Next Article in Journal
Transcriptomic Profiling the Effects of Airway Exposure of Zinc Oxide and Silver Nanoparticles in Mouse Lungs
Next Article in Special Issue
New 4,5-Diarylimidazol-2-ylidene–iodidogold(I) Complexes with High Activity against Esophageal Adenocarcinoma Cells
Previous Article in Journal
Sensing and Stimulation Applications of Carbon Nanomaterials in Implantable Brain-Computer Interface
Previous Article in Special Issue
Synthetic Small Molecule Modulators of Hsp70 and Hsp40 Chaperones as Promising Anticancer Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anticancer Small-Molecule Agents Targeting Eukaryotic Elongation Factor 1A: State of the Art

The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5184; https://doi.org/10.3390/ijms24065184
Submission received: 16 February 2023 / Revised: 5 March 2023 / Accepted: 7 March 2023 / Published: 8 March 2023

Abstract

:
Eukaryotic elongation factor 1A (eEF1A) canonically delivers amino acyl tRNA to the ribosomal A site during the elongation stage of protein biosynthesis. Yet paradoxically, the oncogenic nature of this instrumental protein has long been recognized. Consistently, eEF1A has proven to be targeted by a wide assortment of small molecules with excellent anticancer activity, among which plitidepsin has been granted approval for the treatment of multiple myeloma. Meanwhile, metarrestin is currently under clinical development for metastatic cancers. Bearing these exciting advances in mind, it would be desirable to present a systematic up-to-date account of the title topic, which, to the best of our knowledge, has thus far been unavailable in the literature. The present review summarizes recent advances in eEF1A-targeting anticancer agents, both naturally occurring and synthetically crafted, with regard to their discovery or design, target identification, structure–activity relationship, and mode of action. Their structural diversity and differential eEF1A-targeting mechanisms warrant continuing research in pursuit of curing eEF1A-driven malignancy.

1. Introduction

The majority of cancers arise from accumulated somatic mutations, which over time, transform the cell into a state of malignancy. This is characterized by uncontrolled proliferation, aggressive invasion into surrounding normal tissues, and ultimately lethal metastasis at distant vital organs. Genomic instability and phenotypic heterogeneity inherent in each malignant tissue [1] render the treatment of cancer extremely challenging [2]. Today, apart from surgical operation, a variety of promising therapeutic strategies have been developed, including but not limited to radiotherapy [3], chemotherapy [4], biological therapy [5], immunotherapy [6], and microbial-based therapy [7,8]. From this ever-growing curative armamentarium, targeted chemotherapy with small-molecule or biomacromolecular agents is an indispensable measure [9,10,11].
Eukaryotic elongation factor 1A (eEF1A, formerly termed eEF-1α) is an essential GTPase evolutionarily conserved across diverse eukaryotes [12]. As the second most abundant intracellular protein after actin, it is localized extensively in the cytoplasm and nucleus [13,14,15]. The canonical function of eEF1A is to deliver amino acyl tRNAs to the ribosomal A site during the elongation stage of protein synthesis [16]. Strikingly, beyond this housekeeping role for the translational machinery and many other moonlighting functions [17,18,19,20], mounting evidence has pointed to a causal link between eEF1A and malignancy [21,22,23], suggesting that eEF1A may serve as not only an out-of-control translational cofactor [24,25,26] but also a signal transducer woven into a network of protumorigenic pathways [27,28,29,30,31,32]. Consistent with this revelation, structurally distinct small-molecule anticancer agents continue to emerge with a proven eEF1A-targeting mechanism [33]. Early examples include didemnin B [34], plitidepsin (dehydrodidemnin B) [35], tamandarin A [36], cytotrienin A [37], ansatrienin B [38], narciclasine [39], and synthetic flavonoids [40]. Among them, plitidepsin was approved in Australia for combined treatment of relapsed/refractory multiple myeloma with dexamethasone [41], thus providing initial proof of principle that eEF1A inhibitors can achieve the desired therapeutic efficacy with safety. In recent years, more intriguing compounds of this kind have been discovered and actively investigated. As a notable case, metarrestin is currently in a phase I clinical trial for the treatment of metastatic solid tumors [42]. Though a number of reviews have been published on protein-synthesis inhibitors [43,44,45,46], to the best of our knowledge, no systematic survey has ever been conducted on eEF1A-targeting agents. Hence, the present review delineates the state of the art on eEF1A-targeting small-molecule anticancer agents with a special focus on those actively studied over the recent years, covering their discovery or design, target identification, structure–activity relationship (SAR), and mode of action.

2. Recent Advances in Anticancer eEF1A-Targeting Agents

2.1. Didemnins and Tamandarins

Didemnins are a family of marine cyclic depsipeptides with strong anticancer, antiviral, and immunosuppressive activities [47]. Since their initial discovery in the early 1980s [48], these macrocycles have become the subject of intense research over the last four decades. Among them, didemnin B (1, Figure 1) and plitidepsin (2) have entered multiple clinical trials. Gratifyingly, plitidepsin was approved in Australia for treating multiple myeloma [41]. With nearly identical architectures but subtly different macrocyclic backbones (highlighted red), tamandarins such as tamandarin A (3) were discovered from a different colony of marine ascidian [49]. As already reviewed in multiple comprehensive monographs [36,50,51,52,53,54,55,56], these compounds will not be reiterated here. Instead, a brief update is presented below on their anticancer mode of action.
Having developed functional signature ontology (FUSION) maps for drug discovery and mechanistic elucidation [57], White et al. revisited the mechanism of 1 and found that it induces rapid and extensive apoptosis in sensitive cancer cell lines through concomitant inhibition of palmitoyl-protein thioesterase 1 (PPT1) and eEF1A1 [58]. Independently, Galmarini et al. showed that eEF1A2 is the specific binding target of 2 with a measured dissociation constant (KD) of 80 nM [35]. Since translation inhibition cannot account per se for the observed antiproliferative effect of 2, it was suspected that this drug impacts non-canonical functions of eEF1A2. Indeed, double-stranded RNA (dsRNA)-dependent protein kinase (PKR) was later identified as a novel binding partner of eEF1A2 [30]. In this case, eEF1A2 interacts directly with PKR to block its pro-apoptotic activity and boost tumor survival. In the presence of 2, however, PKR was disengaged from eEF1A2, thereby regaining its kinase activity to initiate extrinsic apoptosis through activation of MAPK and NF-κB signaling cascades [30]. More recently, Martinez-Leal et al. reported that 2 induces endoplasmic reticulum (ER) stress in HeLa cells by activating the multipronged unfolded protein response (UPR) in a characteristic pattern [59]. Working simultaneously as an ER stress inducer and an autophagy inhibitor, 2 was combined with bortezomib to synergistically block proteasomal degradation and autophagy, thereby exacerbating accumulation of misfolded proteins that originate from plitidepsin-induced oxidative stress. Such elevated proteotoxic stress led to apoptosis both in vitro (in MM1S multiple myeloma cells) and in vivo (in SCID mice xenografted with RPMI-8226 multiple myeloma cells). This study shows the promise of combined anticancer therapy using plitidepsin and proteasomal inhibitors such as bortezomib in a clinical setting.

2.2. Cytotrienin A and Ansatrienin B

Cytotrienin A (4, Figure 2) was initially isolated from the culture broth of soil-dwelling Streptomyces sp. RK95-74 [60]; this compound has strong cytotoxicity (IC50 = 7.7 nM) against human leukemia cell line HL-60 [61]. Despite early mechanistic studies [62,63,64], its target remained elusive until Pelletier et al. identified 4 as a translation inhibitor through high-throughput screening [37]. Their finding is that akin to didemnin B, cytotrienin A modulates eEF1A-dependent loading of aa-tRNA to the ribosome, most likely by stabilizing the eEF1A/GTP/aa-tRNA assembly positioned at the ribosomal A site. Thus, without release of eEF1A from the ribosome, translation elongation stops. Further insight into this compound’s mode of action came from a 2015 study led by Taunton, who aimed to seek out the target of a potent antiproliferative ternatin derivative 7 (ternatin-4, cross-refer to the following section) [38]. With the help of photoaffinity labeling, they were able to capture the binding partner with a ternatin-based probe. Interestingly, the photolabeled protein is a ternary complex comprising eEF1A, GTP, and aa-tRNA rather than eEF1A alone. Subsequent competitive-binding experiments noted that the photolabeling efficiency is diminished dose-dependently with the addition of didemnin B or ansatrienin B (5, a close side-chain analogue of cytotrienin A shown in Figure 2). Therefore, it was concluded that ternatin, didemnin B, and cytotrienin A/ansatrienin B may share a binding hot spot on the eEF1A surface, probably located near A399, as indicated by resistance-conferring mutation experiments.

2.3. Ternatin-4

The highly cytotoxic natural product A3, together with several other congeners, was isolated from an Aspergillus fungus [65]. Although its structure was determined to be a partially N-methylated cyclic heptapeptide, the chirality of 7 out of 11 stereo-centers (marked in the structure of A3, Figure 3) remained unknown. The strong structural similarity between A3 and ternatin (6, CAS registry number: 148619-41-4) [66], another natural product with anti-obesity activity [67], inspired Taunton et al. to design ternatin-4 (7) by incorporating the dehydromethyl leucine and pipecolic acid residues of A3 (highlighted red in the structure of 7, Figure 3) into 6 [38]. The resulting hybrid molecule 7 attained more than 10-fold enhancement of potency over the parent compound 6 (IC50 4.6 nM vs. 71 nM against HCT-116 cancer cell line), thus solving all but one stereo-configuration of A3. Wondering the molecular target of ternatins, they developed a bifunctional photoaffinity probe 10. Under UV irradiation, its photolabile diazirine subunit at residue 4 (highlighted red) decomposes into a highly reactive carbene that instantaneously crosslinks to the nearby binding protein. The alkyne at residue 6 (highlighted blue) will then connect to a fluorescent reporter via click cycloaddition, thus tagging the photolabeled target for characterization. In this way, eEF1A was captured and confirmed as the target of ternatins.
Based on the verified structure of ternatin-4 (7), a β-hydroxyl group was introduced at its residue 3 to obtain two epimers of A3, namely SR-A3 (8) and SS-A3 (9). An improved second-generation total synthesis allowed quick access to both compounds [68]. Hence, the identity of natural product A3 was established as SR-A3 (8). Of special note, despite a minor difference in the side chain of residue 3, 8 was found to display a more prolonged duration of growth inhibition action than 7 and 9. Single-molecule fluorescence resonance energy transfer (smFRET) imaging corroborated this observation, while further quantification through in vitro chase experiments confirmed enhanced drug–target residence time (table inset in Figure 3) and rebinding kinetics of 8. Finally, preclinical evaluation of 8 vis-a-vis 7 was carried out in an aggressive Myc-driven mouse lymphoma model. Compared with its des-hydroxyl variant ternatin-4 (7), SR-A3 (8) significantly reduced tumor burden while extending the survival of the treated Eμ-Myc mice [68]. This work highlights the importance of side-chain modification in macrocyclic drug discovery and also makes a good case that the drug–target interaction can be more precisely characterized using the drug–target residence time model [69]. More recently, with the help of smFRET imaging and cryogenic electron microscopy (cryo-EM), Taunton and collaborators demonstrated that in spite of sharing a common eEF1A-binding site, ternatin-4 (7) and didemnin B exhibit differential inhibition dynamics in that the former traps the eEF1A/GDP/aa-tRNA ternary complex on the ribosome in a more reversible fashion than does the latter [70]. Their in-depth mechanistic investigation also revealed that by trapping eEF1A at the ribosomal A site, ternatin-4 induces ubiquitination and degradation of eEF1A on stalled ribosomes through a previously unknown surveillance pathway for translation quality control [71].

