.008. [DOI] [PubMed] [Google Scholar]
  • 21.Hassink G, Slotman J, Oorschot V, Van Der Reijden BA, Monteferrario D, Noordermeer SM, Van Kerkhof P, Klumperman J, Strous GJ. Identification of the ubiquitin ligase Triad1 as a regulator of endosomal transport. Biol Open. 2012;1(6):607–14. doi: 10.1242/bio.2012778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jung JH, Lee SM, Bae S, Lee SJ, Park IC, Jin YW, Lee JH, An S. Triad 1 induces apoptosis by p53 activation. FEBS Lett. 2010;584:1565–70. doi: 10.1016/j.febslet.2010.03.011. [DOI] [PubMed] [Google Scholar]
  • 23.Kelsall IR, Duda DM, Olszewski JL, Hofmann K, Knebel A, Langevin F, Wood N, Wightman M, Schulman BA, Alpi AF. TRIAD1 and HHARI bind to and are activated by distinct neddylated Cullin-RING ligase complexes. EMBO J. 2013;32(21):2848–60. doi: 10.1038/emboj.2013.209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ding VM, Ling L, Natarajan S, Yap MG, Cool SM, Choo AB. FGF-2 modulates Wnt signaling in undifferentiated hESC and iPS cells through activated PI3-K/GSK3B signaling. J Cell Physiol. 2010;225(2):417–28. doi: 10.1002/jcp.22214. [DOI] [PubMed] [Google Scholar]
  • 25.Lin AE, Ebert G, Ow Y, Preston SP, Toe JG, Cooney JP, Scott HW, Sasaki M, Saibil SD, Dissanayake D, Kim RH, Wakeham A, You-Ten A, Shahinian A, Duncan G, Silvester J, Ohashi PS, Mak TW, Pellegrini M. ARIH2 is essential for embryogenesis, and its hematopoietic deficiency causes lethal activation of the immune system. Nat Immunol. 2013;14(1):27–33. doi: 10.1038/ni.2478. [DOI] [PubMed] [Google Scholar]
  • 26.Johansson B, Billström R, Kristoffersson U, Akerman M, Garwicz S, Ahlgren T, Malm C, Mitelman F. Deletion of chromosome arm 3p in hematologic malignancies. Leukemia. 1997;11(8):1207–13. doi: 10.1038/sj.leu.2400718. [DOI] [PubMed] [Google Scholar]
  • 27.Shi G, Weh HJ, Martensen S, Seeger D, Hossfeld DK. 3p21 is a recurrent treatment-related breakpoint in myelodysplastic syndrome and acute myeloid leukemia. Cytogenet Cell Genet. 1996;74(94):295–9. doi: 10.1159/000134439. [DOI] [PubMed] [Google Scholar]
  • 28.Zhu X, He F, Zeng H, Ling S, Chen A, Wang Y, Yan X, Wei W, Pang Y, Cheng H, Hua C, Zhang Y, Yang X, Lu X, Cao L, Hao L, Dong L, Zou W, Wu J, Li X, Zheng S, Yan J, Zhou J, Zhang L, Mi S, Wang X, Zhang L, Zou Y, Chen Y, Geng Z, Wang J, Zhou J, Liu X, Wang J, Yuan W, Huang G, Cheng T, Wang QF. Identification of functional cooperative mutations of SETD2 in human acute leukemia. Nat Genet. 2014;46(3):287–93. doi: 10.1038/ng.2894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Oncomine 4.4 Research Edition Compendia Biosciences. www.oncomine.org/resource/
  • 30.Drabkin HA, Parsy C, Ferguson K, Guilhot F, Lacotte L, Roy L, Zeng C, Baron A, Hunger SP, Varella-Garcia M, Gemmill R, Brizard F, Brizard A, Roche J. Quantitative HOX expression in chromosomally defined subsets of acute myelogenous leukemia. Leukemia. 2002;16(2):186–195. doi: 10.1038/sj.leu.2402354. [DOI] [PubMed] [Google Scholar]
  • 31.Armstrong SA, Staunton JE, Silverman LB, Pieters R, den Boer ML, Minden MD, Sallan SE, Lander ES, Golub TR, Korsmeyer SJ. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Genet. 2002;30(1):41–47. doi: 10.1038/ng765. [DOI] [PubMed] [Google Scholar]
