Next Article in Journal
Origin, Evolution and Diversity of φ29-like Phages—Review and Bioinformatic Analysis
Previous Article in Journal
The Association of IFNL4 Gene Polymorphisms with Hepatitis B Virus (HBV) Infection in the Northern Region of Pará, Brazil
Previous Article in Special Issue
Cordycepin Enhances the Therapeutic Efficacy of Doxorubicin in Treating Triple-Negative Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploiting Translation Machinery for Cancer Therapy: Translation Factors as Promising Targets

by
Urmila Sehrawat
Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
Int. J. Mol. Sci. 2024, 25(19), 10835; https://doi.org/10.3390/ijms251910835
Submission received: 21 August 2024 / Revised: 26 September 2024 / Accepted: 2 October 2024 / Published: 9 October 2024

Abstract

:
Eukaryotic protein translation has slowly gained the scientific community’s attention for its advanced and powerful therapeutic potential. However, recent technical developments in studying ribosomes and global translation have revolutionized our understanding of this complex multistep process. These developments have improved and deepened the current knowledge of mRNA translation, sparking excitement and new possibilities in this field. Translation factors are crucial for maintaining protein synthesis homeostasis. Since actively proliferating cancer cells depend on protein synthesis, dysregulated protein translation is central to tumorigenesis. Translation factors and their abnormal expressions directly affect multiple oncogenes and tumor suppressors. Recently, small molecules have been used to target translation factors, resulting in translation inhibition in a gene-specific manner, opening the door for developing translation inhibitors that can lead to novel chemotherapeutic drugs for treating multiple cancer types caused by dysregulated translation machinery. This review comprehensively summarizes the involvement of translation factors in tumor progression and oncogenesis. Also, it sheds light on the evolution of translation factors as novel drug targets for developing future therapeutic drugs for treating cancer.

1. Introduction

The expression of genetic messages is a crucial fundamental process and provides deep insights into the phenotypes of eukaryotic cells. Gene expression is extensively regulated at all stages, from transcription to protein synthesis during translation [1]. It is evident that mRNA translation is a significant stage of gene expression control [2] and maintains homeostasis for the expression of available cellular mRNA pools [3]. The translation process is generally divided into the following four steps: initiation, elongation, termination, and ribosomal recycling [1]. The primary translation control that regulates protein expression happens during the rate-limiting stage of the translation initiation step, wherein the small subunit of ribosome (40S) associates with translation initiation factors to recognize the AUG start codon on the mRNA template for a systematic sequential assembly of 80S elongation competent ribosome [2,4].
Eukaryotic translation initiation is a multistep process that has been well characterized and believed to be the most prominent mechanism during the translation process. Briefly, during translation initiation, there is the simultaneous formation of 43S pre-initiation complex (PIC) (Figure 1, step 2), which is a multifactorial complex formed by the association of 40S ribosomal subunit with eukaryotic translation initiation factors (eIFs), i.e., eIF1, eIF1A, eIF3, eIF5, and ternary complex (TC). The ternary complex (TC) is a trimeric complex that includes eIF2 (containing subunits, i.e., α, β, and γ), initiator methionyl-tRNA, and GTP (Figure 1, step 1). The recruitment of 43S PIC to the mRNA is enabled by the eIF4F complex (Figure 1, step 3); the eIF4F complex consists of mRNA cap-binding factor eIF4E, a large scaffold factor eIF4G, and the DEAD-box helicase eIF4A. The eIF4F guides the recruitment of 43S PIC onto the mRNA through mRNA cap recognition via eIF4E and eIF4G-eIF3 interactions, forming the 48S initiation complex (Figure 1, step 4). The eIF4A helicase initially interacts with 5’UTR of mRNA and releases mRNA secondary structure, facilitating mRNA scanning by the 43S PIC towards the AUG start codon. The scanning process, which is critical to translation initiation, is stringently controlled by eIF1 and eIF1A; both these factors also facilitate AUG recognition by the 48S PIC; recognition of the start codon triggers the release of eIFs (i.e., initiation factors) and the joining of the 60S ribosomal subunit. In the final step, the formation of translation elongation competent 80S ribosome marks the end of the translation initiation stage and the beginning of the elongation stage (Figure 1).
The most critical step in translation initiation is the selection of the AUG start codon. The efficient translation initiation depends on the context of nucleotides surrounding AUG and the features of the 5’ untranslated region (5’UTR). The canonical context of nucleotides for translation initiation in mammals is KOZAK sequence (A/G)CCAUGG, where purine at position −3 and guanine at position +4 with reference to Adenine at position +1 is essential for stable interaction of mRNA and 48S complex [4,5,6,7]. Besides this, certain other features in 5’UTR of mRNA can influence translation initiation; for instance, 5’ UTR length should be at least 32 nucleotides long for efficient start codon recognition in a consensus context. Any deviations from the consensus context, shortening of 5’UTR length, and presence of additional AUG codons or near-cognate start codons in sub-optimal contexts in 5’UTR may give rise to inefficient translation initiation. For example, the presence of extra AUG in a suboptimal context in 5’UTR and 5’UTR shorter than ~20 nt may give rise to leaky scanning due to stearic clash between the cap complex and 43S complex [6,7], in which 43S PIC bypass the first AUG and translation initiates at the downstream start codon i.e., AUG, leading to formation of truncated proteins or different isoforms of polypeptides [8]. Notably, the secondary structures in the mRNA 5’UTR influence the translation initiation most. These distinct secondary structures include hairpins, long-range interactions, pseudoknots, R-loops, internal ribosomal entry sequences (IRES), and G-quadruplexes. Their presence impacts the initiation process negatively by slowing down the scanning process. The DEAD-box helicase eIF4A is an essential RNA helicase that unwinds these mRNA secondary structures, allowing scanning and thus facilitating translation initiation [9,10]. Most oncogenes, like MYC, RAS, BCL2, PIM1, etc., have secondary structures in their 5’UTR and, therefore, depend on eIF4A for their expression, suggesting a more stringent translation control for their oncogenic expression [11].
Translation dysregulation is considered a hallmark of cancer and is directly linked to abnormal survival, proliferation, angiogenesis, cancer metabolism, and altered immune response [2,12,13,14,15]. Translation control thus maintains the cellular proteome that ultimately regulates the gene expression. Most cancer-related mutations are linked to pathways that merge into translation machinery. Some of the most common oncogenes, like MYC, RAS, and PI3K, and tumor suppressors like TP53 and PTEN, are known to affect translation machinery components [16,17,18,19]. Therefore, it is essential to further deepen our understanding of molecular events of protein synthesis in cancer to develop effective cancer therapeutics. To this end, the mechanism of various translation factors in promoting malignancies and tumorigenesis has triggered wide attention in recent years to target the translation process therapeutically. The promising progress in targeting these factors to develop specific cancer therapeutics is a beacon of hope, providing a therapeutic window for selectively targeting cancer cells. This perspective review delves into the status of translation factors that cause multiple malignancies and the development of small molecules that bind directly to elements of the eukaryotic translational machinery. (see Table 1).

2. Translation Initiation Inhibition

The goal of discovering and developing translation inhibitors is to find mRNA transcript-level selectivity to block the expression of unwanted proteins while maintaining healthy cellular proteome homeostasis. This can be carried out by targeting the specific translational machinery components that differentially regulate protein expression in healthy and disease states or targeting specific mRNA−protein or protein-protein interactions during translation or in translation machinery. Therefore, identifying these targets in translational machinery, which is involved in regulating disease states, is crucial. Targeting these potential therapeutic targets using small molecules, RNA-silencing, or similar state-of-the-art approaches is essential for developing these translation factors as drug candidates with clinical applications.
Since initiation is the essential and highly regulated rate-limiting step, translation initiation factors are the main focus of developing the most targeted therapies targeting translation machinery. The urgency and importance of developing translation inhibitors can be underscored by the attempts to target the components of the eIF4F complex directly [46,67,68,69]. Other translation factors and their interactions have also been targeted since they are dysregulated in multiple malignancies, suggesting their prime role in cell transformation and tumor progression (Table 1). This section discusses individual translation initiation factors, their involvement in cancer onset, and the recent developments toward therapeutically targeting them to develop novel anticancer chemotherapeutic strategies.

2.1. eIF1 and eIF1A

eIF1 and eIF1A facilitate mRNA scanning during translation initiation [8,70]. Ribosomal biochemical and structural analysis has determined that the eIF1 and eIF1A bind 40S ribosomal subunits near P and A sites, respectively, and determine an open 40S conformation that is scanning competent [8]. eIF1 binding to ribosome leads to open conformation, and its dissociation from the pre-initiation complex triggers AUG recognition by the Met-tRNA interface [71,72], stabilizing the ribosomal closed conformation. In the case of eIF1A, the eIF1A C-terminal tail (CTT) enables the ribosomal open conformation. In contrast, closed scanning arrested conformation of the ribosomal 40S subunit is enabled by the N-terminal tail (NTT) of eIF1A. Therefore, the two tails of eIF1A play opposite roles during scanning and AUG recognition [73,74,75]. Additionally, eIF1A interacts with 43S PIC via ribosomal proteins RPS3 and RPS10 located at the ribosomal A site, which undergoes conformational changes during ribosomal subunit joining and 80S ribosome formation [76].
Recently, eIF1 has been implicated in hepatocellular carcinoma, wherein it was found to be overexpressed and linked to poor prognosis [20,21,22]. Targeting eIF1 directly is complicated due to its small size. However, eIF1 has recently been reported to interact with scaffold factor eIF4G1 [77]. The eIF1-eIF4G1 interactions are linked to scanning-dependent mRNA translation and maintaining ER stress response and mTOR-activated gene expression [63]. The eIF1-eIF4G1 complex has been targeted using small-molecule inhibitors, i.e., i14G1-10 and i14G1-12, which are effective in killing multiple cancer cell lines [78]. However, to successfully implement these compounds as anti-cancer therapeutics, further development of these eIF1-eIF4G1 complex inhibitors, i.e., i14G1s, is required.
eIF1A is a 17 KD initiation factor highly conserved among all eukaryotes. eIF1A tails help interact with other initiation factors during the translation initiation process. eIF1A interacts with eIF2, eIF3, and eIF5 through its N-terminal tail (NTT), while the C-terminal tail (CTT) helps interact with eIF5B and ternary complex (GTP-eIF2-initiator tRNA). Somatic mutations in eIF1A are associated with uveal and thyroid cancer and coexist with other mutations like Ras cancer [23,24,25,79]. The most frequent and recurrent hotspots, eIF1A mutations, occur on its exon 2 and intron splice sites. The mutations in exon 2 disrupt the N-terminal tail (NTT) of eIF1A, affecting its ability to promote the closed conformation of pre-initiation complex PIC and preventing leaky scanning [79,80]. On the other hand, 5’ splice site mutations alter the C-strand and C-terminal tail (CTT) domain of eIF1A [24]. Since eIF1A is a small protein, it is difficult to target it alone using small molecules. The best and most promising way to target eIF1A’s activity is perhaps by targeting its interactions with ribosomes and interlocking it in its scanning incompetent state.
Most oncogenic mRNA associated with cell proliferation and cell cycle depend on eIF1A and eIF1 for mRNA scanning and protein expression. Therefore, targeting these translation factors alone or in their interactive complexes provides a platform to block the expression of mRNAs highly dependent on mRNA scanning, suggesting potentially improved cancer therapies for cancer involving upregulation or dependencies on eIF1 and eIF1A.

2.2. eIF4A

eIF4A (eukaryotic initiation factor 4A) is the ATP-dependent DEAD helicase required to unwind the secondary structures in 5’UTR during 43S PIC mRNA scanning [11,81]. eIF4A’s helicase activity undergoes constant cycling between open and closed ribosome conformations. While ATP binding and mRNA induce closed conformation, ATP hydrolysis and mRNA disengagement lead to open conformation. The ratio of eIF4A to the ribosome is 3:1, making it one of the most abundant initiation factors necessary for cap-dependent translation initiation [15,82]. eIF4A has been shown to play a critical role in viral replication and tumor progression, suggesting broader applications of inhibiting eIF4A activity. Most oncogenic mRNA possesses long and highly structured 5’UTR that requires eIF4A for scanning and subsequent translation initiation. Thus, making these oncogenes dependent on eIF4A helicase activity. Some of the most eIF4A-dependent genes include MYC, cyclin D1, PIM1, and BCL2, which lead to tumor survival, excessive cell proliferation, and cell cycle abnormalities [11]. Similarly, eIF4A is required for viral mRNAs with highly structured IRES in their 5’UTR. For example, coronavirus hijacks host eIF4A to translate its highly structured mRNA [83,84]. Similarly, influenza viruses and other positive-strand RNA viruses depend on eIF4A helicase activity [85]. eIF4A expression is higher in proliferating cells [86]. More eIF4A expressions are upregulated in multiple malignancies, including breast, lung, ovarian, endometrial, and cervical cancers [39,40,42,87].
The primary class of eIF4A inhibitors includes rocaglate. Rocaglamide or RocA clamps eIF4A at polypurine sequences with mRNA utilizing ATP, which inhibits the 43S scanning PIC and thus inhibits translation initiation [88,89]. The natural rocaglate silvestrol works by dimerizing eIF4A and RNA, leading to limited loading of eIF4A onto the eIF4F cap complex that, in turn, inhibits global protein translation [90]. Additionally, silvestrol selectively inhibits the translation of mRNA containing G-quadruplex sequence in its 5’UTR, which includes oncogenes like MYC, MDM2, and RUNX1 [9]. Rocaglates, including silvestrol, are highly potent in in-vitro cytotoxicity, but their pharmacokinetics have significant challenges. For example, silvestrol has a short half-life in plasma due to efflux from cell membrane pumps, which limits the clinical development of silvestrol [91,92,93]. Interestingly, the potent cytotoxic activity of eIF4A inhibition led to the development of a cascade of synthetic RocA derivatives, i.e., CR-1-31-B, that binds to eIF4A at Glu195 [94]. Another synthetic rocaglate is zotatifin, or eFT226, which stabilizes eIF4A binding to mRNA at polypurine sequences [95]. It has recently been used for treating breast cancer and implicated in clinical trials in combination with estrogen receptor (ER) inhibitors [96]. Besides being effective in cytotoxicity activity, eIF4A inhibitors have shown antiviral activities. Silvestrol has shown effective against various viruses, including Ebola virus, MERS-CoV (Middle Eastern Respiratory Syndrome coronavirus), and human coronavirus 226E [97,98]. Additionally, many other small molecules are active against eIF4A. A natural marine compound, Peteamine A (PetA), inhibits eIF4A by binding to it irreversibly. PetA induces closed conformation of eIF4A, leading to the loss of its helicase activity [99,100]. Another translation inhibitor associated with eIF4A is hippuristanol, which interacts with the C-terminal domain of eIF4A, blocking its helicase and RNA-dependent ATPase activity [69,101]. Since eIF4A uses ATP hydrolysis to switch between open and closed conformation to release secondary structures in mRNA. This transition is inhibited by hippuristanol, limiting the helicase activity of eIF4A [102].
eIF4A is the most targeted translation factor by small-molecule inhibitors. Although most eIF4A inhibitors have shown potential gene-specific translation inhibition in multiple cancer models, the major setback is the associated cytotoxicity with eIF4A inhibitors, which makes it challenging to develop eIF4A inhibitors as potential drugs. Therefore, making these small molecules more efficient by increasing their on-target efficiency to kill, specifically, cancer cells and limit systemic toxicity is essential for their successful implication as novel and potential chemotherapeutics.