2.4. Nannocystin A

Nannocystin A (11, Figure 4) is a 21-membered cyclic depsipeptide isolated independently by Brönstrup et al. [72] and Hoepfner et al. [73] from the myxobacteria of the Nannocystis genus. Brönstrup et al. found that 11 is a strong inducer of apoptosis, as such inhibiting the growth of multiple cancer cell lines with low nanomolar IC50 values [72]. Meanwhile, another team led by Hoepfner pinned down its target through a combination of genetic and chemoproteomic approaches [73]. In brief, initial haploinsufficiency profiling and mutagenesis experiments implied eEF1A as the most likely target. To verify direct binding, they set up affinity chromatography with the semisynthetic probe 12 (Figure 4). During elution, this immobilized nannocystin sequestered eEF1A1 and eEF1A2 out of the 3644 proteins comprising the HCT-116 cell lysates. Moreover, it competed with unbound nannocystin A (11) and didemnin B for binding to eEF1A. Hence, eEF1A was determined to be the target of nannocystin A [73].
The firsthand structure–activity relationship of nannocystins was derived from isolated and semisynthetic nannocystins [72,73], which indicates that modification at the tyrosine phenol moiety (subdomain I in the structure of 11, Figure 4) or the side chain of N-Me-L-isoleucine (subdomain II) is tolerated. To obtain comprehensive SAR, nevertheless, total synthesis is a must. Thus far, seven routes have been reported for the total syntheses of nannocystin A (11) or its 2E-alkene surrogate nannocystin Ax (16, structure shown in Table 1) [74,75,76,77,78,79,80,81], each involving a distinct macrocyclization reaction as the key strategic step [82]. With the dual purpose of (1) total synthesis and (2) SAR validation concerning the binding role of the polyketide C5-C7 region, Fürstner et al. devised a motif-oriented strategy so that the macrocyclic propargylic alcohol 13 (shown in Figure 4) underwent post-macrocyclization elaboration [83] into an array of novel analogues besides nannocystin Ax (16) [80]. It was found that the 5R-methoxy ether (subdomain VI), instead of the neighboring C6-C7 (E)-alkene (subdomain VII), must be reserved for high activity. As an illustration, Table 1 compares the anticancer activity of four pairs of nannocystin derivatives with or without methylation at the C5-OH group (R = Me or H). Clearly, removal of this moiety causes a drastic reduction in potency (16 vs. 17, 18 vs. 19, 20 vs. 21, 22 vs. 23); on the other hand, changing the C6-methyl (16) to a fluorine (18) or hydrogen (20) atom, or even curtailing the (Z) alkene to an alkyne (22), has insignificant impact on activity.
Following a total synthesis of nannocystin A [76] via Heck macrocyclization [84,85], we prepared a diversity of non-natural nannocystins modified at different sites. Our findings demonstrated that (1) the (2R, 3S)-epoxide (subdomain V) may be substituted for a 2E-alkene without compromising activity [86], (2) the side chain of β-OH-L-valine (subdomain III) is tolerant of minor change [86], and (3) the polyketide C9-C10 segment including its entire (10R, 11S) stereo-chemistry (subdomain VIII) is a key determinant of potency [87,88]. In parallel, He et al. synthesized more variants via Heck macrocyclization too and observed that removal of the N-methyl moiety (subdomain IV) incurred a dramatic loss of activity [89]. Taken together, the SAR of nannocystins is illustrated in the structure of 11 (Figure 4).
Aside from target elucidation and SAR profiling, the exact mechanism of nannocystins is a subject of enduring interest [90,91,92] given the poorly understood role of eEF1A in tumorigenesis. Chen et al. showed that the antimetastatic effect of nannocystin Ax in lung cancer cells is attributable to its interference with the TGF-β/Smad signaling pathway [90]. Their result is in step with another study uncovering the promigratory ability of eEF1A2 to promote lung adenocarcinoma metastasis [32]. The additional finding that the regulation of TGFβI (TGF β receptor I) by nannocystin Ax occurs at the transcriptional rather than the (post-)translational level implied the presence of an alternative mechanism independent of eEF1A inhibition [90]. Therefore, similar to the case of plitidepsin [30], the possibility that nannocystins impact certain protumorigenic pathway(s) mediated by eEF1A cannot be ruled out. Recently, we designed a serine-incorporating nannocystin 14 (Figure 4) to leverage a post-macrocyclization diversification strategy for efficient side-chain variation [92]. Thus obtained SAR concurred with the general trend depicted in Figure 4 and further informed the development of a coumarin-conjugated fluorescent probe 15. With good permeability into the cancer cells, this probe was localized to the ER, as visualized by confocal fluorescence microscopy, which implies that nannocystins act on eEF1A predominantly at the ER-bound ribosome. Our result is in good agreement with the latest work by Förster et al. capturing eEF1A associated with the ribosome at the ER membrane by the use of cryo-electron tomography [93], thereby shedding light on the intracellular mode of action of nannocystins.

2.5. Metarrestin

Perinucleolar compartment (PNC) is a heritable multicomponent dynamic subnuclear organelle located at the periphery of the nucleolus of eukaryotic cells and uniquely associated with metastatic cancer cells [94]. Huang et al. found that PNC prevalence, defined as the percentage of non-apoptotic and non-mitotic cells harboring at least one PNC, is a pan-cancer prognostic marker positively correlated with metastatic capacity [95,96]. Subsequent screening of clinically approved anticancer drugs led to the observation that some of these drugs are capable of reducing PNC prevalence via specific on-target inhibition in lieu of promiscuous toxicity [97]. Having confirmed the existence of mechanism-specific PNC disassemblers with clinical efficacy, this proof-of-concept study supported taking PNC prevalence reduction as a phenotypic screening marker to discover antimetastatic drugs. To this end, a metastatic prostate cancer cell line PC3M with a PNC prevalence of 75% to 85% was engineered to stably express green fluorescent protein (GFP)-fused polypyrimidine-tract-binding protein (PTB) [98]. PTB is an essential PNC marker routinely measured by immunohistochemistry, which is unfortunately inconvenient for automated screening. But now with the self-reporting fluorescent PC3M-GFP-PTB cell line at hand, they were able to establish an image-based high-throughput, high-content assay (HCA) primed for spotting compounds able to reduce PNC prevalence by 50% [99]. Aiming at antimetastasis, the initial hits underwent secondary assays to select for invasion inhibition while excluding those acting via apoptosis induction, DNA intercalation, general cytotoxicity, or cell-cycle arrest [100]. By means of this multistage screening protocol, two leads were eventually identified out of 140,800 structurally diverse compounds from the NIH Molecular Libraries Small Molecule Repository (MLSMR) due to their outstanding PNC-disassembling efficiency and low cytotoxicity, thus setting the stage for the ensuing medicinal chemistry campaign [101].
After a preliminary exploration, pyrrolopyrimidine 24 (Figure 5) was favored over the other lead (structure not shown) for systematic optimization. Robust synthetic methods were next developed to access a broad variety of analogues evaluated for PNC disassembly and drug-like properties as well. The SAR trends are summarized in the structure of 24 (Figure 5). Specifically, (1) the N-3 substitution at the subdomain I prefers a linear alkyl chain bearing a hydroxy, ether, or amine, and conformational constraint with a cyclohexyl ring gives rise to the highest potency; (2) the N-7 position at the subdomain II tolerates a benzyl, phenethyl, or 4-methoxylphenyl group, but the presence of an alkyl substituent diminishes the potency significantly; (3) the unsubstituted C5 and C6 phenyl rings at the subdomain III are indispensable for high potency. While deducing the above trends, multi-round optimization finally yielded metarrestin (25), which possesses a superior selectivity window between PNC reduction and cell viability compared with the classic anticancer drugs doxorubicin and camptothecin (table inset in Figure 5) [101]. The in vitro performance of 25 was smoothly translated into in vivo efficacy in three mouse models of human cancer, where it suppressed metastatic invasion with concomitant reduction in PNC prevalence in the cancer cells of primary and metastasized tumors, offering a remarkable survival advantage to the treated animals [100]. After an in-depth evaluation of its pharmacokinetics [102,103] and safety [104], this drug has been advanced into a phase I clinical trial for the treatment of metastatic solid tumors [42].
The excellent antimetastatic capability of 25 prompted Huang et al. to investigate its mechanism, with the primary conclusion that the drug disrupts PNC assembly by blocking RNA polymerase I transcription [100]. Further seeking the binding target of 25, they designed a biotin-conjugated probe 26 (Figure 5) that is likewise efficacious in disassembling PNC. Affinity purification with 26 combined with competition experiments using untagged metarrestin identified eEF1A2 as the binding target. The metarrestin–eEF1A2 interaction was confirmed by cellular thermal shift assay. Subsequent experiments along this line of research showed that (1) eEF1A2 enhances PNC assembly and metastatic progression; and (2) eEF1A2, at least in part, mediates the PNC-elimination function of metarrestin [100]. Whereas further details await elucidation, it was believed that metarrestin interferes with certain non-translational functions of eEF1A2. Recently, Jin et al. developed a proteolysis-targeting chimera (PROTAC) [105,106,107] strategy by tethering metarrestin with various ligands for the von Hippel–Lindau (VHL) E3 ligase [108]. Thus obtained heterobifunctional molecules were designed to recruit eEF1A2, the binding target of metarrestin, to the ubiquitin/proteasome system (UPS) for selective degradation. As one of these first-in-class eEF1A2 degraders, 27 (Figure 5) was shown to degrade eEF1A2 in three cancer cells in a dose-dependent manner, thus holding promise for the treatment of eEF1A2-mediated carcinogenesis.