  • 32.klund EA, Jalava A, Kakar R. Tyrosine phosphorylation decreases HoxA10 DNA-binding and transcriptional repression during IFNγ differentiation in myeloid cell lines. J Biol Chem. 2000;275(26):20117–26. doi: 10.1074/jbc.M907915199. [DOI] [PubMed] [Google Scholar]
  • 33.Lindsay S, Huang W, Wang H, Horvath E, Zhu C, Eklund EA. Activation of SHP2 Protein-tyrosine Phosphatase Increases HoxA10-induced Repression of the Genes Encoding gp91PHOX and p67PHOX. J Biol Chem. 2007;282(4):2237–49. doi: 10.1074/jbc.M608642200. [DOI] [PubMed] [Google Scholar]
  • 34.Bei L, Lu YF, Eklund EA. HoxA9 activates transcription of the gene encoding gp91PHOX during myeloid differentiation. J. Biol. Chem. 2005;280(13):12359–70. doi: 10.1074/jbc.M408138200. [DOI] [PubMed] [Google Scholar]
  • 35.Camos M, Esteve J, Jares P, Colomer D, Rozman M, Villamor N, Costa D, Carrio A, Nomdedue J, Montserrat E, Campo E. Gene expression profiling of acute myeloid leukemia with translocation t(8;16)(p11;q13) and MYST3-CREBBP rearrangement reveals a distinctive signature with a specific pattern of HOX gene expression. Cancer Res. 2006;66(14):6947–54. doi: 10.1158/0008-5472.CAN-05-4601. [DOI] [PubMed] [Google Scholar]
  • 36.Li Z, Luo RT, Mi S, Sun M, Chen P, Bao J, Neilly MB, Jayathilaka N, Johnson DS, Wang L, Lavau C, Zhang Y, Tseng C, Zhang X, Wang J, Yu J, Yang H, Wang SM, Rowley JD, Chen J, Thirman MJ. Consistent deregulation of gene expression between human and murine MLL rearrangement leukemias. Cancer Res. 2009;69(3):1109–16. doi: 10.1158/0008-5472.CAN-08-3381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Thorsteinsdottir U, Sauvageau G, Hough MR, Dragowska W, Lansdorp PM, Lawrence HJ, Largman C, Humphries RK. Overexpression of HoxA10 in murine hematopoietic cells perturbs both myeloid and lymphoid differentiation and leads to acute myeloid leukemia. Mol. Cell. Biol. 1997;17(1):495–505. doi: 10.1128/mcb.17.1.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bjornsson JM, Andersson E, Lundstrom P, Larsson N, Xu X, Repetowska E, Humphries KR, Karlsson S. Proliferation of primitive myeloid progenitors can be reversibly induced by HOXA10. Blood. 2001;98(12):3301–3308. doi: 10.1182/blood.v98.12.3301. [DOI] [PubMed] [Google Scholar]
  • 39.Balgobind BV, Hollink IH, Arentsen-Peters ST, Zimmermann M, Harbott J, Beverloo HB, von Bergh AR, Cloos J, Kaspers GJ, de Haas V, Zemanova Z, Stary J, Cayuela JM, Baruchel A, Creutzig U, Reinhardt D, Pieters R, Zwaan CM, van den Heuvel-Eibrink MM. Integrative analysis of type-I and type-II aberrations underscores the genetic heterogeneity of pediatric acute myeloid leukemia. Haematologica. 2011;96(10):1478–87. doi: 10.3324/haematol.2010.038976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nabinger SC, Li XJ, Ramdas B, He Y, Zhang X, Zeng L, Richine B, Bowling JD, Fukuda S, Goenka S, Liu Z, Feng GS, Yu M, Sandusky GE, Boswell HS, Zhang ZY, Kapur R, Chan RJ. The protein tyrosine phosphatase, Shp2, positively contributes to FLT3-ITD-induced hematopoietic progenitor hyperproliferation and malignant disease in vivo. Leukemia. 2013;27(2):398–408. doi: 10.1038/leu.2012.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Krivtsov AV, Figueroa ME, Sinha AU, Stubbs MC, Feng Z, Valk PJ, Delwel R, Dohner K, Bullinger L, Kung AL, et al. Cell of origin determines clinically relevant subtypes of MLL-rearranged AML. Leukemia. 