2.3. eIF4E

eIF4E (eukaryotic initiation factor 4E) is an essential factor that binds to mRNA 5’cap [81,103]. It has a concave surface that directs binding to the m7G cap specifically. Regulation of eIF4E is essential to maintain all cap-dependent translation control [67]. eIF4E phosphorylation at S209 by MAP-kinase signal integrating kinases like Mnk1 and Mnk2 enhances its affinity for m7G cap structures, making MnK1/2 inhibitors a target for eIF4E and thus for translation inhibition [68]. On the contrary, eIF4E dephosphorylation causes reduced affinity and thus slows down the global translation and sometimes triggers alternative cap-independent translation initiation [104]. Since eIF4E interacts with small translation repressor proteins, 4E-binding proteins (4E-BPs, i.e., 4E-BP1, 4E-BP2, and 4E-BP3) at the same site that it uses to bind eIF4G [105], therefore, 4E-BPs compete with eIF4G for its binding to eIF4E, thereby inhibiting eIF4F complex assembly. 4E-BPs are phosphorylated upon mTOR activation that dissociates 4EBPs from eIF4E. Upon release, eIF4E associates with eIF4G, thus forming eIF4F complex [67,106,107].
eIF4E is essential for the cap-dependent translation of all nuclear mRNAs, yet there is a subset of mRNAs that are highly dependent on eIF4E activity; these mRNAs are categorized as eIF4E-dependent mRNAs and are involved in cell proliferation, survival, and tumorigenesis. These eIF4E-dependent mRNAs include cyclins, VEGF (vascular endothelial growth factor), PRPS2 (phosphoribosyl-pyrophosphate synthetase 2), MYC, and ODC (ornithine decarboxylase) [108,109,110,111,112]. Hence, eIF4E is one of the most promising targets for translation inhibition. The role of eIF4E in neoplasia is very well studied and established. Not only does eIF4E overexpression exhibit oncogenesis in vitro, but multiple-fold eIF4E overexpression has been documented in various cancers, including breast, lung, leukemias, lymphoma, colon, and head and neck squamous cell carcinoma [28,43,44,45,46,47,48,49,113]. Since eIF4E availability for the eIF4F complex is regulated by 4E-BPs, expression of non-phosphorylated 4E-BP1 mutants has been shown to constitutively bind to eIF4E and suppress cell proliferation and neoplastic growth [114,115,116]. eIF4E has been indirectly targeted through mTOR and MNK inhibition by targeting 4EBPs and eIF4E phosphorylation [68,116,117,118,119,120,121]. However, this discussion focuses on the direct inhibitors of eIF4E developed to target its cap-binding activity using synthetic inhibitors.
Ribavirin is one of the first synthetic nucleosides, initially developed as an antiviral drug, and was one of the first direct inhibitors of eIF4E. Ribavirin was discovered to mimic m7G-cap and compete for direct binding to eIF4E to block eIF4E-cap complex, leading to selective anti-proliferative activity in cancer, e.g., ribavirin selectively inhibits the translation of eIF4E-dependent oncogenes, i.e., BCL2, MYC, MCL1, etc., but it does not affect eIF4E-independent housekeeping genes in acute lymphoblastic leukemia (ALL) [122]. Next, the m7G-cap analogs were developed to inhibit protein synthesis, e.g., 7-BnGMP inhibited m7G-eIF4E cap-binding activity, and to improve its activity, 7Bn-GMP prodrug 4Ei-1 was synthesized as a chemotherapeutic drug [123,124]. One important eIF4E inhibitor is the small-molecule thiazole 4EGI-1, which disrupts interactions between eIF4E and eIF4G1 [125,126]. 4EGI-1 not only inhibited eIF4G1 binding but also stabilized 4E-BP binding, leading to enhanced inhibition of eIF4E activity, which is required for tumor suppression [127]. 4EGI-1 has promising anti-neoplastic properties without systemic toxicity in breast cancer and melanoma xenograft models. It has also been shown to diminish the expression of cyclin D1, BCL2, and MYC oncogenes in non-small-cell cancer cells and glioma cells [128]. Another small molecule that inhibits eIF4E binding to both eIF4G1 and 4E-BP is 4E1RCat and 4E2RCat. Further, 4E1RCat partially prohibits 43S recruitment and thus 80S formation [129,130]. And 4E1RCat was reported to sensitize the doxorubicin-resistant lymphoma mice models in combination therapy with doxorubicin [131]. These attempts to block eIF4E activity alone or in combination with eIF4G have shown promising results and hold the potential to be developed further to eradicate cancer, wherein cap-dependent translation of specific genes plays a critical role. However, ensuring that the eIF4E activity for housekeeping gene expression is not affected while targeting gene-specific translation of oncogenes dependent on eIF4E activity is essential. This might help maintain the targeted effect with the most negligible adverse effects.

2.4. eIF4G1

eIF4G1 is the scaffold protein of the eIF4F complex and facilitates the cap-binding complex and 43S complex assembly to initiate the translation. eIF4G1 associates with eIF4A to activate its helicase activity and direct it to the proximal 5’UTR region [132,133]. Additionally, eIF4G1 possesses RNA binding activity and interacts with eIF4E, which helps tightly bind the eIF4F complex to the 5’cap [3]. eIF4G1 is critical in enhancing the eIF4E affinity for cap structure through allosteric interactions [134]. eIF4G1 actively promotes 43S recruitment by interacting with other initiation factors [135,136]. eIF4G1 actively stimulates the translation of most mRNAs. eIF4G1 is overexpressed and associated with poor prognosis in breast, cervix, lung, and nasopharyngeal carcinomas [50,52]. eIF4G1 interactions have been targeted by translation inhibitor 4EGI-1 to reduce eIF4G1-eIF4E interaction in human melanoma and breast cancer cell lines, showing anti-neoplastic activity, which was also confirmed in mice xenograft models [137]. Recently, as discussed before, novel inhibitors of eIF4G1-eIF1 interactions i14G1-10 and i14G1-12 are reported to inhibit the growth of multiple cancer cell lines in vitro, suggesting enhanced interest in inhibiting translation by inhibiting eIF4G1 activity and interactions with other translation initiation factors [78]. Targeting eIF4G1 could strategically inhibit the expression of not only eIF4E-dependent mRNAs but also scanning-dependent mRNAs by inhibiting its interactions with eIF4E and eIF1.

2.5. eIF2 (α)

eIF2 is the primary factor in forming the ternary complex (TC) consisting of Met-tRNA, which combines with a small ribosomal subunit and forms a 43S pre-initiation complex (PIC). It is a multi-subunit multimeric guanosine triphosphatase comprising eIF2α, eIF2β, and eIF2γ. eIF2 exists in two states, the GTP-bound active and GDP-bound inactive forms, and the GTP/GDP cycle is regulated through reversible phosphorylation of eIF2α (Figure 1; step 1) [103]. eIF2α phosphorylation leads to the inactivation of eIF2, causing translation inhibition and an integrated cellular stress response [138]. Conversely, cellular stress responses like oxidation, heat shock, nutrition deprivation, and hypoxia could lead to phosphorylation of eIF2α [139]. Due to its central role in maintaining cellular stress response, eIF2α is essential for cell survival, proliferation, tumorigenesis, malignancy, and metastasis [139]. One of the ways eIF2α led to cell transformation is by elevating TC levels, leading to uncontrolled translation initiation when mutated [140]. eIF2 has been reported to play a critical role in multiple cancer types like non-small-cell lung carcinoma, melanocytic neoplasm, gastrointestinal, brain, thyroid, lymphoma, and sarcoma [22,26,27,28,29,30,31,32].
eIF2α phosphorylation can promote cell growth or harm cellular fate, depending on the stress response stimulus, duration, and intensity. In mice, eIF2α defected in phosphorylation, leading to cell transformation, whereas its phosphorylation induces apoptosis, suggesting eIF2α phosphorylation as a potential strategy to develop cancer therapeutics [140]. One approach to target and promote eIF2α phosphorylation is through HRI kinase inhibition by BTdCPU, which has shown promising results in both in vitro and in vivo cancer model systems [141,142]. In another strategy, using phosphatase inhibitors such as salubrinal was effective in inhibiting eIF2α dephosphorylation and has shown synthetic lethal phenotype in vitro [143,144]. In early attempts, eIF2 was targeted by TC inhibition using brominated derivatives of fluorescein, NSC119889, and NSC119893, which prevent the binding of Met-tRNAi to eIF2 in vitro [145]. However, the efficacy of direct TC inhibition in vivo has not yet been established.
The implication of eIF2 and TC in cancer makes them potential targets for translation inhibition. However, it is difficult to develop selective inhibitors since both eIF2 and TC are required to translate all nuclear mRNAs. Nevertheless, the careful implication of targeting eIF2α phosphorylation could strategically overcome the risk of global translation inhibition and help develop novel translation inhibitors that can be used for anticancer therapies for eIF2-associated cancer types.

2.6. eIF3 Complex

eIF3 is the largest and most complex translation complex, consisting of 13 subunits, i.e., eIF3a-m, that assemble to constitute the eIF3 complex. eIF3 plays a critical role in all stages of the translation process. eIF3 promotes 43S pre-initiation complex assembly, scanning, and start codon recognition [8,146,147]. Though it is a translation initiation factor, it is implicated in termination and stop codon readthrough [148,149]. eIF3 can interact with IRES of specific viral mRNAs and promote cap-independent translation initiation [150]. Also, eIF3 interacts with 26S proteasome and assists in protein quality control. Abrupt differential expression and amplification of different eIF3 subunits have been implicated in carcinogenesis by increasing cell proliferation, cell cycle progression, and activating signaling pathways involved in cell transformation. Since each eIF3 subunit possesses a differential role in protein synthesis, multiple eIF3 subunits may be used as prognostic markers in the cancers in which they play oncogenic or tumor suppressor roles [151]. Most eIF3 subunits are overexpressed in several cancers except eIF3e and eIF3f. eIF3a is amplified and a prognostic factor in the lung, especially NSCLC (non-small-cell lung cancer). eIF3a is correlated with breast, colon, gastric, and cervical malignancies [34]. eIF3a is associated with bladder and prostate cancer [35]. eIF3d was found to lead to tumor progression in gallbladder cancer [152]. Another subunit, eIF3h, is amplified with MYC in breast cancer [33]. Also, eIF3h has been found to enhance tumorigenicity, invasion, and proliferation in hepatocellular carcinoma via TGF-β and MAPK pathways, making it a prognostic marker for hepatocellular cancer patients [37]. eIF3i overexpressed in multiple colon cancer cell lines, showing its involvement in colon adenocarcinoma [153]. Given the importance of eIF3 subunits in tumorigenesis and neoplasia, attempts have been made to treat them. For example, small molecule mimosine, NCE22, and NCE30 showed cytotoxicity on tumor cells in vitro as the inhibitor of eIF3a, making these compounds candidates for eIF3a regulation with the potential to become anti-cancer agents [154,155]. Similarly, CM16 was reported to target eIF3h and has shown an antiproliferative effect in vitro [156]. eIF3 subunits have versatile functions during translation and protein quality control, making them attractive for developing anti-cancer therapeutics. Yet, they are essential protein subunits for cellular proteome homeostasis, which makes it hard to target them without systemic toxicity. Relevant work targeting eIF3 is underway to develop anti-tumor therapies for clinical studies in different cancers linked with abrupt eIF3 expression.

2.7. eIF6

eIF6 is an essential translation factor required for translation and ribosomal biogenesis. eIF6 is important for anti-association activity; by binding with 60S ribosomal subunit, it prevents premature assembly of 60S and 40S ribosomal subunits [157]. It is partly (~30%) expressed in the nucleus, is associated with pre-ribosomal particles, and is required for 60S ribosomal biogenesis [158]. Multiple studies have reported involvement of eIF6 in tumorigenesis and cancer progression [159,160,161]. Furthermore, eIF6 has been shown to play a vital role as a driver and prognostic marker in multiple cancers, including breast cancer [29], colorectal carcinoma [162], ovarian adenocarcinoma [58], non-small-cell lung cancer [163], lung adenocarcinoma [164], esophageal carcinoma [165], hepatocellular carcinoma, and glioblastoma [166]. Pharmacologic inhibition of eIF6 activity is associated with metabolic rewiring related to hepatocellular carcinoma [167]. eIF6 inhibition is implicated to be effective in RAS mutated and mTOR-sensitive pathways [168,169]. Recently, novel inhibitors of eIF6-60S interactions have been discovered, namely eIFsixty-1 (clofazimine), eIFsixty-4, and eIFsixty-6 [170,171]. These inhibitors have been shown to prohibit cell growth in a dose- and cell-specific manner and affect global translation. eIF6 inhibition using these novel compounds has shown promising results in hepatocellular carcinoma in vitro [171]. eIF6 can thus emerge as another translation factor that could be targeted to improve outcomes in disease states caused by its dysregulation.

3. Translation Elongation Inhibition

Once the formation of 80S elongation-competent ribosome occurs, marking the completion of the translation initiation stage, the peptide synthesis continues when the eukaryotic elongation factor 1A (eEF1A) in its GTP-bound state binds and delivers aminoacyl-tRNAs (aa-tRNAs) to the A-site of the ribosome. The aa-tRNA delivered to the A-site forms a codon-anticodon interaction with the mRNA, followed by GTP hydrolysis, dissociating the eEF1A·GTP complex from the ribosome. In the next step, the A-site and P-site tRNAs are appropriately positioned to undergo peptidyl transfer in the peptidyl transferase center by catalytic bases in the 60S ribosomal subunit. In the following step, elongation factor eEF2, a translocase moves the ribosome along the mRNA to present the next codon so that polypeptide chain elongation can continue. The polypeptide chain is elongated until a stop codon is recognized. Eukaryotic release factors eRF1 and eRF3 are recruited at the stop codon site, which terminates the translation process and releases the new polypeptide chain (Figure 2). Multiple inhibitors that target elongation steps have been identified—most are directed to target eEF1A and ribosomes. Translation elongation inhibition development aims to enhance selective translation inhibition.

3.1. eEF1A

eEF1A is a crucial factor in the translation elongation stage of polypeptide synthesis. Higher vertebrates express two isoforms, eEF1A1 and eEF1A2, respectively. Human eEF1A1 is encoded by chromosome 6q14 and eEF1A2 by 20q13.3 [172]. These isoforms are differentially expressed across the tissues, while eEF1A1 is ubiquitously present in all tissues except the brain, heart, and skeletal muscles, where eEF1A2 is expressed predominantly [173,174]. Multiple malignant tissues express abnormal levels of both eEF1A1 and eEF1A2. Although eEF1A is centric to the elongation step of the translation process, leading to the expression of high-fidelity protein synthesis, it has also been found to be involved in multiple cellular processes, making it a unique translation factor. The cellular process that plays critical roles in signal transduction [175,176,177,178], apoptosis [179,180], cell migration [181], cytoskeletal organization [182], metabolism [183,184,185], neurodevelopment [173,174,186,187,188], post-transcriptional regulation [189], heat-shock response [190], and protein turnover [191,192].
The link between eEF1A and cancer was determined decades ago, with the findings that eEF1A indulgence triggers a transformation [193,194]. Later, eEF1A overexpression was noted in metastasis [195]. Initially, overexpression of eEF1A2 was correlated with anchorage-independent growth and in vivo tumorigenesis in ovarian cancer cell lines, and tumor samples were reported [59,60,64]. Later, many studies reported eEF1A in different cancer types in human cancer cell lines and tumor samples, including breast, lung, liver, prostate, pancreas, thyroid, hematological, kidney, and more (Table 1). Both eEF1A1 and eEF1A2 were reported as prognostic markers for multiple cancers such as renal cell carcinoma [196], pediatric AML [197], and colon adenocarcinoma [198,199]. A recent study used a CRISPR activation screen and reported eEF1A1 as one of the potential biomarkers to predict radiotherapy resistance in lymphoma [200]. All these evident studies highlight eEF1A as an oncogene primarily involved in the elongation step of the mRNA translation process. The oncogenic nature of eEF1A is attributed to its ability to drive malignancy through multiple oncogenic pathways, including PI3K/AKT pathway, mTOR, NF-KB, JAK/STAT, TGF-B, HIF1A, etc. Additionally, eEF1A also has been reported to inhibit tumor suppressor proteins and miRNA. eEF1A negatively impacted chemotherapy-induced apoptosis and p53, leading to chemoresistance to essential anti-cancerous drugs and suggesting strong involvement of eEF1A during anticancer drug resistance.
There have been attempts to target eEF1A using small molecules, i.e., didemnin B and plitidepsin (dehydrodidemin) are cyclic depsipeptide isolated from marine tunicate. The invitro activity of plitidepsin is in the lower nM range in most hematologic cancer cell lines (i.e., leukemia, lymphoma) and solid tumors (i.e., pancreas, breast, lung, and colon). Plitidepsin, in combination with dexamethasone, was approved for treating multiple myeloma in Australia for patients who have failed frontline treatments [201,202,203]. Several potential eEF1A inhibitory compounds have shown potent antiproliferative activity and efficacy in both in vitro and in vivo studies. These compounds include narciclasine, nannocystin, LQ18, and ansatrienin A [204,205,206,207]. Given the role of eEF1A in multiple vital cellular processes and oncogenic mechanisms, targeting eEF1A can significantly impact the development of cancer therapeutics, either alone or in combination therapies, to combat cancer and improve treatment strategies using novel chemotherapeutic approaches.