2.6. 2-Phenyl-3-Hydroxy-4(1H)-Quinolinones

2-Phenyl-3-hydroxy-4(1H)-quinolinones (abbreviated as 3-HQs) such as 28 (Figure 6) are aza-analogues of previously reported eEF1A-targeting anticancer flavonoids [40]. Based on a homology model of human eEF1A1, Hlavac et al. carried out docking calculations to identify the binding site for gamendazole, a known eEF1A1 inhibitor for male contraception [109]. Encouragingly, they found that these 3-HQs fit into the same gamendazole-binding site on the surface of eEF1A1. Such direct interaction between eEF1A1 and 3-HQs was verified through pull-down assay using biotinylated 3-HQ derivatives [110]. Having validated the constructed eEF1A1 model, the authors performed virtual screening of in silico designed 3-HQs with varying substituents R1, R2, and R3 (illustrated in the structure of 28). The six highest-scored and synthetically accessible compounds were chosen for wet-lab preparation. Their binding to eEF1A1 was quantitatively characterized with isothermal titration calorimetry (ITC), which provided thermodynamic information consistent with docking calculation results. Biological evaluation discovered 29, one of these rationally designed eEF1A1 inhibitors, with optimal inhibitory activity against several cancer cell lines but low toxicity toward a normal cell line [110].

2.7. Cordyheptapeptide A

Cordyheptapeptide A (30), a partially N-methylated cyclic heptapeptide with anticancer activity, was originally isolated from the insect pathogenic fungus Cordyceps [111,112]. Although a solution-phase total synthesis of 30 was reported before [113], its SAR and mechanism of action were unclear. Lokey et al. developed a high-throughput solid-phase peptide synthesis (SPPS) to access a library of side-chain- and backbone-modified analogues [114]. They observed the following SAR trends: (1) all side chains are critical to its antiproliferative activity [115]; (2) halogenation at the aromatic side chain of residue 2 or 5 deteriorates activity at varying degrees; (3) whereas removal of the N-methyl moiety at residue 2 or 6 impairs activity, this is not the case for residue 4, for which changing sarcosine to glycine tends to improve activity, and when coupled with ortho-fluorination at residue 5, such N-demethylation brings about equipotent variant 31 with a 39-fold improvement in aqueous solubility (table inset in Figure 7). According to molecular dynamics simulations, the enhancement in activity stems from more conformational flexibility of its glycine-carrying scaffold, which is accordingly more accessible to target-binding conformations than the parent natural product 30.
To find out the mechanism of action of 30, the authors determined its cytotoxicity profile via the NCI60 human tumor cell line assay [114]. Analyzed by the COMPARE algorithm, this profile was best correlated with that of phyllanthoside, a known eukaryotic protein-synthesis inhibitor [116]. Consistently, cytological profiling (CP) [117] indicated that 30 clustered most closely with protein-synthesis inhibitors such as didemnin B and ternatin but deviated significantly from microtubule inhibitors and poly (ADP-ribose) polymerase (PARP) inhibitors. Combining the results from both phenotypic experiments, 30 is quite likely a protein-synthesis inhibitor. This inference was confirmed by bioorthogonal noncanonical amino acid tagging (BONCAT) [118], which proved that the agent primarily blocks protein synthesis and has a secondary influence on DNA synthesis. Suspecting its target to be eEF1A, 30 was evaluated in the HCT-116 cancer cells with a point mutation of eEF1A (A399V). Previously, the same mutation was reported to confer resistance to eEF1A-targeting didemnin B [119], ternatin [38], and nannocystin A [73]. Indeed, the activity dropped remarkably in the mutant cells, thus providing genetic evidence that supports eEF1A as the target of 30.

2.8. BE-43547A2

Isolated from Streptomyces sp. in 1998, BE-43547A1 (32, Figure 8), BE-43547A2 (33), and other congeners are a series of macrocyclic depsipeptides differing in the C21 side chain [120]. These compounds belong to the amidopentadienoate-containing cyclolipodepsipeptide (APD-CLD) natural products that feature an electrophilic 4-amido-2,4-pentadienoate (APD, highlighted red) functionality as well as a lipophilic side chain [121]. Poulsen et al. developed the first total synthesis of ent-32 and re-isolated the authentic 32 from the fermentation broth of a BE-43547-producing microorganism, thereby establishing the absolute stereo-configuration of the BE-43547 family [122]. Importantly, 32 and 33 exhibited superior hypoxia-selective cytotoxicity in PANC-1 pancreatic cancer cells than rakicidin A, another APD-CLD natural product they investigated earlier [123,124,125,126]. Shortly after this work, Chen et al. developed a total synthesis of 33 and reported that this agent selectively targets pancreatic cancer stem cells (PCSCs) [127,128]. Preparation of more analogues [129,130] led to the following SAR results (illustrated in Figure 8): (1) the exocyclic alkene at C8 within the APD unit must be reserved; (2) the macrolide cannot be changed to the corresponding macrolactam, or in other words, the O35 cannot be replaced with a nitrogen atom; (3) the (S)-hydroxyl group at C15 is critical to activity; (4) a lipophilic side chain at C21 is necessary but variable.
The excellent hypoxia-selective toxicity of APD-CLD natural products such as rakicidin A and the BE-43547A members intrigued Poulsen et al. to elucidate their mechanism of action, which is distinguished from conventional hypoxia-activated compounds. They showed that these APD-CLDs induced rapid and hypoxia-selective impairment of mitochondrial structure and function, thereby driving a peculiar form of non-apoptotic cancer cell death in a hypoxic milieu [131]. To discover the molecular target of 33, Chen et al. synthesized a clickable probe 34 [130]. Biotinylation of 34 via in situ click cycloaddition followed by pull-down assay indicated eEF1A as a binding target, and the isoform was determined to be eEF1A1 via immunoblotting. Having located the cysteine234 residue of eEF1A1 as the most probable binding site of 33 according to LC-MS/MS analysis, they engineered three types of pancreatic cancer cells with (1) eEF1A1 knockdown (KD), (2) eEF1A1 recovered from KD (RE-KD), and (3) C234-mutant eEF1A1 constructed from KD (RE-C234S). As shown in Table 2, the in vitro cytotoxicity of 33 against these three and the wild-type (WT) pancreatic cancer cells, as well as its in vivo anticancer efficacy in the four corresponding mouse xenograft models, provided concrete evidence favoring the Cys234 residue of eEF1A1 as the binding site for 33 [130]. Furthermore, it was shown that eEF1A1 plays a significant role in regulating pancreatic cancer cell stemness, its levels positively correlated with pancreatic cancer progression and negatively affecting patient survival.

3. Conclusions and Future Perspectives

Translational control with small-molecule agents represents an emerging direction for anticancer drug discovery [132,133,134]. As proofs of principle, hitherto two such drugs have gained approval for clinical use, namely homoharringtonine and plitidepsin. The former is the first protein translation inhibitor for the treatment of chronic myeloid leukemia, which works by fitting into the ribosomal A site so as to block access by the charged tRNA [135]. Pertaining to the present subject, the latter is a specific inhibitor of eEF1A that, at least in part, disrupts its canonical function of assisting translation elongation to combat multiple myeloma [35]. The whole ensemble of known eEF1A-targeting small-molecule agents is compiled in Table 3, alongside a brief summary of their anticancer effectiveness and selectivity. Intriguingly, though sharing the same molecular target, these compounds manifested differential antiproliferative profiles. For example, narciclasine [39], synthetic flavonoids [40], and ternatin-4 [68] within this category displayed preferential anticancer potency against human melanoma, breast carcinoma, and colorectal carcinoma cells, respectively. Presumably, such contrasting selectivity may partially stem from their distinct chemotypes that influence the drug–target interaction as well as the consequent therapeutic outcome in a subtle but profound way, as revealed very recently by Taunton et al. on the inhibitory mode of action of didemnin B and ternatin-4 at the single-molecule level [70]. Overall, the current review outlines the remarkable progress achieved over the recent years in eEF1A-targeting anticancer agents, which are structurally distinct macrocycles and heterocycles, either naturally occurring, developed based on the hit from high-throughput screening, or rationally designed. Their development status is summarized in Figure 9. For the time being, metarrestin is under clinical development as an unprecedented modality for controlling cancer metastasis [42], whereas metarrestin-based PROTACs have also been disclosed for selective degradation of eEF1A2 [108]. Albeit beyond the scope of this review, it is also worth noting that plitidepsin has now entered a clinical trial as a potential anti-SARS-CoV-2 drug [136] since its target eEF1A turned out to be a crucial host protein co-opted by virus to infect human cells [137,138].
In spite of the aforementioned advances, however, there remains much to learn about the exact mechanisms of action of these targeted agents. A more fundamental question lies in the oncogenic mechanism of eEF1A, a multitalented protein capable of both translation elongation and a myriad of moonlighting duties. As evidenced by an illuminating study on plitidepsin [30], it seems indeed viable for malignant cells to exploit certain non-canonical functions of eEF1A for survival. Moreover, the involvement of eEF1A1 in aggressive castration-resistant prostate cancer (CRPC) [139] and non-small cell lung cancer (NSCLC) metastasis [140] has been demonstrated through its complexation with actin and the eEF1A1/MDM2/MTBP signaling axis, respectively. These latest results showcase the potential opportunities of designing next-generation magic bullets [141] that act upon the manifold oncogenic functions of eEF1A, selectively extinguishing the malignant while sparing the normal. It is foreseeable that ongoing investigation of these mechanistic aspects, along with the expanding repertoire of eEF1A-targeting agents, will position us at a better forefront against cancer and other eEF1A-driven diseases as well.
Table 3. Summary of eEF1A-targeting small-molecule agents on their anticancer efficacy and selectivity.
Table 3. Summary of eEF1A-targeting small-molecule agents on their anticancer efficacy and selectivity.
CompoundAnticancer Efficacy and SelectivityReference
Didemnins
  • potent against human cancer cell lines from different tissues
  • in vivo validated in several preclinical models
  • didemnin B (1) and plitidepsin (2) have been studied in multiple clinical trials
  • plitidepsin (2, aplidin®) has been approved in Australia for combined treatment of relapsed/refractory multiple myeloma with dexamethasone
[36,41,50,51,52,53,54,55,56]
Tamandarins
  • potent against human cancer cell lines from different tissues
[36,49]
Cytotrienin A (4)
  • potent against human leukemia HL-60 cells (IC50 = 7.7 nM) and lung carcinoma A549 cells (IC50 = 0.1 μM)
[61,64]
Ansatrienin B (5)
  • potent against three human pancreatic cancer cell lines (IC50 range, 0.17–1.69 μM)
[142]
Narciclasine
  • potent against five human melanoma cell lines (IC50~40 nM)
  • in vivo validated in a mice xenograft model of brain metastatic melanoma
[39]
Synthetic flavonoids
  • potent against human breast cancer cell lines (IC50 range, 1–50 μM for MDA-MB231)
[40]
Ternatin-4 (7)
  • potent against human colorectal carcinoma HCT-116 cells (IC50 = 4.6 nM)
  • in vivo validated in an aggressive Myc-driven mouse lymphoma model
[68]
Nannocystin A (11)
  • potent against 472 cancer cell lines (IC50 range, 5–500 nM)
  • nannocystin Ax in vivo validated in an HCT-116-derived xenograft zebrafish model
[73,91]
Metarrestin (25)
  • excellent antimetastatic selectivity over cytotoxicity (cytotoxicity IC50/PNC reduction IC50 = 38.3)
  • in vivo validated in several preclinical models
  • currently in a phase I clinical trial for the treatment of metastatic solid tumors
[42,100,101,102,103,104]
Synthetic quinolinones
  • potent against human cancer cell lines from different tissues (IC50 range, 0.56–50 μM)
[110]
Cordyheptapeptide A (30)
  • potent against human colorectal carcinoma HCT-116 cells (IC50 = 0.2 μM)
[114]
BE-43547A2 (33)
  • potent against human pancreatic carcinoma PANC-1 cells (IC50 = 0.87 μM)
  • remarkable hypoxia-selective toxicity against human leukemia K562 cells and breast carcinoma MCF-7 cells (selective index = 28 and 79, respectively)
  • selectively targets pancreatic cancer stem cells (PCSCs)
  • in vivo validated in a pancreatic cancer xenograft mouse model
[127,130]

