The dysregulation of protein synthesis evident in the transformed phenotype has opened up a burgeoning field of research in cancer biology. Translation initiation has recently been shown to be a common downstream target of signal transduction pathways deregulated in cancer and initiated by mutated/overexpressed oncogenes and tumor suppressors. The overexpression and/or activation of proteins involved in translation initiation such as eIF4E, mTOR, and eIF4G have been shown to induce a malignant phenotype. Therefore, understanding the mechanisms that control protein synthesis is emerging as an exciting new research area with significant potential for developing innovative therapies. This review highlights molecules that are activated or dysregulated in hematologic malignancies, and promotes the transformed phenotype through the deregulation of protein synthesis. Targeting these proteins with small molecule inhibitors may constitute a novel therapeutic approach in the treatment of cancer.

Eukaryotic cells have various mechanisms and levels by which gene expression can be regulated including: transcription, export of mRNA messages, mRNA stability, and posttranslational modifications. Protein synthesis is essential for cell viability, and controlling mRNA translation is a critical step in regulation of gene expression. Translation can be divided between 3 stages: initiation, elongation, and termination. Translational control is principally exerted by regulating the formation of the cap-dependent translation initiation complex. Translation initiation comprises a mechanism in which the eIF4F ternary complex (eIF4E, eIF4A, and eIF4G) recruits a 43S preinitiation complex containing a Met-tRNAi and multiple initiation factors (eIFs 1, 1A, 2, 3, and 5) to the 5′ methyl-7-GTP cap complex on mRNA (reviewed in Sonenberg and Hinnebusch).1  Once the preinitiation complex scans the 5′ untranslated region of the mRNA and reaches the AUG start codon, the large (60S) ribosomal subunit joins the small (40S) ribosomal subunit and begins to synthesize the protein.1 

Deregulated protein synthesis plays an important role in human cancer and deregulated translational control has been recognized as an integral part of the malignant state.2,3  In the past several years it has become clear that the efficiency of expression of key proteins involved in cell-growth regulation, proliferation, and apoptosis may be controlled at the translational level by changes in the activity of components of the protein synthesis machinery.4,5  Various classes of mRNAs differ considerably in their translational efficiency. Typically, mRNAs coding for proteins positively involved in regulating cell growth and survival have a high degree of secondary structure in the 5′ untranslated region (UTR). The translation of such messages is particularly sensitive to the activity of the cap-dependent translation-initiation machinery.6  In view of the fact that translation factors are closely regulated by conditions that affect cell growth, it is not surprising that, experimentally, aberrant expression of some of these factors has been shown to induce malignant transformation of cells.

Translation initiation has recently been shown to be a common downstream target of signal transduction pathways deregulated in cancer and initiated by mutated/overexpressed oncogenes and tumor suppressors.7  Several previous publications indicate that aberrant control of protein synthesis contributes to lymphomagenesis3,8  opening up possibilities for innovative therapeutics, that is, targeting the translational machinery. Below, we present an overview of potentially targetable translational machinery components and regulatory signaling pathways that represent a novel approach for the treatment of hematologic malignancies.

A major regulatory step in control of protein synthesis is translation initiation. Translation initiation is modulated by the association of a ternary complex of proteins, eukaryotic translation initiation factor F (eIF4F), composed of eIF4G, eIF4A, and eIF4E.9  The cellular levels of eIF4E molecules are 10- to 30-fold lower than other known initiation factors10,11  and its association with the eIF4F complex is therefore the rate-limiting step in translation initiation (reviewed by Clemens,12  De Benedetti and Graff13 ); however, the stoichiometry of eIF4F components is still debated by some investigators.14  It was previously hypothesized that an increase in the rate of translation would have an impact on the spectrum of mRNAs synthesized.15  Subsequently, it was shown that overexpression of eIF4E could increase the translation of mRNAs with long highly structured 5′ UTR9  such as chloramphenical acetyl transferase and ornithine decarboxylase.16  Early studies established that eIF4E overexpression resulted in the transformation of immortalized cell lines as exemplified by increased proliferation, anchorage independent growth, and invasiveness.17,18  Recently, the overexpression of eIF4E has been observed in primary human malignancies such as colon,19  breast,20,21  non-Hodgkin lymphoma (NHL),22  acute myelogenous leukemia (AML), and chronic myelogenous leukemia (CML).23  These aggregate data prompted several laboratories to investigate the role of eIF4E in neoplastic transformation as well as suggested the feasibility of targeting this molecule as a novel therapeutic approach. Several investigators have examined the impact of overexpressing eIF4E in transgenic mouse models, which can increase the incidence of multiple malignancies including lymphomas.3,24 

The disruption of translation initiation through the modulation of either eIF4E protein levels or formation of the eIF4F ternary complex has been examined using RNA interference.25-28  Graff et al demonstrated that intravenous administration of antisense oligonucleotides in a mouse xenograft model could successfully silence the expression of eIF4E in vivo. This decrease in eIF4E levels led to an inhibition of tumor growth and reduced cell viability. While the decrease in eIF4E levels was not restricted to only the tumor cells (80% reduction in normal tissues), the clinical impact on normal tissues was minimal as there were no significant changes in body weight, liver weight, or hepatic enzymes. These data demonstrated that modulating eIF4E protein levels was a useful approach to disrupt tumor growth in vivo without significant off-target effects in normal tissues. The function of eIF4E may also be interrupted by small molecule inhibitors that mimic the 5′ methyl-7-GTP moiety found within the capped structure of mRNA.29,30  The small molecule Ribavirin has been shown to suppress eIF4E-induced transformation through a mechanism in which the inhibitor competes with the endogenous 5′ methyl-7-GTP cap structure for eIF4E. Through this competitive inhibition, Kentsis et al were able to demonstrate a decrease in the translation of oncogenic messages such as ornithine decarboxylase (ODC) as shown by the absence of its mRNA in the polysomal fraction of ribosomes.29  eIF4E has also been shown to regulate translation of mRNA by promoting the transport of oncogenic messages.16  The use of Ribavirin to interfere with eIF4E association to the 5′ methyl-7-GTP cap structure results in the specific disruption of eIF4E-directed transport of messages to the cytoplasm.29,30  The Ribavirin-mediated inhibition of eIF4E has also been successfully demonstrated in a xenograft mouse model with a 6-fold reduction in tumor volume compared with controls further highlighting the critical role of eIF4E in maintaining the malignant phenotype.

In addition, small molecule inhibitors have been investigated targeting the protein-protein domain that governs binding between eIF4E and eIF4G. These inhibitors work through molecular mimicry of the domain on either eIF4G or 4E-BP1 that promotes binding of eIF4E. These 4E-BP1 functional mimetic inhibitors have been shown to be effective in promoting apoptosis and impairing cell-cycle progression in both lymphoma and leukemia.31,32  Using a high-throughput approach, Moerke et al were the first to describe an eIF4F-specific inhibitor (4EGI-1) that can disrupt the eIF4F complex and inhibit cap-dependent translation. Importantly, treatment of T-cell acute lymphoblastic leukemia (Jurkat T-ALL) and lung cancer (A549) cell lines with 4EGI-1 was able to induce apoptosis as well as inhibit proliferation.31  The authors also demonstrated that 4EGI-1 preferentially inhibits the growth of cells transformed with the Philadelphia chromosome compared with the nontransformed isogenic Ba/F3 cell line. The use of this inhibitor has also been examined in AML.32  While the macrolide rapamycin has been an effective tool to inhibit the activity of eIF4E and cap-dependent translation in certain cell types,33,34  Tamburini et al demonstrated that AML cells are resistant to inhibition of the mammalian target of rapamycin (mTOR) by treatment with the rapamycin derivative RAD001 or Raptor small inhibitory RNA (siRNA). Nevertheless, treatment of cells with 4EGI-1 significantly reduced the clonogenic growth and induced apoptosis of primary AML cells. Furthermore, 4EGI-1 also proved to be effective in disrupting eIF4F complex formation in primary AML samples lending additional credence to targeting of the eIF4F complex.32  Alternatively, an innovative peptidomimetic approach targeting the association between eIF4G and eIF4E was used in an ovarian cancer mouse model. The authors demonstrated that the peptide could be specifically targeted to the ovary by fusing the peptide to gonadotropin-releasing hormone (GnRH). Interference of eIF4F complex formation with a targeted peptide strongly inhibited proliferation and induced apoptosis in tumor cells compared with both saline and GnRH treated controls, with no toxicity observed in normal tissues.35  Interfering with eIF4F activity and translation initiation has also been shown to restore the chemosensitivity of tumors in preclinical lymphoma models.36,37 

The small molecule inhibitor Ribavirin, is currently used in the therapeutic regimen for viral infections such as Lassa fever virus and hepatitis C virus (HCV).38  Treatment of patients with HCV related non-Hodgkin lymphoma39,40  using Ribavirin resulted in a dramatic clinical response in some patients, these responses often correlated with a reduction in HCV viral load. The first clinical trial evaluating the targeting of eIF4E as a monotherapy using Ribavirin was recently conducted in 11 patients with acute myeloid leukemia. The results were striking with complete remission (CR), partial remission (PR), and other hematologic improvements obtained in patients.30  These exciting recent clinical data, along with earlier animal models, together suggest that Ribavirin shows great potential to be an effective new therapeutic agent in the treatment of hematologic malignancies.