3.2. eEF2

eEF2 is the second important factor involved in the elongation of the polypeptide chain during translation. eEF2 is the center for translation control at the elongation step under multiple cellular stress conditions. It is overexpressed in gastric and colorectal cancer and related to cell cycle progression by activating AKT and G2/M-associated pathways [66,208]. In addition, eEF2 has also been found to overexpress in multiple other cancers, including lung, breast, prostate, glioblastoma, and hepatocellular cancer [66,209,210,211]. Interestingly, eEF2 was identified as a tumor-associated antigen, and eEF2-derived polypeptides are immunogenic and induce activated CD8+ T cells or cytotoxic T lymphocytes in vitro, suggesting potential adoptive transfer cancer immunotherapeutic for colon cancer [212]. This suggests that a promising role for eEF2 is being developed as a drug target to treat cancer, where it plays a crucial role.

4. Indirect Inhibitors of Translation

Although this review mainly discusses direct inhibitors of translation factors, several indirect translation inhibitors affect the activity of translation factors and ribosomal machinery during translation. Most of these translation inhibitors can be characterized as upstream signaling inhibitors. These include mTOR and MNK signaling pathway inhibitors, which affect eIF4F complex formation [118], and active inhibitors of mTORC1, including rapamycin, rapalogs, torin1, MLN0128, etc. Since mTOR phosphorylates 4E-BPs and allows eIF4F complex formation by releasing eIF4E from 4E-BPs sequestering. These mTOR inhibitors affect the availability of eIF4E and thus block eIF4F complex formation, leading to the direct effect of global translation [115]. On the other hand, MNK inhibitors (i.e., cercosporamide, CGP57380, retinamides, resorcylic acid lactones) inhibit eIF4E phosphorylation and have antiproliferative activity in multiple cancer cell lines [68,119]. Also, MNK inhibitors can help overcome chemotherapeutic resistance to rapamycin, herceptin, and gemcitabine [213,214,215,216]. Some other translation inhibitors that block ribosomal translation and have shown clinical evidence include homoharrigtonine and its analogs [217]. Homoharrigtonine prevents the formation of the first peptide bond and blocks the translation at the initiating ribosome. It has been approved for chronic myeloid leukemia [218]. It should be noted that translation factors can be targeted in combination therapies, where using indirect targets, such as MNK, mTOR, or ribosomes, could lead to synergistic effects and better outcomes in combating tumor progression.

5. Perspective

mRNA translation is a substantial process that has been shown to play a critical role in cancer development. Recently, the development in understanding the role of translation factors in malignancies has provided essential evidence for developing translation inhibitors as promising therapeutics. Since most translation factors show aberrant expression and associated dysregulated translation, they are exceptional novel targets for cancer therapeutic development. Encouragingly, most translation factors in translation initiation and elongation can be targeted using small-molecule inhibitors (Figure 3 and Figure 4). Notably, most translation inhibitors include RNA in binding with their targeting factors, suggesting an essential role of RNA intercalators to be developed and tested towards inhibiting translation; for example, silvestrol targets eIF4A bound to mRNA polypurine sequence. Recent advancement of novel strategies to study translation, including ribosome profiling, cryo-electron microscopy, and single-molecule FRET (fluorescence resonance energy transfer), has played a vital role in the understanding and discovery of translation mechanisms using small-molecule inhibitors, which has eventually opened the door for these small molecules to be developed as potential drugs in cancer therapeutics, as it appears that the use of such small-molecule inhibitors to treat cancer is imminent and holds promise to minimize the problem of acquired resistance to available cancer chemotherapies.
However, transitioning translation inhibitors to the clinic will present new challenges unique to direct translation inhibitors due to associated systemic cytotoxicity. Because most translation factors are essential genes required for normal cellular homeostasis, the imbalance in their expression and activity leads to disease states like cancer development. However, targeting these factors could lead to overall cellular toxicity. With an enhanced understanding of the translation mechanism of each translation factor, it has become easier to develop novel strategies to target them or their interactions, both of which have shown promise in different in vitro and in vivo cancer models. Substantially, small-molecule translation inhibitors targeting different translation factors have demonstrated promising outcomes in preclinical and clinical stages (Table 2.). Interestingly, some translation inhibitors have gained clinical successes, such as Aplidin (Plitidepsin) in combination therapy with dexamethasone in multiple myeloma, and Zotatiffin (eFT226) has shown impressive results in solid tumors clinical trials. These advancements present a new avenue to develop and accelerate small molecules targeting translation. It is also evident that translation factors can be used in novel combination chemotherapeutic approaches by targeting them along with other cellular processes like immune checkpoints, signaling factors, cell cycle, etc. As discussed previously, a significant challenge to overcome in targeting translation machinery is the associated systemic toxicity, off-target activity, and inappropriate plasma half-life, which limits the development of these drug-like molecules in preclinical murine model systems (e.g., CR-31-B) and clinically in patients (e.g., Plitidepsin). These hurdles make it hard for these drugs to be advanced to the clinics. Therefore, it is crucial to determine additional ways to deliver these compounds to the local tumor site in optimal amounts, sufficient for killing cancer cells and eliminating systemic toxicity. The exhaustion of available drug targets in cancer and other disease states underscores the importance of leveraging new cellular mechanisms and pharmacologic modalities such as translation factors. Thus, targeting them could open new avenues for research and development in cancer therapeutics. The available data targeting translation factors suggest a promising strategy to tackle the therapeutic challenges in developing cancer therapeutics and tackle the ongoing quest for drug resistance towards available chemotherapeutic drugs. The upcoming successes of therapeutics that target dysregulated translation in cancer predict their transition from the bench to the bedside well.

Funding

This review research received no external funding. I acknowledge the general support from MSK funding CCSG P30-CA08748.

Data Availability Statement

This study did not create or analyze new data, and data sharing does not apply to this article.