Author Contributions

Conceptualization and funding acquisition, W.Z.; data curation, H.Z. and J.C.; resources, S.Y.; visualization, B.S.; writing—original draft preparation, W.Z.; writing—review and editing, W.Z., H.Z., J.C., S.Y. and B.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work is generously supported by the National Natural Science Foundation of China (No. 21772101) and the Natural Science Foundation of Tianjin City (Grant No. 17JCYBJC28400).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Burrell, R.A.; McGranahan, N.; Bartek, J.; Swanton, C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature 2013, 501, 338–345. [Google Scholar] [CrossRef] [PubMed]
  2. Kitano, H. Cancer as a robust system: Implications for anticancer therapy. Nat. Rev. Cancer 2004, 4, 227–235. [Google Scholar] [CrossRef] [PubMed]
  3. Barker, H.E.; Paget, J.T.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Avendaño, C.; Menendez, J.C. Medicinal Chemistry of Anticancer Drugs, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2015. [Google Scholar]
  5. Papież, M.A.; Krzyściak, W. Biological therapies in the treatment of cancer—Update and new directions. Int. J. Mol. Sci. 2021, 22, 11694. [Google Scholar] [CrossRef]
  6. Kaiser, M.; Semeraro, M.D.; Herrmann, M.; Absenger, G.; Gerger, A.; Renner, W. Immune aging and immunotherapy in cancer. Int. J. Mol. Sci. 2021, 22, 7016. [Google Scholar] [CrossRef]
  7. Diwan, D.; Cheng, L.; Usmani, Z.; Sharma, M.; Holden, N.; Willoughby, N.; Sangwan, N.; Baadhe, R.R.; Liu, C.; Gupta, V.K. Microbial cancer therapeutics: A promising approach. Semin. Cancer Biol. 2022, 86, 931–950. [Google Scholar] [CrossRef]
  8. Sawant, S.S.; Patil, S.M.; Gupta, V.; Kunda, N.K. Microbes as medicines: Harnessing the power of bacteria in advancing cancer treatment. Int. J. Mol. Sci. 2020, 21, 7575. [Google Scholar] [CrossRef]
  9. Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target Ther. 2021, 6, 201. [Google Scholar] [CrossRef]
  10. Ohishi, T.; Kaneko, M.K.; Yoshida, Y.; Takashima, A.; Kato, Y.; Kawada, M. Current Targeted Therapy for Metastatic Colorectal Cancer. Int. J. Mol. Sci. 2023, 24, 1702. [Google Scholar] [CrossRef]
  11. Lau, K.H.; Tan, A.M.; Shi, Y. New and emerging targeted therapies for advanced breast cancer. Int. J. Mol. Sci. 2022, 23, 2288. [Google Scholar] [CrossRef]
  12. Negrutskii, B.S.; El’skaya, A.V. Eukaryotic translation elongation factor 1α: Structure, expression, functions, and possible role in aminoacyl-tRNA channeling. Prog. Nucleic Acid Res. Mol. Biol. 1998, 60, 47–78. [Google Scholar]
  13. Sanders, J.; Brandsma, M.; Janssen, G.M.C.; Dijk, J.; Moeller, W. Immunofluorescence studies of human fibroblasts demonstrate the presence of the complex of elongation factor-1βγδ in the endoplasmic reticulum. J. Cell Sci. 1996, 109, 1113–1117. [Google Scholar] [CrossRef] [PubMed]
  14. Kjær, S.; Wind, T.; Ravn, P.; Østergaard, M.; Clark, B.F.C.; Nissim, A. Generation and epitope mapping of high-affinity scFv to eukaryotic elongation factor 1A by dual application of phage display. Eur. J. Biochem. 2001, 268, 3407–3415. [Google Scholar] [CrossRef]
  15. Migliaccio, N.; Ruggiero, I.; Martucci, N.M.; Sanges, C.; Arbucci, S.; Tatè, R.; Rippa, E.; Arcari, P.; Lamberti, A. New insights on the interaction between the isoforms 1 and 2 of human translation elongation factor 1A. Biochimie 2015, 118, 1–7. [Google Scholar] [CrossRef] [PubMed]
  16. Dever, T.E.; Dinman, J.D.; Green, R. Translation elongation and recoding in eukaryotes. Cold Spring Harb. Perspect. Biol. 2018, 10, a032649. [Google Scholar] [CrossRef] [PubMed]
  17. Mateyak, M.K.; Kinzy, T.G. eEF1A: Thinking outside the ribosome. J. Biol. Chem. 2010, 285, 21209–21213. [Google Scholar] [CrossRef] [Green Version]
  18. Sasikumar, A.N.; Perez, W.B.; Kinzy, T.G. The many roles of the eukaryotic elongation factor 1 complex. Wiley Interdiscip. Rev. RNA 2012, 3, 543–555. [Google Scholar] [CrossRef] [Green Version]
  19. Li, D.; Wei, T.; Abbott, C.M.; Harrich, D. The unexpected roles of eukaryotic translation elongation factors in RNA virus replication and pathogenesis. Microbiol. Mol. Biol. Rev. 2013, 77, 253–266. [Google Scholar] [CrossRef] [Green Version]
  20. Carriles, A.A.; Mills, A.; Muñoz-Alonso, M.-J.; Gutiérrez, D.; Domínguez, J.M.; Hermoso, J.A.; Gago, F. Structural Cues for Understanding eEF1A2 Moonlighting. ChemBioChem 2021, 22, 374–391. [Google Scholar] [CrossRef]
  21. Thornton, S.; Anand, N.; Purcell, D.; Lee, J. Not just for housekeeping: Protein initiation and elongation factors in cell growth and tumorigenesis. J. Mol. Med. 2003, 81, 536–548. [Google Scholar] [CrossRef]
  22. Lamberti, A.; Caraglia, M.; Longo, O.; Marra, M.; Abbruzzese, A.; Arcari, P. The translation elongation factor 1A in tumorigenesis, signal transduction and apoptosis: Review article. Amino Acids 2004, 26, 443–448. [Google Scholar] [CrossRef]
  23. Abbas, W.; Kumar, A.; Herbein, G. The eEF1A Proteins: At the Crossroads of Oncogenesis, Apoptosis, and Viral Infections. Front. Oncol. 2015, 5, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hussey, G.S.; Chaudhury, A.; Dawson, A.E.; Lindner, D.J.; Knudsen, C.R.; Wilce, M.C.; Merrick, W.C.; Howe, P.H. Identification of an mRNP Complex Regulating Tumorigenesis at the Translational Elongation Step. Mol. Cell 2011, 41, 419–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Liu, S.; Hausmann, S.; Carlson, S.M.; Fuentes, M.E.; Francis, J.W.; Pillai, R.; Lofgren, S.M.; Hulea, L.; Tandoc, K.; Lu, J.; et al. METTL13 Methylation of eEF1A Increases Translational Output to Promote Tumorigenesis. Cell 2019, 176, 491–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kobayashi, D.; Tokuda, T.; Sato, K.; Okanishi, H.; Nagayama, M.; Hirayama-Kurogi, M.; Ohtsuki, S.; Araki, N. Identification of a specific translational machinery via TCTP-EF1A2 interaction regulating NF1-associated tumor growth by affinity purification and data-independent mass spectrometry acquisition (AP-DIA). Mol. Cell. Proteom. 2019, 18, 245–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Amiri, A.; Noei, F.; Jeganathan, S.; Kulkarni, G.; Pinke, D.E.; Lee, J.M. eEF1A2 activates Akt and stimulates Akt-dependent actin remodeling, invasion and migration. Oncogene 2007, 26, 3027–3040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Li, Z.; Qi, C.F.; Shin, D.M.; Zingone, A.; Newbery, H.J.; Kovalchuk, A.L.; Abbott, C.M.; Morse, H.C.I. Eef1a2 promotes cell growth, inhibits apoptosis and activates JAK/STAT and AKT signaling in mouse plasmacytomas. PLoS ONE 2010, 5, e10755. [Google Scholar] [CrossRef] [Green Version]
  29. Pellegrino, R.; Calvisi, D.F.; Neumann, O.; Kolluru, V.; Wesely, J.; Chen, X.; Wang, C.; Wuestefeld, T.; Ladu, S.; Elgohary, N.; et al. EEF1A2 inactivates p53 by way of PI3K/AKT/mTOR-dependent stabilization of MDM4 in hepatocellular carcinoma. Hepatology 2014, 59, 1886–1899. [Google Scholar] [CrossRef] [Green Version]
  30. Losada, A.; Munoz-Alonso, M.J.; Martinez-Diez, M.; Gago, F.; Dominguez, J.M.; Martinez-Leal, J.F.; Galmarini, C.M. Binding of eEF1A2 to the RNA-dependent protein kinase PKR modulates its activity and promotes tumor cell survival. Br. J. Cancer 2018, 119, 1410–1420. [Google Scholar] [CrossRef] [Green Version]
  31. Itagaki, K.; Sasada, M.; Iyoda, T.; Imaizumi, T.; Haga, M.; Kuga, A.; Inomata, H.; Fukai, F.; Miyazaki, S.; Kondo, Y.; et al. Exposure of the cryptic de-adhesive site FNIII14 in fibronectin molecule and its binding to membrane-type eEF1A induce migration and invasion of cancer cells via β1-integrin inactivation. Am. J. Cancer Res. 2020, 10, 3990–4004. [Google Scholar]
  32. Jia, L.; Ge, X.; Du, C.; Chen, L.; Zhou, Y.; Xiong, W.; Xiang, J.; Li, G.; Xiao, G.; Fang, L.; et al. EEF1A2 interacts with HSP90AB1 to promote lung adenocarcinoma metastasis via enhancing TGF-β/SMAD signalling. Brit. J. Cancer 2021, 124, 1301–1311. [Google Scholar] [CrossRef]
  33. 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, Human eFE1A has two predominant isoforms eEF1A1 and eEF1A2. According to the literature, some agents were reported to target eEF1A without knowing whether it is eEF1A1, eEF1A2, or both, whereas others target only one isoform. For a recent discussion about these two isoforms. [Google Scholar] [CrossRef]