2013 Apr;27(4):852–60. doi: 10.1038/leu.2012.363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Neff T, Armstrong SA. Recent progress toward epigenetic therapies: the example of mixed lineage leukemia. Blood. 2013 Jun 13;121(24):4847–53. doi: 10.1182/blood-2013-02-474833. PMCID:PMC3682337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.de BJ, Walf-Vorderwulbecke V, Williams O. In focus: MLL-rearranged leukemia. Leukemia. 2013 Jun;27(6):1224–8. doi: 10.1038/leu.2013.78. [DOI] [PubMed] [Google Scholar]
  • 44.Del PG, Bruno A, Del Principe, Venditti A, Maurillo L, Buccisano F, Stasi R, Neri B, Luciano F, Siniscalchi A, et al. Deregulation of the mitochondrial apoptotic machinery and development of molecular _targeted drugs in acute myeloid leukemia. Curr.Cancer Drug _targets. 2008 May;8(3):207–22. doi: 10.2174/156800908784293640. [DOI] [PubMed] [Google Scholar]
  • 45.Beurlet S, Omidvar N, Gorombei P, Krief P, Le PC, Setterblad N, de la Grange P, Leboeuf C, Janin A, Noguera ME, et al. BCL-2 inhibition with ABT-737 prolongs survival in an NRAS/BCL-2 mouse model of AML by _targeting primitive LSK and progenitor cells. Blood. 2013 Oct 17;122(16):2864–76. doi: 10.1182/blood-2012-07-445635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pan R, Hogdal LJ, Benito JM, Bucci D, Han L, Borthakur G, Cortes J, DeAngelo DJ, Debose L, Mu H, et al. Selective BCL-2 inhibition by ABT-199 causes on-_target cell death in acute myeloid leukemia. Cancer Discov. 2014 Mar;4(3):362–75. doi: 10.1158/2159-8290.CD-13-0609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Vo TT, Ryan J, Carrasco R, Neuberg D, Rossi DJ, Stone RM, DeAngelo DJ, Frattini MG, Letai A. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell. 2012 Oct 12;151(2):344–55. doi: 10.1016/j.cell.2012.08.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Niu X, Wang G, Wang Y, Caldwell JT, Edwards H, Xie C, Taub JW, Li C, Lin H, Ge Y. Acute myeloid leukemia cells harboring MLL fusion genes or with the acute promyelocytic leukemia phenotype are sensitive to the Bcl-2-selective inhibitor ABT-199. Leukemia. 2014 Jul;28(7):1557–60. doi: 10.1038/leu.2014.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ewings KE, Hadfield-Moorhouse K, Wiggins CM, Wickenden JA, Balmanno K, Gilley R, Degenhardt K, White E, Cook SJ. ERK1/2-dependent phosphorylation of Bim(EL) promotes its rapid dissociation from Mcl-1 and Bcl-x(L) EMBO J. 2007 May 24;26:2856–67. doi: 10.1038/sj.emboj.7601723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B, McQueen T, Bornmann W, Tsao T, Bergamo P, et al. MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia. 2012;26(4):778–87. doi: 10.1038/leu.2011.287. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 51.Martelli AM, Evangelisti C, Chiarini F, McCubrey JA. The phosphatidylinositol 3-kinase/Akt/mTOR signaling network as a therapeutic _target in acute myelogenous leukemia patients. Onco_target. 2010 Jun;1(2):89–103. doi: 10.18632/onco_target.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Maurer U, Charvet C, Wagman AS, Dejardin E, Green DR. Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol.Cell. 2006 Mar 17;21(6):749–60. doi: 10.1016/j.molcel.2006.02.009. [DOI] [PubMed] [Google Scholar]