Acknowledgments

The figures were prepared using Biorender and Adobe Illustrator.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Kozak, M. Initiation of translation in prokaryotes and eukaryotes. Gene 1999, 234, 187–208. [Google Scholar] [CrossRef] [PubMed]
  2. Sonenberg, N.; Hinnebusch, A.G. Regulation of translation initiation in eukaryotes: Mechanisms and biological targets. Cell 2009, 136, 731–745. [Google Scholar] [CrossRef] [PubMed]
  3. Hinnebusch, A.G.; Lorsch, J.R. The mechanism of eukaryotic translation initiation: New insights and challenges. Cold Spring Harb. Perspect. Biol. 2012, 4, a011544. [Google Scholar] [CrossRef] [PubMed]
  4. Aitken, C.E.; Lorsch, J.R. A mechanistic overview of translation initiation in eukaryotes. Nat. Struct. Mol. Biol. 2012, 19, 568–576. [Google Scholar] [CrossRef]
  5. Ivanov, I.P.; Loughran, G.; Sachs, M.S.; Atkins, J.F. Initiation context modulates autoregulation of eukaryotic translation initiation factor 1 (eIF1). Proc. Natl. Acad. Sci. USA 2010, 107, 18056–18060. [Google Scholar] [CrossRef]
  6. Kozak, M. Point mutations define a sequence flanking the AUG initiator codon that modulates translation by eukaryotic ribosomes. Cell 1986, 44, 283–292. [Google Scholar] [CrossRef]
  7. Haimov, O.; Sinvani, H.; Dikstein, R. Cap-dependent, scanning-free translation initiation mechanisms. Biochim. Biophys. Acta 2015, 1849, 1313–1318. [Google Scholar] [CrossRef]
  8. Hinnebusch, A.G. The scanning mechanism of eukaryotic translation initiation. Annu. Rev. Biochem. 2014, 83, 779–812. [Google Scholar] [CrossRef]
  9. Wolfe, A.L.; Singh, K.; Zhong, Y.; Drewe, P.; Rajasekhar, V.K.; Sanghvi, V.R.; Mavrakis, K.J.; Jiang, M.; Roderick, J.E.; Van der Meulen, J.; et al. RNA G-quadruplexes cause eIF4A-dependent oncogene translation in cancer. Nature 2014, 513, 65–70. [Google Scholar] [CrossRef]
  10. Georgakopoulos-Soares, I.; Parada, G.E.; Hemberg, M. Secondary structures in RNA synthesis, splicing and translation. Comput. Struct. Biotechnol. J. 2022, 20, 2871–2884. [Google Scholar] [CrossRef]
  11. Raza, F.; Waldron, J.A.; Quesne, J.L. Translational dysregulation in cancer: eIF4A isoforms and sequence determinants of eIF4A dependence. Biochem. Soc. Trans. 2015, 43, 1227–1233. [Google Scholar] [CrossRef] [PubMed]
  12. Polunovsky, V.A.; Rosenwald, I.B.; Tan, A.T.; White, J.; Chiang, L.; Sonenberg, N.; Bitterman, P.B. Translational control of programmed cell death: Eukaryotic translation initiation factor 4E blocks apoptosis in growth-factor-restricted fibroblasts with physiologically expressed or deregulated Myc. Mol. Cell Biol. 1996, 16, 6573–6581. [Google Scholar] [CrossRef] [PubMed]
  13. Holland, E.C.; Sonenberg, N.; Pandolfi, P.P.; Thomas, G. Signaling control of mRNA translation in cancer pathogenesis. Oncogene 2004, 23, 3138–3144. [Google Scholar] [CrossRef] [PubMed]
  14. Alboushi, L.; Hackett, A.P.; Naeli, P.; Bakhti, M.; Jafarnejad, S.M. Multifaceted control of mRNA translation machinery in cancer. Cell Signal 2021, 84, 110037. [Google Scholar] [CrossRef] [PubMed]
  15. Chu, J.; Cargnello, M.; Topisirovic, I.; Pelletier, J. Translation Initiation Factors: Reprogramming Protein Synthesis in Cancer. Trends Cell Biol. 2016, 26, 918–933. [Google Scholar] [CrossRef]
  16. Burns, D.M.; Richter, J.D. CPEB regulation of human cellular senescence, energy metabolism, and p53 mRNA translation. Genes Dev. 2008, 22, 3449–3460. [Google Scholar] [CrossRef]
  17. Topisirovic, I.; Sonenberg, N. mRNA translation and energy metabolism in cancer: The role of the MAPK and mTORC1 pathways. Cold Spring Harb. Symp. Quant. Biol. 2011, 76, 355–367. [Google Scholar] [CrossRef]
  18. Ravitz, M.J.; Chen, L.; Lynch, M.; Schmidt, E.V. c-myc Repression of TSC2 contributes to control of translation initiation and Myc-induced transformation. Cancer Res. 2007, 67, 11209–11217. [Google Scholar] [CrossRef]
  19. Liu, L.; Cash, T.P.; Jones, R.G.; Keith, B.; Thompson, C.B.; Simon, M.C. Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol. Cell 2006, 21, 521–531. [Google Scholar] [CrossRef]
  20. Chen, Y.; Zhou, Y.; Qiu, S.; Wang, K.; Liu, S.; Peng, X.X.; Li, J.; Tan, E.M.; Zhang, J.Y. Autoantibodies to tumor-associated antigens combined with abnormal alpha-fetoprotein enhance immunodiagnosis of hepatocellular carcinoma. Cancer Lett. 2010, 289, 32–39. [Google Scholar] [CrossRef]
  21. Lian, Z.; Pan, J.; Liu, J.; Zhang, S.; Zhu, M.; Arbuthnot, P.; Kew, M.; Feitelson, M.A. The translation initiation factor, hu-Sui1 may be a target of hepatitis B X antigen in hepatocarcinogenesis. Oncogene 1999, 18, 1677–1687. [Google Scholar] [CrossRef] [PubMed]
  22. Golob-Schwarzl, N.; Puchas, P.; Gogg-Kamerer, M.; Weichert, W.; Goppert, B.; Haybaeck, J. New Pancreatic Cancer Biomarkers eIF1, eIF2D, eIF3C and eIF6 Play a Major Role in Translational Control in Ductal Adenocarcinoma. Anticancer. Res. 2020, 40, 3109–3118. [Google Scholar] [CrossRef] [PubMed]
  23. Ewens, K.G.; Kanetsky, P.A.; Richards-Yutz, J.; Purrazzella, J.; Shields, C.L.; Ganguly, T.; Ganguly, A. Chromosome 3 status combined with BAP1 and EIF1AX mutation profiles are associated with metastasis in uveal melanoma. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5160–5167. [Google Scholar] [CrossRef] [PubMed]
  24. Karunamurthy, A.; Panebianco, F.; Hsiao, S.J.; Vorhauer, J.; Nikiforova, M.N.; Chiosea, S.; Nikiforov, Y.E. Prevalence and phenotypic correlations of EIF1AX mutations in thyroid nodules. Endocr. Relat. Cancer 2016, 23, 295–301. [Google Scholar] [CrossRef]
  25. Etemadmoghadam, D.; Azar, W.J.; Lei, Y.; Moujaber, T.; Garsed, D.W.; Kennedy, C.J.; Fereday, S.; Mitchell, C.; Chiew, Y.E.; Hendley, J.; et al. EIF1AX and NRAS Mutations Co-occur and Cooperate in Low-Grade Serous Ovarian Carcinomas. Cancer Res. 2017, 77, 4268–4278. [Google Scholar] [CrossRef]
  26. He, Y.; Correa, A.M.; Raso, M.G.; Hofstetter, W.L.; Fang, B.; Behrens, C.; Roth, J.A.; Zhou, Y.; Yu, L.; Wistuba, I.I.; et al. The role of PKR/eIF2alpha signaling pathway in prognosis of non-small cell lung cancer. PLoS ONE 2011, 6, e24855. [Google Scholar] [CrossRef]
  27. Lobo, M.V.; Martin, M.E.; Perez, M.I.; Alonso, F.J.; Redondo, C.; Alvarez, M.I.; Salinas, M. Levels, phosphorylation status and cellular localization of translational factor eIF2 in gastrointestinal carcinomas. Histochem. J. 2000, 32, 139–150. [Google Scholar] [CrossRef]
  28. Tejada, S.; Lobo, M.V.; Garcia-Villanueva, M.; Sacristan, S.; Perez-Morgado, M.I.; Salinas, M.; Martin, M.E. Eukaryotic initiation factors (eIF) 2alpha and 4E expression, localization, and phosphorylation in brain tumors. J. Histochem. Cytochem. 2009, 57, 503–512. [Google Scholar] [CrossRef] [PubMed]
  29. Gatza, M.L.; Silva, G.O.; Parker, J.S.; Fan, C.; Perou, C.M. An integrated genomics approach identifies drivers of proliferation in luminal-subtype human breast cancer. Nat. Genet. 2014, 46, 1051–1059. [Google Scholar] [CrossRef]
  30. Rosenwald, I.B.; Koifman, L.; Savas, L.; Chen, J.J.; Woda, B.A.; Kadin, M.E. Expression of the translation initiation factors eIF-4E and eIF-2* is frequently increased in neoplastic cells of Hodgkin lymphoma. Hum. Pathol. 2008, 39, 910–916. [Google Scholar] [CrossRef]
  31. Lee, S.; Jee, H.Y.; Lee, Y.G.; Shin, J.I.; Jeon, Y.J.; Kim, J.B.; Seo, H.E.; Lee, J.Y.; Lee, K. PKR-Mediated Phosphorylation of eIF2a and CHK1 Is Associated with Doxorubicin-Mediated Apoptosis in HCC1143 Triple-Negative Breast Cancer Cells. Int. J. Mol. Sci. 2022, 23, 15872. [Google Scholar] [CrossRef] [PubMed]
  32. Tanaka, I.; Sato, M.; Kato, T.; Goto, D.; Kakumu, T.; Miyazawa, A.; Yogo, N.; Hase, T.; Morise, M.; Sekido, Y.; et al. eIF2beta, a subunit of translation-initiation factor EIF2, is a potential therapeutic target for non-small cell lung cancer. Cancer Sci. 2018, 109, 1843–1852. [Google Scholar] [CrossRef] [PubMed]
  33. Nupponen, N.N.; Porkka, K.; Kakkola, L.; Tanner, M.; Persson, K.; Borg, A.; Isola, J.; Visakorpi, T. Amplification and overexpression of p40 subunit of eukaryotic translation initiation factor 3 in breast and prostate cancer. Am. J. Pathol. 1999, 154, 1777–1783. [Google Scholar] [CrossRef] [PubMed]
  34. Pincheira, R.; Chen, Q.; Zhang, J.T. Identification of a 170-kDa protein over-expressed in lung cancers. Br. J. Cancer 2001, 84, 1520–1527. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, H.; Ru, Y.; Sanchez-Carbayo, M.; Wang, X.; Kieft, J.S.; Theodorescu, D. Translation initiation factor eIF3b expression in human cancer and its role in tumor growth and lung colonization. Clin. Cancer Res. 2013, 19, 2850–2860. [Google Scholar] [CrossRef]
  36. Shen, J.; Yin, J.Y.; Li, X.P.; Liu, Z.Q.; Wang, Y.; Chen, J.; Qu, J.; Xu, X.J.; McLeod, H.L.; He, Y.J.; et al. The prognostic value of altered eIF3a and its association with p27 in non-small cell lung cancers. PLoS ONE 2014, 9, e96008. [Google Scholar] [CrossRef]
  37. Zhu, Q.; Qiao, G.L.; Zeng, X.C.; Li, Y.; Yan, J.J.; Duan, R.; Du, Z.Y. Elevated expression of eukaryotic translation initiation factor 3H is associated with proliferation, invasion and tumorigenicity in human hepatocellular carcinoma. Oncotarget 2016, 7, 49888–49901. [Google Scholar] [CrossRef]
  38. Shi, J.; Kahle, A.; Hershey, J.W.; Honchak, B.M.; Warneke, J.A.; Leong, S.P.; Nelson, M.A. Decreased expression of eukaryotic initiation factor 3f deregulates translation and apoptosis in tumor cells. Oncogene 2006, 25, 4923–4936. [Google Scholar] [CrossRef]
  39. Liang, S.; Zhou, Y.; Chen, Y.; Ke, G.; Wen, H.; Wu, X. Decreased expression of EIF4A1 after preoperative brachytherapy predicts better tumor-specific survival in cervical cancer. Int. J. Gynecol. Cancer 2014, 24, 908–915. [Google Scholar] [CrossRef]
  40. Modelska, A.; Turro, E.; Russell, R.; Beaton, J.; Sbarrato, T.; Spriggs, K.; Miller, J.; Graf, S.; Provenzano, E.; Blows, F.; et al. The malignant phenotype in breast cancer is driven by eIF4A1-mediated changes in the translational landscape. Cell Death Dis. 2015, 6, e1603. [Google Scholar] [CrossRef]
  41. Kong, T.; Xue, Y.; Cencic, R.; Zhu, X.; Monast, A.; Fu, Z.; Pilon, V.; Sangwan, V.; Guiot, M.C.; Foulkes, W.D.; et al. eIF4A Inhibitors Suppress Cell-Cycle Feedback Response and Acquired Resistance to CDK4/6 Inhibition in Cancer. Mol. Cancer Ther. 2019, 18, 2158–2170. [Google Scholar] [CrossRef] [PubMed]
  42. Casacuberta-Serra, S.; Gonzalez-Larreategui, I.; Soucek, L. eIF4A dependency: The hidden key to unlock KRAS mutant non-small cell lung cancer’s vulnerability. Transl. Lung Cancer Res. 2023, 12, 2570–2575. [Google Scholar] [CrossRef] [PubMed]
  43. Nathan, C.O.; Liu, L.; Li, B.D.; Abreo, F.W.; Nandy, I.; De Benedetti, A. Detection of the proto-oncogene eIF4E in surgical margins may predict recurrence in head and neck cancer. Oncogene 1997, 15, 579–584. [Google Scholar] [CrossRef] [PubMed]
  44. Berkel, H.J.; Turbat-Herrera, E.A.; Shi, R.; de Benedetti, A. Expression of the translation initiation factor eIF4E in the polyp-cancer sequence in the colon. Cancer Epidemiol. Biomark. Prev. 2001, 10, 663–666. [Google Scholar]
  45. DeFatta, R.J.; Turbat-Herrera, E.A.; Li, B.D.; Anderson, W.; De Benedetti, A. Elevated expression of eIF4E in confined early breast cancer lesions: Possible role of hypoxia. Int. J. Cancer 1999, 80, 516–522. [Google Scholar] [CrossRef]
  46. Borden, K.L. Tissue targeting in cancer: eIF4E’s tale. Clin. Cancer Res. 2009, 15, 4254–4255. [Google Scholar] [CrossRef] [PubMed]
  47. Zheng, J.; Li, X.; Zhang, C.; Zhang, Y. eIF4E Overexpression Is Associated with Poor Prognoses of Ovarian Cancer. Anal. Cell Pathol. 2020, 2020, 8984526. [Google Scholar] [CrossRef]
  48. Li, F.; Sun, H.; Li, Y.; Bai, X.; Dong, X.; Zhao, N.; Meng, J.; Sun, B.; Zhang, D. High expression of eIF4E is associated with tumor macrophage infiltration and leads to poor prognosis in breast cancer. BMC Cancer 2021, 21, 1305. [Google Scholar] [CrossRef]
  49. Pisera, A.; Campo, A.; Campo, S. Structure and functions of the translation initiation factor eIF4E and its role in cancer development and treatment. J. Genet. Genom. 2018, 45, 13–24. [Google Scholar] [CrossRef]
  50. Bauer, C.; Brass, N.; Diesinger, I.; Kayser, K.; Grasser, F.A.; Meese, E. Overexpression of the eukaryotic translation initiation factor 4G (eIF4G-1) in squamous cell lung carcinoma. Int. J. Cancer 2002, 98, 181–185. [Google Scholar] [CrossRef]
  51. Harris, M.N.; Ozpolat, B.; Abdi, F.; Gu, S.; Legler, A.; Mawuenyega, K.G.; Tirado-Gomez, M.; Lopez-Berestein, G.; Chen, X. Comparative proteomic analysis of all-trans-retinoic acid treatment reveals systematic posttranscriptional control mechanisms in acute promyelocytic leukemia. Blood 2004, 104, 1314–1323. [Google Scholar] [CrossRef]
  52. Silvera, D.; Arju, R.; Darvishian, F.; Levine, P.H.; Zolfaghari, L.; Goldberg, J.; Hochman, T.; Formenti, S.C.; Schneider, R.J. Essential role for eIF4GI overexpression in the pathogenesis of inflammatory breast cancer. Nat. Cell Biol. 2009, 11, 903–908. [Google Scholar] [CrossRef]
  53. Tu, L.; Liu, Z.; He, X.; He, Y.; Yang, H.; Jiang, Q.; Xie, S.; Xiao, G.; Li, X.; Yao, K.; et al. Over-expression of eukaryotic translation initiation factor 4 gamma 1 correlates with tumor progression and poor prognosis in nasopharyngeal carcinoma. Mol. Cancer 2010, 9, 78. [Google Scholar] [CrossRef] [PubMed]
  54. Shek, F.H.; Fatima, S.; Lee, N.P. Implications of the Use of Eukaryotic Translation Initiation Factor 5A (eIF5A) for Prognosis and Treatment of Hepatocellular Carcinoma. Int. J. Hepatol. 2012, 2012, 760928. [Google Scholar] [CrossRef]
  55. Cracchiolo, B.M.; Heller, D.S.; Clement, P.M.; Wolff, E.C.; Park, M.H.; Hanauske-Abel, H.M. Eukaryotic initiation factor 5A-1 (eIF5A-1) as a diagnostic marker for aberrant proliferation in intraepithelial neoplasia of the vulva. Gynecol. Oncol. 2004, 94, 217–222. [Google Scholar] [CrossRef]