  34. Crews, C.M.; Collins, J.L.; Lane, W.S.; Snapper, M.L.; Schreiber, S.L. GTP-dependent binding of the antiproliferative agent didemnin to elongation factor 1α. J. Biol. Chem. 1994, 269, 15411–15414. [Google Scholar] [CrossRef]
  35. Losada, A.; Munoz-Alonso, M.J.; Garcia, C.; Sanchez-Murcia, P.A.; Martinez-Leal, J.F.; Dominguez, J.M.; Lillo, M.P.; Gago, F.; Galmarini, C.M. Translation Elongation Factor eEF1A2 is a Novel Anticancer Target for the Marine Natural Product Plitidepsin. Sci. Rep. 2016, 6, 35100. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, J.; Currano, J.N.; Carroll, P.J.; Joullie, M.M. Didemnins, tamandarins and related natural products. Nat. Prod. Rep. 2012, 29, 404–424. [Google Scholar] [CrossRef] [PubMed]
  37. Lindqvist, L.; Robert, F.; Merrick, W.; Kakeya, H.; Fraser, C.; Osada, H.; Pelletier, J. Inhibition of translation by cytotrienin A--a member of the ansamycin family. RNA 2010, 16, 2404–2413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Carelli, J.D.; Sethofer, S.G.; Smith, G.A.; Miller, H.R.; Simard, J.L.; Merrick, W.C.; Jain, R.K.; Ross, N.T.; Taunton, J. Ternatin and improved synthetic variants kill cancer cells by targeting the elongation factor-1A ternary complex. eLife 2015, 4, e10222. [Google Scholar] [CrossRef] [PubMed]
  39. 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] [Green Version]
  40. Yao, N.; Chen, C.Y.; Wu, C.Y.; Motonishi, K.; Kung, H.J.; Lam, K.S. Novel flavonoids with antiproliferative activities against breast cancer cells. J. Med. Chem. 2011, 54, 4339–4349. [Google Scholar] [CrossRef] [Green Version]
  41. 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. Brit. J. Pharmacol. 2020, 177, 3–27. [Google Scholar] [CrossRef] [Green Version]
  42. Metarrestin (ML-246) in Subjects with Metastatic Solid Tumors. 2020. Available online: https://ClinicalTrials.gov/show/NCT04222413 (accessed on 2 March 2023).
  43. Dmitriev, S.E.; Vladimirov, D.O.; Lashkevich, K.A. A Quick Guide to Small-Molecule Inhibitors of Eukaryotic Protein Synthesis. Biochemistry (Moscow) 2020, 85, 1389–1421. [Google Scholar] [CrossRef] [PubMed]
  44. Brönstrup, M.; Sasse, F. Natural products targeting the elongation phase of eukaryotic protein biosynthesis. Nat. Prod. Rep. 2020, 37, 752–762. [Google Scholar] [CrossRef]
  45. Burgers, L.D.; Fuerst, R. Natural products as drugs and tools for influencing core processes of eukaryotic mRNA translation. Pharmacol. Res. 2021, 170, 105535. [Google Scholar] [CrossRef]
  46. Fan, A.; Sharp, P.P. Inhibitors of Eukaryotic Translational Machinery as Therapeutic Agents. J. Med. Chem. 2021, 64, 2436–2465. [Google Scholar] [CrossRef]
  47. Zhang, J.-N.; Xia, Y.-X.; Zhang, H.-J. Natural cyclopeptides as anticancer agents in the last 20 years. Int. J. Mol. Sci. 2021, 22, 3973. [Google Scholar] [CrossRef] [PubMed]
  48. Rinehart, K.L., Jr.; Gloer, J.B.; Hughes, R.G., Jr.; Renis, H.E.; McGovren, J.P.; Swynenberg, E.B.; Stringfellow, D.A.; Kuentzel, S.L.; Li, L.H. Didemnins: Antiviral and antitumor depsipeptides from a caribbean tunicate. Science 1981, 212, 933–935. [Google Scholar] [CrossRef] [PubMed]
  49. Vervoort, H.; Fenical, W.; de Epifanio, R. Tamandarins A and B: New cytotoxic depsipeptides from a Brazilian ascidian of the family Didemnidae. J. Org. Chem. 2000, 65, 782–792. [Google Scholar] [CrossRef]
  50. 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]
  51. 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]
  52. Vera, M.D.; Joullie, M.M. Natural products as probes of cell biology: 20 years of didemnin research. Med. Res. Rev. 2002, 22, 102–145. [Google Scholar] [CrossRef]
  53. Le Tourneau, C.; Raymond, E.; Faivre, S. Aplidine: A paradigm of how to handle the activity and toxicity of a novel marine anticancer poison. Curr. Pharm. Des. 2007, 13, 3427–3439. [Google Scholar] [CrossRef]
  54. Muñoz-Alonso, M.J.; González-Santiago, L.; Martínez, T.; Losada, A.; Galmarini, C.M.; Muñoz, A. The mechanism of action of plitidepsin. Curr. Opin. Investig. Drugs 2009, 10, 536–542. [Google Scholar] [PubMed]
  55. Danu, A.; Willekens, C.; Ribrag, V. Plitidepsin: An orphan drug. Expert Opin. Orphan Drugs 2013, 1, 569–580. [Google Scholar] [CrossRef]
  56. Alonso-Alvarez, S.; Pardal, E.; Sanchez-Nieto, D.; Navarro, M.; Caballero, M.D.; Mateos, M.V.; Martin, A. Plitidepsin: Design, development, and potential place in therapy. Drug Des. Dev. Ther. 2017, 11, 253–264. [Google Scholar] [CrossRef] [Green Version]
  57. Potts, M.B.; Kim, H.S.; Fisher, K.W.; Hu, Y.; Carrasco, Y.P.; Bulut, G.B.; Ou, Y.-H.; Herrera-Herrera, M.L.; Cubillos, F.; Mendiratta, S.; et al. Using functional signature ontology (FUSION) to identify mechanisms of action for natural products. Sci. Signal. 2013, 6, ra90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Potts, M.B.; McMillan, E.A.; Rosales, T.I.; Kim, H.S.; Ou, Y.-H.; Toombs, J.E.; Brekken, R.A.; Minden, M.D.; MacMillan, J.B.; White, M.A. Mode of action and pharmacogenomic biomarkers for exceptional responders to didemnin B. Nat. Chem. Biol. 2015, 11, 401–408. [Google Scholar] [CrossRef] [Green Version]
  59. Losada, A.; Berlanga, J.J.; Molina-Guijarro, J.M.; Jimenez-Ruiz, A.; Gago, F.; Aviles, P.; de Haro, C.; Martinez-Leal, J.F. Generation of endoplasmic reticulum stress and inhibition of autophagy by plitidepsin induces proteotoxic apoptosis in cancer cells. Biochem. Pharmacol. 2020, 172, 113744. [Google Scholar] [CrossRef]
  60. Kakeya, H.; Zhang, H.P.; Kobinata, K.; Onose, R.; Onozawa, C.; Kudo, T.; Osada, H. Cytotrienin A, a novel apoptosis inducer in human leukemia HL-60 cells. J. Antibiot. 1997, 50, 370–372. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, H.P.; Kakeya, H.; Osada, H. Novel triene-ansamycins, cytotrienins A and B, inducing apoptosis on human leukemia HL-60 cells. Tetrahedron Lett. 1997, 38, 1789–1792. [Google Scholar] [CrossRef]
  62. Kakeya, H.; Onose, R.; Osada, H. Caspase-mediated activation of a 36-kDa myelin basic protein kinase during anticancer drug-induced apoptosis. Cancer Res. 1998, 58, 4888–4894. [Google Scholar] [PubMed]
  63. Watabe, M.; Kakeya, H.; Onose, R.; Osada, H. Activation of MST/Krs and c-Jun N-terminal kinases by different signaling pathways during cytotrienin A-induced apoptosis. J. Biol. Chem. 2000, 275, 8766–8771. [Google Scholar] [CrossRef] [Green Version]
  64. Yamada, Y.; Taketani, S.; Osada, H.; Kataoka, T. Cytotrienin A, a translation inhibitor that induces ectodomain shedding of TNF receptor 1 via activation of ERK and p38 MAP kinase. Eur. J. Pharmacol. 2011, 667, 113–119. [Google Scholar] [CrossRef]
  65. Blunt, J.; Cole, T.; Munro, M.; Sun, L.; Weber, J.-F.R.; Ramasamy, K.; Abu Bakar, H.; Abdul Majeed, A.B.B. Bioactive Compounds Derived from Endophytic Aspergillus Fungus Strain Isolated from Garcinia Scortechinii. WO2010062159A1, 3 June 2010. [Google Scholar]
  66. Briggs, L.H.; Locker, R.H. 459. Flavonols from the bark of Melicope ternata. Part I. The isolation of four new flavonols, meliternatin, meliternin, ternatin, and wharangin. J. Chem. Soc. 1949, 2157–2162, The term ternatin also refers to a flavone-type natural product, 5-dihydroxy-3,3ʹ,7,8-tetramethoxyflavone (CAS registry number: 571-71-1) first reported in 1949. [Google Scholar] [CrossRef]
  67. Shimokawa, K.; Mashima, I.; Asai, A.; Yamada, K.; Kita, M.; Uemura, D. (−)-Ternatin, a highly N-methylated cyclic heptapeptide that inhibits fat accumulation: Structure and synthesis. Tetrahedron Lett. 2006, 47, 4445–4448. [Google Scholar] [CrossRef]
  68. Wang, H.-Y.; Yang, H.; Holm, M.; Tom, H.; Oltion, K.; Al-Khdhairawi, A.A.Q.; Weber, J.-F.F.; Blanchard, S.C.; Ruggero, D.; Taunton, J. Synthesis and single-molecule imaging reveal stereospecific enhancement of binding kinetics by the antitumour eEF1A antagonist SR-A3. Nat. Chem. 2022, 14, 1443–1450. [Google Scholar] [CrossRef]
  69. Copeland, R.A. The drug–target residence time model: A 10-year retrospective. Nat. Rev. Drug Discov. 2016, 15, 87–95. [Google Scholar] [CrossRef]