The evolutionarily conserved target of rapamycin (TOR) is a large serine/threonine protein kinase that belongs to a family of kinases known as the phosphatidylinositol kinase-related kinase family.41  TOR was originally identified in the yeast species, Saccharomyces cerevisiae, because of its sensitivity to the compound rapamycin (sirolimus), a macrocyclic lactone with immunosuppressive properties initially isolated from the bacterial strain Streptomyces hygroscopicus.42  The mammalian homologue, mTOR, is a key component of 2 functionally and structurally distinct multiprotein complexes, mTOR complex 1 (mTORC1) and mTORC2. While mTORC1 is responsive to the inhibitory effects of rapamycin, mTORC2 appears to be insensitive in a tissue-specific manner.43  Interestingly, prolonged treatment with rapamycin derivatives reduces mTORC2 signaling in AML cells resulting in a decrease in AKT signaling.44  Compelling evidence demonstrates that mTORC1 is a prime signaling node in many pathways that regulate cell growth and metabolism by transducing signals associated with environmental, nutritional, growth factor, and other bioenergetic cues.

mTORC1 is a multimeric protein complex comprising mTOR together with the associated proteins raptor (regulatory associated protein of mTOR), mLST8,45  and PRAS40.46  Upon activation, the catalytic subunit of mTOR directly controls protein synthesis by phosphorylating key regulators of the translational machinery such as the eukaryotic initiation factor 4E-binding protein-1 (4EBP1), an inhibitor of cap-dependent translation, and the ribosomal p70 S6 kinase (S6K), a critical factor that may participate in ribosomal biogenesis through activation of ribosomal protein S6 (RPS6).47  In this regard, S6K is phosphorylated by mTOR at threonine 389, which in concert with another phosphorylating event carried out by the serine/threonine kinase phosphoinositide-dependent kinase-1 (PDK1) promotes full activation of S6K through phosphorylation of threonine 229. Once activated, S6K phosphorylates the RPS6 to enhance ribosomal biogenesis and overall protein synthesis (see “RPS6”). While mTOR phosphorylation of p70 S6 kinase results in the activation of the kinase, mTOR controls cap-dependent translation by phosphorylating 4EBP1, which is a suppressor of eIF4E.43  Because the strong binding of a hypophosphorylated 4EBP1 prevents the assembly of the translation initiation complex to eIF4E, mTOR-mediated phosphorylation of 4EBP1 facilitates the release of eIF4E from its inactive state to form, together with eIF4G and eIF4A, the eIF4F complex. Therefore, by inhibiting mTOR activity, multiple components of protein translation are suppressed, resulting in cell-growth retardation and eventually cell death.48 

Cellular processes commonly deregulated in human cancer, such as cell proliferation, cell-cycle progression, survival, and motility are closely related to mTORC1 function.49  In addition, several upstream (PTEN, AKT, TSC1/2) and downstream (eIF4E) mediators within the mTORC1 signaling axis are often mutated or activated in several human malignancies including cancers of the hematopoietic system.50,51  These findings have triggered an intense search for novel therapeutic strategies to target the mTOR signaling pathway in conditions such as leukemia, lymphomas, and multiple myeloma. Indeed, this is evident by the increasing number of preclinical studies and the approximately 20 clinical trials that are either active or about to be initiated on the use of sirolimus, rapamycin analogs “rapalogs,” or second-generation derivatives, such as temsirolimus, everolimus, and deferolimus alone or in combination with other agents on hematologic malignancies, as described on www.ClinicalTrials.gov at the time this review was being written.

Early studies using mTOR inhibitors in hematologic conditions provided key positive findings to launch further investigations in the field. Brown et al initially reported on the proapoptotic activity of sirolimus on B-precursor acute lymphoblastic leukemia cell lines. They were also able to demonstrate that by using sirolimus as monotherapy, survival of Eμ-RET transgenic mice, an animal model of leukemia/lymphoma, could be extended compared with littermate controls.52  Later, the same group investigated whether the use of a rapamycin analog, temsirolimus, was effective in preventing the in vitro growth of primary adult acute lymphoblastic leukemia cells. Compared with untreated cells, temsirolimus-treated cells showed a marked decrease in cell proliferation and an increase in apoptosis. They further validated the in vitro data in a nonobese diabetic severe combined immunodeficiency (NOD/SCID) xenograft model by establishing tumors with the same patient samples. A significant reduction in peripheral blood blasts and splenomegaly was observed in the temsirolimus-treated animals compared with the control group.53  The effectiveness of rapamycin in chronic lymphocytic leukemia (CLL) has been investigated in an Eμ-TCL1 transgenic mouse model, which gives rise to a CD5+ B-cell lymphoproliferative disorder similar to human CLL. The authors demonstrated that rapamycin could significantly extend the life of rapamycin-treated mice compared with controls.54  Xu et al performed the definitive work that provided the rationale for combining rapamycin with chemotherapeutic agents. The authors demonstrated that mTOR was responsible for cell survival after primary AML cells were treated with etoposide and that addition of rapamycin to the treatment could dramatically increase the cytotoxic effects of etoposide.55  Overall, these studies were critical to establishing the promising role mTOR inhibitors may exert against hematologic cancers.

In this regard, the results of a phase 2 clinical trial on the use of deferolimus as a single agent in patients with relapsed or refractory hematologic malignancies such as ALL, CML, CLL, and mantle cell lymphoma (MCL) have recently been reported.56  Of the 52 evaluable patients with various types of leukemia/lymphoma who were previously heavily pretreated, 40% responded positively to deferolimus treatment through hematologic improvement or stable disease. The effects of deferolimus on the translational machinery controlled by mTOR activity was demonstrated by the presence of decreased levels in the phosphorylated status of 4EBP1 in tumor cells from a subset of 9 of 11 patients with acute myelogenous leukemia, confirming the inhibition of mTOR. Similar positive outcomes and biochemical end points relative to the mTOR pathway were obtained in another phase 1/2 clinical study evaluating responses to everolimus, another second-generation mTOR inhibitor, in patients with relapsed or recurrent hematologic cancers. Overall, everolimus was well tolerated at a daily dose of 10 mg. As expected, the phosphorylation status of downstream targets of mTOR, 4EBP1, and S6K was inhibited in 6 of 9 patient samples. The authors concluded that everolimus in combination with other agents targeting components of the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway are warranted.57  Rapamycin has also been shown to be effective in imatinib-resistant chronic myeloid leukemia. A major hematologic response was reported in 2 of 7 patients. The investigators also demonstrated in Ba/F3 cells, which expressed multiple forms of imatinib-resistant BCR-ABL kinases, that rapamycin could inhibit the cell-cycle progression.58 

As a matter of fact, more recent preclinical and clinical studies have been addressing the use of combination therapies in hematologic malignancies by adding other targeted agents or cytotoxic chemotherapeutics to protocols that include mTOR inhibitors.50,59-61  Recently, multiple groups have developed and described mTOR kinase-specific small molecule inhibitors Torin1, PP242, and PP3034,62  (Figure 1). Recently, the Gartenhaus laboratory has established a paradigm in which chronic ethanol exposure inhibits mTOR signaling in lymphocytes with a significant repression of cap-dependent translation and reduction in the tumorigenic capacity of NHL in a human lymphoma xenograft system.63  The observed ethanol-mediated repression of mTOR signaling coupled with a decrease in lymphoma growth presented underscore the critical role of mTOR signaling and translation in lymphoma.63 

Figure 1

Targeting the translational machinery. Multiple drugs have been developed to target molecules involved in the regulation of protein translation. Rapamycin and “rapalogs” (temsirolimus, everolimus, and deferolimus) restrict the recruitment of the FKBP12 protein to the mTOR complex inhibiting mTORC1 signaling. Other small molecule inhibitors have been developed to target the mTOR kinase domain (Torin1, PP242, and PP30), which may inhibit both mTORC1 and mTORC2 signaling pathways. CGP57380 has been developed as an ATP competitive inhibitor of the MNK kinases, which may prevent the release of eIF4E from the 5′ methyl-7-GTP cap structure preventing a subsequent round of translation on the same mRNA. Other drugs have been found to block the recruitment of eIF4E to the eIF4F ternary complex including 4EGI-1 and Ribavirin to inhibit both translation initiation and eIF4E-mediated transport of mRNA. The PUA dominant negative has also been shown to block the interaction of MCT-1 with the translation initiation complex (TIC).

Figure 1

Targeting the translational machinery. Multiple drugs have been developed to target molecules involved in the regulation of protein translation. Rapamycin and “rapalogs” (temsirolimus, everolimus, and deferolimus) restrict the recruitment of the FKBP12 protein to the mTOR complex inhibiting mTORC1 signaling. Other small molecule inhibitors have been developed to target the mTOR kinase domain (Torin1, PP242, and PP30), which may inhibit both mTORC1 and mTORC2 signaling pathways. CGP57380 has been developed as an ATP competitive inhibitor of the MNK kinases, which may prevent the release of eIF4E from the 5′ methyl-7-GTP cap structure preventing a subsequent round of translation on the same mRNA. Other drugs have been found to block the recruitment of eIF4E to the eIF4F ternary complex including 4EGI-1 and Ribavirin to inhibit both translation initiation and eIF4E-mediated transport of mRNA. The PUA dominant negative has also been shown to block the interaction of MCT-1 with the translation initiation complex (TIC).

Close modal

The role of rapamycin and its derivatives and their mechanism of action are still under investigation. A recent report demonstrated that rapamycin treatment increased the cap-dependent translation of cervical carcinoma and embryonic kidney cells lines33  illustrating how rapamycin can have cell type–specific effects. Rapamycin is also reportedly effective in inhibiting mTOR signaling in either 4E-BP1 or p70 S6 kinase 1 null cells. However, another publication demonstrated that RPS6 phosphorylation was still present in MEF cells (p70 S6 kinase 1−/−) and was likely due to a compensatory signal from p70 S6 kinase 2.47  A possible explanation may be that related family member proteins may be differentially inhibited by rapamycin treatments. Certain cell types have been observed to be resistant to rapamycin-mediated mTOR inhibition; the mechanism(s) of resistance are still to be elucidated (reviewed in Kurmasheva et al).64  The earliest resistance mechanism observed was mutations in the FKBP12 gene that prevented the formation of FKBP12-rapamycin complex formation and rendered cells rapamycin resistant.65  Decreased AKT activity has also been shown to induce rapamycin resistance. This may be due to continued cap-independent protein synthesis of c-MYC and cyclin D1 through internal ribosomal entry sites (IRES) that are negatively regulated by AKT signaling and that this ongoing translation permits cell-cycle progression.66  New strategies are being developed to overcome resistance to small molecule inhibitors including the targeting of multiple proteins within the same pathway (reviewed in Weinstein and Joe).67 

The further development and understanding of novel molecules specifically inhibiting mTOR are likely to be even more effective at suppressing tumor growth than rapamycin and rapamycin derivatives. It is anticipated that results obtained from ongoing preclinical and clinical trials will provide valuable information on how to optimize the incorporation of mTOR inhibitors into therapeutic regimens.

As discussed, mTOR plays a major role governing the rate and efficiency of translation by signaling through the p70 S6 kinase (S6K) family members. S6K is generally implicated in ribosomal biogenesis and activity by phosphorylating RPS6, which is thought to be the rate-limiting step in the synthesis of new ribosomal RNA.43  However, the role(s) of p70 S6 kinase and RPS6 has become an area of keen investigation recently as multiple publications have presented conflicting data as to the function of these proteins in ribosomal biogenesis (see “RPS6”). Other S6K substrates have been identified including the eukaryotic elongation factor 2 kinase (eEF2K) and the eukaryotic translation initiation factor 4B (eIF4B). The phosphorylation of eEF2K results in inhibition of kinase activity, which leads to an increase in translation elongation. The phosphorylation of eIF4B by S6K leads to an increase of eIF4B protein levels recruited to eIF4A (reviewed in Ruvinsky and Meyuhas).68  This increased association promotes the helicase function of eIF4A and potentially increases the scanning ability of the ribosome. Because p70 S6 kinase can regulate components of the translational machinery, the targeting of S6K should have a detrimental effect on leukemias and lymphomas that are dependent upon an abnormal rate of protein synthesis. Early indications of a plausible role of S6K in hematologic malignancies, in particular T-cell lymphoma, were first described by Dumont et al nearly 15 years ago. They observed that rapamycin inhibited several biologic responses in the YAC-1 T-cell lymphoma, including proliferation and cell-cycle progression. By generating stable somatic mutants with altered sensitivities to rapamycin, they further demonstrated that the growth inhibitory effects of the drug were in fact associated with the inhibition of S6K activity.69 

More recent studies have validated these earlier findings by demonstrating that inhibition of S6K after rapamycin treatment results in both growth inhibition and apoptotic cell death of B-precursor acute lymphoblastic leukemia cell lines in vitro and improved survival in a leukemia/lymphoma transgenic mouse model.52  S6K hyperactivation has also been suggested as a critical mediator underlying the pathogenesis of the malignant Hodgkin/Reed-Sternberg cells of Hodgkin lymphoma (HL). Dutton et al showed that constitutive activation of the PI3K/AKT pathway was common in HL primary tumors and cell lines. As a consequence of this activation, the mTOR substrates 4E-BP1 and S6K were also hyperphosphorylated. This relationship was demonstrated not only by the clear inhibitory actions of rapamycin on these proteins but also by the use of LY294002, a commonly used inhibitor that targets PIKK family members, including mTOR (reviewed in Liu et al).70,71  These results highlight the critical role of the PI3K/AKT/mTOR pathway in sustaining the growth of leukemia/lymphoma cells. The inhibition of p70 S6 kinase has been shown to activate other multiple signaling pathways through a S6K/PI3K feedback mechanism. Therefore, the targeting of p70 S6 kinase with small molecule inhibitors would optimally be incorporated with other therapies to disrupt signaling pathways known to be affected by this feedback such as mitogen-activated protein kinase (MAPK), MAPK signal-integrating kinase (MNK), and AKT.72-74 

Ribosomes in higher eukaryotic cells are large multimeric ribonucleoprotein complexes consisting of 4 ribosomal RNA (rRNA) molecules and approximately 80 ribosomal proteins which make up a large (60S) and small (40S) subunit.75,76  In view of the fact that a significant proportion of the cell's energy is spent in ribosomal biogenesis,77  it is reasonable to expect that the proteins and rRNA that are the constituents of ribosomes be tightly regulated. Ribosomal biogenesis and assembly is extremely complex with ribosomal proteins themselves being involved in the regulation of other ribosomal proteins. Experimental evidence has recently demonstrated that the lack of a specific ribosomal protein, RPS6 can be catastrophic for the formation of a nascent ribosomal subunit. Due to the interdependency of ribosomal protein regulation, the loss of RPS6 can lead to an overabundance of other ribosomal proteins and the activation of extraribosomal functions of these proteins secondary to loss of regulation.78  Of all the essential ribosomal proteins, RPS6 has attracted the most attention since it was the first ribosomal protein shown to be phosphorylated in response to stimuli.79-81  The significant body of research conducted on RPS6 over the past few decades has implicated RPS6 in the regulation of protein synthesis (through the regulation of 5′ terminal oligopyrimidine [5′ TOP] RNA synthesis) and cell size. These messages containing an oligopyrimidine tract are important for ribosomal biogenesis as they encode most of the translational apparatus.82,83  A selective increase in the translation of these messages can be induced in mitogen-stimulated cells80,81,84  and can be selectively decreased through treatment with rapamycin or its derivatives,85  suggesting that this group of mRNAs may have a important role in the control of cellular growth. However, the role of RPS6 and its upstream regulatory molecules have come into question in recent years with publications demonstrating that 5′ TOP mRNA are still regulated in mouse embryonic fibroblasts (MEF) in which all 5 possible phosphorylation sites on RPS6 had been mutated to alanines86  or in p70 S6 kinase 1 null mice.87  Conversely, another report demonstrated that RPS6 phosphorylation was still present in MEF cells (p70 S6 kinase 1−/−) due to a compensatory signal from p70 S6 kinase 2.47  To resolve these discrepancies a more thorough molecular understanding of the phosphorylation status of RPS6 is needed.

The haploinsufficiency of RPS6 has been linked to multiple different phenotypes including cancer,88,89  and specifically cancers of the hematopoietic system.90  In contrast, the Gartenhaus laboratory has recently found that RPS6 is highly expressed in primary diffuse large B-cell lymphoma (DLBCL) samples compared with normal lymph node samples (P.R.H., Krystyna Mazan-Mamczarz, Bojie Dai, Frank Zhao, R.B.G., manuscript in preparation). As RPS6 may regulate the stoichiometry of other ribosomal proteins, it is plausible that RPS6 levels are increased in lymphoma to confer a larger number of ribosomes and therefore greater capacity to translate oncogenic messages. The impact on the cellular phenotype vis-à-vis the absence or decrease of RPS6 has previously been investigated using genetic methods such as siRNA or conditional gene knockouts supporting RPS6 as a negative regulator of 5′ TOP message translation. Several publications have revealed a mechanism in which an increase in the ribosomal protein RPL11, after suppression of RPS6 function, leads to an inhibition in proliferation and an increase in cell apoptosis through a p53-dependent pathway.78,91,92  Because the inactivation of p53 function in non-Hodgkin lymphoma is relatively low,93,94  targeting RPS6 function in non-Hodgkin lymphoma may represent a potential therapeutic paradigm.

The oncogene MCT-1 (multiple copies in T-cell lymphoma-1) is found on chromosome Xq22-24 and has been shown to be overexpressed in several B-cell and T-cell lymphoma lines and a small subset of diffuse large B-cell lymphoma (DLBCL), the most common form of non-Hodgkin lymphoma.95,96  Recent data demonstrate that MCT-1 protein levels are highly elevated in the majority of primary DLBCL samples compared with indolent lymphoid malignancies when examined by tissue microarray (TMA) analysis.97  The overexpression of MCT-1 has been previously implicated in cellular transformation through its ability to stimulate cell proliferation, suppress apoptosis, and enhance signaling pathways involved in cell survival.95,96,98  Knocking down MCT-1 protein levels in DLBCL significantly reduced cell viability through an apoptotic mechanism,97  providing the first direct genetic evidence that interfering with MCT-1 function was able to induce apoptosis in lymphoma cells with high endogenous levels of MCT-1 protein.

MCT-1 belongs to a family of PUA (pseudouridine synthase and archaeosine transglycosylase)–containing proteins, which have the ability to interact with RNA.98,99  MCT-1 protein interacts with the cap complex, m7GpppN (where N is any nucleotide), found at the 5′-terminus of all cellular eukaryotic mRNAs which recruits the density-regulated protein (DENR/DRP) in an eIF4E-dependent manner.100  DENR/DRP contains a SUI1 domain and is also found in the translation initiation factor eIF1.98  The recruitment of DENR can increase the translation initiation of a subset of mRNAs, containing a long and highly structured 5′ UTR, typically found in cancer-related messages.100  The mechanism(s) underlying this selective increase in translation has been described and is attributed to the increased scanning capability of the 40S ribosome and increased recognition of the correct initiation codon conferred by the DENR.98,100,101  This increased scanning ability and AUG start codon recognition allows for these selectively up-regulated messages to increase their presence in heavier polysomes and facilitate subsequent rounds of translation leading to significantly increased levels of oncogenic proteins present in the cell.

Earlier work demonstrated that a MCT-1 deletion mutant containing only the PUA domain still retained the ability to interact with the eIF4F/5′ methyl-7-GTP capped structure, but not enhance translation.100  A recent report demonstrated that the PUA domain mutant protein could act in a dominant-negative (DN) manner competing with wild-type MCT-1 for the translation initiation complex.102  The DN-mutant protein attenuated the malignant phenotype of lymphoma cells through modulation of the translational profile and sensitized cells to both irradiation and anthracyclines, 2 common therapeutic modalities used for the treatment of NHL.102  Intriguingly, MCT-1 is unique in that it is the only known PUA-domain containing protein to also not contain a SUI1 motif.98  using a peptide-based or small molecule approach to interrupt the physical association of MCT-1 and DENR proteins is a potentially useful approach to prevent the recruitment of SUI1 domain-containing DENR/DRP protein to the cap complex. Currently, there are no specific small molecule inhibitors that can directly inhibit MCT-1 protein function. Nandi et al previously demonstrated that phosphorylation of MCT-1 protein on threonine 81 by ERK1/2 is critical for protein stability and for its ability to promote cell proliferation.103  Taking advantage of a recently identified ERK1/2 small molecule inhibitor104  MCT-1 function was disrupted by targeting its upstream kinase with significant impact on lymphoma cell viability and clonogenic capacity. Importantly, using a human lymphoma xenograft model Dai et al were able to demonstrate significant antilymphoma activity in vivo using this approach, thereby supporting the development of novel small inhibitory molecules directed toward MCT-1 as a promising approach in DLBCL.97 

These exciting findings support both the clinical relevance of potentially targeting the MCT-1 oncogene and provide additional evidence linking MCT-1 with lymphoma development and/or progression. The current challenge is to identify the complete repertoire of translated mRNAs regulated by MCT-1 and how they participate in lymphomagenesis.

In addition to the availability of eIF4E protein to form the eIF4F translation initiation complex, eIF4E activity is also regulated by phosphorylation events. Phosphorylation of eIF4E is know to be enhanced when cells are treated with specific stimuli including; serum, phorbol esters and growth factors.105  The signals from these stimuli were determined to be transduced through the MAPK pathway,105  specifically MNK.106,107 

The exact mechanism as to how the phosphorylation of eIF4E regulates protein translation is still unclear. Prior data suggested that the phosphorylation of eIF4E enhanced the 5′ methyl-7-GTP cap binding potential of the protein.108  However, more recent experimental evidence has since shown that the phosphorylation of eIF4E actually decreases its affinity for a capped substrate.109,110  In contrast, the physiologic role of MNK phosphorylation of eIF4E may be better defined. Wendel et al have demonstrated that the insertion of a constitutively activated MNK1 into an Eμ-Myc mouse model produced lymphomas of a mature B-cell phenotype.111  Ueda et al created a MNK1/2 double knockout mice and demonstrated that there was no phosphorylated eIF4E levels in all tissues examined. These results demonstrated that eIF4E phosphorylation is not necessary for general protein synthesis and cap-dependent translation.112 

With the advent of a novel small molecule inhibitor specific to the MNK kinases, CGP57380, these data may become more easily reconciled. Treatment of prostate cancer cell lines with CGP57380 reduced the levels of phospho-eIF4E and reduced the amount of mRNA in the polysomal fraction of translating ribosomes and inhibited proliferation.113  CGP57380 has also been shown to be effective in inhibiting polysome associations with mRNA in chronic myelogenous leukemia cells and that treatment with this inhibitor enhances the activity of imatinib and induces apoptosis in imatinib-resistant CML cells.114  The inhibition of MNK has also been shown to affect the ability of eIF4E to transport oncogenic messages from the nucleus to the cytoplasm.115,116  Topisirovic et al convincingly demonstrated that phosphorylation of eIF4E contributes to the oncogenic potential of eIF4E by mutating the residues which can be phosphorylated. These mutants were less able to form anchorage-dependent foci compared with the wild-type eIF4E.115  A theory proposed by Morley and Naegele could explain the molecular mechanism and the phenotypic evidence presented here, they propose that phosphorylation of eIF4E and its subsequent release from the 5′ methyl-7-GTP cap complex could allow for a second molecule of eIF4E to interact with the cap and prime another round of translation initiation seen in polysomes.117 

The overexpression of ERK1/2 and p38 kinases, which are responsible for the activation of MNK, has been demonstrated in follicular lymphoma118  and DLBCL.97  In addition, the absence of microRNAs (miRNA) that regulate the MAPK pathway has also been observed in primary DLBCL samples while still present in matched normal B-cell lymphocytes from the same patients.119  Dai et al also found increased levels of ERK protein in a large percentage of human DLBCL samples.97  These observations in conjunction with the experimental evidence of using small molecule inhibitors in CML cells demonstrate that the MNK kinases are viable targets for future treatment strategies in hematologic malignancies.

Although significant data have been accumulated to date, the potential of targeting components of the translational machinery in hematologic malignancies has yet to be fully realized. The seminal work accomplished in targeting the eIF4F complex28-31  demonstrates that inhibition of translation initiation should be given high priority for future development of new and more effective small molecule inhibitors. While the work on rapamycin and its analogs is broad and has finally reached the clinical settings in several cases, we are excited to see work from laboratories that are working to develop molecules that target the mTOR kinase domain directly34,62  (Table 1). We have shown that specific targeting of MCT-1 has significant preclinical activity and experiments are in progress designed to disrupt the MCT-1/DENR association observed at the cap complex during translation initiation. Finally, the improved understanding and discovery of novel molecules involved in translation will provide innovative clinical approaches toward the treatment of lymphoma and leukemia.

Table 1

Small molecule inhibitors involved in the suppression of translation

DrugBinding SiteMechanism of actionReference(s)
Ribavirin 5′ methyl-7-GTP cap Competitive inhibitor of eIF4E binding to capped mRNA 29,30  
4EGI-1 eIF4G Competitive inhibitor of eIF4E binding to eIF4G 31,32  
GnRH-4EBP fusion peptide eIF4E Peptidomimetic inhibitor of eIF4E binding to eIF4G 35  
Rapamycin, analogs and derivatives FKBP-rapamycin-binding (FRB) domain of mTOR Inhibits recruitment of FKBP12 protein to mTOR Reviewed in 45  
Torin1 mTOR kinase domain Inhibition of ATP binding to kinase domain 34  
PP242 mTOR kinase domain Inhibition of ATP binding to kinase domain 62  
PP30 mTOR kinase domain Inhibition of ATP binding to kinase domain 62  
CGP57380 MNK kinase domain Inhibition of ATP binding to kinase domain 113,,116  
DrugBinding SiteMechanism of actionReference(s)
Ribavirin 5′ methyl-7-GTP cap Competitive inhibitor of eIF4E binding to capped mRNA 29,30  
4EGI-1 eIF4G Competitive inhibitor of eIF4E binding to eIF4G 31,32  
GnRH-4EBP fusion peptide eIF4E Peptidomimetic inhibitor of eIF4E binding to eIF4G 35  
Rapamycin, analogs and derivatives FKBP-rapamycin-binding (FRB) domain of mTOR Inhibits recruitment of FKBP12 protein to mTOR Reviewed in 45  
Torin1 mTOR kinase domain Inhibition of ATP binding to kinase domain 34  
PP242 mTOR kinase domain Inhibition of ATP binding to kinase domain 62  
PP30 mTOR kinase domain Inhibition of ATP binding to kinase domain 62  
CGP57380 MNK kinase domain Inhibition of ATP binding to kinase domain 113,,116  

We apologize to those authors whose original articles could not be cited due to space constraints.

This work was supported in part by a Merit Review Award from the Department of Veterans Affairs (R.B.G.) and R01AA017972 from the National Institutes of Health (R.B.G.).

National Institutes of Health

Contribution: R.B.G. designed the review; and all authors contributed to the writing and proofreading of the paper.

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

Correspondence: Ronald B. Gartenhaus, Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201; e-mail: rgartenhaus@som.umaryland.edu.

1
Sonenberg
 
N
Hinnebusch
 
AG
Regulation of translation initiation in eukaryotes: mechanisms and biological targets.
Cell
2009
, vol. 
136
 
4
(pg. 
731
-
745
)
2
Ruggero
 
D
Pandolfi
 
PP
Does the ribosome translate cancer?
Nat Rev Cancer
2003
, vol. 
3
 
3
(pg. 
179
-
192
)
3
Ruggero
 
D
Montanaro
 
L
Ma
 
L
, et al. 
The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis.
Nat Med
2004
, vol. 
10
 
5
(pg. 
484
-
486
)
4
Cormier
 
P
Pyronnet
 
S
Salaün
 
P
Mulner-Lorillon
 
O
Sonenberg
 
N
Cap-dependent translation and control of the cell cycle.
Prog Cell Cycle Res
2003
, vol. 
5
 (pg. 
469
-
475
)
5
Holcik
 
M
Sonenberg
 
N
Translational control in stress and apoptosis.
Nat Rev Mol Cell Biol
2005
, vol. 
6
 
4
(pg. 
318
-
327
)
6
Pickering
 
BM
Willis
 
AE
The implications of structured 5′ untranslated regions on translation and disease.
Semin Cell Dev Biol
2005
, vol. 
16
 
1
(pg. 
39
-
47
)
7
Bilanges
 
B
Stokoe
 
D
Mechanisms of translational deregulation in human tumors and therapeutic intervention strategies.
Oncogene
2007
, vol. 
26
 
41
(pg. 
5973
-
5990
)
8
Smith
 
SM
van Besien
 
K
mTOR inhibition in lymphoma: a rational and promising strategy.
Lett Drug Des Discov
2007
, vol. 
4
 (pg. 
224
-
231
)
9
Koromilas
 
AE
Lazaris-Karatzas
 
A
Sonenberg
 
N
mRNAs containing extensive secondary structure in their 5′ non-coding region translate efficiently in cells overexpressing initiation factor eIF-4E.
EMBO J
1992
, vol. 
11
 
11
(pg. 
4153
-
4158
)
10
Hiremath
 
LS
Webb
 
NR
Rhoads
 
RE
Immunological detection of the messenger RNA cap-binding protein.
J Biol Chem
1985
, vol. 
260
 
13
(pg. 
7843
-
7849
)
11
Duncan
 
R
Milburn
 
SC
Hershey
 
JW
Regulated phosphorylation and low abundance of HeLa cell initiation factor eIF-4F suggest a role in translational control: heat shock effects on eIF-4F.
J Biol Chem
1987
, vol. 
262
 
1
(pg. 
380
-
388
)
12
Clemens
 
MJ
Targets and mechanisms for the regulation of translation in malignant transformation.
Oncogene
2004
, vol. 
23
 
18
(pg. 
3180
-
3188
)
13
De Benedetti
 
A
Graff
 
JR
eIF-4E expression and its role in malignancies and metastases.
Oncogene
2004
, vol. 
23
 
18
(pg. 
3189
-
3199
)
14
Rau
 
M
Ohlmann
 
T
Morley
 
SJ
Pain
 
VM
A reevaluation of the cap-binding protein, eIF4E, as a rate-limiting factor for initiation of translation in reticulocyte lysate.
J Biol Chem
1996
, vol. 
271
 
15
(pg. 
8983
-
8990
)
15
Lodish
 
HF
Model for the regulation of mRNA translation applied to haemoglobin synthesis.
Nature
1974
, vol. 
251
 
5474
(pg. 
385
-
388
)
16
Rousseau
 
D
Kaspar
 
R
Rosenwald
 
I
Gehrke
 
L
Sonenberg
 
N
Translation initiation of ornithine decarboxylase and nucleocytoplasmic transport of cyclin D1 mRNA are increased in cells overexpressing eukaryotic initiation factor 4E.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 
3
(pg. 
1065
-
1070
)
17
Lazaris-Karatzas
 
A
Montine
 
KS
Sonenberg
 
N
Malignant transformation by a eukaryotic initiation factor subunit that binds to mRNA 5′ cap.
Nature
1990
, vol. 
345
 
6275
(pg. 
544
-
547
)
18
Zimmer
 
SG
De Benedetti
 
A
Graff
 
JR
Translational control of malignancy: the mRNA cap-binding protein, eIF-4E, as a central regulator of tumor formation, growth, invasion and metastasis.
Anticancer Res
2000
, vol. 
20
 
3A
(pg. 
1343
-
1351
)
19
Rosenwald
 
IB
Chen
 
JJ
Wang
 
S
Savas
 
L
London
 
IM
Pullman
 
J
Upregulation of protein synthesis initiation factor eIF-4E is an early event during colon carcinogenesis.
Oncogene
1999
, vol. 
18
 
15
(pg. 
2507
-
2517
)
20
Kerekatte
 
V
Smiley
 
K
Hu
 
B
Smith
 
A
Gelder
 
F
De Benedetti
 
A
The proto-oncogene/translation factor eIF4E: a survey of its expression in breast carcinomas.
Int J Cancer
1995
, vol. 
64
 
1
(pg. 
27
-
31
)
21
Coleman
 
LJ
Peter
 
MB
Teall
 
TJ
, et al. 
Combined analysis of eIF4E and 4E-binding protein expression predicts breast cancer survival and estimates eIF4E activity.
Br J Cancer
2009
, vol. 
100
 
9
(pg. 
1393
-
1399
)
22
Wang
 
S
Rosenwald
 
IB
Hutzler
 
MJ
, et al. 
Expression of the eukaryotic translation initiation factors 4E and 2alpha in non-Hodgkin's lymphomas.
Am J Pathol
1999
, vol. 
155
 
1
(pg. 
247
-
255
)
23
Topisirovic
 
I
Guzman
 
ML
McConnell
 
MJ
, et al. 
Aberrant eukaryotic translation initiation factor 4E-dependent mRNA transport impedes hematopoietic differentiation and contributes to leukemogenesis.
Mol Cell Biol
2003
, vol. 
23
 
24
(pg. 
8992
-
9002
)
24
Wendel
 
HG
De Stanchina
 
E
Fridman
 
JS
, et al. 
Survival signaling by Akt and eIF4E in oncogenesis and cancer therapy.
Nature
2004
, vol. 
428
 
6980
(pg. 
332
-
337
)
25
De Benedetti
 
A
Joshi-Barve
 
S
Rinker-Schaeffer
 
C
Rhoads
 
RE
Expression of antisense RNA against initiation factor eIF-4E mRNA in HeLa cells results in lengthened cell division times, diminished translation rates, and reduced levels of both eIF-4E and the p220 component of eIF-4F.
Mol Cell Biol
1991
, vol. 
11
 
11
(pg. 
5435
-
5445
)
26
Rinker-Schaeffer
 
CW
Graff
 
JR
De Benedetti
 
A
Zimmer
 
SG
Rhoads
 
RE
Decreasing the level of translation initiation factor 4E with antisense RNA causes reversal of ras-mediated transformation and tumorigenesis of cloned rat embryo fibroblasts.
Int J Cancer
1993
, vol. 
55
 
5
(pg. 
841
-
847
)
27
Graff
 
JR
Boghaert
 
ER
De Benedetti
 
A
, et al. 
Reduction of translation initiation factor 4E decreases the malignancy of ras-transformed cloned rat embryo fibroblasts.
Int J Cancer
1995
, vol. 
60
 
2
(pg. 
255
-
263
)
28
Graff
 
JR
Konicek
 
BW
Vincent
 
TM
, et al. 
Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity.
J Clin Invest
2007
, vol. 
117
 
9
(pg. 
2638
-
2648
)
29
Kentsis
 
A
Topisirovic
 
I
Culjkovic
 
B
Shao
 
L
Borden
 
KL
Ribavirin suppresses eIF4E-mediated oncogenic transformation by physical mimicry of the 7-methyl guanosine mRNA cap.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
52
(pg. 
18105
-
18110
)
30
Assouline
 
S
Culjkovic
 
B
Cocolakis
 
E
, et al. 
Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): a proof-of-principle clinical trial with ribavirin.
Blood
2009
, vol. 
114
 
2
(pg. 
257
-
260
)
31
Moerke
 
NJ
Aktas
 
H
Chen
 
H
, et al. 
Small-molecule inhibition of the interaction between the translation initiation factors eIF4E and eIF4G.
Cell
2007
, vol. 
128
 
2
(pg. 
257
-
267
)
32
Tamburini
 
J
Green
 
AS
Bardet
 
V
, et al. 
Protein synthesis is resistant to rapamycin and constitutes a promising therapeutic target in acute myeloid leukemia.
Blood
2009
, vol. 
114
 
8
(pg. 
1618
-
1627
)
33
Choo
 
AY
Yoon
 
SO
Kim
 
SG
Roux
 
PP
Blenis
 
J
Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
45
(pg. 
17414
-
17419
)
34
Thoreen
 
CC
Kang
 
SA
Chang
 
JW
, et al. 
An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1.
J Biol Chem
2009
, vol. 
284
 
12
(pg. 
8023
-
8032
)
35
Ko
 
SY
Guo
 
H
Barengo
 
N
Naora
 
H
Inhibition of ovarian cancer growth by a tumor-targeting peptide that binds eukaryotic translation initiation factor 4E.
Clin Cancer Res
2009
, vol. 
15
 
13
(pg. 
4336
-
4347
)
36
Bordeleau
 
ME
Robert
 
F
Gerard
 
B
, et al. 
Therapeutic suppression of translation initiation modulates chemosensitivity in a mouse lymphoma model.
J Clin Invest
2008
, vol. 
118
 
7
(pg. 
2651
-
2660
)
37
Robert
 
F
Carrier
 
M
Rawe
 
S
Chen
 
S
Lowe
 
S
Pelletier
 
J
Altering chemosensitivity by modulating translation elongation.
PLoS ONE
2009
, vol. 
4
 
5
pg. 
e5428
 
38
Crotty
 
S
Cameron
 
CE
Andino
 
R
RNA virus error catastrophe: direct molecular test by using ribavirin.
Proc Natl Acad Sci U S A
2001
, vol. 
98
 
12
(pg. 
6895
-
6900
)
39
Vallisa
 
D
Bernuzzi
 
P
Arcaini
 
L
, et al. 
Role of anti-hepatitis C virus (HCV) treatment in HCV-related, low-grade, B-cell, non-Hodgkin's lymphoma: a multicenter Italian experience.
J Clin Oncol
2005
, vol. 
23
 
3
(pg. 
468
-
473
)
40
Mazzaro
 
C
De Re
 
V
Spina
 
M
, et al. 
Pegylated-interferon plus ribavirin for HCV-positive indolent non-Hodgkin lymphomas.
Br J Haematol
2009
, vol. 
145
 
2
(pg. 
255
-
257
)
41
Sabatini
 
DM
mTOR and cancer: insights into a complex relationship.
Nat Rev Cancer
2006
, vol. 
6
 
9
(pg. 
729
-
734
)
42
Heitman
 
J
Movva
 
NR
Hiestand
 
PC
Hall
 
MN
FK 506-binding protein proline rotamase is a target for the immunosuppressive agent FK 506 in Saccharomyces cerevisiae.
Proc Natl Acad Sci U S A
1991
, vol. 
88
 
5
(pg. 
1948
-
1952
)
43
Wullschleger
 
S
Loewith
 
R
Hall
 
MN
TOR signaling in growth and metabolism.
Cell
2006
, vol. 
124
 
3
(pg. 
471
-
484
)
44
Zeng
 
Z
Sarbassov dos
 
D
Samudio
 
IJ
, et al. 
Rapamycin derivatives reduce mTORC2 signaling and inhibit AKT activation in AML.
Blood
2007
, vol. 
109
 
8
(pg. 
3509
-
3512
)
45
Guertin
 
DA
Sabatini
 
DM
Defining the role of mTOR in cancer.
Cancer Cell
2007
, vol. 
12
 
1
(pg. 
9
-
22
)
46
Sancak
 
Y
Thoreen
 
CC
Peterson
 
TR
, et al. 
PRAS40 is an insulin-regulated inhibitor of the mTORC1 protein kinase.
Mol Cell
2007
, vol. 
25
 
6
(pg. 
903
-
915
)
47
Lee-Fruman
 
KK
Kuo
 
CJ
Lippincott
 
J
Terada
 
N
Blenis
 
J
Characterization of S6K2, a novel kinase homologous to S6K1.
Oncogene
1999
, vol. 
18
 
36
(pg. 
5108
-
5114
)
48
Easton
 
JB
Houghton
 
PJ
mTOR and cancer therapy.
Oncogene
2006
, vol. 
25
 
48
(pg. 
6436
-
6446
)
49
Abraham
 
RT
Eng
 
CH
Mammalian target of rapamycin as a therapeutic target in oncology.
Expert Opin Ther Targets
2008
, vol. 
12
 
2
(pg. 
209
-
222
)
50
Teachey
 
DT
Grupp
 
SA
Brown
 
VI
Mammalian target of rapamycin inhibitors and their potential role in therapy in leukaemia and other haematological malignancies.
Br J Haematol
2009
, vol. 
145
 
5
(pg. 
569
-
580
)
51
Martelli
 
AM
Evangelisti
 
C
Chiarini
 
F
Grimaldi
 
C
Manzoli
 
L
McCubrey
 
JA
Targeting the PI3K/AKT/mTOR signaling network in acute myelogenous leukemia.
Expert Opin Investig Drugs
2009
, vol. 
18
 
9
(pg. 
1333
-
1349
)
52
Brown
 
VI
Fang
 
J
Alcorn
 
K
, et al. 
Rapamycin is active against B-precursor leukemia in vitro and in vivo, an effect that is modulated by IL-7-mediated signaling.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
25
(pg. 
15113
-
15118
)
53
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
)
54
Zanesi
 
N
Aqeilan
 
R
Drusco
 
A
, et al. 
Effect of rapamycin on mouse chronic lymphocytic leukemia and the development of nonhematopoietic malignancies in Emu-TCL1 transgenic mice.
Cancer Res
2006
, vol. 
66
 
2
(pg. 
915
-
920
)
55
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
)
56
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
)
57
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
)
58
Sillaber
 
C
Mayerhofer
 
M
Böhm
 
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
)
59
Ikezoe
 
T
Yang
 
Y
Nishioka
 
C
, et al. 
Effect of SU11248 on gastrointestinal stromal tumor-T1 cells: enhancement of growth inhibition via inhibition of 3-kinase/Akt/mammalian target of rapamycin signaling.
Cancer Sci
2006
, vol. 
97
 
9
(pg. 
945
-
951
)
60
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
 
Prepublished on July 28, 2009 as DOI 10.1016/j.leukres.2009.07.012
61
Chiarini
 
F
Falà
 
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
)
62
Feldman
 
ME
Apsel
 
B
Uotila
 
A
, et al. 
Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2.
PLoS Biol
2009
, vol. 
7
 
2
pg. 
e38
 
63
Hagner
 
PR
Mazan-Mamczarz
 
K
Dai
 
B
Corl
 
S
Zhao
 
XF
Gartenhaus
 
RB
Alcohol consumption and decreased risk of non-Hodgkin lymphoma: role of mTOR dysfunction.
Blood
2009
, vol. 
113
 
22
(pg. 
5526
-
5535
)
64
Kurmasheva
 
RT
Huang
 
S
Houghton
 
PJ
Predicted mechanisms of resistance to mTOR inhibitors.
Br J Cancer
2006
, vol. 
95
 
8
(pg. 
955
-
960
)
65
Fruman
 
DA
Wood
 
MA
Gjertson
 
CK
Katz
 
HR
Burakoff
 
SJ
Bierer
 
BE
FK506 binding protein 12 mediates sensitivity to both FK506 and rapamycin in murine mast cells.
Eur J Immunol
1995
, vol. 
25
 
2
(pg. 
563
-
571
)
66
Shi
 
Y
Sharma
 
A
Wu
 
H
Lichtenstein
 
A
Gera
 
J
Cyclin D1 and c-myc internal ribosome entry site (IRES)-dependent translation is regulated by AKT activity and enhanced by rapamycin through a p38 MAPK- and ERK-dependent pathway.
J Biol Chem
2005
, vol. 
280
 
12
(pg. 
10964
-
10973
)
67
Weinstein
 
IB
Joe
 
AK
Mechanisms of disease: Oncogene addiction–a rationale for molecular targeting in cancer therapy.
Nat Clin Pract Oncol
2006
, vol. 
3
 
8
(pg. 
448
-
457
)
68
Ruvinsky
 
I
Meyuhas
 
O
Ribosomal protein S6 phosphorylation: from protein synthesis to cell size.
Trends Biochem Sci
2006
, vol. 
31
 
6
(pg. 
342
-
348
)
69
Dumont
 
FJ
Altmeyer
 
A
Kastner
 
C
, et al. 
Relationship between multiple biologic effects of rapamycin and the inhibition of pp70S6 protein kinase activity: analysis in mutant clones of a T cell lymphoma.
J Immunol
1994
, vol. 
152
 
3
(pg. 
992
-
1003
)
70
Liu
 
P
Cheng
 
H
Roberts
 
TM
Zhao
 
JJ
Targeting the phosphoinositide 3-kinase pathway in cancer.
Nat Rev Drug Discov
2009
, vol. 
8
 
8
(pg. 
627
-
644
)
71
Dutton
 
A
Reynolds
 
GM
Dawson
 
CW
Young
 
LS
Murray
 
PG
Constitutive activation of phosphatidyl-inositide 3 kinase contributes to the survival of Hodgkin's lymphoma cells through a mechanism involving Akt kinase and mTOR.
J Pathol
2005
, vol. 
205
 
4
(pg. 
498
-
506
)
72
Wang
 
X
Yue
 
P
Chan
 
CB
, et al. 
Inhibition of mammalian target of rapamycin induces phosphatidylinositol 3-kinase-dependent and Mnk-mediated eukaryotic translation initiation factor 4E phosphorylation.
Mol Cell Biol
2007
, vol. 
27
 
21
(pg. 
7405
-
7413
)
73
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
)
74
Carracedo
 
A
Ma
 
L
Teruya-Feldstein
 
J
, et al. 
Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer.
J Clin Invest
2008
, vol. 
118
 
9
(pg. 
3065
-
3074
)
75
Fatica
 
A
Tollervey
 
D
Making ribosomes.
Curr Opin Cell Biol
2002
, vol. 
14
 
3
(pg. 
313
-
318
)
76
Perry
 
RP
Balanced production of ribosomal proteins.
Gene
2007
, vol. 
401
 
1–2
(pg. 
1
-
3
)
77
Warner
 
JR
The economics of ribosome biosynthesis in yeast.
Trends Biochem Sci
1999
, vol. 
24
 
11
(pg. 
437
-
440
)
78
Fumagalli
 
S
Di Cara
 
A
Neb-Gulati
 
A
, et al. 
Absence of nucleolar disruption after impairment of 40S ribosome biogenesis reveals an rpL11-translation-dependent mechanism of p53 induction.
Nat Cell Biol
2009
, vol. 
11
 
4
(pg. 
501
-
508
)
79
Gross
 
T
Nischt
 
R
Gatermann
 
K
Swida
 
U
Käufer
 
NF
Primary structure of the ribosomal protein gene S6 from Schizosaccharomyces pombe.
Curr Genet
1988
, vol. 
13
 
1
(pg. 
57
-
63
)
80
Franco
 
R
Rosenfeld
 
MG
Hormonally inducible phosphorylation of a nuclear pool of ribosomal protein S6.
J Biol Chem
1990
, vol. 
265
 
8
(pg. 
4321
-
4325
)
81
Pende
 
M
Um
 
SH
Mieulet
 
V
, et al. 
S6K1(−/−)/S6K2(−/−) mice exhibit perinatal lethality and rapamycin-sensitive 5′-terminal oligopyrimidine mRNA translation and reveal a mitogen-activated protein kinase-dependent S6 kinase pathway.
Mol Cell Biol
2004
, vol. 
24
 
8
(pg. 
3112
-
3124
)
82
Avni
 
D
Biberman
 
Y
Meyuhas
 
O
The 5′ terminal oligopyrimidine tract confers translational control on TOP mRNAs in a cell type- and sequence context-dependent manner.
Nucleic Acids Res
1997
, vol. 
25
 
5
(pg. 
995
-
1001
)
83
Jefferies
 
HB
Fumagalli
 
S
Dennis
 
PB
Reinhard
 
C
Pearson
 
RB
Thomas
 
G
Rapamycin suppresses 5′TOP mRNA translation through inhibition of p70s6k.
EMBO J
1997
, vol. 
16
 
12
(pg. 
3693
-
3704
)
84
Patursky-Polischuk
 
I
Stolovich-Rain
 
M
Hausner-Hanochi
 
M
, et al. 
The TSC-mTOR pathway mediates translational activation of TOP mRNAs by insulin largely in a raptor- or rictor-independent manner.
Mol Cell Biol
2009
, vol. 
29
 
3
(pg. 
640
-
649
)
85
Jefferies
 
HB
Reinhard
 
C
Kozma
 
SC
Thomas
 
G
Rapamycin selectively represses translation of the “polypyrimidine tract” mRNA family.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 
10
(pg. 
4441
-
4445
)
86
Ruvinsky
 
I
Sharon
 
N
Lerer
 
T
, et al. 
Ribosomal protein S6 phosphorylation is a determinant of cell size and glucose homeostasis.
Genes Dev
2005
, vol. 
19
 
18
(pg. 
2199
-
2211
)
87
Stolovich
 
M
Tang
 
H
Hornstein
 
E
, et al. 
Transduction of growth or mitogenic signals into translational activation of TOP mRNAs is fully reliant on the phosphatidylinositol 3-kinase-mediated pathway but requires neither S6K1 nor rpS6 phosphorylation.
Mol Cell Biol
2002
, vol. 
22
 
23
(pg. 
8101
-
8113
)
88
Nakamura
 
JL
Garcia
 
E
Pieper
 
RO
S6K1 plays a key role in glial transformation.
Cancer Res
2008
, vol. 
68
 
16
(pg. 
6516
-
6523
)
89
Ruvinsky
 
I
Katz
 
M
Dreazen
 
A
, et al. 
Mice deficient in ribosomal protein S6 phosphorylation suffer from muscle weakness that reflects a growth defect and energy deficit.
PLoS ONE
2009
, vol. 
4
 
5
pg. 
e5618
 
90
Watson
 
KL
Konrad
 
KD
Woods
 
DF
Bryant
 
PJ
Drosophila homolog of the human S6 ribosomal protein is required for tumor suppression in the hematopoietic system.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 
23
(pg. 
11302
-
11306
)
91
Volarevic
 
S
Stewart
 
MJ
Ledermann
 
B
, et al. 
Proliferation, but not growth, blocked by conditional deletion of 40S ribosomal protein S6.
Science
2000
, vol. 
288
 
5473
(pg. 
2045
-
2047
)
92
Sulic
 
S
Panic
 
L
Barkic
 
M
Mercep
 
M
Uzelac
 
M
Volarevic
 
S
Inactivation of S6 ribosomal protein gene in T lymphocytes activates a p53-dependent checkpoint response.
Genes Dev
2005
, vol. 
19
 
24
(pg. 
3070
-
3082
)
93
Koduru
 
PR
Raju
 
K
Vadmal
 
V
, et al. 
Correlation between mutation in P53, p53 expression, cytogenetics, histologic type, and survival in patients with B-cell non-Hodgkin's lymphoma.
Blood
1997
, vol. 
90
 
10
(pg. 
4078
-
4091
)
94
Sánchez-Beato
 
M
Sánchez-Aguilera
 
A
Piris
 
MA
Cell cycle deregulation in B-cell lymphomas.
Blood
2003
, vol. 
101
 
4
(pg. 
1220
-
1235
)
95
Prosniak
 
M
Dierov
 
J
Okami
 
K
, et al. 
A novel candidate oncogene, MCT-1, is involved in cell cycle progression.
Cancer Res
1998
, vol. 
58
 
19
(pg. 
4233
-
4237
)
96
Shi
 
B
Hsu
 
HL
Evens
 
AM
Gordon
 
LI
Gartenhaus
 
RB
Expression of the candidate MCT-1 oncogene in B- and T-cell lymphoid malignancies.
Blood
2003
, vol. 
102
 
1
(pg. 
297
-
302
)
97
Dai
 
B
Zhao
 
XF
Hagner
 
P
, et al. 
Extracellular signal-regulated kinase positively regulates the oncogenic activity of MCT-1 in diffuse large B-cell lymphoma.
Cancer Res
2009
, vol. 
1:69
 
19
(pg. 
7835
-
7843
)
98
Pérez-Arellano
 
I
Gallego
 
J
Cervera
 
J
The PUA domain–a structural and functional overview.
FEBS J
2007
, vol. 
274
 
19
(pg. 
4972
-
4984
)
99
Duan
 
J
Li
 
L
Lu
 
J
Wang
 
W
Ye
 
K
Structural mechanism of substrate RNA recruitment in H/ACA RNA-guided pseudouridine synthase.
Mol Cell
2009
, vol. 
34
 
4
(pg. 
427
-
439
)
100
Reinert
 
LS
Shi
 
B
Nandi
 
S
, et al. 
MCT-1 protein interacts with the cap complex and modulates messenger RNA translational profiles.
Cancer Res
2006
, vol. 
66
 
18
(pg. 
8994
-
9001
)
101
Fleischer
 
TC
Weaver
 
CM
McAfee
 
KJ
Jennings
 
JL
Link
 
AJ
Systematic identification and functional screens of uncharacterized proteins associated with eukaryotic ribosomal complexes.
Genes Dev
2006
, vol. 
20
 
10
(pg. 
1294
-
1307
)
102
Hagner
 
PR
Hagner
 
P
Dai
 
B
Corl
 
S
Liu
 
Z
Gartenhaus
 
RB
Targeted suppression of MCT-1 attenuates the malignant phenotype through a translational mechanism.
Leuk Res
2009
, vol. 
33
 
3
(pg. 
474
-
482
)
103
Nandi
 
S
Reinert
 
LS
Hachem
 
A
, et al. 
Phosphorylation of MCT-1 by p44/42 MAPK is required for its stabilization in response to DNA damage.
Oncogene
2007
, vol. 
26
 
16
(pg. 
2283
-
2289
)
104
Hancock
 
CN
Macias
 
A
Lee
 
EK
Yu
 
SY
Mackerell
 
AD
Shapiro
 
P
Identification of novel extracellular signal-regulated kinase docking domain inhibitors.
J Med Chem
2005
, vol. 
48
 
14
(pg. 
4586
-
4595
)
105
Wang
 
X
Flynn
 
A
Waskiewicz
 
AJ
, et al. 
The phosphorylation of eukaryotic initiation factor eIF4E in response to phorbol esters, cell stresses, and cytokines is mediated by distinct MAP kinase pathways.
J Biol Chem
1998
, vol. 
273
 
16
(pg. 
9373
-
9377
)
106
Fukunaga
 
R
Hunter
 
T
MNK1, a new MAP kinase-activated protein kinase, isolated by a novel expression screening method for identifying protein kinase substrates.
EMBO J
1997
, vol. 
16
 
8
(pg. 
1921
-
1933
)
107
Waskiewicz
 
AJ
Flynn
 
A
Proud
 
CG
Cooper
 
JA
Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2.
EMBO J
1997
, vol. 
16
 
8
(pg. 
1909
-
1920
)
108
Minich
 
WB
Balasta
 
ML
Goss
 
DJ
Rhoads
 
RE
Chromatographic resolution of in vivo phosphorylated and nonphosphorylated eukaryotic translation initiation factor eIF-4E: increased cap affinity of the phosphorylated form.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 
16
(pg. 
7668
-
7672
)
109
von Der Haar
 
T
Ball
 
PD
McCarthy
 
JE
Stabilization of eukaryotic initiation factor 4E binding to the mRNA 5′-Cap by domains of eIF4G.
J Biol Chem
2000
, vol. 
275
 
39
(pg. 
30551
-
30555
)
110
Scheper
 
GC
Proud
 
CG
Does phosphorylation of the cap-binding protein eIF4E play a role in translation initiation?
Eur J Biochem
2002
, vol. 
269
 
22
(pg. 
5350
-
5359
)
111
Wendel
 
HG
Silva
 
RL
Malina
 
A
, et al. 
Dissecting eIF4E action in tumorigenesis.
Genes Dev
2007
, vol. 
21
 
24
(pg. 
3232
-
3237
)
112
Ueda
 
T
Watanabe-Fukunaga
 
R
Fukuyama
 
H
Nagata
 
S
Fukunaga
 
R
Mnk2 and Mnk1 are essential for constitutive and inducible phosphorylation of eukaryotic initiation factor 4E but not for cell growth or development.
Mol Cell Biol
2004
, vol. 
24
 
15
(pg. 
6539
-
6549
)
113
Bianchini
 
A
Loiarro
 
M
Bielli
 
P
, et al. 
Phosphorylation of eIF4E by MNKs supports protein synthesis, cell cycle progression and proliferation in prostate cancer cells.
Carcinogenesis
2008
, vol. 
29
 
12
(pg. 
2279
-
2288
)
114
Zhang
 
M
, et al. 
Inhibition of polysome assembly enhances imatinib activity against chronic myelogenous leukemia and overcomes imatinib resistance.
Mol Cell Biol
2008
, vol. 
28
 
20
(pg. 
6496
-
6509
)
115
Topisirovic
 
I
Ruiz-Gutierrez
 
M
Borden
 
KL
Phosphorylation of the eukaryotic translation initiation factor eIF4E contributes to its transformation and mRNA transport activities.
Cancer Res
2004
, vol. 
64
 
23
(pg. 
8639
-
8642
)
116
Phillips
 
A
Blaydes
 
JP
MNK1 and EIF4E are downstream effectors of MEKs in the regulation of the nuclear export of HDM2 mRNA.
Oncogene
2008
, vol. 
27
 
11
(pg. 
1645
-
1649
)
117
Morley
 
SJ
Naegele
 
S
Phosphorylation of eukaryotic initiation factor (eIF) 4E is not required for de novo protein synthesis following recovery from hypertonic stress in human kidney cells.
J Biol Chem
2002
, vol. 
277
 
36
(pg. 
32855
-
32859
)
118
Elenitoba-Johnson
 
KS
Jenson
 
SD
Abbott
 
RT
, et al. 
Involvement of multiple signaling pathways in follicular lymphoma transformation: p38-mitogen-activated protein kinase as a target for therapy.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 
12
(pg. 
7259
-
7264
)
119
Sánchez
 
A
Setién
 
F
Martinez
 
N
, et al. 
Epigenetic inactivation of the ERK inhibitor Spry2 in B-cell diffuse lymphomas.
Oncogene
2008
, vol. 
27
 
36
(pg. 
4969
-
4972
)
Sign in via your Institution