  56. Cai, S.X.; Chen, W.S.; Zeng, W.; Cheng, X.F.; Lin, M.B.; Wang, J.S. Roles of HDAC2, eIF5, and eIF6 in Lung Cancer Tumorigenesis. Curr. Med. Sci. 2021, 41, 764–769. [Google Scholar] [CrossRef]
  57. Preukschas, M.; Hagel, C.; Schulte, A.; Weber, K.; Lamszus, K.; Sievert, H.; Pallmann, N.; Bokemeyer, C.; Hauber, J.; Braig, M.; et al. Expression of eukaryotic initiation factor 5A and hypusine forming enzymes in glioblastoma patient samples: Implications for new targeted therapies. PLoS ONE 2012, 7, e43468. [Google Scholar] [CrossRef] [PubMed]
  58. Flavin, R.J.; Smyth, P.C.; Finn, S.P.; Laios, A.; O’Toole, S.A.; Barrett, C.; Ring, M.; Denning, K.M.; Li, J.; Aherne, S.T.; et al. Altered eIF6 and Dicer expression is associated with clinicopathological features in ovarian serous carcinoma patients. Mod. Pathol. 2008, 21, 676–684. [Google Scholar] [CrossRef] [PubMed]
  59. Anand, N.; Murthy, S.; Amann, G.; Wernick, M.; Porter, L.A.; Cukier, I.H.; Collins, C.; Gray, J.W.; Diebold, J.; Demetrick, D.J.; et al. Protein elongation factor EEF1A2 is a putative oncogene in ovarian cancer. Nat. Genet. 2002, 31, 301–305. [Google Scholar] [CrossRef]
  60. Lee, J.M. The role of protein elongation factor eEF1A2 in ovarian cancer. Reprod. Biol. Endocrinol. 2003, 1, 69. [Google Scholar] [CrossRef]
  61. Dapas, B.; Tell, G.; Scaloni, A.; Pines, A.; Ferrara, L.; Quadrifoglio, F.; Scaggiante, B. Identification of different isoforms of eEF1A in the nuclear fraction of human T-lymphoblastic cancer cell line specifically binding to aptameric cytotoxic GT oligomers. Eur. J. Biochem. 2003, 270, 3251–3262. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, L.; Madura, K. Increased proteasome activity, ubiquitin-conjugating enzymes, and eEF1A translation factor detected in breast cancer tissue. Cancer Res. 2005, 65, 5599–5606. [Google Scholar] [CrossRef]
  63. Scaggiante, B.; Dapas, B.; Grassi, G.; Manzini, G. Interaction of G-rich GT oligonucleotides with nuclear-associated eEF1A is correlated with their antiproliferative effect in haematopoietic human cancer cell lines. FEBS J. 2006, 273, 1350–1361. [Google Scholar] [CrossRef] [PubMed]
  64. Pinke, D.E.; Kalloger, S.E.; Francetic, T.; Huntsman, D.G.; Lee, J.M. The prognostic significance of elongation factor eEF1A2 in ovarian cancer. Gynecol. Oncol. 2008, 108, 561–568. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, W.; Wang, J.; Shan, C. The eEF1A protein in cancer: Clinical significance, oncogenic mechanisms, and targeted therapeutic strategies. Pharmacol. Res. 2024, 204, 107195. [Google Scholar] [CrossRef]
  66. Oji, Y.; Tatsumi, N.; Fukuda, M.; Nakatsuka, S.; Aoyagi, S.; Hirata, E.; Nanchi, I.; Fujiki, F.; Nakajima, H.; Yamamoto, Y.; et al. The translation elongation factor eEF2 is a novel tumor-associated antigen overexpressed in various types of cancers. Int. J. Oncol. 2014, 44, 1461–1469. [Google Scholar] [CrossRef] [PubMed]
  67. Richter, J.D.; Sonenberg, N. Regulation of cap-dependent translation by eIF4E inhibitory proteins. Nature 2005, 433, 477–480. [Google Scholar] [CrossRef]
  68. Silva, R.L.; Wendel, H.G. MNK, EIF4E and targeting translation for therapy. Cell Cycle 2008, 7, 553–555. [Google Scholar] [CrossRef] [PubMed]
  69. Lindqvist, L.; Oberer, M.; Reibarkh, M.; Cencic, R.; Bordeleau, M.E.; Vogt, E.; Marintchev, A.; Tanaka, J.; Fagotto, F.; Altmann, M.; et al. Selective pharmacological targeting of a DEAD box RNA helicase. PLoS ONE 2008, 3, e1583. [Google Scholar] [CrossRef]
  70. Jackson, R.J.; Hellen, C.U.; Pestova, T.V. The mechanism of eukaryotic translation initiation and principles of its regulation. Nat. Rev. Mol. Cell Biol. 2010, 11, 113–127. [Google Scholar] [CrossRef]
  71. Maag, D.; Fekete, C.A.; Gryczynski, Z.; Lorsch, J.R. A conformational change in the eukaryotic translation preinitiation complex and release of eIF1 signal recognition of the start codon. Mol. Cell 2005, 17, 265–275. [Google Scholar] [CrossRef] [PubMed]
  72. Passmore, L.A.; Schmeing, T.M.; Maag, D.; Applefield, D.J.; Acker, M.G.; Algire, M.A.; Lorsch, J.R.; Ramakrishnan, V. The eukaryotic translation initiation factors eIF1 and eIF1A induce an open conformation of the 40S ribosome. Mol. Cell 2007, 26, 41–50. [Google Scholar] [CrossRef] [PubMed]
  73. Llacer, J.L.; Hussain, T.; Marler, L.; Aitken, C.E.; Thakur, A.; Lorsch, J.R.; Hinnebusch, A.G.; Ramakrishnan, V. Conformational Differences between Open and Closed States of the Eukaryotic Translation Initiation Complex. Mol. Cell 2015, 59, 399–412. [Google Scholar] [CrossRef] [PubMed]
  74. Fekete, C.A.; Mitchell, S.F.; Cherkasova, V.A.; Applefield, D.; Algire, M.A.; Maag, D.; Saini, A.K.; Lorsch, J.R.; Hinnebusch, A.G. N- and C-terminal residues of eIF1A have opposing effects on the fidelity of start codon selection. EMBO J. 2007, 26, 1602–1614. [Google Scholar] [CrossRef] [PubMed]
  75. Fekete, C.A.; Applefield, D.J.; Blakely, S.A.; Shirokikh, N.; Pestova, T.; Lorsch, J.R.; Hinnebusch, A.G. The eIF1A C-terminal domain promotes initiation complex assembly, scanning and AUG selection in vivo. EMBO J. 2005, 24, 3588–3601. [Google Scholar] [CrossRef]
  76. Haimov, O.; Sinvani, H.; Martin, F.; Ulitsky, I.; Emmanuel, R.; Tamarkin-Ben-Harush, A.; Vardy, A.; Dikstein, R. Efficient and Accurate Translation Initiation Directed by TISU Involves RPS3 and RPS10e Binding and Differential Eukaryotic Initiation Factor 1A Regulation. Mol. Cell Biol. 2017, 37, e00150-17. [Google Scholar] [CrossRef]
  77. Haimov, O.; Sehrawat, U.; Tamarkin-Ben Harush, A.; Bahat, A.; Uzonyi, A.; Will, A.; Hiraishi, H.; Asano, K.; Dikstein, R. Dynamic Interaction of Eukaryotic Initiation Factor 4G1 (eIF4G1) with eIF4E and eIF1 Underlies Scanning-Dependent and -Independent Translation. Mol. Cell Biol. 2018, 38, e00139-18. [Google Scholar] [CrossRef]
  78. Sehrawat, U.; Haimov, O.; Weiss, B.; Tamarkin-Ben Harush, A.; Ashkenazi, S.; Plotnikov, A.; Noiman, T.; Leshkowitz, D.; Stelzer, G.; Dikstein, R. Inhibitors of eIF4G1-eIF1 uncover its regulatory role of ER/UPR stress-response genes independent of eIF2alpha-phosphorylation. Proc. Natl. Acad. Sci. USA 2022, 119, e2120339119. [Google Scholar] [CrossRef]
  79. Martin-Marcos, P.; Zhou, F.; Karunasiri, C.; Zhang, F.; Dong, J.; Nanda, J.; Kulkarni, S.D.; Sen, N.D.; Tamame, M.; Zeschnigk, M.; et al. eIF1A residues implicated in cancer stabilize translation preinitiation complexes and favor suboptimal initiation sites in yeast. Elife 2017, 6, e31250. [Google Scholar] [CrossRef]
  80. Sehrawat, U.; Koning, F.; Ashkenazi, S.; Stelzer, G.; Leshkowitz, D.; Dikstein, R. Cancer-Associated Eukaryotic Translation Initiation Factor 1A Mutants Impair Rps3 and Rps10 Binding and Enhance Scanning of Cell Cycle Genes. Mol. Cell Biol. 2019, 39, e00441-18. [Google Scholar] [CrossRef]
  81. Pelletier, J.; Sonenberg, N. The Organizing Principles of Eukaryotic Ribosome Recruitment. Annu. Rev. Biochem. 2019, 88, 307–335. [Google Scholar] [CrossRef]
  82. Bhat, M.; Robichaud, N.; Hulea, L.; Sonenberg, N.; Pelletier, J.; Topisirovic, I. Targeting the translation machinery in cancer. Nat. Rev. Drug Discov. 2015, 14, 261–278. [Google Scholar] [CrossRef] [PubMed]
  83. Taroncher-Oldenburg, G.; Muller, C.; Obermann, W.; Ziebuhr, J.; Hartmann, R.K.; Grunweller, A. Targeting the DEAD-Box RNA Helicase eIF4A with Rocaglates-A Pan-Antiviral Strategy for Minimizing the Impact of Future RNA Virus Pandemics. Microorganisms 2021, 9, 540. [Google Scholar] [CrossRef] [PubMed]
  84. Madhugiri, R.; Fricke, M.; Marz, M.; Ziebuhr, J. RNA structure analysis of alphacoronavirus terminal genome regions. Virus Res. 2014, 194, 76–89. [Google Scholar] [CrossRef] [PubMed]
  85. Montero, H.; Perez-Gil, G.; Sampieri, C.L. Eukaryotic initiation factor 4A (eIF4A) during viral infections. Virus Genes 2019, 55, 267–273. [Google Scholar] [CrossRef]
  86. Williams-Hill, D.M.; Duncan, R.F.; Nielsen, P.J.; Tahara, S.M. Differential expression of the murine eukaryotic translation initiation factor isogenes eIF4A(I) and eIF4A(II) is dependent upon cellular growth status. Arch. Biochem. Biophys. 1997, 338, 111–120. [Google Scholar] [CrossRef] [PubMed]
  87. Lomnytska, M.I.; Becker, S.; Gemoll, T.; Lundgren, C.; Habermann, J.; Olsson, A.; Bodin, I.; Engstrom, U.; Hellman, U.; Hellman, K.; et al. Impact of genomic stability on protein expression in endometrioid endometrial cancer. Br. J. Cancer 2012, 106, 1297–1305. [Google Scholar] [CrossRef]
  88. Iwasaki, S.; Floor, S.N.; Ingolia, N.T. Rocaglates convert DEAD-box protein eIF4A into a sequence-selective translational repressor. Nature 2016, 534, 558–561. [Google Scholar] [CrossRef]
  89. Iwasaki, S.; Iwasaki, W.; Takahashi, M.; Sakamoto, A.; Watanabe, C.; Shichino, Y.; Floor, S.N.; Fujiwara, K.; Mito, M.; Dodo, K.; et al. The Translation Inhibitor Rocaglamide Targets a Bimolecular Cavity between eIF4A and Polypurine RNA. Mol. Cell 2019, 73, 738–748.e739. [Google Scholar] [CrossRef]
  90. Cencic, R.; Carrier, M.; Galicia-Vazquez, G.; Bordeleau, M.E.; Sukarieh, R.; Bourdeau, A.; Brem, B.; Teodoro, J.G.; Greger, H.; Tremblay, M.L.; et al. Antitumor activity and mechanism of action of the cyclopenta[b]benzofuran, silvestrol. PLoS ONE 2009, 4, e5223. [Google Scholar] [CrossRef]
  91. Gupta, S.V.; Sass, E.J.; Davis, M.E.; Edwards, R.B.; Lozanski, G.; Heerema, N.A.; Lehman, A.; Zhang, X.; Jarjoura, D.; Byrd, J.C.; et al. Resistance to the translation initiation inhibitor silvestrol is mediated by ABCB1/P-glycoprotein overexpression in acute lymphoblastic leukemia cells. AAPS J. 2011, 13, 357–364. [Google Scholar] [CrossRef] [PubMed]
  92. Saradhi, U.V.; Gupta, S.V.; Chiu, M.; Wang, J.; Ling, Y.; Liu, Z.; Newman, D.J.; Covey, J.M.; Kinghorn, A.D.; Marcucci, G.; et al. Characterization of silvestrol pharmacokinetics in mice using liquid chromatography-tandem mass spectrometry. AAPS J. 2011, 13, 347–356. [Google Scholar] [CrossRef] [PubMed]
  93. Cunningham, T.A.; Chapman, E.; Schatz, J.H. eIF4A inhibition: Ready for primetime? Oncotarget 2018, 9, 35515–35516. [Google Scholar] [CrossRef] [PubMed]
  94. Chu, J.; Zhang, W.; Cencic, R.; Devine, W.G.; Beglov, D.; Henkel, T.; Brown, L.E.; Vajda, S.; Porco, J.A., Jr.; Pelletier, J. Amidino-Rocaglates: A Potent Class of eIF4A Inhibitors. Cell Chem. Biol. 2019, 26, 1586–1593.e1583. [Google Scholar] [CrossRef]
  95. Ernst, J.T.; Thompson, P.A.; Nilewski, C.; Sprengeler, P.A.; Sperry, S.; Packard, G.; Michels, T.; Xiang, A.; Tran, C.; Wegerski, C.J.; et al. Design of Development Candidate eFT226, a First in Class Inhibitor of Eukaryotic Initiation Factor 4A RNA Helicase. J. Med. Chem. 2020, 63, 5879–5955. [Google Scholar] [CrossRef] [PubMed]
  96. Gerson-Gurwitz, A.; Young, N.P.; Goel, V.K.; Eam, B.; Stumpf, C.R.; Chen, J.; Fish, S.; Barrera, M.; Sung, E.; Staunton, J.; et al. Zotatifin, an eIF4A-Selective Inhibitor, Blocks Tumor Growth in Receptor Tyrosine Kinase Driven Tumors. Front. Oncol. 2021, 11, 766298. [Google Scholar] [CrossRef]
  97. Biedenkopf, N.; Lange-Grunweller, K.; Schulte, F.W.; Weisser, A.; Muller, C.; Becker, D.; Becker, S.; Hartmann, R.K.; Grunweller, A. The natural compound silvestrol is a potent inhibitor of Ebola virus replication. Antivir. Res. 2017, 137, 76–81. [Google Scholar] [CrossRef]
  98. Muller, C.; Schulte, F.W.; Lange-Grunweller, K.; Obermann, W.; Madhugiri, R.; Pleschka, S.; Ziebuhr, J.; Hartmann, R.K.; Grunweller, A. Broad-spectrum antiviral activity of the eIF4A inhibitor silvestrol against corona- and picornaviruses. Antivir. Res. 2018, 150, 123–129. [Google Scholar] [CrossRef]
  99. Low, W.K.; Dang, Y.; Bhat, S.; Romo, D.; Liu, J.O. Substrate-dependent targeting of eukaryotic translation initiation factor 4A by pateamine A: Negation of domain-linker regulation of activity. Chem. Biol. 2007, 14, 715–727. [Google Scholar] [CrossRef]
  100. Bordeleau, M.E.; Matthews, J.; Wojnar, J.M.; Lindqvist, L.; Novac, O.; Jankowsky, E.; Sonenberg, N.; Northcote, P.; Teesdale-Spittle, P.; Pelletier, J. Stimulation of mammalian translation initiation factor eIF4A activity by a small molecule inhibitor of eukaryotic translation. Proc. Natl. Acad. Sci. USA 2005, 102, 10460–10465. [Google Scholar] [CrossRef]
  101. Somaiah, R.; Ravindar, K.; Cencic, R.; Pelletier, J.; Deslongchamps, P. Synthesis of the antiproliferative agent hippuristanol and its analogues from hydrocortisone via Hg(II)-catalyzed spiroketalization: Structure-activity relationship. J. Med. Chem. 2014, 57, 2511–2523. [Google Scholar] [CrossRef] [PubMed]
  102. Sun, Y.; Atas, E.; Lindqvist, L.M.; Sonenberg, N.; Pelletier, J.; Meller, A. Single-molecule kinetics of the eukaryotic initiation factor 4AI upon RNA unwinding. Structure 2014, 22, 941–948. [Google Scholar] [CrossRef]
  103. Sonenberg, N.; Dever, T.E. Eukaryotic translation initiation factors and regulators. Curr. Opin. Struct. Biol. 2003, 13, 56–63. [Google Scholar] [CrossRef] [PubMed]
  104. Dyer, J.R.; Michel, S.; Lee, W.; Castellucci, V.F.; Wayne, N.L.; Sossin, W.S. An activity-dependent switch to cap-independent translation triggered by eIF4E dephosphorylation. Nat. Neurosci. 2003, 6, 219–220. [Google Scholar] [CrossRef] [PubMed]
  105. Pause, A.; Belsham, G.J.; Gingras, A.C.; Donze, O.; Lin, T.A.; Lawrence, J.C., Jr.; Sonenberg, N. Insulin-dependent stimulation of protein synthesis by phosphorylation of a regulator of 5′-cap function. Nature 1994, 371, 762–767. [Google Scholar] [CrossRef] [PubMed]
  106. Gingras, A.C.; Gygi, S.P.; Raught, B.; Polakiewicz, R.D.; Abraham, R.T.; Hoekstra, M.F.; Aebersold, R.; Sonenberg, N. Regulation of 4E-BP1 phosphorylation: A novel two-step mechanism. Genes Dev. 1999, 13, 1422–1437. [Google Scholar] [CrossRef]
  107. Constantinou, C.; Clemens, M.J. Regulation of the phosphorylation and integrity of protein synthesis initiation factor eIF4GI and the translational repressor 4E-BP1 by p53. Oncogene 2005, 24, 4839–4850. [Google Scholar] [CrossRef]