  70. Juette, M.F.; Carelli, J.D.; Rundlet, E.J.; Brown, A.; Shao, S.; Ferguson, A.; Wasserman, M.R.; Holm, M.; Taunton, J.; Blanchard, S.C. Didemnin B and ternatin-4 differentially inhibit conformational changes in eEF1A required for aminoacyl-tRNA accommodation into mammalian ribosomes. eLife 2022, 11, e81608. [Google Scholar] [CrossRef] [PubMed]
  71. Oltion, K.; Carelli, J.D.; Yang, T.; See, S.K.; Wang, H.-Y.; Kampmann, M.; Taunton, J. An E3 ligase network engages GCN1 to promote the degradation of translation factors on stalled ribosomes. Cell 2023, 186, 346–362. [Google Scholar] [CrossRef] [PubMed]
  72. Hoffmann, H.; Kogler, H.; Heyse, W.; Matter, H.; Caspers, M.; Schummer, D.; Klemke-Jahn, C.; Bauer, A.; Penarier, G.; Debussche, L.; et al. Discovery, Structure Elucidation, and Biological Characterization of Nannocystin A, a Macrocyclic Myxobacterial Metabolite with Potent Antiproliferative Properties. Angew. Chem. Int. Ed. 2015, 54, 10145–10148. [Google Scholar] [CrossRef]
  73. Krastel, P.; Roggo, S.; Schirle, M.; Ross, N.T.; Perruccio, F.; Aspesi, P., Jr.; Aust, T.; Buntin, K.; Estoppey, D.; Liechty, B.; et al. Nannocystin A: An Elongation Factor 1 Inhibitor from Myxobacteria with Differential Anti-Cancer Properties. Angew. Chem. Int. Ed. 2015, 54, 10149–10154. [Google Scholar] [CrossRef]
  74. Liao, L.; Zhou, J.; Xu, Z.; Ye, T. Concise Total Synthesis of Nannocystin A. Angew. Chem. Int. Ed. 2016, 55, 13263–13266. [Google Scholar] [CrossRef] [PubMed]
  75. Huang, J.; Wang, Z. Total Syntheses of Nannocystins A and A0, Two Elongation Factor 1 Inhibitors. Org. Lett. 2016, 18, 4702–4705. [Google Scholar] [CrossRef]
  76. Yang, Z.; Xu, X.; Yang, C.H.; Tian, Y.; Chen, X.; Lian, L.; Pan, W.; Su, X.; Zhang, W.; Chen, Y. Total Synthesis of Nannocystin A. Org. Lett. 2016, 18, 5768–5770. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, Y.H.; Liu, R.; Liu, B. Total synthesis of nannocystin Ax. Chem. Commun. 2017, 53, 5549–5552. [Google Scholar] [CrossRef] [PubMed]
  78. Poock, C.; Kalesse, M. Total Synthesis of Nannocystin Ax. Org. Lett. 2017, 19, 4536–4539. [Google Scholar] [CrossRef]
  79. Liu, Q.; Hu, P.; He, Y. Asymmetric Total Synthesis of Nannocystin A. J. Org. Chem. 2017, 82, 9217–9222. [Google Scholar] [CrossRef]
  80. Meng, Z.; Souillart, L.; Monks, B.; Huwyler, N.; Herrmann, J.; Mueller, R.; Furstner, A. A “Motif-Oriented” Total Synthesis of Nannocystin Ax. Preparation and Biological Assessment of Analogues. J. Org. Chem. 2018, 83, 6977–6994. [Google Scholar] [CrossRef]
  81. Wang, Z. The Chemical Syntheses of Nannocystins. Synthesis 2019, 51, 2252–2260. [Google Scholar] [CrossRef]
  82. Zhang, W. From Target-Oriented to Motif-Oriented: A Case Study on Nannocystin Total Synthesis. Molecules 2020, 25, 5327. [Google Scholar] [CrossRef]
  83. Fürstner, A. Lessons from Natural Product Total Synthesis: Macrocyclization and Postcyclization Strategies. Acc. Chem. Res. 2021, 54, 861–874. [Google Scholar] [CrossRef]
  84. Paul, D.; Das, S.; Saha, S.; Sharma, H.; Goswami, R.K. Intramolecular Heck Reaction in Total Synthesis of Natural Products: An Update. Eur. J. Org. Chem. 2021, 2021, 2057–2076. [Google Scholar] [CrossRef]
  85. Zhang, W. Heck macrocyclization in natural product total synthesis. Nat. Prod. Rep. 2021, 38, 1109–1135. [Google Scholar] [CrossRef]
  86. Tian, Y.; Xu, X.; Ding, Y.; Hao, X.; Bai, Y.; Tang, Y.; Zhang, X.; Li, Q.; Yang, Z.; Zhang, W.; et al. Synthesis and biological evaluation of nannocystin analogues toward understanding the binding role of the (2R,3S)-Epoxide in nannocystin A. Eur. J. Med. Chem. 2018, 150, 626–632. [Google Scholar] [CrossRef] [PubMed]
  87. Tian, Y.; Ding, Y.; Xu, X.; Bai, Y.; Tang, Y.; Hao, X.; Zhang, W.; Chen, Y. Total synthesis and biological evaluation of nannocystin analogues modified at the polyketide phenyl moiety. Tetrahedron Lett. 2018, 59, 3206–3209. [Google Scholar] [CrossRef]
  88. Tian, Y.; Wang, J.; Liu, W.; Yuan, X.; Tang, Y.; Li, J.; Chen, Y.; Zhang, W. Stereodivergent total synthesis of Br-nannocystins underpinning the polyketide (10R,11S) configuration as a key determinant of potency. J. Mol. Struct. 2019, 1181, 568–578. [Google Scholar] [CrossRef]
  89. Liu, Q.; Yang, X.; Ji, J.; Zhang, S.-L.; He, Y. Novel nannocystin A analogues as anticancer therapeutics: Synthesis, biological evaluations and structure-activity relationship studies. Eur. J. Med. Chem. 2019, 170, 99–111. [Google Scholar] [CrossRef]
  90. Sun, C.; Liu, R.; Xia, M.; Hou, Y.; Wang, X.; Lu, J.-J.; Liu, B.; Chen, X. Nannocystin Ax, a natural elongation factor 1α inhibitor from Nannocystis sp., suppresses epithelial-mesenchymal transition, adhesion and migration in lung cancer cells. Toxicol. Appl. Pharmacol. 2021, 420, 115535. [Google Scholar] [CrossRef]
  91. 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]
  92. Zhang, H.; Tian, Y.; Yuan, X.; Xie, F.; Yu, S.; Cai, J.; Sun, B.; Shan, C.; Zhang, W. Site-directed Late-Stage Diversification of Macrocyclic Nannocystins Facilitating Anticancer SAR and Mode of Action Studies. RSC Med. Chem. 2023, 14, 299–312. [Google Scholar] [CrossRef]
  93. Gemmer, M.; Chaillet, M.L.; van Loenhout, J.; Cuevas Arenas, R.; Vismpas, D.; Gröllers-Mulderij, M.; Koh, F.A.; Albanese, P.; Scheltema, R.A.; Howes, S.C.; et al. Visualization of translation and protein biogenesis at the ER membrane. Nature 2023, 614, 160–167. [Google Scholar] [CrossRef]
  94. Pollock, C.; Huang, S. The perinucleolar compartment. Cold Spring Harbor Perspect. Biol. 2010, 2, a000679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Kamath, R.V.; Thor, A.D.; Wang, C.; Edgerton, S.M.; Slusarczyk, A.; Leary, D.J.; Wang, J.; Wiley, E.L.; Jovanovic, B.; Wu, Q.; et al. Perinucleolar compartment prevalence has an independent prognostic value for breast cancer. Cancer Res. 2005, 65, 246–253. [Google Scholar] [CrossRef] [PubMed]
  96. Norton, J.T.; Pollock, C.B.; Wang, C.; Schink, J.C.; Kim, J.J.; Huang, S. Perinucleolar compartment prevalence is a phenotypic pancancer marker of malignancy. Cancer 2008, 113, 861–869. [Google Scholar] [CrossRef] [Green Version]
  97. Norton, J.T.; Wang, C.; Gjidoda, A.; Henry, R.W.; Huang, S. The Perinucleolar Compartment Is Directly Associated with DNA. J. Biol. Chem. 2009, 284, 4090–4101. [Google Scholar] [CrossRef] [Green Version]
  98. Huang, S.; Deerinck, T.J.; Ellisman, M.H.; Spector, D.L. The Dynamic Organization of the Perinucleolar Compartment in the Cell Nucleus. J. Cell Biol. 1997, 137, 965–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Norton, J.T.; Titus, S.A.; Dexter, D.; Austin, C.P.; Zheng, W.; Huang, S. Automated High-Content Screening for Compounds That Disassemble the Perinucleolar Compartment. J. Biomol. Screen. 2009, 14, 1045–1053. [Google Scholar] [CrossRef] [Green Version]
  100. Frankowski, K.J.; Wang, C.; Patnaik, S.; Schoenen, F.J.; Southall, N.; Li, D.; Teper, Y.; Sun, W.; Kandela, I.; Hu, D.; et al. Metarrestin, a perinucleolar compartment inhibitor, effectively suppresses metastasis. Sci. Transl. Med. 2018, 10, eaap8307. [Google Scholar] [CrossRef] [Green Version]
  101. Frankowski, K.J.; Patnaik, S.; Wang, C.; Southall, N.; Dutta, D.; De, S.; Li, D.; Dextras, C.; Lin, Y.-H.; Bryant-Connah, M.; et al. Discovery and Optimization of Pyrrolopyrimidine Derivatives as Selective Disruptors of the Perinucleolar Compartment, a Marker of Tumor Progression toward Metastasis. J. Med. Chem. 2022, 65, 8303–8331. [Google Scholar] [CrossRef]
  102. Vilimas, T.; Wang, A.Q.; Patnaik, S.; Hughes, E.A.; Singleton, M.D.; Knotts, Z.; Li, D.; Frankowski, K.; Schlomer, J.J.; Guerin, T.M.; et al. Pharmacokinetic evaluation of the PNC disassembler metarrestin in wild-type and Pdx1-Cre; LSL-KrasG12D/+; Tp53R172H/+ (KPC) mice, a genetically engineered model of pancreatic cancer. Cancer Chemoth. Pharm. 2018, 82, 1067–1080. [Google Scholar] [CrossRef] [Green Version]
  103. Padilha, E.C.; Shah, P.; Wang, A.Q.; Singleton, M.D.; Hughes, E.A.; Li, D.; Rice, K.A.; Konrath, K.M.; Patnaik, S.; Marugan, J. Metabolism and pharmacokinetics characterization of metarrestin in multiple species. Cancer Chemother. Pharm. 2020, 85, 805–816. [Google Scholar] [CrossRef]
  104. Bourdi, M.; Rudloff, U.; Patnaik, S.; Marugan, J.; Terse, P.S. Safety assessment of metarrestin in dogs: A clinical candidate targeting a subnuclear structure unique to metastatic cancer cells. Regul. Toxicol. Pharm. 2020, 116, 104716. [Google Scholar] [CrossRef]