  • 53.Rahmani M, Aust MM, Attkisson E, Williams DC, Jr., Ferreira-Gonzalez A, Grant S. Dual inhibition of Bcl-2 and Bcl-xL strikingly enhances PI3K inhibition-induced apoptosis in human myeloid leukemia cells through a. Cancer Res. 2013 Feb 15;73(4):1340–51. doi: 10.1158/0008-5472.CAN-12-1365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Forman SJ, Rowe JM. The myth of the second remission of acute leukemia in the adult. Blood. 2013;121(7):1077–82. doi: 10.1182/blood-2012-08-234492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ding L, Ley TJ, Larson DE, Miller CA, Koboldt DC, Welch JS, Ritchey JK, Young MA, Lamprecht T, McLellan MD, McMichael JF, Wallis JW, Lu C, Shen D, Harris CC, Dooling DJ, Fulton RS, Fulton LL, Chen K, Schmidt H, Kalicki-Veizer J, Magrini VJ, Cook L, McGrath SD, Vickery TL, Wendl MC, Heath S, Watson MA, Link DC, Tomasson MH, Shannon WD, Payton JE, Kulkarni S, Westervelt P, Walter MJ, Graubert TA, Mardis ER, Wilson RK, DiPersio JF. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–10. doi: 10.1038/nature10738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Parkin B, Ouillette P, Li Y, Keller J, Lam C, Roulston D, Li C, Shedden K, Malek SN. Clonal evolution and devolution after chemotherapy in adult acute myelogenous leukemia. Blood. 2013;121(2):369–77. doi: 10.1182/blood-2012-04-427039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Corces-Zimmerman MR, Hong WJ, Weissman IL, Medeiros BC, Majeti R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proc Natl Acad Sci U S A. 2014;111(7):2548–53. doi: 10.1073/pnas.1324297111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Shlush LI, Zandi S, Mitchell A, Chen WC, Brandwein JM, Gupta V, Kennedy JA, Schimmer AD, Schuh AC, Yee KW, McLeod JL, Doedens M, Medeiros JJ, Marke R, Kim HJ, Lee K, McPherson JD, Hudson TJ, HALT Pan-Leukemia Gene Panel Consortium. Brown AM, Yousif F, Trinh QM, Stein LD, Minden MD, Wang JC, Dick JE. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014;506(7488):328–33. doi: 10.1038/nature13038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Koboldt DC, Wartman LD, Lamprecht TL, Liu F, Xia J, Kandoth C, Fulton RS, McLellan MD, Dooling DJ, Wallis JW, Chen K, Harris CC, Schmidt HK, Kalicki-Veizer JM, Lu C, Zhang Q, Lin L, O'Laughlin MD, McMichael JF, Delehaunty KD, Fulton LA, Magrini VJ, McGrath SD, Demeter RT, Vickery TL, Hundal J, Cook LL, Swift GW, Reed JP, Alldredge PA, Wylie TN, Walker JR, Watson MA, Heath SE, Shannon WD, Varghese N, Nagarajan R, Payton JE, Baty JD, Kulkarni S, Klco JM, Tomasson MH, Westervelt P, Walter MJ, Graubert TA, DiPersio JF, Ding L, Mardis ER, Wilson RK. The origin and evolution of mutations in acute myeloid leukemia. Cell. 2012;150(2):264–78. doi: 10.1016/j.cell.2012.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell. 2012;149(2):274–93. doi: 10.1016/j.cell.2012.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Beauchamp EM, Platanias LC. The evolution of the TOR pathway and its role in cancer. Oncogene. 2013;32(34):3923–32. doi: 10.1038/onc.2012.567. doi: 10.1038/onc.2012.567. [DOI] [PubMed] [Google Scholar]
  • 62.Aoki Y, Matsubara Y. Ras/MAPK syndromes and childhood hemato-oncological diseases. Int J Hematol. 2013;97(1):30–6. doi: 10.1007/s12185-012-1239-y. [DOI] [PubMed] [Google Scholar]