  108. Rosenwald, I.B.; Rhoads, D.B.; Callanan, L.D.; Isselbacher, K.J.; Schmidt, E.V. Increased expression of eukaryotic translation initiation factors eIF-4E and eIF-2 alpha in response to growth induction by c-myc. Proc. Natl. Acad. Sci. USA 1993, 90, 6175–6178. [Google Scholar] [CrossRef]
  109. Shantz, L.M.; Hu, R.H.; Pegg, A.E. Regulation of ornithine decarboxylase in a transformed cell line that overexpresses translation initiation factor eIF-4E. Cancer Res. 1996, 56, 3265–3269. [Google Scholar]
  110. Ruggero, D.; Montanaro, L.; Ma, L.; Xu, W.; Londei, P.; Cordon-Cardo, C.; Pandolfi, P.P. The translation factor eIF-4E promotes tumor formation and cooperates with c-Myc in lymphomagenesis. Nat. Med. 2004, 10, 484–486. [Google Scholar] [CrossRef]
  111. Scott, P.A.; Smith, K.; Poulsom, R.; De Benedetti, A.; Bicknell, R.; Harris, A.L. Differential expression of vascular endothelial growth factor mRNA vs protein isoform expression in human breast cancer and relationship to eIF-4E. Br. J. Cancer 1998, 77, 2120–2128. [Google Scholar] [CrossRef] [PubMed]
  112. Cunningham, J.T.; Moreno, M.V.; Lodi, A.; Ronen, S.M.; Ruggero, D. Protein and nucleotide biosynthesis are coupled by a single rate-limiting enzyme, PRPS2, to drive cancer. Cell 2014, 157, 1088–1103. [Google Scholar] [CrossRef]
  113. De Benedetti, A.; Graff, J.R. eIF-4E expression and its role in malignancies and metastases. Oncogene 2004, 23, 3189–3199. [Google Scholar] [CrossRef] [PubMed]
  114. Hinnebusch, A.G. Translational homeostasis via eIF4E and 4E-BP1. Mol. Cell 2012, 46, 717–719. [Google Scholar] [CrossRef] [PubMed]
  115. Hsieh, A.C.; Costa, M.; Zollo, O.; Davis, C.; Feldman, M.E.; Testa, J.R.; Meyuhas, O.; Shokat, K.M.; Ruggero, D. Genetic dissection of the oncogenic mTOR pathway reveals druggable addiction to translational control via 4EBP-eIF4E. Cancer Cell 2010, 17, 249–261. [Google Scholar] [CrossRef] [PubMed]
  116. Batool, A.; Majeed, S.T.; Aashaq, S.; Majeed, R.; Shah, G.; Nazir, N.; Andrabi, K.I. Eukaryotic Initiation Factor 4E (eIF4E) sequestration mediates 4E-BP1 response to rapamycin. Int. J. Biol. Macromol. 2019, 125, 651–659. [Google Scholar] [CrossRef]
  117. Livingstone, M.; Bidinosti, M. Rapamycin-insensitive mTORC1 activity controls eIF4E:4E-BP1 binding. F1000Research 2012, 1, 4. [Google Scholar] [CrossRef]
  118. Li, S.; Chen, J.S.; Li, X.; Bai, X.; Shi, D. MNK, mTOR or eIF4E-selecting the best anti-tumor target for blocking translation initiation. Eur. J. Med. Chem. 2023, 260, 115781. [Google Scholar] [CrossRef]
  119. Hay, N. Mnk earmarks eIF4E for cancer therapy. Proc. Natl. Acad. Sci. USA 2010, 107, 13975–13976. [Google Scholar] [CrossRef]
  120. Grosso, S.; Pesce, E.; Brina, D.; Beugnet, A.; Loreni, F.; Biffo, S. Sensitivity of global translation to mTOR inhibition in REN cells depends on the equilibrium between eIF4E and 4E-BP1. PLoS ONE 2011, 6, e29136. [Google Scholar] [CrossRef]
  121. Hu, K.; Zhang, J.; Yu, M.; Xiong, C. Inhibition of Mnk-eIF4E pathway sensitizes the efficacy to chemotherapy in anaplastic thyroid cancer. Future Oncol. 2017, 13, 489–498. [Google Scholar] [CrossRef] [PubMed]
  122. Urtishak, K.A.; Wang, L.S.; Culjkovic-Kraljacic, B.; Davenport, J.W.; Porazzi, P.; Vincent, T.L.; Teachey, D.T.; Tasian, S.K.; Moore, J.S.; Seif, A.E.; et al. Targeting EIF4E signaling with ribavirin in infant acute lymphoblastic leukemia. Oncogene 2019, 38, 2241–2262. [Google Scholar] [CrossRef] [PubMed]
  123. Darzynkiewicz, E.; Stepinski, J.; Ekiel, I.; Goyer, C.; Sonenberg, N.; Temeriusz, A.; Jin, Y.; Sijuwade, T.; Haber, D.; Tahara, S.M. Inhibition of eukaryotic translation by nucleoside 5′-monophosphate analogues of mRNA 5′-cap: Changes in N7 substituent affect analogue activity. Biochemistry 1989, 28, 4771–4778. [Google Scholar] [CrossRef] [PubMed]
  124. Ghosh, B.; Benyumov, A.O.; Ghosh, P.; Jia, Y.; Avdulov, S.; Dahlberg, P.S.; Peterson, M.; Smith, K.; Polunovsky, V.A.; Bitterman, P.B.; et al. Nontoxic chemical interdiction of the epithelial-to-mesenchymal transition by targeting cap-dependent translation. ACS Chem. Biol. 2009, 4, 367–377. [Google Scholar] [CrossRef]
  125. Moerke, N.J.; Aktas, H.; Chen, H.; Cantel, S.; Reibarkh, M.Y.; Fahmy, A.; Gross, J.D.; Degterev, A.; Yuan, J.; Chorev, M.; et al. Small-molecule inhibition of the interaction between the translation initiation factors eIF4E and eIF4G. Cell 2007, 128, 257–267. [Google Scholar] [CrossRef]
  126. Takrouri, K.; Chen, T.; Papadopoulos, E.; Sahoo, R.; Kabha, E.; Chen, H.; Cantel, S.; Wagner, G.; Halperin, J.A.; Aktas, B.H.; et al. Structure-activity relationship study of 4EGI-1, small molecule eIF4E/eIF4G protein-protein interaction inhibitors. Eur. J. Med. Chem. 2014, 77, 361–377. [Google Scholar] [CrossRef] [PubMed]
  127. Sekiyama, N.; Arthanari, H.; Papadopoulos, E.; Rodriguez-Mias, R.A.; Wagner, G.; Leger-Abraham, M. Molecular mechanism of the dual activity of 4EGI-1: Dissociating eIF4G from eIF4E but stabilizing the binding of unphosphorylated 4E-BP1. Proc. Natl. Acad. Sci. USA 2015, 112, E4036–E4045. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, W.; Li, J.; Wen, Q.; Luo, J.; Chu, S.; Chen, L.; Qing, Z.; Xie, G.; Xu, L.; Alnemah, M.M.; et al. 4EGI-1 induces apoptosis and enhances radiotherapy sensitivity in nasopharyngeal carcinoma cells via DR5 induction on 4E-BP1 dephosphorylation. Oncotarget 2016, 7, 21728–21741. [Google Scholar] [CrossRef]
  129. Cencic, R.; Desforges, M.; Hall, D.R.; Kozakov, D.; Du, Y.; Min, J.; Dingledine, R.; Fu, H.; Vajda, S.; Talbot, P.J.; et al. Blocking eIF4E-eIF4G interaction as a strategy to impair coronavirus replication. J. Virol. 2011, 85, 6381–6389. [Google Scholar] [CrossRef]
  130. Kardos, G.R.; Gowda, R.; Dinavahi, S.S.; Kimball, S.; Robertson, G.P. Salubrinal in Combination With 4E1RCat Synergistically Impairs Melanoma Development by Disrupting the Protein Synthetic Machinery. Front. Oncol. 2020, 10, 834. [Google Scholar] [CrossRef]
  131. Cencic, R.; Hall, D.R.; Robert, F.; Du, Y.; Min, J.; Li, L.; Qui, M.; Lewis, I.; Kurtkaya, S.; Dingledine, R.; et al. Reversing chemoresistance by small molecule inhibition of the translation initiation complex eIF4F. Proc. Natl. Acad. Sci. USA 2011, 108, 1046–1051. [Google Scholar] [CrossRef] [PubMed]
  132. Schutz, P.; Bumann, M.; Oberholzer, A.E.; Bieniossek, C.; Trachsel, H.; Altmann, M.; Baumann, U. Crystal structure of the yeast eIF4A-eIF4G complex: An RNA-helicase controlled by protein-protein interactions. Proc. Natl. Acad. Sci. USA 2008, 105, 9564–9569. [Google Scholar] [CrossRef] [PubMed]
  133. Rogers, G.W., Jr.; Komar, A.A.; Merrick, W.C. eIF4A: The godfather of the DEAD box helicases. Prog. Nucleic Acid. Res. Mol. Biol. 2002, 72, 307–331. [Google Scholar] [CrossRef] [PubMed]
  134. Gross, J.D.; Moerke, N.J.; von der Haar, T.; Lugovskoy, A.A.; Sachs, A.B.; McCarthy, J.E.; Wagner, G. Ribosome loading onto the mRNA cap is driven by conformational coupling between eIF4G and eIF4E. Cell 2003, 115, 739–750. [Google Scholar] [CrossRef]
  135. Hinton, T.M.; Coldwell, M.J.; Carpenter, G.A.; Morley, S.J.; Pain, V.M. Functional analysis of individual binding activities of the scaffold protein eIF4G. J. Biol. Chem. 2007, 282, 1695–1708. [Google Scholar] [CrossRef] [PubMed]
  136. Park, E.H.; Zhang, F.; Warringer, J.; Sunnerhagen, P.; Hinnebusch, A.G. Depletion of eIF4G from yeast cells narrows the range of translational efficiencies genome-wide. BMC Genom. 2011, 12, 68. [Google Scholar] [CrossRef]
  137. Chen, L.; Aktas, B.H.; Wang, Y.; He, X.; Sahoo, R.; Zhang, N.; Denoyelle, S.; Kabha, E.; Yang, H.; Freedman, R.Y.; et al. Tumor suppression by small molecule inhibitors of translation initiation. Oncotarget 2012, 3, 869–881. [Google Scholar] [CrossRef]
  138. Kedersha, N.; Chen, S.; Gilks, N.; Li, W.; Miller, I.J.; Stahl, J.; Anderson, P. Evidence that ternary complex (eIF2-GTP-tRNA(i)(Met))-deficient preinitiation complexes are core constituents of mammalian stress granules. Mol. Biol. Cell 2002, 13, 195–210. [Google Scholar] [CrossRef]
  139. Clemens, M.J. Initiation factor eIF2 alpha phosphorylation in stress responses and apoptosis. Prog. Mol. Subcell. Biol. 2001, 27, 57–89. [Google Scholar] [CrossRef]
  140. Donze, O.; Jagus, R.; Koromilas, A.E.; Hershey, J.W.; Sonenberg, N. Abrogation of translation initiation factor eIF-2 phosphorylation causes malignant transformation of NIH 3T3 cells. EMBO J. 1995, 14, 3828–3834. [Google Scholar] [CrossRef]
  141. Chen, T.; Ozel, D.; Qiao, Y.; Harbinski, F.; Chen, L.; Denoyelle, S.; He, X.; Zvereva, N.; Supko, J.G.; Chorev, M.; et al. Chemical genetics identify eIF2alpha kinase heme-regulated inhibitor as an anticancer target. Nat. Chem. Biol. 2011, 7, 610–616. [Google Scholar] [CrossRef] [PubMed]
  142. Denoyelle, S.; Chen, T.; Chen, L.; Wang, Y.; Klosi, E.; Halperin, J.A.; Aktas, B.H.; Chorev, M. In vitro inhibition of translation initiation by N,N′-diarylureas–potential anti-cancer agents. Bioorg. Med. Chem. Lett. 2012, 22, 402–409. [Google Scholar] [CrossRef] [PubMed]
  143. Boyce, M.; Bryant, K.F.; Jousse, C.; Long, K.; Harding, H.P.; Scheuner, D.; Kaufman, R.J.; Ma, D.; Coen, D.M.; Ron, D.; et al. A selective inhibitor of eIF2alpha dephosphorylation protects cells from ER stress. Science 2005, 307, 935–939. [Google Scholar] [CrossRef] [PubMed]
  144. Schewe, D.M.; Aguirre-Ghiso, J.A. Inhibition of eIF2alpha dephosphorylation maximizes bortezomib efficiency and eliminates quiescent multiple myeloma cells surviving proteasome inhibitor therapy. Cancer Res. 2009, 69, 1545–1552. [Google Scholar] [CrossRef]
  145. Robert, F.; Kapp, L.D.; Khan, S.N.; Acker, M.G.; Kolitz, S.; Kazemi, S.; Kaufman, R.J.; Merrick, W.C.; Koromilas, A.E.; Lorsch, J.R.; et al. Initiation of protein synthesis by hepatitis C virus is refractory to reduced eIF2.GTP.Met-tRNA(i)(Met) ternary complex availability. Mol. Biol. Cell 2006, 17, 4632–4644. [Google Scholar] [CrossRef]
  146. des Georges, A.; Dhote, V.; Kuhn, L.; Hellen, C.U.; Pestova, T.V.; Frank, J.; Hashem, Y. Structure of mammalian eIF3 in the context of the 43S preinitiation complex. Nature 2015, 525, 491–495. [Google Scholar] [CrossRef]
  147. Sokabe, M.; Fraser, C.S. Human eukaryotic initiation factor 2 (eIF2)-GTP-Met-tRNAi ternary complex and eIF3 stabilize the 43 S preinitiation complex. J. Biol. Chem. 2014, 289, 31827–31836. [Google Scholar] [CrossRef]
  148. Beznoskova, P.; Cuchalova, L.; Wagner, S.; Shoemaker, C.J.; Gunisova, S.; von der Haar, T.; Valasek, L.S. Translation initiation factors eIF3 and HCR1 control translation termination and stop codon read-through in yeast cells. PLoS Genet. 2013, 9, e1003962. [Google Scholar] [CrossRef]
  149. Beznoskova, P.; Wagner, S.; Jansen, M.E.; von der Haar, T.; Valasek, L.S. Translation initiation factor eIF3 promotes programmed stop codon readthrough. Nucleic Acids Res. 2015, 43, 5099–5111. [Google Scholar] [CrossRef]
  150. Lopez de Quinto, S.; Lafuente, E.; Martinez-Salas, E. IRES interaction with translation initiation factors: Functional characterization of novel RNA contacts with eIF3, eIF4B, and eIF4GII. RNA 2001, 7, 1213–1226. [Google Scholar] [CrossRef]
  151. Zhang, L.; Pan, X.; Hershey, J.W. Individual overexpression of five subunits of human translation initiation factor eIF3 promotes malignant transformation of immortal fibroblast cells. J. Biol. Chem. 2007, 282, 5790–5800. [Google Scholar] [CrossRef] [PubMed]
  152. Zhang, F.; Xiang, S.; Cao, Y.; Li, M.; Ma, Q.; Liang, H.; Li, H.; Ye, Y.; Zhang, Y.; Jiang, L.; et al. EIF3D promotes gallbladder cancer development by stabilizing GRK2 kinase and activating PI3K-AKT signaling pathway. Cell Death Dis. 2017, 8, e2868. [Google Scholar] [CrossRef] [PubMed]
  153. Qi, J.; Dong, Z.; Liu, J.; Zhang, J.T. EIF3i promotes colon oncogenesis by regulating COX-2 protein synthesis and beta-catenin activation. Oncogene 2014, 33, 4156–4163. [Google Scholar] [CrossRef] [PubMed]
  154. Li, X.W.; Hu, C.P.; Li, Y.J.; Gao, Y.X.; Wang, X.M.; Yang, J.R. Inhibitory effect of l-mimosine on bleomycin-induced pulmonary fibrosis in rats: Role of eIF3a and p27. Int. Immunopharmacol. 2015, 27, 53–64. [Google Scholar] [CrossRef] [PubMed]
  155. Yin, J.Y.; Zhang, J.T.; Zhang, W.; Zhou, H.H.; Liu, Z.Q. eIF3a: A new anticancer drug target in the eIF family. Cancer Lett. 2018, 412, 81–87. [Google Scholar] [CrossRef]
  156. Carvalho, A.; Chu, J.; Meinguet, C.; Kiss, R.; Vandenbussche, G.; Masereel, B.; Wouters, J.; Kornienko, A.; Pelletier, J.; Mathieu, V. A harmine-derived beta-carboline displays anti-cancer effects in vitro by targeting protein synthesis. Eur. J. Pharmacol. 2017, 805, 25–35. [Google Scholar] [CrossRef]
  157. Miluzio, A.; Beugnet, A.; Volta, V.; Biffo, S. Eukaryotic initiation factor 6 mediates a continuum between 60S ribosome biogenesis and translation. EMBO Rep. 2009, 10, 459–465. [Google Scholar] [CrossRef]
  158. Ceci, M.; Gaviraghi, C.; Gorrini, C.; Sala, L.A.; Offenhauser, N.; Marchisio, P.C.; Biffo, S. Release of eIF6 (p27BBP) from the 60S subunit allows 80S ribosome assembly. Nature 2003, 426, 579–584. [Google Scholar] [CrossRef]
  159. Brina, D.; Miluzio, A.; Ricciardi, S.; Biffo, S. eIF6 anti-association activity is required for ribosome biogenesis, translational control and tumor progression. Biochim. Biophys. Acta 2015, 1849, 830–835. [Google Scholar] [CrossRef]
  160. Miluzio, A.; Beugnet, A.; Grosso, S.; Brina, D.; Mancino, M.; Campaner, S.; Amati, B.; de Marco, A.; Biffo, S. Impairment of cytoplasmic eIF6 activity restricts lymphomagenesis and tumor progression without affecting normal growth. Cancer Cell 2011, 19, 765–775. [Google Scholar] [CrossRef]