  105. Békés, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200. [Google Scholar] [CrossRef]
  106. Pedrucci, F.; Pappalardo, C.; Marzaro, G.; Ferri, N.; Ferlin, A.; De Toni, L. Proteolysis Targeting Chimeric Molecules: Tuning Molecular Strategies for a Clinically Sound Listening. Int. J. Mol. Sci. 2022, 23, 6630. [Google Scholar] [CrossRef] [PubMed]
  107. Yao, T.; Xiao, H.; Wang, H.; Xu, X. Recent Advances in PROTACs for Drug Targeted Protein Research. Int. J. Mol. Sci. 2022, 23, 10328. [Google Scholar] [CrossRef]
  108. Jin, J.; Kabir, M.; Sun, N.; Kaniskan, H.U. Preparation of Heterobifunctional Compounds as Degraders of eEF1A2. WO2022159650A1, 28 July 2022. [Google Scholar]
  109. Tash, J.S.; Chakrasali, R.; Jakkaraj, S.R.; Hughes, J.; Smith, S.K.; Hornbaker, K.; Heckert, L.L.; Ozturk, S.B.; Hadden, M.K.; Kinzy, T.G.; et al. Gamendazole, an orally active indazole carboxylic acid male contraceptive agent, targets HSP90AB1 (HSP90BETA) and EEF1A1 (eEF1A), and stimulates Il1a transcription in rat sertoli cells. Biol. Reprod. 2008, 78, 1139–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Burglová, K.; Rylová, G.; Markos, A.; Prichystalova, H.; Soural, M.; Petracek, M.; Medvedikova, M.; Tejral, G.; Sopko, B.; Hradil, P.; et al. Identification of Eukaryotic Translation Elongation Factor 1-α 1 Gamendazole-Binding Site for Binding of 3-Hydroxy-4(1H)-quinolinones as Novel Ligands with Anticancer Activity. J. Med. Chem. 2018, 61, 3027–3036. [Google Scholar] [CrossRef] [PubMed]
  111. Isaka, M.; Srisanoh, U.; Lartpornmatulee, N.; Boonruangprapa, T. ES-242 derivatives and cycloheptapeptides from Cordyceps sp. strains BCC 16173 and BCC 16176. J. Nat. Prod. 2007, 70, 1601–1604. [Google Scholar] [CrossRef]
  112. Rukachaisirikul, V.; Chantaruk, S.; Tansakul, C.; Saithong, S.; Chaicharernwimonkoon, L.; Pakawatchai, C.; Isaka, M.; Intereya, K. A cyclopeptide from the insect pathogenic fungus Cordyceps sp. BCC 1788. J. Nat. Prod. 2006, 69, 305–307. [Google Scholar] [CrossRef]
  113. Kumar, S.; Dahiya, R.; Khokra, S.L.; Mourya, R.; Chennupati, S.V.; Maharaj, S. Total synthesis and pharmacological investigation of cordyheptapeptide A. Molecules 2017, 22, 682. [Google Scholar] [CrossRef] [Green Version]
  114. Klein, V.G.; Bray, W.M.; Wang, H.-Y.; Edmondson, Q.; Schwochert, J.; Ono, S.; Naylor, M.R.; Turmon, A.C.; Faris, J.H.; Okada, O.; et al. Identifying the Cellular Target of Cordyheptapeptide A and Synthetic Derivatives. ACS Chem. Biol. 2021, 16, 1354–1364. [Google Scholar] [CrossRef] [PubMed]
  115. Naylor, M.R.; Ly, A.M.; Handford, M.J.; Ramos, D.P.; Pye, C.R.; Furukawa, A.; Klein, V.G.; Noland, R.P.; Edmondson, Q.; Turmon, A.C.; et al. Lipophilic Permeability Efficiency Reconciles the Opposing Roles of Lipophilicity in Membrane Permeability and Aqueous Solubility. J. Med. Chem. 2018, 61, 11169–11182. [Google Scholar] [CrossRef]
  116. Chan, J.; Khan, S.N.; Harvey, I.; Merrick, W.; Pelletier, J. Eukaryotic protein synthesis inhibitors identified by comparison of cytotoxicity profiles. RNA 2004, 10, 528–543. [Google Scholar] [CrossRef] [Green Version]
  117. Schulze, C.J.; Bray, W.M.; Woerhmann, M.H.; Stuart, J.; Lokey, R.S.; Linington, R.G. “Function-first” lead discovery: Mode of action profiling of natural product libraries using image-based screening. Chem. Biol. 2013, 20, 285–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Dieterich, D.C.; Link, A.J.; Graumann, J.; Tirrell, D.A.; Schuman, E.M. Selective identification of newly synthesized proteins in mammalian cells using bioorthogonal noncanonical amino acid tagging (BONCAT). Proc. Natl. Acad. Sci. USA 2006, 103, 9482–9487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Sánchez-Murcia, P.A.; Cortés-Cabrera, Á.; 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]
  120. Nishioka, H.; Nakajima, S.; Nagashima, M.; Kojiri, K.; Suda, H. BE-43547 Series Substances, Their Manufacture with Streptomyces Species, and Their Use as Antitumor Agents. JP10147594A, 2 June 1998. [Google Scholar]
  121. Poulsen, T.B. Total Synthesis of Natural Products Containing Enamine or Enol Ether Derivatives. Acc. Chem. Res. 2021, 54, 1830–1842. [Google Scholar] [CrossRef]
  122. Villadsen, N.L.; Jacobsen, K.M.; Keiding, U.B.; Weibel, E.T.; Christiansen, B.; Vosegaard, T.; Bjerring, M.; Jensen, F.; Johannsen, M.; Tørring, T.; et al. Synthesis of ent-BE-43547A1 reveals a potent hypoxia-selective anticancer agent and uncovers the biosynthetic origin of the APD-CLD natural products. Nat. Chem. 2017, 9, 264–272. [Google Scholar] [CrossRef] [Green Version]
  123. Poulsen, T.B. A concise route to the macrocyclic core of the rakicidins. Chem. Commun. 2011, 47, 12837–12839. [Google Scholar] [CrossRef] [PubMed]
  124. Clement, L.L.; Tsakos, M.; Schaffert, E.S.; Scavenius, C.; Enghild, J.J.; Poulsen, T.B. The amido-pentadienoate-functionality of the rakicidins is a thiol reactive electrophile–development of a general synthetic strategy. Chem. Commun. 2015, 51, 12427–12430. [Google Scholar] [CrossRef] [PubMed]
  125. Tsakos, M.; Clement, L.L.; Schaffert, E.S.; Olsen, F.N.; Rupiani, S.; Djurhuus, R.; Yu, W.; Jacobsen, K.M.; Villadsen, N.L.; Poulsen, T.B. Total synthesis and biological evaluation of rakicidin A and discovery of a simplified bioactive analogue. Angew. Chem. Int. Ed. 2016, 55, 1030–1035. [Google Scholar] [CrossRef]
  126. Tsakos, M.; Jacobsen, K.M.; Yu, W.; Villadsen, N.L.; Poulsen, T.B. The rakicidin family of anticancer natural products–synthetic strategies towards a new class of hypoxia-selective cytotoxins. Synlett 2016, 27, 1898–1906. [Google Scholar] [CrossRef]
  127. Sun, Y.; Ding, Y.; Li, D.; Zhou, R.; Su, X.; Yang, J.; Guo, X.; Chong, C.; Wang, J.; Zhang, W.; et al. Cyclic Depsipeptide BE-43547A2: Synthesis and Activity against Pancreatic Cancer Stem Cells. Angew. Chem. Int. Ed. 2017, 56, 14627–14631. [Google Scholar] [CrossRef]
  128. Sun, Y.; Su, X.; Zhou, R.; Wang, D.; Zhao, Y.; Jiang, Y.; Wang, L.; Chen, Y. Total synthesis of BE-43547A2. Tetrahedron 2018, 74, 5955–5964. [Google Scholar] [CrossRef]
  129. Sun, Y.; Zhou, R.; Xu, H.; Wang, D.; Su, X.; Wang, C.; Ding, Y.; Wang, L.; Chen, Y. Syntheses and biological evaluation of BE-43547A2 analogs modified at O35 ester and C15-OH sites. Tetrahedron 2019, 75, 1808–1818. [Google Scholar] [CrossRef]
  130. Liu, C.; Wang, L.; Sun, Y.; Zhao, X.; Chen, T.; Su, X.; Guo, H.; Wang, Q.; Xi, X.; Ding, Y.; et al. Probe Synthesis Reveals Eukaryotic Translation Elongation Factor 1 Alpha 1 as the Anti-Pancreatic Cancer Target of BE-43547A2. Angew. Chem. Int. Ed. 2022, 61, e202206953. [Google Scholar] [CrossRef] [PubMed]
  131. Jacobsen, K.M.; Villadsen, N.L.; Toerring, T.; Nielsen, C.B.; Salomon, T.; Nielsen, M.M.; Tsakos, M.; Sibbersen, C.; Scavenius, C.; Nielsen, R.; et al. APD-Containing Cyclolipodepsipeptides Target Mitochondrial Function in Hypoxic Cancer Cells. Cell Chem. Biol. 2018, 25, 1337–1349. [Google Scholar] [CrossRef] [Green Version]
  132. Silvera, D.; Formenti, S.C.; Schneider, R.J. Translational control in cancer. Nat. Rev. Cancer 2010, 10, 254–266. [Google Scholar] [CrossRef]
  133. 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]
  134. Chu, J.; Pelletier, J. Therapeutic opportunities in eukaryotic translation. Cold Spring Harbor Perspect. Biol. 2018, 10, a032995. [Google Scholar] [CrossRef] [PubMed]
  135. Gandhi, V.; Plunkett, W.; Cortes, J.E. Omacetaxine: A Protein Translation Inhibitor for Treatment of Chronic Myelogenous Leukemia. Clin. Cancer Res. 2014, 20, 1735–1740. [Google Scholar] [CrossRef] [Green Version]
  136. Varona, J.F.; Landete, P.; Lopez-Martin, J.A.; Estrada, V.; Paredes, R.; Guisado-Vasco, P.; Fernandez de Orueta, L.; Torralba, M.; Fortun, J.; Vates, R.; et al. Preclinical and randomized phase I studies of plitidepsin in adults hospitalized with COVID-19. Life Sci. Alliance 2022, 5, e202101200. [Google Scholar] [CrossRef] [PubMed]
  137. White, K.M.; Rosales, R.; Yildiz, S.; Kehrer, T.; Miorin, L.; Moreno, E.; Jangra, S.; Uccellini, M.B.; Rathnasinghe, R.; Coughlan, L.; et al. Plitidepsin has potent preclinical efficacy against SARS-CoV-2 by targeting the host protein eEF1A. Science 2021, 371, 926–931. [Google Scholar] [CrossRef] [PubMed]
  138. Sachse, M.; Tenorio, R.; de Castro, I.F.; Muñoz-Basagoiti, J.; Perez-Zsolt, D.; Raïch-Regué, D.; Rodon, J.; Losada, A.; Avilés, P.; Cuevas, C. Unraveling the antiviral activity of plitidepsin against SARS-CoV-2 by subcellular and morphological analysis. Antivir. Res. 2022, 200, 105270. [Google Scholar] [CrossRef]
  139. Bosutti, A.; Dapas, B.; Grassi, G.; Bussani, R.; Zanconati, F.; Giudici, F.; Bottin, C.; Pavan, N.; Trombetta, C.; Scaggiante, B. High eEF1A1 Protein Levels Mark Aggressive Prostate Cancers and the In Vitro Targeting of eEF1A1 Reveals the eEF1A1–actin Complex as a New Potential Target for Therapy. Int. J. Mol. Sci. 2022, 23, 4143. [Google Scholar] [CrossRef] [PubMed]
  140. Wu, A.; Tang, J.; Guo, Z.; Dai, Y.; Nie, J.; Hu, W.; Liu, N.; Ye, C.; Li, S.; Pei, H. Long non-coding RNA CRYBG3 promotes lung cancer metastasis via activating the eEF1A1/MDM2/MTBP axis. Int. J. Mol. Sci. 2021, 22, 3211. [Google Scholar] [CrossRef] [PubMed]
  141. Dugger, S.A.; Platt, A.; Goldstein, D.B. Drug development in the era of precision medicine. Nat. Rev. Drug Discov. 2018, 17, 183–196. [Google Scholar] [CrossRef]
  142. Li, H.; Chen, S.; Wang, J.; Zhang, M.; Wu, W.; Liu, W.; Sun, P. Ansafurantrienins, Unprecedented Ansatrienin Derivatives Formed via Photocatalytic Intramolecular [3+ 2] Oxidative Cycloaddition. Org. Lett. 2022, 24, 592–596. [Google Scholar] [CrossRef]
Figure 1. Structures of didemnin B (1), plitidepsin (2), and tamandarin A (3).
Figure 1. Structures of didemnin B (1), plitidepsin (2), and tamandarin A (3).
Ijms 24 05184 g001
Figure 2. Structures of cytotrienin A (4) and ansatrienin B (5).
Figure 2. Structures of cytotrienin A (4) and ansatrienin B (5).
Ijms 24 05184 g002
Figure 3. Structures of natural products A3 and ternatin (6), and synthetic analogues ternatin-4 (7), SR-A3 (8), SS-A3 (9), and photoaffinity probe 10. The inset table compares anticancer activity and target residence time of 7–9.
Figure 3. Structures of natural products A3 and ternatin (6), and synthetic analogues ternatin-4 (7), SR-A3 (8), SS-A3 (9), and photoaffinity probe 10. The inset table compares anticancer activity and target residence time of 7–9.
Ijms 24 05184 g003
Figure 4. Structures of naturally occurring nannocystin A (11), semisynthetic affinity probe 12, as well as synthetic macrocyclic propargylic alcohol 13, serine-containing intermediate 14, and coumarin-tagged fluorescent probe 15. The SAR illustrated in the structure of 11 consists of inhibitive (red) and permissive (blue) sites: the former must be reserved for high activity, whereas the latter can tolerate moderate change.
Figure 4. Structures of naturally occurring nannocystin A (11), semisynthetic affinity probe 12, as well as synthetic macrocyclic propargylic alcohol 13, serine-containing intermediate 14, and coumarin-tagged fluorescent probe 15. The SAR illustrated in the structure of 11 consists of inhibitive (red) and permissive (blue) sites: the former must be reserved for high activity, whereas the latter can tolerate moderate change.
Ijms 24 05184 g004
Figure 5. Structures of the initial hit 24, final drug candidate metarrestin (25), biotin-labeled affinity probe 26, and metarrestin-based PROTAC 27. The inset table compares the selectivity window between PNC reduction and cytotoxicity of 24, 25, doxorubicin, and camptothecin.
Figure 5. Structures of the initial hit 24, final drug candidate metarrestin (25), biotin-labeled affinity probe 26, and metarrestin-based PROTAC 27. The inset table compares the selectivity window between PNC reduction and cytotoxicity of 24, 25, doxorubicin, and camptothecin.
Ijms 24 05184 g005
Figure 6. Structures of eEF1A1-targeting anticancer 3-HQ derivative 29 and its prototype 28.
Figure 6. Structures of eEF1A1-targeting anticancer 3-HQ derivative 29 and its prototype 28.
Ijms 24 05184 g006
Figure 7. Structures of natural cordyheptapeptide A (30) and synthetic analogue 31, with the inset table comparing their IC50 values against HCT-116 cancer cell line and aqueous solubility.
Figure 7. Structures of natural cordyheptapeptide A (30) and synthetic analogue 31, with the inset table comparing their IC50 values against HCT-116 cancer cell line and aqueous solubility.
Ijms 24 05184 g007
Figure 8. Structures of BE-43547A1 (32), BE-43547A2 (33), and the clickable probe 34. The SAR illustrated in their structures consists of inhibitive (red) and permissive (blue) sites: the former must be reserved for high activity, whereas the latter can tolerate moderate change.
Figure 8. Structures of BE-43547A1 (32), BE-43547A2 (33), and the clickable probe 34. The SAR illustrated in their structures consists of inhibitive (red) and permissive (blue) sites: the former must be reserved for high activity, whereas the latter can tolerate moderate change.
Ijms 24 05184 g008
Figure 9. Development status of eEF1A-targeting small-molecule anticancer agents, among which narciclasine, ternatin-4, nannocystin Ax, and BE-43547A2 have been validated in at least one in vivo preclinical model, didemnin B and metarrestin have been or currently are being studied in clinical trials, and plitidepsin has gained approval for clinical use. These three groups of compounds are at more advanced development stages than the leftmost group, making up over half of the whole collection of anticancer eEF1A inhibitors discernible from the accompanying pie chart. Their validated efficacy in preclinical models or clinical cohorts lends concrete support to the principle of targeting eEF1A with viable anticancer selectivity.
Figure 9. Development status of eEF1A-targeting small-molecule anticancer agents, among which narciclasine, ternatin-4, nannocystin Ax, and BE-43547A2 have been validated in at least one in vivo preclinical model, didemnin B and metarrestin have been or currently are being studied in clinical trials, and plitidepsin has gained approval for clinical use. These three groups of compounds are at more advanced development stages than the leftmost group, making up over half of the whole collection of anticancer eEF1A inhibitors discernible from the accompanying pie chart. Their validated efficacy in preclinical models or clinical cohorts lends concrete support to the principle of targeting eEF1A with viable anticancer selectivity.
Ijms 24 05184 g009
Table 1. Antiproliferative activity of nannocystin derivatives 1623 against human colorectal carcinoma HCT-116 cells [80].
Table 1. Antiproliferative activity of nannocystin derivatives 1623 against human colorectal carcinoma HCT-116 cells [80].
Ijms 24 05184 i001
CompoundXRIC50 (nM)
Nannocystin Ax (16)MeMe0.8
17H198
18FMe1.5
19H1345
20HMe4.3
21H1549
22---Me22.2
23H1761
Table 2. In vitro cytotoxicity of 33 against different types of pancreatic cancer cells (WT, KD, RE-KD, RE-C234S) and its in vivo anticancer efficacy in the corresponding mouse xenograft models [130].
Table 2. In vitro cytotoxicity of 33 against different types of pancreatic cancer cells (WT, KD, RE-KD, RE-C234S) and its in vivo anticancer efficacy in the corresponding mouse xenograft models [130].
Pancreatic Cancer CellsIn Vitro Cytotoxicity
IC50 (μM)
In Vivo Tumor
Inhibition Rate (%)
WT1.3398.8
KD11.6220.7
RE-KD0.8093.2
RE-C234S11.6118.3
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

Zhang, H.; Cai, J.; Yu, S.; Sun, B.; Zhang, W. Anticancer Small-Molecule Agents Targeting Eukaryotic Elongation Factor 1A: State of the Art. Int. J. Mol. Sci. 2023, 24, 5184. https://doi.org/10.3390/ijms24065184

AMA Style

Zhang H, Cai J, Yu S, Sun B, Zhang W. Anticancer Small-Molecule Agents Targeting Eukaryotic Elongation Factor 1A: State of the Art. International Journal of Molecular Sciences. 2023; 24(6):5184. https://doi.org/10.3390/ijms24065184

Chicago/Turabian Style

Zhang, Han, Jiayou Cai, Siqi Yu, Bin Sun, and Weicheng Zhang. 2023. "Anticancer Small-Molecule Agents Targeting Eukaryotic Elongation Factor 1A: State of the Art" International Journal of Molecular Sciences 24, no. 6: 5184. https://doi.org/10.3390/ijms24065184

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

Article Metrics

Back to TopTop