  • 63.Pritchard AL, Hayward NK. Molecular pathways: mitogen-activated protein kinase pathway mutations and drug resistance. Clin Cancer Res. 2013;19(9):2301–9. doi: 10.1158/1078-0432.CCR-12-0383. [DOI] [PubMed] [Google Scholar]
  • 64.Geest CR, Coffer PJ. MAPK signaling pathways in the regulation of hematopoiesis. J Leukoc Biol. 2009;86(2):237–50. doi: 10.1189/jlb.0209097. [DOI] [PubMed] [Google Scholar]
  • 65.Volk A, Li J, Xin J, You D, Zhang J, Liu X, Xiao Y, Breslin P, Li Z, Wei W, Schmidt R, Li X, Zhang Z, Kuo PC, Nand S, Zhang J, Chen J, Zhang J. Co-inhibition of NF-κB and JNK is synergistic in TNF-expressing human AML. J Exp Med. 2014;211(6):1093–108. doi: 10.1084/jem.20130990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Altman JK, Glaser H, Sassano A, Joshi S, Ueda T, Watanabe-Fukunaga R, Fukunaga R, Tallman MS, Platanias LC. Negative regulatory effects of Mnk kinases in the generation of chemotherapy-induced antileukemic responses. Mol Pharmacol. 2010;78(4):778–84. doi: 10.1124/mol.110.064642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Altman JK, Szilard A, Konicek BW, Iversen PW, Kroczynska B, Glaser H, Sassano A, Vakana E, Graff JR, Platanias LC. Inhibition of Mnk kinase activity by cercosporamide and suppressive effects on acute myeloid leukemia precursors. Blood. 2013;121(18):3675–81. doi: 10.1182/blood-2013-01-477216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Altman JK, Platanias LC. Acute myeloid leukemia: potential for new therapeutic approaches _targeting mRNA translation pathways. Int J Hematol Oncol. 2013;2(3) doi: 10.2217/ijh.13.23. doi: 10.2217/ijh.13.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Altman JK, Sassano A, Kaur S, Glaser H, Kroczynska B, Redig AJ, Russo S, Barr S, Platanias LC. Dual mTORC2/mTORC1 _targeting results in potent suppressive effects on acute myeloid leukemia (AML) progenitors. Clin Cancer Res. 2011;17(13):4378–88. doi: 10.1158/1078-0432.CCR-10-2285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Zeng Z, Shi YX, Tsao T, Qiu Y, Kornblau SM, Baggerly KA, Liu W, Jessen K, Liu Y, Kantarjian H, Rommel C, Fruman DA, Andreeff M, Konopleva M. _targeting of mTORC1/2 by the mTOR kinase inhibitor PP242 induces apoptosis in AML cells under conditions mimicking the bone marrow microenvironment. Blood. 2012;120(13):2679–89. doi: 10.1182/blood-2011-11-393934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Willems L, Chapuis N, Puissant A, Maciel TT, Green AS, Jacque N, Vignon C, Park S, Guichard S, Herault O, Fricot A, Hermine O, Moura IC, Auberger P, Ifrah N, Dreyfus F, Bonnet D, Lacombe C, Mayeux P, Bouscary D, Tamburini J. The dual mTORC1 and mTORC2 inhibitor AZD8055 has anti-tumor activity in acute myeloid leukemia. Leukemia. 2012;26(6):1195–202. doi: 10.1038/leu.2011.339. [DOI] [PubMed] [Google Scholar]
  • 72.Kampa-Schittenhelm KM, Heinrich MC, Akmut F, Rasp KH, Illing B, Döhner H, Döhner K, Schittenhelm MM. Cell cycle-dependent activity of the novel dual PI3K-MTORC1/2 inhibitor NVP-BGT226 in acute leukemia. Mol Cancer. 2013;12:46. doi: 10.1186/1476-4598-12-46. doi: 10.1186/1476-4598-12-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chapuis N, Tamburini J, Green AS, Vignon C, Bardet V, Neyret A, Pannetier M, Willems L, Park S, Macone A, Maira SM, Ifrah N, Dreyfus F, Herault O, Lacombe C, Mayeux P, Bouscary D. Dual inhibition of PI3K and mTORC1/2 signaling by NVP-BEZ235 as a new therapeutic strategy for acute myeloid leukemia. Clin Cancer Res. 2010;16(22):5424–35. doi: 10.1158/1078-0432.CCR-10-1102. doi: 10.1158/1078-0432.CCR-10-1102. [DOI] [PubMed] [Google Scholar]
  • 74.McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Franklin RA, Montalto G, Cervello M, Libra M, Candido S, Malaponte G, Mazzarino MC, Fagone P, Nicoletti F, Bäsecke J, Mijatovic S, Maksimovic-Ivanic D, Milella M, Tafuri A, Chiarini F, Evangelisti C, Cocco L, Martelli AM. Mutations and deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascades, which alter therapy response. Onco_target. 2012;3(9):954–87. doi: 10.18632/onco_target.652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stead RL, Proud CG. Rapamycin enhances eIF4E phosphorylation by activating MAP kinase-interacting kinase 2a (Mnk2a) FEBS Lett. 2013;587(16):2623–8. doi: 10.1016/j.febslet.2013.06.045. [DOI] [PubMed] [Google Scholar]
  • 76.Greger JG, Eastman SD, Zhang V, Bleam MR, Hughes AM, Smitheman KN, Dickerson SH, Laquerre SG, Liu L, Gilmer TM. Combinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutations. Mol Cancer Ther. 2012;11(4):909–20. doi: 10.1158/1535-7163.MCT-11-0989. [DOI] [PubMed] [Google Scholar]
  • 77.Brundage ME, Tandon P, Eaves DW, Williams JP, Miller SJ, Hennigan RH, Jegga A, Cripe TP, Ratner N. MAF mediates crosstalk between Ras-MAPK and mTOR signaling in NF1. Oncogene. 2014 doi: 10.1038/onc.2013.506. doi: 10.1038/onc.2013.506. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zhang W, Ruvolo VR, Gao C, Zhou L, Bornmann W, Tsao T, Schober WD, Smith P, Guichard S, Konopleva M, Andreeff M. Evaluation of apoptosis induction by concomitant inhibition of MEK, mTOR, and Bcl-2 in human acute myelogenous leukemia cells. Mol Cancer Ther. 2014;13(7):1848–59. doi: 10.1158/1535-7163.MCT-13-0576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Levine RL, Pardanani A, Tefferi A, Gilliland DG. Role of JAK2 in the pathogenesis and therapy of myeloproliferative disorders. Nat Rev Cancer. 2007;7(9):673–83. doi: 10.1038/nrc2210. [DOI] [PubMed] [Google Scholar]
  • 80.Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NC, Berg T, Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant'Antonio E, Ferretti V, Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schönegger A, Bock C, Malcovati L, Pascutto C, Superti-Furga G, Cazzola M, Kralovics R. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379–90. doi: 10.1056/NEJMoa1311347. [DOI] [PubMed] [Google Scholar]
  • 81.Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA, Estrov Z, Fridman JS, Bradley EC, Erickson-Viitanen S, Vaddi K, Levy R, Tefferi A. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010;363(12):1117–27. doi: 10.1056/NEJMoa1002028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, Winton EF, Harvey JH, Jr, Arcasoy MO, Hexner EO, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Sun W, Sandor V, Kantarjian HM. Efficacy, safety and survival with ruxolitinib in patients with myelofibrosis: results of a median 2-year follow-up of COMFORT-I. Haematologica. 2013;98(12):1865–71. doi: 10.3324/haematol.2013.092155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tefferi A. JAK inhibitors for myeloproliferative neoplasms: clarifying facts from myths. Blood. 2012;119(12):2721–30. doi: 10.1182/blood-2011-11-395228. [DOI] [PubMed] [Google Scholar]
  • 84.Koppikar P, Bhagwat N, Kilpivaara O, Manshouri T, Adli M, Hricik T, Liu F, Saunders LM, Mullally A, Abdel-Wahab O, Leung L, Weinstein A, Marubayashi S, Goel A, Gönen M, Estrov Z, Ebert BL, Chiosis G, Nimer SD, Bernstein BE, Verstovsek S, Levine RL. Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2 inhibitor therapy. Nature. 2012;489(7414):155–9. doi: 10.1038/nature11303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kalota A, Jeschke GR, Carroll M, Hexner EO. Intrinsic resistance to JAK2 inhibition in myelofibrosis. Clin Cancer Res. 2013;19(7):1729–39. doi: 10.1158/1078-0432.CCR-12-1907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Vicente C, Vázquez I, Marcotegui N, Conchillo A, Carranza C, Rivell G, Bandrés E, Cristobal I, Lahortiga I, Calasanz MJ, Odero MD. JAK2-V617F activating mutation in acute myeloid leukemia: prognostic impact and association with other molecular markers. Leukemia. 2007;21(11):2386–90. doi: 10.1038/sj.leu.2404812. [DOI] [PubMed] [Google Scholar]
  • 87.Steensma DP, McClure RF, Karp JE, Tefferi A, Lasho TL, Powell HL, DeWald GW, Kaufmann SH. JAK2 V617F is a rare finding in de novo acute myeloid leukemia, but STAT3 activation is common and remains unexplained. Leukemia. 2006;20(6):971–8. doi: 10.1038/sj.leu.2404206. [DOI] [PubMed] [Google Scholar]
  • 88.Lo MC, Peterson LF, Yan M, Cong X, Hickman JH, Dekelver RC, Niewerth D, Zhang DE. JAK inhibitors suppress t(8;21) fusion protein-induced leukemia. Leukemia. 2013;27(12):2272–9. doi: 10.1038/leu.2013.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Mizuki M, Fenski R, Halfter H, Matsumura I, Schmidt R, Müller C, Grüning W, Kratz-Albers K, Serve S, Steur C, Büchner T, Kienast J, Kanakura Y, Berdel WE, Serve H. Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways. Blood. 2000;96(12):3907–14. [PubMed] [Google Scholar]
  • 90.Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C, Faderl S, Ferrajoli A, Borthakur G, George S, Scherle PA, Newton RC, Kantarjian HM, Ravandi F. Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood. 2012;119(20):4614–8. doi: 10.1182/blood-2011-12-400051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Furqan M, Akinleye A, Mukhi N, Mittal V, Chen Y, Liu D. STAT inhibitors for cancer therapy. J Hematol Oncol. 2013;6:90. doi: 10.1186/1756-8722-6-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Redell MS, Ruiz MJ, Alonzo TA, Gerbing RB, Tweardy DJ. Stat3 signaling in acute myeloid leukemia: ligand-dependent and -independent activation and induction of apoptosis by a novel small-molecule Stat3 inhibitor. Blood. 2011;117(21):5701–9. doi: 10.1182/blood-2010-04-280123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hayakawa F1, Sugimoto K, Harada Y, Hashimoto N, Ohi N, Kurahashi S, Naoe T. A novel STAT inhibitor, OPB-31121, has a significant antitumor effect on leukemia with STAT-addictive oncokinases. Blood Cancer J. 2013 Nov 29;3:e166. doi: 10.1038/bcj.2013.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lee HJ, Zhuang G, Cao Y, Du P, Kim HJ, Settleman J. Drug resistance via feedback activation of Stat3 in oncogene-addicted cancer cells. Cancer Cell. 2014;26(2):207–21. doi: 10.1016/j.ccr.2014.05.019. [DOI] [PubMed] [Google Scholar]

RESOURCES

  NODES
Association 1
INTERN 3
Note 3
twitter 2