  161. Gandin, V.; Miluzio, A.; Barbieri, A.M.; Beugnet, A.; Kiyokawa, H.; Marchisio, P.C.; Biffo, S. Eukaryotic initiation factor 6 is rate-limiting in translation, growth and transformation. Nature 2008, 455, 684–688. [Google Scholar] [CrossRef] [PubMed]
  162. Golob-Schwarzl, N.; Schweiger, C.; Koller, C.; Krassnig, S.; Gogg-Kamerer, M.; Gantenbein, N.; Toeglhofer, A.M.; Wodlej, C.; Bergler, H.; Pertschy, B.; et al. Separation of low and high grade colon and rectum carcinoma by eukaryotic translation initiation factors 1, 5 and 6. Oncotarget 2017, 8, 101224–101243. [Google Scholar] [CrossRef]
  163. Gantenbein, N.; Bernhart, E.; Anders, I.; Golob-Schwarzl, N.; Krassnig, S.; Wodlej, C.; Brcic, L.; Lindenmann, J.; Fink-Neuboeck, N.; Gollowitsch, F.; et al. Influence of eukaryotic translation initiation factor 6 on non-small cell lung cancer development and progression. Eur. J. Cancer 2018, 101, 165–180. [Google Scholar] [CrossRef] [PubMed]
  164. Shen, Y.; Zhang, R.; Li, X. Identification of eIF6 as a prognostic factor that drives tumor progression and predicts arsenic trioxide efficacy in lung adenocarcinoma. Mol. Biol. Rep. 2023, 50, 1167–1180. [Google Scholar] [CrossRef] [PubMed]
  165. Gao, Y.; Yuan, L.; Zeng, J.; Li, F.; Li, X.; Tan, F.; Liu, X.; Wan, H.; Kui, X.; Liu, X.; et al. eIF6 is potential diagnostic and prognostic biomarker that associated with (18)F-FDG PET/CT features and immune signatures in esophageal carcinoma. J. Transl. Med. 2022, 20, 303. [Google Scholar] [CrossRef]
  166. Liang, J.; Liu, F.; Yang, Y.; Li, X.; Cai, G.; Cao, J.; Zhang, B. Diagnostic and prognostic utility of eIF6 in glioblastoma: A study based on TCGA and CGGA databases. Am. J. Transl. Res. 2022, 14, 5040–5049. [Google Scholar]
  167. Scagliola, A.; Miluzio, A.; Ventura, G.; Oliveto, S.; Cordiglieri, C.; Manfrini, N.; Cirino, D.; Ricciardi, S.; Valenti, L.; Baselli, G.; et al. Targeting of eIF6-driven translation induces a metabolic rewiring that reduces NAFLD and the consequent evolution to hepatocellular carcinoma. Nat. Commun. 2021, 12, 4878. [Google Scholar] [CrossRef]
  168. Miluzio, A.; Ricciardi, S.; Manfrini, N.; Alfieri, R.; Oliveto, S.; Brina, D.; Biffo, S. Translational control by mTOR-independent routes: How eIF6 organizes metabolism. Biochem. Soc. Trans. 2016, 44, 1667–1673. [Google Scholar] [CrossRef]
  169. Sun, L.; Liu, S.; Wang, X.; Zheng, X.; Chen, Y.; Shen, H. eIF6 promotes the malignant progression of human hepatocellular carcinoma via the mTOR signaling pathway. J. Transl. Med. 2021, 19, 216. [Google Scholar] [CrossRef]
  170. Pesce, E.; Miluzio, A.; Turcano, L.; Minici, C.; Cirino, D.; Calamita, P.; Manfrini, N.; Oliveto, S.; Ricciardi, S.; Grifantini, R.; et al. Discovery and Preliminary Characterization of Translational Modulators that Impair the Binding of eIF6 to 60S Ribosomal Subunits. Cells 2020, 9, 172. [Google Scholar] [CrossRef]
  171. Scagliola, A.; Miluzio, A.; Mori, G.; Ricciardi, S.; Oliveto, S.; Manfrini, N.; Biffo, S. Inhibition of eIF6 Activity Reduces Hepatocellular Carcinoma Growth: An In Vivo and In Vitro Study. Int. J. Mol. Sci. 2022, 23, 7720. [Google Scholar] [CrossRef] [PubMed]
  172. Lund, A.; Knudsen, S.M.; Vissing, H.; Clark, B.; Tommerup, N. Assignment of human elongation factor 1alpha genes: EEF1A maps to chromosome 6q14 and EEF1A2 to 20q13.3. Genomics 1996, 36, 359–361. [Google Scholar] [CrossRef]
  173. Davies, F.C.J.; Marshall, G.F.; Pegram, E.; Gadd, D.; Abbott, C.M. Endogenous epitope tagging of eEF1A2 in mice reveals early embryonic expression of eEF1A2 and subcellular compartmentalisation of neuronal eEF1A1 and eEF1A2. Mol. Cell Neurosci. 2023, 126, 103879. [Google Scholar] [CrossRef] [PubMed]
  174. Mills, A.; Gago, F. On the Need to Tell Apart Fraternal Twins eEF1A1 and eEF1A2, and Their Respective Outfits. Int. J. Mol. Sci. 2021, 22, 6973. [Google Scholar] [CrossRef] [PubMed]
  175. Ramesh, R.; Sattlegger, E. Domain II of the translation elongation factor eEF1A is required for Gcn2 kinase inhibition. FEBS Lett. 2020, 594, 2266–2281. [Google Scholar] [CrossRef]
  176. Visweswaraiah, J.; Lageix, S.; Castilho, B.A.; Izotova, L.; Kinzy, T.G.; Hinnebusch, A.G.; Sattlegger, E. Evidence that eukaryotic translation elongation factor 1A (eEF1A) binds the Gcn2 protein C terminus and inhibits Gcn2 activity. J. Biol. Chem. 2011, 286, 36568–36579. [Google Scholar] [CrossRef]
  177. Leclercq, T.M.; Moretti, P.A.; Vadas, M.A.; Pitson, S.M. Eukaryotic elongation factor 1A interacts with sphingosine kinase and directly enhances its catalytic activity. J. Biol. Chem. 2008, 283, 9606–9614. [Google Scholar] [CrossRef]
  178. Schulz, I.; Engel, C.; Niestroj, A.J.; Kehlen, A.; Rahfeld, J.U.; Kleinschmidt, M.; Lehmann, K.; Rossner, S.; Demuth, H.U. A non-canonical function of eukaryotic elongation factor 1A1: Regulation of interleukin-6 expression. Biochim. Biophys. Acta 2014, 1843, 965–975. [Google Scholar] [CrossRef]
  179. Akintade, D.D.; Chaudhuri, B. Identification of proteins involved in transcription/translation (eEF 1A1) as an inhibitor of Bax induced apoptosis. Mol. Biol. Rep. 2020, 47, 6785–6792. [Google Scholar] [CrossRef]
  180. Chen, E.; Proestou, G.; Bourbeau, D.; Wang, E. Rapid up-regulation of peptide elongation factor EF-1alpha protein levels is an immediate early event during oxidative stress-induced apoptosis. Exp. Cell Res. 2000, 259, 140–148. [Google Scholar] [CrossRef]
  181. Zhong, D.; Zhang, J.; Yang, S.; Soh, U.J.; Buschdorf, J.P.; Zhou, Y.T.; Yang, D.; Low, B.C. The SAM domain of the RhoGAP DLC1 binds EF1A1 to regulate cell migration. J. Cell Sci. 2009, 122, 414–424. [Google Scholar] [CrossRef] [PubMed]
  182. Gross, S.R.; Kinzy, T.G. Translation elongation factor 1A is essential for regulation of the actin cytoskeleton and cell morphology. Nat. Struct. Mol. Biol. 2005, 12, 772–778. [Google Scholar] [CrossRef] [PubMed]
  183. Guo, M.; Li, Y.; Wang, Y.; Li, Z.; Li, X.; Zhao, P.; Li, C.; Lv, J.; Liu, X.; Du, X.; et al. eEF1A2 exacerbated insulin resistance in male skeletal muscle via PKCbeta and ER stress. J. Endocrinol. 2020, 244, 25–40. [Google Scholar] [CrossRef] [PubMed]
  184. Wilson, R.B.; Kozlov, A.M.; Hatam Tehrani, H.; Twumasi-Ankrah, J.S.; Chen, Y.J.; Borrelli, M.J.; Sawyez, C.G.; Maini, S.; Shepherd, T.G.; Cumming, R.C.; et al. Elongation factor 1A1 regulates metabolic substrate preference in mammalian cells. J. Biol. Chem. 2024, 300, 105684. [Google Scholar] [CrossRef] [PubMed]
  185. Borradaile, N.M.; Buhman, K.K.; Listenberger, L.L.; Magee, C.J.; Morimoto, E.T.; Ory, D.S.; Schaffer, J.E. A critical role for eukaryotic elongation factor 1A-1 in lipotoxic cell death. Mol. Biol. Cell 2006, 17, 770–778. [Google Scholar] [CrossRef] [PubMed]
  186. Khwanraj, K.; Madlah, S.; Grataitong, K.; Dharmasaroja, P. Comparative mRNA Expression of eEF1A Isoforms and a PI3K/Akt/mTOR Pathway in a Cellular Model of Parkinson’s Disease. Park. Dis. 2016, 2016, 8716016. [Google Scholar] [CrossRef]
  187. Gao, G.; Duan, Y.; Chang, F.; Zhang, T.; Huang, X.; Yu, C. METTL14 promotes apoptosis of spinal cord neurons by inducing EEF1A2 m6A methylation in spinal cord injury. Cell Death Discov. 2022, 8, 15. [Google Scholar] [CrossRef]
  188. Liu, Y.; Deng, S.; Song, Z.; Zhang, Q.; Guo, Y.; Yu, Y.; Wang, Y.; Li, T.; Megahed, F.A.K.; Addissouky, T.A.; et al. MLIF Modulates Microglia Polarization in Ischemic Stroke by Targeting eEF1A1. Front. Pharmacol. 2021, 12, 725268. [Google Scholar] [CrossRef]
  189. Yan, G.; You, B.; Chen, S.P.; Liao, J.K.; Sun, J. Tumor necrosis factor-alpha downregulates endothelial nitric oxide synthase mRNA stability via translation elongation factor 1-alpha 1. Circ. Res. 2008, 103, 591–597. [Google Scholar] [CrossRef]
  190. Vera, M.; Pani, B.; Griffiths, L.A.; Muchardt, C.; Abbott, C.M.; Singer, R.H.; Nudler, E. The translation elongation factor eEF1A1 couples transcription to translation during heat shock response. Elife 2014, 3, e03164. [Google Scholar] [CrossRef]
  191. Chuang, S.M.; Chen, L.; Lambertson, D.; Anand, M.; Kinzy, T.G.; Madura, K. Proteasome-mediated degradation of cotranslationally damaged proteins involves translation elongation factor 1A. Mol. Cell Biol. 2005, 25, 403–413. [Google Scholar] [CrossRef] [PubMed]
  192. Gandin, V.; Gutierrez, G.J.; Brill, L.M.; Varsano, T.; Feng, Y.; Aza-Blanc, P.; Au, Q.; McLaughlan, S.; Ferreira, T.A.; Alain, T.; et al. Degradation of newly synthesized polypeptides by ribosome-associated RACK1/c-Jun N-terminal kinase/eukaryotic elongation factor 1A2 complex. Mol. Cell Biol. 2013, 33, 2510–2526. [Google Scholar] [CrossRef] [PubMed]
  193. Tatsuka, M.; Mitsui, H.; Wada, M.; Nagata, A.; Nojima, H.; Okayama, H. Elongation factor-1 alpha gene determines susceptibility to transformation. Nature 1992, 359, 333–336. [Google Scholar] [CrossRef] [PubMed]
  194. Sanders, J.; Maassen, J.A.; Moller, W. Elongation factor-1 messenger-RNA levels in cultured cells are high compared to tissue and are not drastically affected further by oncogenic transformation. Nucleic Acids Res. 1992, 20, 5907–5910. [Google Scholar] [CrossRef] [PubMed]
  195. Edmonds, B.T.; Wyckoff, J.; Yeung, Y.G.; Wang, Y.; Stanley, E.R.; Jones, J.; Segall, J.; Condeelis, J. Elongation factor-1 alpha is an overexpressed actin binding protein in metastatic rat mammary adenocarcinoma. J. Cell Sci. 1996, 109 Pt 11, 2705–2714. [Google Scholar] [CrossRef]
  196. Zhong, W.; Huang, C.; Lin, J.; Zhu, M.; Zhong, H.; Chiang, M.H.; Chiang, H.S.; Hui, M.S.; Lin, Y.; Huang, J. Development and Validation of Nine-RNA Binding Protein Signature Predicting Overall Survival for Kidney Renal Clear Cell Carcinoma. Front. Genet. 2020, 11, 568192. [Google Scholar] [CrossRef]
  197. Cai, Z.; Wu, Y.; Zhang, F.; Wu, H. A three-gene signature and clinical outcome in pediatric acute myeloid leukemia. Clin. Transl. Oncol. 2021, 23, 866–873. [Google Scholar] [CrossRef]
  198. Hu, G.; Li, J.; Zeng, Y.; Liu, L.; Yu, Z.; Qi, X.; Liu, K.; Yao, H. The anoikis-related gene signature predicts survival accurately in colon adenocarcinoma. Sci. Rep. 2023, 13, 13919. [Google Scholar] [CrossRef]
  199. Yuan, Q.; Zhang, W.; Shang, W. Identification and validation of a prognostic risk-scoring model based on sphingolipid metabolism-associated cluster in colon adenocarcinoma. Front. Endocrinol. 2022, 13, 1045167. [Google Scholar] [CrossRef]
  200. Luo, B.H.; Huang, J.Q.; Huang, C.Y.; Tian, P.; Chen, A.Z.; Wu, W.H.; Ma, X.M.; Yuan, Y.X.; Yu, L. Screening of Lymphoma Radiotherapy-Resistant Genes with CRISPR Activation Library. Pharmgenomics Pers. Med. 2023, 16, 67–80. [Google Scholar] [CrossRef]
  201. Chun, H.G.; Davies, B.; Hoth, D.; Suffness, M.; Plowman, J.; Flora, K.; Grieshaber, C.; Leyland-Jones, B. Didemnin B. The first marine compound entering clinical trials as an antineoplastic agent. Investig. New Drugs 1986, 4, 279–284. [Google Scholar] [CrossRef] [PubMed]
  202. Leisch, M.; Egle, A.; Greil, R. Plitidepsin: A potential new treatment for relapsed/refractory multiple myeloma. Future Oncol. 2019, 15, 109–120. [Google Scholar] [CrossRef] [PubMed]
  203. Jimenez, P.C.; Wilke, D.V.; Branco, P.C.; Bauermeister, A.; Rezende-Teixeira, P.; Gaudencio, S.P.; Costa-Lotufo, L.V. Enriching cancer pharmacology with drugs of marine origin. Br. J. Pharmacol. 2020, 177, 3–27. [Google Scholar] [CrossRef] [PubMed]
  204. Van Goietsenoven, G.; Hutton, J.; Becker, J.P.; Lallemand, B.; Robert, F.; Lefranc, F.; Pirker, C.; Vandenbussche, G.; Van Antwerpen, P.; Evidente, A.; et al. Targeting of eEF1A with Amaryllidaceae isocarbostyrils as a strategy to combat melanomas. FASEB J. 2010, 24, 4575–4584. [Google Scholar] [CrossRef]
  205. Sanchez-Murcia, P.A.; Cortes-Cabrera, A.; Gago, F. Structural rationale for the cross-resistance of tumor cells bearing the A399V variant of elongation factor eEF1A1 to the structurally unrelated didemnin B, ternatin, nannocystin A and ansatrienin B. J. Comput. Aided Mol. Des. 2017, 31, 915–928. [Google Scholar] [CrossRef]
  206. Hou, Y.; Liu, R.; Xia, M.; Sun, C.; Zhong, B.; Yu, J.; Ai, N.; Lu, J.J.; Ge, W.; Liu, B.; et al. Nannocystin ax, an eEF1A inhibitor, induces G1 cell cycle arrest and caspase-independent apoptosis through cyclin D1 downregulation in colon cancer in vivo. Pharmacol. Res. 2021, 173, 105870. [Google Scholar] [CrossRef] [PubMed]
  207. Sun, C.; Liu, R.; Xia, M.; Hou, Y.; Wang, X.; Lu, J.J.; Liu, B.; Chen, X. Nannocystin Ax, a natural elongation factor 1alpha inhibitor from Nannocystis sp., suppresses epithelial-mesenchymal transition, adhesion and migration in lung cancer cells. Toxicol. Appl. Pharmacol. 2021, 420, 115535. [Google Scholar] [CrossRef]
  208. Nakamura, J.; Aoyagi, S.; Nanchi, I.; Nakatsuka, S.; Hirata, E.; Shibata, S.; Fukuda, M.; Yamamoto, Y.; Fukuda, I.; Tatsumi, N.; et al. Overexpression of eukaryotic elongation factor eEF2 in gastrointestinal cancers and its involvement in G2/M progression in the cell cycle. Int. J. Oncol. 2009, 34, 1181–1189. [Google Scholar]
  209. Sun, H.G.; Dong, X.J.; Lu, T.; Yang, M.F.; Wang, X.M. Clinical value of eukaryotic elongation factor 2 (eEF2) in non-small cell lung cancer patients. Asian Pac. J. Cancer Prev. 2014, 14, 6533–6535. [Google Scholar] [CrossRef]
  210. Raspin, K.; Marthick, J.R.; Donovan, S.; Blizzard, L.; Malley, R.C.; Jung, C.H.; Banks, A.; Redwig, F.; Skala, M.; Dickinson, J.L.; et al. Identification of a novel recurrent EEF2 gene amplification in familial prostate tumors. Genes Chromosomes Cancer 2023, 62, 247–255. [Google Scholar] [CrossRef]
  211. Pott, L.L.; Hagemann, S.; Reis, H.; Lorenz, K.; Bracht, T.; Herold, T.; Skryabin, B.V.; Megger, D.A.; Kalsch, J.; Weber, F.; et al. Eukaryotic elongation factor 2 is a prognostic marker and its kinase a potential therapeutic target in HCC. Oncotarget 2017, 8, 11950–11962. [Google Scholar] [CrossRef] [PubMed]
  212. Sun, W.; Wei, X.; Niu, A.; Ma, X.; Li, J.J.; Gao, D. Enhanced anti-colon cancer immune responses with modified eEF2-derived peptides. Cancer Lett. 2015, 369, 112–123. [Google Scholar] [CrossRef] [PubMed]
  213. Sun, S.Y.; Rosenberg, L.M.; Wang, X.; Zhou, Z.; Yue, P.; Fu, H.; Khuri, F.R. Activation of Akt and eIF4E survival pathways by rapamycin-mediated mammalian target of rapamycin inhibition. Cancer Res. 2005, 65, 7052–7058. [Google Scholar] [CrossRef]
  214. Astanehe, A.; Finkbeiner, M.R.; Krzywinski, M.; Fotovati, A.; Dhillon, J.; Berquin, I.M.; Mills, G.B.; Marra, M.A.; Dunn, S.E. MKNK1 is a YB-1 target gene responsible for imparting trastuzumab resistance and can be blocked by RSK inhibition. Oncogene 2012, 31, 4434–4446. [Google Scholar] [CrossRef]
  215. Adesso, L.; Calabretta, S.; Barbagallo, F.; Capurso, G.; Pilozzi, E.; Geremia, R.; Delle Fave, G.; Sette, C. Gemcitabine triggers a pro-survival response in pancreatic cancer cells through activation of the MNK2/eIF4E pathway. Oncogene 2013, 32, 2848–2857. [Google Scholar] [CrossRef] [PubMed]
  216. Kwegyir-Afful, A.K.; Murigi, F.N.; Purushottamachar, P.; Ramamurthy, V.P.; Martin, M.S.; Njar, V.C.O. Galeterone and its analogs inhibit Mnk-eIF4E axis, synergize with gemcitabine, impede pancreatic cancer cell migration, invasion and proliferation and inhibit tumor growth in mice. Oncotarget 2017, 8, 52381–52402. [Google Scholar] [CrossRef]
  217. Huang, M.T. Harringtonine, an inhibitor of initiation of protein biosynthesis. Mol. Pharmacol. 1975, 11, 511–519. [Google Scholar]
  218. O’Brien, S.; Kantarjian, H.; Keating, M.; Beran, M.; Koller, C.; Robertson, L.E.; Hester, J.; Rios, M.B.; Andreeff, M.; Talpaz, M. Homoharringtonine therapy induces responses in patients with chronic myelogenous leukemia in late chronic phase. Blood 1995, 86, 3322–3326. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of eukaryotic translation initiation. Translation initiation is a multi-step process. In step 1, the ternary complex (TC) is formed wherein eIF2 (α, β, and γ), initiator methionyl tRNA (Met-tRNAMeti), and GTP assemble. In step 2, 40S ribosomal subunit, ternary complex, eIF1, eIF1A, eIF3, and eIF5 assembles to form 43S pre-initiation complex PIC). Simultaneously, the eIF4F complex consisting of eIF4E, eIF4A and eIF4G, binds to 5’cap-mRNA (step 3). Next, the 43S PIC is recruited onto the mRNA (step 4). eIF4A helps release mRNA secondary structure in 5’UTR and help facilitate pre-initiation 43S complex scanning of 5’UTR in 5’ to 3’ direction. Upon the AUG start codon recognition, all the translation factors (i.e., eIFs) are released that follows the joining of a 60S large subunit, leading to the formation of elongation-competent 80S ribosome marking the end of the translation initiation stage of translation.
Figure 1. Schematic representation of eukaryotic translation initiation. Translation initiation is a multi-step process. In step 1, the ternary complex (TC) is formed wherein eIF2 (α, β, and γ), initiator methionyl tRNA (Met-tRNAMeti), and GTP assemble. In step 2, 40S ribosomal subunit, ternary complex, eIF1, eIF1A, eIF3, and eIF5 assembles to form 43S pre-initiation complex PIC). Simultaneously, the eIF4F complex consisting of eIF4E, eIF4A and eIF4G, binds to 5’cap-mRNA (step 3). Next, the 43S PIC is recruited onto the mRNA (step 4). eIF4A helps release mRNA secondary structure in 5’UTR and help facilitate pre-initiation 43S complex scanning of 5’UTR in 5’ to 3’ direction. Upon the AUG start codon recognition, all the translation factors (i.e., eIFs) are released that follows the joining of a 60S large subunit, leading to the formation of elongation-competent 80S ribosome marking the end of the translation initiation stage of translation.
Ijms 25 10835 g001
Figure 2. Schematic representation of eukaryotic translation elongation followed by termination. Translation elongation marks the peptidyl transfer reaction happening in the core of the ribosome. eEF1A brings amino-acyl-tRNA to the A-site of the 80S ribosome. eEF1A is released following GTP hydrolysis. In the next step, a peptidyl transfer reaction occurs by eEF2, wherein nucleophilic tRNA on the A site attacks the electrophilic peptidyl-tRNA in the P-site. Upon reaching the stop codon, eukaryotic release factors (RFs) bind to the A-site and allow the release of the completed polypeptide chain, marking the termination of the translation process.
Figure 2. Schematic representation of eukaryotic translation elongation followed by termination. Translation elongation marks the peptidyl transfer reaction happening in the core of the ribosome. eEF1A brings amino-acyl-tRNA to the A-site of the 80S ribosome. eEF1A is released following GTP hydrolysis. In the next step, a peptidyl transfer reaction occurs by eEF2, wherein nucleophilic tRNA on the A site attacks the electrophilic peptidyl-tRNA in the P-site. Upon reaching the stop codon, eukaryotic release factors (RFs) bind to the A-site and allow the release of the completed polypeptide chain, marking the termination of the translation process.
Ijms 25 10835 g002
Figure 3. Collection of selected translation inhibitors targeting different translation factors. (A) eIF4A inhibitors: silvestrol and other synthetic Rocaglamide analog CR-1-31-B, Zotatiffin, and marine compound eIF4A inhibitor Pateamine A. (B) eIF4E inhibitors: ribavirin, 4EG1-I, 4E1Rcat, and 4E2RCat. (C) eEF1A inhibitors: didemnin A, plitidepsin, narciclasine, and nannocyctin.
Figure 3. Collection of selected translation inhibitors targeting different translation factors. (A) eIF4A inhibitors: silvestrol and other synthetic Rocaglamide analog CR-1-31-B, Zotatiffin, and marine compound eIF4A inhibitor Pateamine A. (B) eIF4E inhibitors: ribavirin, 4EG1-I, 4E1Rcat, and 4E2RCat. (C) eEF1A inhibitors: didemnin A, plitidepsin, narciclasine, and nannocyctin.
Ijms 25 10835 g003
Figure 4. Potential direct inhibitors of translation factors are being explored for developing novel cancer therapeutics. Essential and potential drug target translation factors in the translation machinery and the compounds that target them are shown. Red bar-headed lines indicate inhibition. As shown, almost every translation factor in translation initiation can be targeted by small-molecule inhibitors, including eukaryotic translation initiation factor 2α (eIF2α) phosphorylation and ternary complex formation (upper left), eIF4E and eIF4F complex formation, enzymatic activity and cap-binding (upper right), eIF4A (center), eIF4G1 (center right), eIF3 (upper middle) as well as first peptide bond formation or initiating ribosome (center left) and translation elongation by eEF1A (bottom left).
Figure 4. Potential direct inhibitors of translation factors are being explored for developing novel cancer therapeutics. Essential and potential drug target translation factors in the translation machinery and the compounds that target them are shown. Red bar-headed lines indicate inhibition. As shown, almost every translation factor in translation initiation can be targeted by small-molecule inhibitors, including eukaryotic translation initiation factor 2α (eIF2α) phosphorylation and ternary complex formation (upper left), eIF4E and eIF4F complex formation, enzymatic activity and cap-binding (upper right), eIF4A (center), eIF4G1 (center right), eIF3 (upper middle) as well as first peptide bond formation or initiating ribosome (center left) and translation elongation by eEF1A (bottom left).
Ijms 25 10835 g004
Table 1. A comprehensive overview of translation factors, their functions, and their aberrant expression in human cancer. This summary provides an understanding of the key players in translation and their potential as targets for cancer therapeutics.
Table 1. A comprehensive overview of translation factors, their functions, and their aberrant expression in human cancer. This summary provides an understanding of the key players in translation and their potential as targets for cancer therapeutics.
Translation FactorFunctionExpression DiscrepancyType of Cancer
eIF1 Maintains the integrity and stringency of start codon selection and mRNA scanning Low expressionHepatocellular Carcinoma, Pancreas Cancer [20,21,22]
eIF1AMaintains mRNA scanning in cooperation with eIF1, helps start codon recognition, and prevents premature 40S joining.MutationsUveal Melanomas, Thyroid, Ovarian, and Leptomeningeal Melanocytic Neoplasms [23,24,25]
eIF2A/B Ternary complex (TC) formation with initiator tRNA, pre-initiation complex maintenance, and preventing 40S ribosomal subunit premature joining. OverexpressionNon-small-cell Lung Carcinoma, Melanocytic Neoplasm, Gastrointestinal, Brain, Thyroid, Lymphoma, Sarcoma [26,27,28,29,30,31,32]
eIF3 complexOrganization and maintenance of the stability of 43S pre-initiation complex, prevention of premature binding between small (40S) and large (60S)ribosomal subunits.OverexpressionBreast, Bladder, Cervix, Esophagus, Lung, Stomach, Colon, Ovary, Pancreas, Vulva, Prostrate, Hepatocellular Carcinoma [22,33,34,35,36,37,38]
eIF4AIt plays a crucial part in the eIF4F complex, is a critical player in translation initiation, and has an RNA helicase activity that unwinds secondary structures in mRNA’s 5’UTR. OverexpressionHepatocellular Carcinoma, Melanoma, Lymphoma, Breast, Lung [11,39,40,41,42]
eIF4EPart of the eIF4F complex binds to mRNA 5’Cap. It stimulates eIF4A unwinding activity and recruitment of the pre-initiation complex. Overexpression and hyper-phosphorylationBreast, Bladder, Brain, Cervix, Colon, Liver, Lung, Head & Neck, Prostrate, Skin [28,43,44,45,46,47,48,49]
eIF4G1It is the scaffold factor of the eIF4F complex, crucial for its assembly. It stimulates the helicase activity of eIF4A and the cap-binding efficiency of eIF4E. Its interactions with eIF1 facilitate mRNA scanning.OverexpressionBreast, Cervix, Lung,
Nasopharyngeal [50,51,52,53]
eIF5This guanine transferase is required to activate multiple translation factors. It maintains a stable connection of ribosomal subunits during subunit joining or 80S ribosome formation.Overexpression Hepatocellular carcinoma, Lung, Glioblastoma, Cervix, Colorectal [54,55,56,57]
eIF6 Regulation of ribosomal biogenesis and premature binding of ribosomal subunits. OverexpressionColorectal, Ovarian, Leukemia, Head & Neck, Colon, Rectum, Pancreas, Lung [58]
eEF1A Delivers all aminoacyl-tRNA to the ribosome and catalyzes the peptidyl transferase step during the elongation. OverexpressionBlood, Breast, Lung, Liver, Prostrate, Pancreas, Thyroid, Kidney [51,59,60,61,62,63,64,65]
eEF2 Catalysis of ribosomal translocation step during elongation. OverexpressionGastric, Colorectal Carcinoma, Breast, Prostrate, Lung, Hepatocellular Cancer [66]
Table 2. Therapeutic direct inhibitors of translation in cancer. The table below lists translation inhibitors specific to translation factors and their development stage.
Table 2. Therapeutic direct inhibitors of translation in cancer. The table below lists translation inhibitors specific to translation factors and their development stage.
Translation FactorMechanismInhibitorDevelopment Stage
eIF1eIF1 interaction with eIF4G1,
blocking scanning and AUG recognition
i14G1-10
i14G1-12
Preclinical: activity in in vitro multiple cancer cell lines
eIF2α eIF2α phosphorylation activation by HRI BTdCPUPreclinical: activity in xenograft models
eIF2α eIF2α dephosphorylation inhibition SalubrinalClinical: synthetic lethal in combination with proteasome inhibitor; Phase II clinical trials in combination with Carfilzomib (NCT01775553)
Ternary Complex Met-tRNAi-eIF2 interaction inhibition NSC119889, NSC119893Preclinical: in vitro translation inhibition
eIF3aReduction in eIF3a expressionMimosine, NCE22, and NCE30 Preclinical: in vitro cytotoxic activity
eIF3hInhibition of eIF3h activityCM16Preclinical: in vitro cytotoxic activity
eIF4AInhibition of eIF4A helicase activitySilvestrol
CR-1-31-B
Zotatiffin
Pateamine A
Hippuristanol
Preclinical: excellent efficacy in multiple cancer xenograft models for silvestrol, CR-1-31-B, Pateamine A, and Hippuristanol
Clinical: Zotatiffin (eFT226) is being studied in solid tumor malignancies (NCT04092673)
eIF4EInhibition of eIF4E-Cap interactionsRibavirin
7-BnGMP (4Ei-1)
Preclinical: antiproliferative activity in vitro
eIF4EInhibition of eIF4E-eIF4G1 interactions4EGI-1
4E1RCat
4E2RCat
Preclinical: antiproliferative activity in vitro and in vivo xenograft models
eIF4G1Interaction with eIF1 and eIF4E
blocking scanning and AUG recognition
i14G1-10
i14G1-12
4EGI-1
Preclinical: antiproliferative activity in vitro
eIF6Interaction with eIF1 and 60SeIF1sixty-1
eIF1sixty-4
eIF1sixty-6
Preclinical: antiproliferative activity in vitro and in vivo cancer models
eEF1A Irreversible binding to eEF1A, blocking its peptidyl transferase activity Didemnin B
Plitidepsin
Narsiclasine
Nannocystin
Ansatrienin A
LQ18
Preclinical: antiproliferative activity in vitro and in vivo cancer models
Clinical: Plitidepsin has been approved for treatment in multiple myeloma in combination with dexamethasone
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sehrawat, U. Exploiting Translation Machinery for Cancer Therapy: Translation Factors as Promising Targets. Int. J. Mol. Sci. 2024, 25, 10835. https://doi.org/10.3390/ijms251910835

AMA Style

Sehrawat U. Exploiting Translation Machinery for Cancer Therapy: Translation Factors as Promising Targets. International Journal of Molecular Sciences. 2024; 25(19):10835. https://doi.org/10.3390/ijms251910835

Chicago/Turabian Style

Sehrawat, Urmila. 2024. "Exploiting Translation Machinery for Cancer Therapy: Translation Factors as Promising Targets" International Journal of Molecular Sciences 25, no. 19: 10835. https://doi.org/10.3390/ijms251910835

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop