Ribosome Biogenesis and Function in Cancer: From Mechanisms to Therapy
Simple Summary
Abstract
1. Introduction
2. Ribosome Synthesis and Function
2.1. Transcription of rRNA Genes and Synthesis of RPs and RBFs
2.2. Pre-40S and -60S Ribosomal Subunits Processing and Modification
2.3. Nuclear Export of Pre-Ribosomal Particles and Assembly into a Mature 80S Ribosome
2.4. mRNA Translation
3. The Nucleolus and Nucleolar Stress Response
4. Dysregulation of Ribosome Biogenesis and Function in Cancer
4.1. From Ribosomopathies to Cancer
4.2. Oncoribosomes
4.3. Oncogenic Signalling Modulates Ribosome Synthesis and Function
5. Targeting Ribosome Biogenesis and mRNA Translation in Cancer
5.1. Existing Chemotherapeutics Affect Ribosome Synthesis
5.1.1. Alkylating-like Platinum Compounds
5.1.2. DNA Intercalating Agents
5.1.3. Antimetabolites
5.1.4. Topoisomerase Inhibitors
5.2. Pol I Transcription Inhibitors
5.3. Direct Inhibitors of the Translation Machinery
5.3.1. Inhibitors of eIF4E-eIF4G Interaction
5.3.2. Inhibitors of eIF4E Cap-Binding Activity
5.3.3. Antisense Oligonucleotides Targeting eIF4E
5.3.4. Targeting RNA Helicase eIF4A
5.3.5. Translation Elongation Inhibitor: Omacetaxine
5.4. Inhibitors of Signalling Pathways Regulating Translation
5.4.1. mTOR Inhibitors
5.4.2. MNK Inhibitors
5.5. Nuclear Export Inhibitor: Selinexor (KPT-330)
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Unuma, T.; Busch, H. Formation of microspherules in nucleoli of tumor cells treated with high doses of actinomycin D. Cancer Res. 1967, 27, 1232–1242. [Google Scholar]
- Daskal, Y.; Woodard, C.; Crooke, S.T.; Busch, H. Comparative ultrastructural studies of nucleoli of tumor cells treated with adriamycin and the newer anthracyclines, carminomycin and marcellomycin. Cancer Res. 1978, 38, 467–473. [Google Scholar] [PubMed]
- Derenzini, M.; Montanaro, L.; Trere, D. Ribosome biogenesis and cancer. Acta Histochem. 2017, 119, 190–197. [Google Scholar] [CrossRef] [PubMed]
- Derenzini, M.; Montanaro, L.; Trere, D. What the nucleolus says to a tumour pathologist. Histopathology 2009, 54, 753–762. [Google Scholar] [CrossRef] [PubMed]
- Dhillon, A.S.; Hagan, S.; Rath, O.; Kolch, W. MAP kinase signalling pathways in cancer. Oncogene 2007, 26, 3279–3290. [Google Scholar] [CrossRef]
- Noorolyai, S.; Shajari, N.; Baghbani, E.; Sadreddini, S.; Baradaran, B. The relation between PI3K/AKT signalling pathway and cancer. Gene 2019, 698, 120–128. [Google Scholar] [CrossRef]
- Bailey, M.H.; Tokheim, C.; Porta-Pardo, E.; Sengupta, S.; Bertrand, D.; Weerasinghe, A.; Colaprico, A.; Wendl, M.C.; Kim, J.; Reardon, B.; et al. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 2018, 174, 1034–1035. [Google Scholar] [CrossRef]
- Jiao, L.; Liu, Y.; Yu, X.Y.; Pan, X.; Zhang, Y.; Tu, J.; Song, Y.H.; Li, Y. Ribosome biogenesis in disease: New players and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 15. [Google Scholar] [CrossRef]
- Kang, J.; Brajanovski, N.; Chan, K.T.; Xuan, J.; Pearson, R.B.; Sanij, E. Ribosomal proteins and human diseases: Molecular mechanisms and targeted therapy. Signal Transduct. Target. Ther. 2021, 6, 323. [Google Scholar] [CrossRef]
- Sulima, S.O.; Kampen, K.R.; De Keersmaecker, K. Cancer Biogenesis in Ribosomopathies. Cells 2019, 8, 229. [Google Scholar] [CrossRef]
- Thomson, E.; Ferreira-Cerca, S.; Hurt, E. Eukaryotic ribosome biogenesis at a glance. J. Cell Sci. 2013, 126, 4815–4821. [Google Scholar] [CrossRef] [PubMed]
- Henras, A.K.; Soudet, J.; Gerus, M.; Lebaron, S.; Caizergues-Ferrer, M.; Mougin, A.; Henry, Y. The post-transcriptional steps of eukaryotic ribosome biogenesis. Cell. Mol. Life Sci. 2008, 65, 2334–2359. [Google Scholar] [CrossRef] [PubMed]
- Dorner, K.; Ruggeri, C.; Zemp, I.; Kutay, U. Ribosome biogenesis factors-from names to functions. EMBO J. 2023, 42, e112699. [Google Scholar] [CrossRef] [PubMed]
- Lafontaine, D.L.J.; Riback, J.A.; Bascetin, R.; Brangwynne, C.P. The nucleolus as a multiphase liquid condensate. Nat. Rev. Mol. Cell Biol. 2021, 22, 165–182. [Google Scholar] [CrossRef]
- Derenzini, M.; Thiry, M.; Goessens, G. Ultrastructural cytochemistry of the mammalian cell nucleolus. J. Histochem. Cytochem. 1990, 38, 1237–1256. [Google Scholar] [CrossRef]
- Henderson, A.S.; Warburton, D.; Atwood, K.C. Location of ribosomal DNA in the human chromosome complement. Proc. Natl. Acad. Sci. USA 1972, 69, 3394–3398. [Google Scholar] [CrossRef]
- McStay, B. Nucleolar organizer regions: Genomic ‘dark matter’ requiring illumination. Genes. Dev. 2016, 30, 1598–1610. [Google Scholar] [CrossRef]
- McStay, B.; Grummt, I. The epigenetics of rRNA genes: From molecular to chromosome biology. Annu. Rev. Cell Dev. Biol. 2008, 24, 131–157. [Google Scholar] [CrossRef]
- Caburet, S.; Conti, C.; Schurra, C.; Lebofsky, R.; Edelstein, S.J.; Bensimon, A. Human ribosomal RNA gene arrays display a broad range of palindromic structures. Genome Res. 2005, 15, 1079–1085. [Google Scholar] [CrossRef]
- Nurk, S.; Koren, S.; Rhie, A.; Rautiainen, M.; Bzikadze, A.V.; Mikheenko, A.; Vollger, M.R.; Altemose, N.; Uralsky, L.; Gershman, A.; et al. The complete sequence of a human genome. Science 2022, 376, 44–53. [Google Scholar] [CrossRef]
- Liao, W.W.; Asri, M.; Ebler, J.; Doerr, D.; Haukness, M.; Hickey, G.; Lu, S.; Lucas, J.K.; Monlong, J.; Abel, H.J.; et al. A draft human pangenome reference. Nature 2023, 617, 312–324. [Google Scholar] [CrossRef]
- Sanij, E.; Hannan, R.D. The role of UBF in regulating the structure and dynamics of transcriptionally active rDNA chromatin. Epigenetics 2009, 4, 374–382. [Google Scholar] [CrossRef]
- Sanij, E.; Poortinga, G.; Sharkey, K.; Hung, S.; Holloway, T.P.; Quin, J.; Robb, E.; Wong, L.H.; Thomas, W.G.; Stefanovsky, V.; et al. UBF levels determine the number of active ribosomal RNA genes in mammals. J. Cell Biol. 2008, 183, 1259–1274. [Google Scholar] [CrossRef]
- Hamperl, S.; Wittner, M.; Babl, V.; Perez-Fernandez, J.; Tschochner, H.; Griesenbeck, J. Chromatin states at ribosomal DNA loci. Biochim. Biophys. Acta 2013, 1829, 405–417. [Google Scholar] [CrossRef]
- Hamdane, N.; Stefanovsky, V.Y.; Tremblay, M.G.; Nemeth, A.; Paquet, E.; Lessard, F.; Sanij, E.; Hannan, R.; Moss, T. Conditional inactivation of Upstream Binding Factor reveals its epigenetic functions and the existence of a somatic nucleolar precursor body. PLoS Genet. 2014, 10, e1004505. [Google Scholar] [CrossRef] [PubMed]
- Zomerdijk, J.C.; Beckmann, H.; Comai, L.; Tjian, R. Assembly of transcriptionally active RNA polymerase I initiation factor SL1 from recombinant subunits. Science 1994, 266, 2015–2018. [Google Scholar] [CrossRef] [PubMed]
- Comai, L.; Tanese, N.; Tjian, R. The TATA-binding protein and associated factors are integral components of the RNA polymerase I transcription factor, SL1. Cell 1992, 68, 965–976. [Google Scholar] [CrossRef] [PubMed]
- Knutson, B.A.; Hahn, S. TFIIB-related factors in RNA polymerase I transcription. Biochim. Biophys. Acta 2013, 1829, 265–273. [Google Scholar] [CrossRef]
- Bywater, M.J.; Pearson, R.B.; McArthur, G.A.; Hannan, R.D. Dysregulation of the basal RNA polymerase transcription apparatus in cancer. Nat. Rev. Cancer 2013, 13, 299–314. [Google Scholar] [CrossRef]
- Engel, C.; Neyer, S.; Cramer, P. Distinct Mechanisms of Transcription Initiation by RNA Polymerases I and II. Annu Rev. Biophys. 2018, 47, 425–446. [Google Scholar] [CrossRef]
- Girbig, M.; Misiaszek, A.D.; Muller, C.W. Structural insights into nuclear transcription by eukaryotic DNA-dependent RNA polymerases. Nat. Rev. Mol. Cell Biol. 2022, 23, 603–622. [Google Scholar] [CrossRef] [PubMed]
- Russell, J.; Zomerdijk, J.C. The RNA polymerase I transcription machinery. Biochem. Soc. Symp. 2006, 73, 203–216. [Google Scholar] [CrossRef] [PubMed]
- Ray, S.; Panova, T.; Miller, G.; Volkov, A.; Porter, A.C.; Russell, J.; Panov, K.I.; Zomerdijk, J.C. Topoisomerase IIalpha promotes activation of RNA polymerase I transcription by facilitating pre-initiation complex formation. Nat. Commun. 2013, 4, 1598. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Wang, J.C.; Liu, L.F. Involvement of DNA topoisomerase I in transcription of human ribosomal RNA genes. Proc. Natl. Acad. Sci. USA 1988, 85, 1060–1064. [Google Scholar] [CrossRef]
- Ciganda, M.; Williams, N. Eukaryotic 5S rRNA biogenesis. Wiley Interdiscip. Rev. RNA 2011, 2, 523–533. [Google Scholar] [CrossRef]
- Layat, E.; Probst, A.V.; Tourmente, S. Structure, function and regulation of Transcription Factor IIIA: From Xenopus to Arabidopsis. Biochim. Biophys. Acta 2013, 1829, 274–282. [Google Scholar] [CrossRef]
- Hannan, K.M.; Soo, P.; Wong, M.S.; Lee, J.K.; Hein, N.; Poh, P.; Wysoke, K.D.; Williams, T.D.; Montellese, C.; Smith, L.K.; et al. Nuclear stabilization of p53 requires a functional nucleolar surveillance pathway. Cell Rep. 2022, 41, 111571. [Google Scholar] [CrossRef]
- Kressler, D.; Bange, G.; Ogawa, Y.; Stjepanovic, G.; Bradatsch, B.; Pratte, D.; Amlacher, S.; Strauss, D.; Yoneda, Y.; Katahira, J.; et al. Synchronizing nuclear import of ribosomal proteins with ribosome assembly. Science 2012, 338, 666–671. [Google Scholar] [CrossRef]
- Jorjani, H.; Kehr, S.; Jedlinski, D.J.; Gumienny, R.; Hertel, J.; Stadler, P.F.; Zavolan, M.; Gruber, A.R. An updated human snoRNAome. Nucleic Acids Res. 2016, 44, 5068–5082. [Google Scholar] [CrossRef]
- Hirose, T.; Steitz, J.A. Position within the host intron is critical for efficient processing of box C/D snoRNAs in mammalian cells. Proc. Natl. Acad. Sci. USA 2001, 98, 12914–12919. [Google Scholar] [CrossRef]
- Marz, M.; Gruber, A.R.; Honer Zu Siederdissen, C.; Amman, F.; Badelt, S.; Bartschat, S.; Bernhart, S.H.; Beyer, W.; Kehr, S.; Lorenz, R.; et al. Animal snoRNAs and scaRNAs with exceptional structures. RNA Biol. 2011, 8, 938–946. [Google Scholar] [CrossRef] [PubMed]
- Grandi, P.; Rybin, V.; Bassler, J.; Petfalski, E.; Strauss, D.; Marzioch, M.; Schafer, T.; Kuster, B.; Tschochner, H.; Tollervey, D.; et al. 90S pre-ribosomes include the 35S pre-rRNA, the U3 snoRNP, and 40S subunit processing factors but predominantly lack 60S synthesis factors. Mol. Cell 2002, 10, 105–115. [Google Scholar] [CrossRef] [PubMed]
- Kiss-Laszlo, Z.; Henry, Y.; Bachellerie, J.P.; Caizergues-Ferrer, M.; Kiss, T. Site-specific ribose methylation of preribosomal RNA: A novel function for small nucleolar RNAs. Cell 1996, 85, 1077–1088. [Google Scholar] [CrossRef] [PubMed]
- Albanese, V.; Reissmann, S.; Frydman, J. A ribosome-anchored chaperone network that facilitates eukaryotic ribosome biogenesis. J. Cell Biol. 2010, 189, 69–81. [Google Scholar] [CrossRef]
- Leidig, C.; Bange, G.; Kopp, J.; Amlacher, S.; Aravind, A.; Wickles, S.; Witte, G.; Hurt, E.; Beckmann, R.; Sinning, I. Structural characterization of a eukaryotic chaperone--the ribosome-associated complex. Nat. Struct. Mol. Biol. 2013, 20, 23–28. [Google Scholar] [CrossRef]
- Klinge, S.; Woolford, J.L., Jr. Ribosome assembly coming into focus. Nat. Rev. Mol. Cell Biol. 2019, 20, 116–131. [Google Scholar] [CrossRef]
- Vanden Broeck, A.; Klinge, S. An emerging mechanism for the maturation of the Small Subunit Processome. Curr. Opin. Struct. Biol. 2022, 73, 102331. [Google Scholar] [CrossRef]
- Singh, S.; Vanden Broeck, A.; Miller, L.; Chaker-Margot, M.; Klinge, S. Nucleolar maturation of the human small subunit processome. Science 2021, 373, eabj5338. [Google Scholar] [CrossRef]
- Vanden Broeck, A.; Klinge, S. Eukaryotic Ribosome Assembly. Annu. Rev. Biochem. 2024, 93, 189–210. [Google Scholar] [CrossRef]
- Bohnsack, K.E.; Bohnsack, M.T. Uncovering the assembly pathway of human ribosomes and its emerging links to disease. EMBO J. 2019, 38, e100278. [Google Scholar] [CrossRef]
- Bailey, A.D.; Talkish, J.; Ding, H.; Igel, H.; Duran, A.; Mantripragada, S.; Paten, B.; Ares, M. Concerted modification of nucleotides at functional centers of the ribosome revealed by single-molecule RNA modification profiling. Elife 2022, 11, e76562. [Google Scholar] [CrossRef]
- Meyer, B.; Wurm, J.P.; Sharma, S.; Immer, C.; Pogoryelov, D.; Kotter, P.; Lafontaine, D.L.; Wohnert, J.; Entian, K.D. Ribosome biogenesis factor Tsr3 is the aminocarboxypropyl transferase responsible for 18S rRNA hypermodification in yeast and humans. Nucleic Acids Res. 2016, 44, 4304–4316. [Google Scholar] [CrossRef] [PubMed]
- Schosserer, M.; Minois, N.; Angerer, T.B.; Amring, M.; Dellago, H.; Harreither, E.; Calle-Perez, A.; Pircher, A.; Peter Gerstl, M.; Pfeifenberger, S.; et al. Corrigendum: Methylation of ribosomal RNA by NSUN5 is a conserved mechanism modulating organismal lifespan. Nat. Commun. 2016, 7, 11530. [Google Scholar] [CrossRef] [PubMed]
- Sharma, S.; Yang, J.; van Nues, R.; Watzinger, P.; Kotter, P.; Lafontaine, D.L.J.; Granneman, S.; Entian, K.D. Specialized box C/D snoRNPs act as antisense guides to target RNA base acetylation. PLoS Genet. 2017, 13, e1006804. [Google Scholar] [CrossRef] [PubMed]
- Haruehanroengra, P.; Zheng, Y.Y.; Zhou, Y.; Huang, Y.; Sheng, J. RNA modifications and cancer. RNA Biol. 2020, 17, 1560–1575. [Google Scholar] [CrossRef]
- Cui, L.; Zheng, J.; Lin, Y.; Lin, P.; Lu, Y.; Zheng, Y.; Guo, B.; Zhao, X. Decoding the ribosome's hidden language: rRNA modifications as key players in cancer dynamics and targeted therapies. Clin. Transl. Med. 2024, 14, e1705. [Google Scholar] [CrossRef]
- Johnson, A.W.; Lund, E.; Dahlberg, J. Nuclear export of ribosomal subunits. Trends Biochem. Sci. 2002, 27, 580–585. [Google Scholar] [CrossRef]
- Thomas, F.; Kutay, U. Biogenesis and nuclear export of ribosomal subunits in higher eukaryotes depend on the CRM1 export pathway. J. Cell Sci. 2003, 116, 2409–2419. [Google Scholar] [CrossRef]
- Wild, T.; Horvath, P.; Wyler, E.; Widmann, B.; Badertscher, L.; Zemp, I.; Kozak, K.; Csucs, G.; Lund, E.; Kutay, U. A protein inventory of human ribosome biogenesis reveals an essential function of exportin 5 in 60S subunit export. PLoS Biol. 2010, 8, e1000522. [Google Scholar] [CrossRef]
- 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]
- Fabbri, L.; Chakraborty, A.; Robert, C.; Vagner, S. The plasticity of mRNA translation during cancer progression and therapy resistance. Nat. Rev. Cancer 2021, 21, 558–577. [Google Scholar] [CrossRef]
- Hinnebusch, A.G. Structural Insights into the Mechanism of Scanning and Start Codon Recognition in Eukaryotic Translation Initiation. Trends Biochem. Sci. 2017, 42, 589–611. [Google Scholar] [CrossRef]
- Hinnebusch, A.G.; Ivanov, I.P.; Sonenberg, N. Translational control by 5’-untranslated regions of eukaryotic mRNAs. Science 2016, 352, 1413–1416. [Google Scholar] [CrossRef]
- Knight, J.R.P.; Garland, G.; Poyry, T.; Mead, E.; Vlahov, N.; Sfakianos, A.; Grosso, S.; De-Lima-Hedayioglu, F.; Mallucci, G.R.; von der Haar, T.; et al. Control of translation elongation in health and disease. Dis. Model Mech. 2020, 13, dmm043208. [Google Scholar] [CrossRef]
- Hellen, C.U.T. Translation Termination and Ribosome Recycling in Eukaryotes. Cold Spring Harb. Perspect. Biol. 2018, 10, a032656. [Google Scholar] [CrossRef] [PubMed]
- Dever, T.E.; Green, R. The elongation, termination, and recycling phases of translation in eukaryotes. Cold Spring Harb. Perspect. Biol. 2012, 4, a013706. [Google Scholar] [CrossRef] [PubMed]
- Shore, D.; Albert, B. Ribosome biogenesis and the cellular energy economy. Curr. Biol. 2022, 32, R611–R617. [Google Scholar] [CrossRef] [PubMed]
- Carotenuto, P.; Pecoraro, A.; Palma, G.; Russo, G.; Russo, A. Therapeutic Approaches Targeting Nucleolus in Cancer. Cells 2019, 8, 1090. [Google Scholar] [CrossRef]
- Pederson, T. The nucleolus. Cold Spring Harb. Perspect. Biol. 2011, 3, a000638. [Google Scholar] [CrossRef]
- Iarovaia, O.V.; Minina, E.P.; Sheval, E.V.; Onichtchouk, D.; Dokudovskaya, S.; Razin, S.V.; Vassetzky, Y.S. Nucleolus: A Central Hub for Nuclear Functions. Trends Cell Biol. 2019, 29, 647–659. [Google Scholar] [CrossRef]
- Gavrilova, A.A.; Neklesova, M.V.; Zagryadskaya, Y.A.; Kuznetsova, I.M.; Turoverov, K.K.; Fonin, A.V. Stress-Induced Evolution of the Nucleolus: The Role of Ribosomal Intergenic Spacer (rIGS) Transcripts. Biomolecules 2024, 14, 1333. [Google Scholar] [CrossRef]
- Sakthivel, D.; Brown-Suedel, A.; Bouchier-Hayes, L. The role of the nucleolus in regulating the cell cycle and the DNA damage response. Adv. Protein. Chem. Struct. Biol. 2023, 135, 203–241. [Google Scholar] [CrossRef]
- Gonzalez-Arzola, K. The nucleolus: Coordinating stress response and genomic stability. Biochim. Biophys. Acta Gene Regul. Mech. 2024, 1867, 195029. [Google Scholar] [CrossRef]
- King, M.R.; Ruff, K.M.; Pappu, R.V. Emergent microenvironments of nucleoli. Nucleus 2024, 15, 2319957. [Google Scholar] [CrossRef]
- Brown, I.N.; Lafita-Navarro, M.C.; Conacci-Sorrell, M. Regulation of Nucleolar Activity by MYC. Cells 2022, 11, 574. [Google Scholar] [CrossRef]
- Corman, A.; Sirozh, O.; Lafarga, V.; Fernandez-Capetillo, O. Targeting the nucleolus as a therapeutic strategy in human disease. Trends Biochem. Sci. 2023, 48, 274–287. [Google Scholar] [CrossRef] [PubMed]
- Potapova, T.A.; Gerton, J.L. Ribosomal DNA and the nucleolus in the context of genome organization. Chromosome Res. 2019, 27, 109–127. [Google Scholar] [CrossRef] [PubMed]
- Feric, M.; Vaidya, N.; Harmon, T.S.; Mitrea, D.M.; Zhu, L.; Richardson, T.M.; Kriwacki, R.W.; Pappu, R.V.; Brangwynne, C.P. Coexisting Liquid Phases Underlie Nucleolar Subcompartments. Cell 2016, 165, 1686–1697. [Google Scholar] [CrossRef] [PubMed]
- Yoneda, M.; Nakagawa, T.; Hattori, N.; Ito, T. The nucleolus from a liquid droplet perspective. J. Biochem. 2021, 170, 153–162. [Google Scholar] [CrossRef]
- Lam, Y.W.; Trinkle-Mulcahy, L. New insights into nucleolar structure and function. F1000Prime Rep 2015, 7, 48. [Google Scholar] [CrossRef]
- Boulon, S.; Westman, B.J.; Hutten, S.; Boisvert, F.M.; Lamond, A.I. The nucleolus under stress. Mol. Cell 2010, 40, 216–227. [Google Scholar] [CrossRef] [PubMed]
- Munoz-Velasco, I.; Herrera-Escamilla, A.K.; Vazquez-Salazar, A. Nucleolar origins: Challenging perspectives on evolution and function. Open Biol. 2025, 15, 240330. [Google Scholar] [CrossRef]
- Sirri, V.; Urcuqui-Inchima, S.; Roussel, P.; Hernandez-Verdun, D. Nucleolus: The fascinating nuclear body. Histochem. Cell Biol. 2008, 129, 13–31. [Google Scholar] [CrossRef] [PubMed]
- Boisvert, F.M.; van Koningsbruggen, S.; Navascues, J.; Lamond, A.I. The multifunctional nucleolus. Nat. Rev. Mol. Cell Biol. 2007, 8, 574–585. [Google Scholar] [CrossRef] [PubMed]
- Mayer, C.; Grummt, I. Cellular stress and nucleolar function. Cell Cycle 2005, 4, 1036–1038. [Google Scholar] [CrossRef]
- Yung, B.Y.; Busch, R.K.; Busch, H.; Mauger, A.B.; Chan, P.K. Effects of actinomycin D analogs on nucleolar phosphoprotein B23 (37,000 daltons/pI 5.1). Biochem. Pharmacol. 1985, 34, 4059–4063. [Google Scholar] [CrossRef]
- Pestov, D.G.; Strezoska, Z.; Lau, L.F. Evidence of p53-dependent cross-talk between ribosome biogenesis and the cell cycle: Effects of nucleolar protein Bop1 on G(1)/S transition. Mol. Cell Biol. 2001, 21, 4246–4255. [Google Scholar] [CrossRef]
- Yang, K.; Yang, J.; Yi, J. Nucleolar Stress: Hallmarks, sensing mechanism and diseases. Cell Stress 2018, 2, 125–140. [Google Scholar] [CrossRef]
- Nicolas, E.; Parisot, P.; Pinto-Monteiro, C.; de Walque, R.; De Vleeschouwer, C.; Lafontaine, D.L. Involvement of human ribosomal proteins in nucleolar structure and p53-dependent nucleolar stress. Nat. Commun. 2016, 7, 11390. [Google Scholar] [CrossRef]
- Xuan, J.; Gitareja, K.; Brajanovski, N.; Sanij, E. Harnessing the Nucleolar DNA Damage Response in Cancer Therapy. Genes 2021, 12, 1156. [Google Scholar] [CrossRef]
- Maehama, T.; Nishio, M.; Otani, J.; Mak, T.W.; Suzuki, A. Nucleolar stress: Molecular mechanisms and related human diseases. Cancer Sci. 2023, 114, 2078–2086. [Google Scholar] [CrossRef]
- Rubbi, C.P.; Milner, J. Disruption of the nucleolus mediates stabilization of p53 in response to DNA damage and other stresses. EMBO J. 2003, 22, 6068–6077. [Google Scholar] [CrossRef]
- Liu, Y.; Deisenroth, C.; Zhang, Y. RP-MDM2-p53 Pathway: Linking Ribosomal Biogenesis and Tumor Surveillance. Trends Cancer 2016, 2, 191–204. [Google Scholar] [CrossRef] [PubMed]
- Sulima, S.O.; Hofman, I.J.F.; De Keersmaecker, K.; Dinman, J.D. How Ribosomes Translate Cancer. Cancer Discov. 2017, 7, 1069–1087. [Google Scholar] [CrossRef] [PubMed]
- Kampen, K.R.; Sulima, S.O.; Vereecke, S.; De Keersmaecker, K. Hallmarks of ribosomopathies. Nucleic Acids Res. 2020, 48, 1013–1028. [Google Scholar] [CrossRef] [PubMed]
- Da Costa, L.; Leblanc, T.; Mohandas, N. Diamond-Blackfan anemia. Blood 2020, 136, 1262–1273. [Google Scholar] [CrossRef]
- Da Costa, L.; Mohandas, N.; David, N.L.; Platon, J.; Marie, I.; O'Donohue, M.F.; Leblanc, T.; Gleizes, P.E. Diamond-Blackfan anemia, the archetype of ribosomopathy: How distinct is it from the other constitutional ribosomopathies? Blood Cells Mol. Dis. 2024, 106, 102838. [Google Scholar] [CrossRef]
- Ulirsch, J.C.; Verboon, J.M.; Kazerounian, S.; Guo, M.H.; Yuan, D.; Ludwig, L.S.; Handsaker, R.E.; Abdulhay, N.J.; Fiorini, C.; Genovese, G.; et al. The Genetic Landscape of Diamond-Blackfan Anemia. Am. J. Hum. Genet. 2019, 104, 356. [Google Scholar] [CrossRef]
- Liu, Y.; Karlsson, S. Perspectives of current understanding and therapeutics of Diamond-Blackfan anemia. Leukemia 2024, 38, 1–9. [Google Scholar] [CrossRef]
- Ludwig, L.S.; Gazda, H.T.; Eng, J.C.; Eichhorn, S.W.; Thiru, P.; Ghazvinian, R.; George, T.I.; Gotlib, J.R.; Beggs, A.H.; Sieff, C.A.; et al. Altered translation of GATA1 in Diamond-Blackfan anemia. Nat. Med. 2014, 20, 748–753. [Google Scholar] [CrossRef]
- Gripp, K.W.; Curry, C.; Olney, A.H.; Sandoval, C.; Fisher, J.; Chong, J.X.; Genomics, U.W.C.f.M.; Pilchman, L.; Sahraoui, R.; Stabley, D.L.; et al. Diamond-Blackfan anemia with mandibulofacial dystostosis is heterogeneous, including the novel DBA genes TSR2 and RPS28. Am. J. Med. Genet. A. 2014, 164, 2240–2249. [Google Scholar] [CrossRef] [PubMed]
- O’Donohue, M.F.; Da Costa, L.; Lezzerini, M.; Unal, S.; Joret, C.; Bartels, M.; Brilstra, E.; Scheijde-Vermeulen, M.; Wacheul, L.; De Keersmaecker, K.; et al. HEATR3 variants impair nuclear import of uL18 (RPL5) and drive Diamond-Blackfan anemia. Blood 2022, 139, 3111–3126. [Google Scholar] [CrossRef] [PubMed]
- Vlachos, A.; Muir, E. How I treat Diamond-Blackfan anemia. Blood 2010, 116, 3715–3723. [Google Scholar] [CrossRef] [PubMed]
- Iskander, D.; Wang, G.; Heuston, E.F.; Christodoulidou, C.; Psaila, B.; Ponnusamy, K.; Ren, H.; Mokhtari, Z.; Robinson, M.; Chaidos, A.; et al. Single-cell profiling of human bone marrow progenitors reveals mechanisms of failing erythropoiesis in Diamond-Blackfan anemia. Sci. Transl. Med. 2021, 13, eabf0113. [Google Scholar] [CrossRef]
- Wang, B.; Wang, C.; Wan, Y.; Gao, J.; Ma, Y.; Zhang, Y.; Tong, J.; Zhang, Y.; Liu, J.; Chang, L.; et al. Decoding the pathogenesis of Diamond-Blackfan anemia using single-cell RNA-seq. Cell Discov. 2022, 8, 41. [Google Scholar] [CrossRef]
- Ashley, R.J.; Yan, H.; Wang, N.; Hale, J.; Dulmovits, B.M.; Papoin, J.; Olive, M.E.; Udeshi, N.D.; Carr, S.A.; Vlachos, A.; et al. Steroid resistance in Diamond Blackfan anemia associates with p57Kip2 dysregulation in erythroid progenitors. J. Clin. Investig. 2020, 130, 2097–2110. [Google Scholar] [CrossRef]
- Maceckova, Z.; Kubickova, A.; De Sanctis, J.B.; Hajduch, M. Effect of Glucocorticosteroids in Diamond-Blackfan Anaemia: Maybe Not as Elusive as It Seems. Int. J. Mol. Sci. 2022, 23, 1886. [Google Scholar] [CrossRef]
- Vlachos, A.; Rosenberg, P.S.; Atsidaftos, E.; Alter, B.P.; Lipton, J.M. Incidence of neoplasia in Diamond Blackfan anemia: A report from the Diamond Blackfan Anemia Registry. Blood 2012, 119, 3815–3819. [Google Scholar] [CrossRef]
- Vlachos, A.; Rosenberg, P.S.; Atsidaftos, E.; Kang, J.; Onel, K.; Sharaf, R.N.; Alter, B.P.; Lipton, J.M. Increased risk of colon cancer and osteogenic sarcoma in Diamond-Blackfan anemia. Blood 2018, 132, 2205–2208. [Google Scholar] [CrossRef]
- Thompson, A.S.; Giri, N.; Gianferante, D.M.; Jones, K.; Savage, S.A.; Alter, B.P.; McReynolds, L.J. Shwachman Diamond syndrome: Narrow genotypic spectrum and variable clinical features. Pediatr. Res. 2022, 92, 1671–1680. [Google Scholar] [CrossRef]
- Mason, P.J.; Bessler, M. The genetics of dyskeratosis congenita. Cancer Genet. 2011, 204, 635–645. [Google Scholar] [CrossRef]
- Goldfarb, K.C.; Cech, T.R. Targeted CRISPR disruption reveals a role for RNase MRP RNA in human preribosomal RNA processing. Genes Dev. 2017, 31, 59–71. [Google Scholar] [CrossRef] [PubMed]
- Valdez, B.C.; Henning, D.; So, R.B.; Dixon, J.; Dixon, M.J. The Treacher Collins syndrome (TCOF1) gene product is involved in ribosomal DNA gene transcription by interacting with upstream binding factor. Proc. Natl. Acad. Sci. USA 2004, 101, 10709–10714. [Google Scholar] [CrossRef] [PubMed]
- Reilly, C.R.; Shimamura, A. Predisposition to myeloid malignancies in Shwachman-Diamond syndrome: Biological insights and clinical advances. Blood 2023, 141, 1513–1523. [Google Scholar] [CrossRef] [PubMed]
- Ajore, R.; Raiser, D.; McConkey, M.; Joud, M.; Boidol, B.; Mar, B.; Saksena, G.; Weinstock, D.M.; Armstrong, S.; Ellis, S.R.; et al. Deletion of ribosomal protein genes is a common vulnerability in human cancer, especially in concert with TP53 mutations. EMBO Mol. Med. 2017, 9, 498–507. [Google Scholar] [CrossRef]
- Fancello, L.; Kampen, K.R.; Hofman, I.J.; Verbeeck, J.; De Keersmaecker, K. The ribosomal protein gene RPL5 is a haploinsufficient tumor suppressor in multiple cancer types. Oncotarget 2017, 8, 14462–14478. [Google Scholar] [CrossRef]
- Hofman, I.J.F.; van Duin, M.; De Bruyne, E.; Fancello, L.; Mulligan, G.; Geerdens, E.; Garelli, E.; Mancini, C.; Lemmens, H.; Delforge, M.; et al. RPL5 on 1p22.1 is recurrently deleted in multiple myeloma and its expression is linked to bortezomib response. Leukemia 2017, 31, 1706–1714. [Google Scholar] [CrossRef]
- Rao, S.; Lee, S.Y.; Gutierrez, A.; Perrigoue, J.; Thapa, R.J.; Tu, Z.; Jeffers, J.R.; Rhodes, M.; Anderson, S.; Oravecz, T.; et al. Inactivation of ribosomal protein L22 promotes transformation by induction of the stemness factor, Lin28B. Blood 2012, 120, 3764–3773. [Google Scholar] [CrossRef]
- Ferreira, A.M.; Tuominen, I.; van Dijk-Bos, K.; Sanjabi, B.; van der Sluis, T.; van der Zee, A.G.; Hollema, H.; Zazula, M.; Sijmons, R.H.; Aaltonen, L.A.; et al. High frequency of RPL22 mutations in microsatellite-unstable colorectal and endometrial tumors. Hum. Mutat. 2014, 35, 1442–1445. [Google Scholar] [CrossRef]
- De Keersmaecker, K.; Atak, Z.K.; Li, N.; Vicente, C.; Patchett, S.; Girardi, T.; Gianfelici, V.; Geerdens, E.; Clappier, E.; Porcu, M.; et al. Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia. Nat. Genet. 2013, 45, 186–190. [Google Scholar] [CrossRef]
- Liu, Y.; Easton, J.; Shao, Y.; Maciaszek, J.; Wang, Z.; Wilkinson, M.R.; McCastlain, K.; Edmonson, M.; Pounds, S.B.; Shi, L.; et al. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat. Genet. 2017, 49, 1211–1218. [Google Scholar] [CrossRef] [PubMed]
- Orsolic, I.; Bursac, S.; Jurada, D.; Drmic Hofman, I.; Dembic, Z.; Bartek, J.; Mihalek, I.; Volarevic, S. Cancer-associated mutations in the ribosomal protein L5 gene dysregulate the HDM2/p53-mediated ribosome biogenesis checkpoint. Oncogene 2020, 39, 3443–3457. [Google Scholar] [CrossRef] [PubMed]
- Landau, D.A.; Tausch, E.; Taylor-Weiner, A.N.; Stewart, C.; Reiter, J.G.; Bahlo, J.; Kluth, S.; Bozic, I.; Lawrence, M.; Bottcher, S.; et al. Mutations driving CLL and their evolution in progression and relapse. Nature 2015, 526, 525–530. [Google Scholar] [CrossRef] [PubMed]
- Ljungstrom, V.; Cortese, D.; Young, E.; Pandzic, T.; Mansouri, L.; Plevova, K.; Ntoufa, S.; Baliakas, P.; Clifford, R.; Sutton, L.A.; et al. Whole-exome sequencing in relapsing chronic lymphocytic leukemia: Clinical impact of recurrent RPS15 mutations. Blood 2016, 127, 1007–1016. [Google Scholar] [CrossRef]
- Nieminen, T.T.; O’Donohue, M.F.; Wu, Y.; Lohi, H.; Scherer, S.W.; Paterson, A.D.; Ellonen, P.; Abdel-Rahman, W.M.; Valo, S.; Mecklin, J.P.; et al. Germline mutation of RPS20, encoding a ribosomal protein, causes predisposition to hereditary nonpolyposis colorectal carcinoma without DNA mismatch repair deficiency. Gastroenterology 2014, 147, 595–598. [Google Scholar] [CrossRef]
- Dutton-Regester, K.; Gartner, J.J.; Emmanuel, R.; Qutob, N.; Davies, M.A.; Gershenwald, J.E.; Robinson, W.; Robinson, S.; Rosenberg, S.A.; Scolyer, R.A.; et al. A highly recurrent RPS27 5'UTR mutation in melanoma. Oncotarget 2014, 5, 2912–2917. [Google Scholar] [CrossRef]
- Gay, D.M.; Lund, A.H.; Jansson, M.D. Translational control through ribosome heterogeneity and functional specialization. Trends Biochem. Sci. 2022, 47, 66–81. [Google Scholar] [CrossRef]
- Zhang, Z.; Xu, A.; Bai, Y.; Chen, Y.; Cates, K.; Kerr, C.; Bermudez, A.; Susanto, T.T.; Wysong, K.; Garcia Marques, F.J.; et al. A subcellular map of translational machinery composition and regulation at the single-molecule level. Science 2025, 387, eadn2623. [Google Scholar] [CrossRef]
- Kampen, K.R.; Sulima, S.O.; Verbelen, B.; Girardi, T.; Vereecke, S.; Fancello, L.; Rinaldi, G.; Verbeeck, J.; Op de Beeck, J.; Uyttebroeck, A.; et al. Correction: The ribosomal RPL10 R98S mutation drives IRES-dependent BCL-2 translation in T-ALL. Leukemia 2019, 33, 1055–1062. [Google Scholar] [CrossRef]
- Girardi, T.; Vereecke, S.; Sulima, S.O.; Khan, Y.; Fancello, L.; Briggs, J.W.; Schwab, C.; de Beeck, J.O.; Verbeeck, J.; Royaert, J.; et al. The T-cell leukemia-associated ribosomal RPL10 R98S mutation enhances JAK-STAT signaling. Leukemia 2018, 32, 809–819. [Google Scholar] [CrossRef]
- Marcel, V.; Ghayad, S.E.; Belin, S.; Therizols, G.; Morel, A.P.; Solano-Gonzalez, E.; Vendrell, J.A.; Hacot, S.; Mertani, H.C.; Albaret, M.A.; et al. p53 acts as a safeguard of translational control by regulating fibrillarin and rRNA methylation in cancer. Cancer Cell 2013, 24, 318–330. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Gao, C.; Xu, X.; Li, M.; Zhao, X.; Wang, Y.; Wang, Y.; Zhang, S.; Yan, Z.; Liu, X.; et al. FBL promotes cancer cell resistance to DNA damage and BRCA1 transcription via YBX1. EMBO Rep. 2023, 24, e56230. [Google Scholar] [CrossRef]
- Yoon, A.; Peng, G.; Brandenburger, Y.; Zollo, O.; Xu, W.; Rego, E.; Ruggero, D. Impaired control of IRES-mediated translation in X-linked dyskeratosis congenita. Science 2006, 312, 902–906. [Google Scholar] [CrossRef] [PubMed]
- Bellodi, C.; Kopmar, N.; Ruggero, D. Deregulation of oncogene-induced senescence and p53 translational control in X-linked dyskeratosis congenita. EMBO J. 2010, 29, 1865–1876. [Google Scholar] [CrossRef]
- Kampen, K.R.; Fancello, L.; Girardi, T.; Rinaldi, G.; Planque, M.; Sulima, S.O.; Loayza-Puch, F.; Verbelen, B.; Vereecke, S.; Verbeeck, J.; et al. Translatome analysis reveals altered serine and glycine metabolism in T-cell acute lymphoblastic leukemia cells. Nat. Commun. 2019, 10, 2542. [Google Scholar] [CrossRef]
- Sulima, S.O.; Kampen, K.R.; Vereecke, S.; Pepe, D.; Fancello, L.; Verbeeck, J.; Dinman, J.D.; De Keersmaecker, K. Ribosomal Lesions Promote Oncogenic Mutagenesis. Cancer Res. 2019, 79, 320–327. [Google Scholar] [CrossRef] [PubMed]
- Casacuberta-Serra, S.; Gonzalez-Larreategui, I.; Capitan-Leo, D.; Soucek, L. MYC and KRAS cooperation: From historical challenges to therapeutic opportunities in cancer. Signal Transduct. Target. Ther. 2024, 9, 205. [Google Scholar] [CrossRef]
- Schaub, F.X.; Dhankani, V.; Berger, A.C.; Trivedi, M.; Richardson, A.B.; Shaw, R.; Zhao, W.; Zhang, X.; Ventura, A.; Liu, Y.; et al. Pan-cancer Alterations of the MYC Oncogene and Its Proximal Network across the Cancer Genome Atlas. Cell Syst. 2018, 6, 282–300. [Google Scholar] [CrossRef]
- Campbell, K.J.; White, R.J. MYC regulation of cell growth through control of transcription by RNA polymerases I and III. Cold Spring Harb. Perspect. Med. 2014, 4, a018408. [Google Scholar] [CrossRef]
- Stine, Z.E.; Walton, Z.E.; Altman, B.J.; Hsieh, A.L.; Dang, C.V. MYC, Metabolism, and Cancer. Cancer Discov. 2015, 5, 1024–1039. [Google Scholar] [CrossRef]
- Dhanasekaran, R.; Deutzmann, A.; Mahauad-Fernandez, W.D.; Hansen, A.S.; Gouw, A.M.; Felsher, D.W. The MYC oncogene—the grand orchestrator of cancer growth and immune evasion. Nat. Rev. Clin. Oncol. 2022, 19, 23–36. [Google Scholar] [CrossRef]
- Martini, M.; De Santis, M.C.; Braccini, L.; Gulluni, F.; Hirsch, E. PI3K/AKT signaling pathway and cancer: An updated review. Ann. Med 2014, 46, 372–383. [Google Scholar] [CrossRef]
- Piazzi, M.; Bavelloni, A.; Gallo, A.; Faenza, I.; Blalock, W.L. Signal Transduction in Ribosome Biogenesis: A Recipe to Avoid Disaster. Int. J. Mol. Sci. 2019, 20, 2718. [Google Scholar] [CrossRef]
- Papa, A.; Pandolfi, P.P. The PTEN(-)PI3K Axis in Cancer. Biomolecules 2019, 9, 153. [Google Scholar] [CrossRef]
- Mendoza, M.C.; Er, E.E.; Blenis, J. The Ras-ERK and PI3K-mTOR pathways: Cross-talk and compensation. Trends Biochem. Sci. 2011, 36, 320–328. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Thoreen, C.C.; Chantranupong, L.; Keys, H.R.; Wang, T.; Gray, N.S.; Sabatini, D.M. A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 2012, 485, 109–113. [Google Scholar] [CrossRef] [PubMed]
- Sonenberg, N.; Hinnebusch, A.G. Regulation of translation initiation in eukaryotes: Mechanisms and biological targets. Cell 2009, 136, 731–745. [Google Scholar] [CrossRef]
- Holz, M.K.; Ballif, B.A.; Gygi, S.P.; Blenis, J. mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events. Cell 2005, 123, 569–580. [Google Scholar] [CrossRef]
- Wang, X.; Li, W.; Williams, M.; Terada, N.; Alessi, D.R.; Proud, C.G. Regulation of elongation factor 2 kinase by p90(RSK1) and p70 S6 kinase. EMBO J. 2001, 20, 4370–4379. [Google Scholar] [CrossRef]
- 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]
- Silva, R.L.; Wendel, H.G. MNK, EIF4E and targeting translation for therapy. Cell Cycle 2008, 7, 553–555. [Google Scholar] [CrossRef]
- Burger, K.; Muhl, B.; Harasim, T.; Rohrmoser, M.; Malamoussi, A.; Orban, M.; Kellner, M.; Gruber-Eber, A.; Kremmer, E.; Holzel, M.; et al. Chemotherapeutic drugs inhibit ribosome biogenesis at various levels. J. Biol. Chem. 2010, 285, 12416–12425. [Google Scholar] [CrossRef]
- Siddik, Z.H. Cisplatin: Mode of cytotoxic action and molecular basis of resistance. Oncogene 2003, 22, 7265–7279. [Google Scholar] [CrossRef] [PubMed]
- Dilruba, S.; Kalayda, G.V. Platinum-based drugs: Past, present and future. Cancer Chemother. Pharmacol. 2016, 77, 1103–1124. [Google Scholar] [CrossRef] [PubMed]
- Jordan, P.; Carmo-Fonseca, M. Cisplatin inhibits synthesis of ribosomal RNA in vivo. Nucleic Acids Res. 1998, 26, 2831–2836. [Google Scholar] [CrossRef] [PubMed]
- Bruno, P.M.; Liu, Y.; Park, G.Y.; Murai, J.; Koch, C.E.; Eisen, T.J.; Pritchard, J.R.; Pommier, Y.; Lippard, S.J.; Hemann, M.T. A subset of platinum-containing chemotherapeutic agents kills cells by inducing ribosome biogenesis stress. Nat. Med. 2017, 23, 461–471. [Google Scholar] [CrossRef]
- Sutton, E.C.; DeRose, V.J. Early nucleolar responses differentiate mechanisms of cell death induced by oxaliplatin and cisplatin. J. Biol. Chem. 2021, 296, 100633. [Google Scholar] [CrossRef]
- Ferreira, R.; Schneekloth, J.S., Jr.; Panov, K.I.; Hannan, K.M.; Hannan, R.D. Targeting the RNA Polymerase I Transcription for Cancer Therapy Comes of Age. Cells 2020, 9, 266. [Google Scholar] [CrossRef]
- Perry, R.P.; Kelley, D.E. Inhibition of RNA synthesis by actinomycin D: Characteristic dose-response of different RNA species. J. Cell. Physiol. 1970, 76, 127–139. [Google Scholar] [CrossRef]
- Tomasz, M. Mitomycin C: Small, fast and deadly (but very selective). Chem. Biol. 1995, 2, 575–579. [Google Scholar] [CrossRef] [PubMed]
- Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar] [CrossRef] [PubMed]
- Avendaño, C.; Menéndez, J.C. Medicinal Chemistry of Anticancer Drugs; Elsevier Science: San Diego, CA, USA, 2023. [Google Scholar]
- Longley, D.B.; Harkin, D.P.; Johnston, P.G. 5-fluorouracil: Mechanisms of action and clinical strategies. Nat. Rev. Cancer 2003, 3, 330–338. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.Y.; Bacanu, S.; Sreekumar, L.; Ramos, A.D.; Dai, L.; Michaelis, M.; Cinatl, J.; Seki, T.; Cao, Y.; Coffill, C.R.; et al. CETSA interaction proteomics define specific RNA-modification pathways as key components of fluorouracil-based cancer drug cytotoxicity. Cell Chem. Biol. 2022, 29, 572–585. [Google Scholar] [CrossRef]
- Sun, X.X.; Dai, M.S.; Lu, H. 5-fluorouracil activation of p53 involves an MDM2-ribosomal protein interaction. J. Biol. Chem. 2007, 282, 8052–8059. [Google Scholar] [CrossRef]
- Therizols, G.; Bash-Imam, Z.; Panthu, B.; Machon, C.; Vincent, A.; Ripoll, J.; Nait-Slimane, S.; Chalabi-Dchar, M.; Gaucherot, A.; Garcia, M.; et al. Alteration of ribosome function upon 5-fluorouracil treatment favors cancer cell drug-tolerance. Nat. Commun. 2022, 13, 173. [Google Scholar] [CrossRef]
- Thomas, C.J.; Rahier, N.J.; Hecht, S.M. Camptothecin: Current perspectives. Bioorg. Med. Chem. 2004, 12, 1585–1604. [Google Scholar] [CrossRef]
- Martino, E.; Della Volpe, S.; Terribile, E.; Benetti, E.; Sakaj, M.; Centamore, A.; Sala, A.; Collina, S. The long story of camptothecin: From traditional medicine to drugs. Bioorg. Med. Chem. Lett. 2017, 27, 701–707. [Google Scholar] [CrossRef]
- Wu, R.S.; Kumar, A.; Warner, J.R. Ribosome formation is blocked by camptothecin, a reversible inhibitor of RNA synthesis. Proc. Natl. Acad. Sci. USA 1971, 68, 3009–3014. [Google Scholar] [CrossRef]
- Pietrzak, M.; Smith, S.C.; Geralds, J.T.; Hagg, T.; Gomes, C.; Hetman, M. Nucleolar disruption and apoptosis are distinct neuronal responses to etoposide-induced DNA damage. J. Neurochem. 2011, 117, 1033–1046. [Google Scholar] [CrossRef]
- Drygin, D.; Lin, A.; Bliesath, J.; Ho, C.B.; O'Brien, S.E.; Proffitt, C.; Omori, M.; Haddach, M.; Schwaebe, M.K.; Siddiqui-Jain, A.; et al. Targeting RNA polymerase I with an oral small molecule CX-5461 inhibits ribosomal RNA synthesis and solid tumor growth. Cancer Res. 2011, 71, 1418–1430. [Google Scholar] [CrossRef]
- Bywater, M.J.; Poortinga, G.; Sanij, E.; Hein, N.; Peck, A.; Cullinane, C.; Wall, M.; Cluse, L.; Drygin, D.; Anderes, K.; et al. Inhibition of RNA polymerase I as a therapeutic strategy to promote cancer-specific activation of p53. Cancer Cell 2012, 22, 51–65. [Google Scholar] [CrossRef]
- Quin, J.; Chan, K.T.; Devlin, J.R.; Cameron, D.P.; Diesch, J.; Cullinane, C.; Ahern, J.; Khot, A.; Hein, N.; George, A.J.; et al. Inhibition of RNA polymerase I transcription initiation by CX-5461 activates non-canonical ATM/ATR signaling. Oncotarget 2016, 7, 49800–49818. [Google Scholar] [CrossRef]
- Sanij, E.; Hannan, K.M.; Xuan, J.; Yan, S.; Ahern, J.E.; Trigos, A.S.; Brajanovski, N.; Son, J.; Chan, K.T.; Kondrashova, O.; et al. CX-5461 activates the DNA damage response and demonstrates therapeutic efficacy in high-grade serous ovarian cancer. Nat. Commun. 2020, 11, 2641. [Google Scholar] [CrossRef]
- Cameron, D.P.; Sornkom, J.; Alsahafi, S.; Drygin, D.; Poortinga, G.; McArthur, G.A.; Hein, N.; Hannan, R.; Panov, K.I. CX-5461 Preferentially Induces Top2alpha-Dependent DNA Breaks at Ribosomal DNA Loci. Biomedicines 2024, 12, 1514. [Google Scholar] [CrossRef]
- Bruno, P.M.; Lu, M.; Dennis, K.A.; Inam, H.; Moore, C.J.; Sheehe, J.; Elledge, S.J.; Hemann, M.T.; Pritchard, J.R. The primary mechanism of cytotoxicity of the chemotherapeutic agent CX-5461 is topoisomerase II poisoning. Proc. Natl. Acad. Sci. USA 2020, 117, 4053–4060. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Di Antonio, M.; McKinney, S.; Mathew, V.; Ho, B.; O'Neil, N.J.; Santos, N.D.; Silvester, J.; Wei, V.; Garcia, J.; et al. CX-5461 is a DNA G-quadruplex stabilizer with selective lethality in BRCA1/2 deficient tumours. Nat. Commun. 2017, 8, 14432. [Google Scholar] [CrossRef] [PubMed]
- Bossaert, M.; Pipier, A.; Riou, J.F.; Noirot, C.; Nguyen, L.T.; Serre, R.F.; Bouchez, O.; Defrancq, E.; Calsou, P.; Britton, S.; et al. Transcription-associated topoisomerase 2alpha (TOP2A) activity is a major effector of cytotoxicity induced by G-quadruplex ligands. Elife 2021, 10, e65184. [Google Scholar] [CrossRef] [PubMed]
- Yan, S.; Xuan, J.; Brajanovski, N.; Tancock, M.R.C.; Madhamshettiwar, P.B.; Simpson, K.J.; Ellis, S.; Kang, J.; Cullinane, C.; Sheppard, K.E.; et al. The RNA polymerase I transcription inhibitor CX-5461 cooperates with topoisomerase 1 inhibition by enhancing the DNA damage response in homologous recombination-proficient high-grade serous ovarian cancer. Br. J. Cancer 2021, 124, 616–627. [Google Scholar] [CrossRef]
- Masud, T.; Soong, C.; Xu, H.; Biele, J.; Bjornson, S.; McKinney, S.; Aparicio, S. Ubiquitin-mediated DNA damage response is synthetic lethal with G-quadruplex stabilizer CX-5461. Sci. Rep. 2021, 11, 9812. [Google Scholar] [CrossRef]
- Son, J.; Hannan, K.M.; Poortinga, G.; Hein, N.; Cameron, D.P.; Ganley, A.R.D.; Sheppard, K.E.; Pearson, R.B.; Hannan, R.D.; Sanij, E. rDNA Chromatin Activity Status as a Biomarker of Sensitivity to the RNA Polymerase I Transcription Inhibitor CX-5461. Front Cell Dev. Biol. 2020, 8, 568. [Google Scholar] [CrossRef]
- Devlin, J.R.; Hannan, K.M.; Hein, N.; Cullinane, C.; Kusnadi, E.; Ng, P.Y.; George, A.J.; Shortt, J.; Bywater, M.J.; Poortinga, G.; et al. Combination Therapy Targeting Ribosome Biogenesis and mRNA Translation Synergistically Extends Survival in MYC-Driven Lymphoma. Cancer Discov. 2016, 6, 59–70. [Google Scholar] [CrossRef]
- Lee, H.C.; Wang, H.; Baladandayuthapani, V.; Lin, H.; He, J.; Jones, R.J.; Kuiatse, I.; Gu, D.; Wang, Z.; Ma, W.; et al. RNA Polymerase I Inhibition with CX-5461 as a Novel Therapeutic Strategy to Target MYC in Multiple Myeloma. Br. J. Haematol. 2017, 177, 80–94. [Google Scholar] [CrossRef] [PubMed]
- Lawrence, M.G.; Porter, L.H.; Choo, N.; Pook, D.; Grummet, J.P.; Pezaro, C.J.; Sandhu, S.; Ramm, S.; Luu, J.; Bakshi, A.; et al. CX-5461 Sensitizes DNA Damage Repair-proficient Castrate-resistant Prostate Cancer to PARP Inhibition. Mol. Cancer Ther. 2021, 20, 2140–2150. [Google Scholar] [CrossRef] [PubMed]
- Rebello, R.J.; Kusnadi, E.; Cameron, D.P.; Pearson, H.B.; Lesmana, A.; Devlin, J.R.; Drygin, D.; Clark, A.K.; Porter, L.; Pedersen, J.; et al. The Dual Inhibition of RNA Pol I Transcription and PIM Kinase as a New Therapeutic Approach to Treat Advanced Prostate Cancer. Clin. Cancer Res. 2016, 22, 5539–5552. [Google Scholar] [CrossRef] [PubMed]
- Kusnadi, E.P.; Trigos, A.S.; Cullinane, C.; Goode, D.L.; Larsson, O.; Devlin, J.R.; Chan, K.T.; De Souza, D.P.; McConville, M.J.; McArthur, G.A.; et al. Reprogrammed mRNA translation drives resistance to therapeutic targeting of ribosome biogenesis. EMBO J. 2020, 39, e105111. [Google Scholar] [CrossRef]
- Maclachlan, K.H.; Gitareja, K.; Kang, J.; Cuddihy, A.; Cao, Y.; Hein, N.; Cullinane, C.; Ang, C.S.; Brajanovski, N.; Pearson, R.B.; et al. Targeting the ribosome to treat multiple myeloma. Mol. Ther. Oncol. 2024, 32, 200771. [Google Scholar] [CrossRef]
- Pan, M.; Wright, W.C.; Chapple, R.H.; Zubair, A.; Sandhu, M.; Batchelder, J.E.; Huddle, B.C.; Low, J.; Blankenship, K.B.; Wang, Y.; et al. The chemotherapeutic CX-5461 primarily targets TOP2B and exhibits selective activity in high-risk neuroblastoma. Nat. Commun. 2021, 12, 6468. [Google Scholar] [CrossRef]
- Khot, A.; Brajanovski, N.; Cameron, D.P.; Hein, N.; Maclachlan, K.H.; Sanij, E.; Lim, J.; Soong, J.; Link, E.; Blombery, P.; et al. First-in-Human RNA Polymerase I Transcription Inhibitor CX-5461 in Patients with Advanced Hematologic Cancers: Results of a Phase I Dose-Escalation Study. Cancer Discov. 2019, 9, 1036–1049. [Google Scholar] [CrossRef]
- Hilton, J.; Gelmon, K.; Bedard, P.L.; Tu, D.; Xu, H.; Tinker, A.V.; Goodwin, R.; Laurie, S.A.; Jonker, D.; Hansen, A.R.; et al. Results of the phase I CCTG IND.231 trial of CX-5461 in patients with advanced solid tumors enriched for DNA-repair deficiencies. Nat. Commun. 2022, 13, 3607. [Google Scholar] [CrossRef]
- Soberanis Pina, P.D.; Lheureux, S.; Han, H.; Shapiro, G.; Provencher, D.M.; Rosen, L.S.; Sardesai, S.; Taylor, S.; Cescon, D.; Alqaisi, H.A.; et al. 631P Phase Ib expansion study of CX-5461 in patients with solid tumours and BRCA2 and/or PALB2 mutation. Ann. Oncol. 2024, 35, S503. [Google Scholar] [CrossRef]
- Koh, G.C.C.; Boushaki, S.; Zhao, S.J.; Pregnall, A.M.; Sadiyah, F.; Badja, C.; Memari, Y.; Georgakopoulos-Soares, I.; Nik-Zainal, S. The chemotherapeutic drug CX-5461 is a potent mutagen in cultured human cells. Nat. Genet. 2024, 56, 23–26. [Google Scholar] [CrossRef]
- Ferreira, R.; Hannah, K.M.; Panov, K.; Udumanne, T.; George, A.J.; Closa, A.; Yuen, Z.; Eyras, E.; Poh, P.; Maclachlan, K.; et al. The novel RNA polymerase I transcription inhibitor PMR-116 exploits a critical therapeutic vulnerability in a broad-spectrum of high MYC malignancies. bioRxiv 2025. [Google Scholar] [CrossRef]
- Peltonen, K.; Colis, L.; Liu, H.; Jaamaa, S.; Moore, H.M.; Enback, J.; Laakkonen, P.; Vaahtokari, A.; Jones, R.J.; af Hallstrom, T.M.; et al. Identification of novel p53 pathway activating small-molecule compounds reveals unexpected similarities with known therapeutic agents. PLoS ONE 2010, 5, e12996. [Google Scholar] [CrossRef]
- Colis, L.; Peltonen, K.; Sirajuddin, P.; Liu, H.; Sanders, S.; Ernst, G.; Barrow, J.C.; Laiho, M. DNA intercalator BMH-21 inhibits RNA polymerase I independent of DNA damage response. Oncotarget 2014, 5, 4361–4369. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, R.Q.; Huffines, A.K.; Laiho, M.; Schneider, D.A. The small-molecule BMH-21 directly inhibits transcription elongation and DNA occupancy of RNA polymerase I in vivo and in vitro. J. Biol. Chem. 2022, 298, 101450. [Google Scholar] [CrossRef] [PubMed]
- Peltonen, K.; Colis, L.; Liu, H.; Jaamaa, S.; Zhang, Z.; Af Hallstrom, T.; Moore, H.M.; Sirajuddin, P.; Laiho, M. Small molecule BMH-compounds that inhibit RNA polymerase I and cause nucleolar stress. Mol. Cancer Ther. 2014, 13, 2537–2546. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Papadopoulos, E.; Jenni, S.; Kabha, E.; Takrouri, K.J.; Yi, T.; Salvi, N.; Luna, R.E.; Gavathiotis, E.; Mahalingam, P.; Arthanari, H.; et al. Structure of the eukaryotic translation initiation factor eIF4E in complex with 4EGI-1 reveals an allosteric mechanism for dissociating eIF4G. Proc. Natl. Acad. Sci. USA 2014, 111, E3187–E3195. [Google Scholar] [CrossRef]
- 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]
- De, A.; Jacobson, B.A.; Peterson, M.S.; Stelzner, M.E.; Jay-Dixon, J.; Kratzke, M.G.; Patel, M.R.; Bitterman, P.B.; Kratzke, R.A. Inhibition of oncogenic cap-dependent translation by 4EGI-1 reduces growth, enhances chemosensitivity and alters genome-wide translation in non-small cell lung cancer. Cancer Gene Ther. 2019, 26, 157–165. [Google Scholar] [CrossRef] [PubMed]
- Descamps, G.; Gomez-Bougie, P.; Tamburini, J.; Green, A.; Bouscary, D.; Maiga, S.; Moreau, P.; Le Gouill, S.; Pellat-Deceunynck, C.; Amiot, M. The cap-translation inhibitor 4EGI-1 induces apoptosis in multiple myeloma through Noxa induction. Br. J. Cancer 2012, 106, 1660–1667. [Google Scholar] [CrossRef] [PubMed]
- Willimott, S.; Beck, D.; Ahearne, M.J.; Adams, V.C.; Wagner, S.D. Cap-translation inhibitor, 4EGI-1, restores sensitivity to ABT-737 apoptosis through cap-dependent and -independent mechanisms in chronic lymphocytic leukemia. Clin. Cancer Res. 2013, 19, 3212–3223. [Google Scholar] [CrossRef]
- 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]
- 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]
- Wang, L.; Guo, C.; Li, X.; Yu, X.; Li, X.; Xu, K.; Jiang, B.; Jia, X.; Li, C.; Shi, D. Design, synthesis and biological evaluation of bromophenol-thiazolylhydrazone hybrids inhibiting the interaction of translation initiation factors eIF4E/eIF4G as multifunctional agents for cancer treatment. Eur. J. Med. Chem. 2019, 177, 153–170. [Google Scholar] [CrossRef]
- Qi, X.; Zhang, S.; Chen, Z.; Wang, L.; Zhu, W.; Yin, C.; Fan, J.; Wu, X.; Wang, J.; Guo, C. EGPI-1, a novel eIF4E/eIF4G interaction inhibitor, inhibits lung cancer cell growth and angiogenesis through Ras/MNK/ERK/eIF4E signaling pathway. Chem. Biol. Interact. 2022, 352, 109773. [Google Scholar] [CrossRef]
- Ziemniak, M.; Strenkowska, M.; Kowalska, J.; Jemielity, J. Potential therapeutic applications of RNA cap analogs. Future Med. Chem. 2013, 5, 1141–1172. [Google Scholar] [CrossRef]
- O’Rourke, R.L.; Garner, A.L. Chemical Probes for Studying the Eukaryotic Translation Initiation Factor 4E (eIF4E)-Regulated Translatome in Cancer. ACS Pharmacol. Transl. Sci. 2025, 8, 621–635. [Google Scholar] [CrossRef]
- 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]
- Chen, E.Z.; Jacobson, B.A.; Patel, M.R.; Okon, A.M.; Li, S.; Xiong, K.; Vaidya, A.J.; Bitterman, P.B.; Wagner, C.R.; Kratzke, R.A. Small-molecule inhibition of oncogenic eukaryotic protein translation in mesothelioma cells. Investig. New Drugs 2014, 32, 598–603. [Google Scholar] [CrossRef]
- Smith, K.A.; Zhou, B.; Avdulov, S.; Benyumov, A.; Peterson, M.; Liu, Y.; Okon, A.; Hergert, P.; Braziunas, J.; Wagner, C.R.; et al. Transforming Growth Factor-beta1 Induced Epithelial Mesenchymal Transition is blocked by a chemical antagonist of translation factor eIF4E. Sci. Rep. 2015, 5, 18233. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Jia, Y.; Jacobson, B.; McCauley, J.; Kratzke, R.; Bitterman, P.B.; Wagner, C.R. Treatment of breast and lung cancer cells with a N-7 benzyl guanosine monophosphate tryptamine phosphoramidate pronucleotide (4Ei-1) results in chemosensitization to gemcitabine and induced eIF4E proteasomal degradation. Mol. Pharm. 2013, 10, 523–531. [Google Scholar] [CrossRef] [PubMed]
- Golojuch, S.; Kopcial, M.; Strzelecka, D.; Kasprzyk, R.; Baran, N.; Sikorski, P.J.; Kowalska, J.; Jemielity, J. Exploring tryptamine conjugates as pronucleotides of phosphate-modified 7-methylguanine nucleotides targeting cap-dependent translation. Bioorg. Med. Chem. 2020, 28, 115523. [Google Scholar] [CrossRef]
- Cardenas, E.L.; O’Rourke, R.L.; Menon, A.; Vega-Hernandez, G.; Meagher, J.; Stuckey, J.; Garner, A.L. Second-Generation Cap Analogue Prodrugs for Targeting Aberrant Eukaryotic Translation Initiation Factor 4E Activity in Cancer. ACS Med. Chem. Lett. 2025, 16, 96–100. [Google Scholar] [CrossRef] [PubMed]
- Westman, B.; Beeren, L.; Grudzien, E.; Stepinski, J.; Worch, R.; Zuberek, J.; Jemielity, J.; Stolarski, R.; Darzynkiewicz, E.; Rhoads, R.E.; et al. The antiviral drug ribavirin does not mimic the 7-methylguanosine moiety of the mRNA cap structure in vitro. RNA 2005, 11, 1505–1513. [Google Scholar] [CrossRef]
- Yan, Y.; Svitkin, Y.; Lee, J.M.; Bisaillon, M.; Pelletier, J. Ribavirin is not a functional mimic of the 7-methyl guanosine mRNA cap. RNA 2005, 11, 1238–1244. [Google Scholar] [CrossRef]
- Pettersson, F.; Yau, C.; Dobocan, M.C.; Culjkovic-Kraljacic, B.; Retrouvey, H.; Puckett, R.; Flores, L.M.; Krop, I.E.; Rousseau, C.; Cocolakis, E.; et al. Ribavirin treatment effects on breast cancers overexpressing eIF4E, a biomarker with prognostic specificity for luminal B-type breast cancer. Clin. Cancer Res. 2011, 17, 2874–2884. [Google Scholar] [CrossRef]
- 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]
- Assouline, S.; Culjkovic, B.; Cocolakis, E.; Rousseau, C.; Beslu, N.; Amri, A.; Caplan, S.; Leber, B.; Roy, D.C.; Miller, W.H., Jr.; et al. Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): A proof-of-principle clinical trial with ribavirin. Blood 2009, 114, 257–260. [Google Scholar] [CrossRef]
- Dunn, L.A.; Fury, M.G.; Sherman, E.J.; Ho, A.A.; Katabi, N.; Haque, S.S.; Pfister, D.G. Phase I study of induction chemotherapy with afatinib, ribavirin, and weekly carboplatin and paclitaxel for stage IVA/IVB human papillomavirus-associated oropharyngeal squamous cell cancer. Head Neck 2018, 40, 233–241. [Google Scholar] [CrossRef]
- Graff, J.R.; Konicek, B.W.; Vincent, T.M.; Lynch, R.L.; Monteith, D.; Weir, S.N.; Schwier, P.; Capen, A.; Goode, R.L.; Dowless, M.S.; et al. Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity. J. Clin. Investig. 2007, 117, 2638–2648. [Google Scholar] [CrossRef]
- Hong, D.S.; Kurzrock, R.; Oh, Y.; Wheler, J.; Naing, A.; Brail, L.; Callies, S.; Andre, V.; Kadam, S.K.; Nasir, A.; et al. A phase 1 dose escalation, pharmacokinetic, and pharmacodynamic evaluation of eIF-4E antisense oligonucleotide LY2275796 in patients with advanced cancer. Clin. Cancer Res. 2011, 17, 6582–6591. [Google Scholar] [CrossRef]
- Duffy, A.G.; Makarova-Rusher, O.V.; Ulahannan, S.V.; Rahma, O.E.; Fioravanti, S.; Walker, M.; Abdullah, S.; Raffeld, M.; Anderson, V.; Abi-Jaoudeh, N.; et al. Modulation of tumor eIF4E by antisense inhibition: A phase I/II translational clinical trial of ISIS 183750-an antisense oligonucleotide against eIF4E-in combination with irinotecan in solid tumors and irinotecan-refractory colorectal cancer. Int. J. Cancer 2016, 139, 1648–1657. [Google Scholar] [CrossRef] [PubMed]
- Bordeleau, M.E.; Mori, A.; Oberer, M.; Lindqvist, L.; Chard, L.S.; Higa, T.; Belsham, G.J.; Wagner, G.; Tanaka, J.; Pelletier, J. Functional characterization of IRESes by an inhibitor of the RNA helicase eIF4A. Nat. Chem. Biol. 2006, 2, 213–220. [Google Scholar] [CrossRef] [PubMed]
- Tsumuraya, T.; Ishikawa, C.; Machijima, Y.; Nakachi, S.; Senba, M.; Tanaka, J.; Mori, N. Effects of hippuristanol, an inhibitor of eIF4A, on adult T-cell leukemia. Biochem. Pharmacol. 2011, 81, 713–722. [Google Scholar] [CrossRef] [PubMed]
- Cencic, R.; Robert, F.; Galicia-Vazquez, G.; Malina, A.; Ravindar, K.; Somaiah, R.; Pierre, P.; Tanaka, J.; Deslongchamps, P.; Pelletier, J. Modifying chemotherapy response by targeted inhibition of eukaryotic initiation factor 4A. Blood Cancer J. 2013, 3, e128. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Zhang, W.; Chu, J.; Cyr, A.M.; Yueh, H.; Brown, L.E.; Wang, T.T.; Pelletier, J.; Porco, J.A., Jr. Intercepted Retro-Nazarov Reaction: Syntheses of Amidino-Rocaglate Derivatives and Their Biological Evaluation as eIF4A Inhibitors. J. Am. Chem. Soc. 2019, 141, 12891–12900. [Google Scholar] [CrossRef]
- Sadlish, H.; Galicia-Vazquez, G.; Paris, C.G.; Aust, T.; Bhullar, B.; Chang, L.; Helliwell, S.B.; Hoepfner, D.; Knapp, B.; Riedl, R.; et al. Evidence for a functionally relevant rocaglamide binding site on the eIF4A-RNA complex. ACS Chem. Biol. 2013, 8, 1519–1527. [Google Scholar] [CrossRef]
- Hwang, B.Y.; Su, B.N.; Chai, H.; Mi, Q.; Kardono, L.B.; Afriastini, J.J.; Riswan, S.; Santarsiero, B.D.; Mesecar, A.D.; Wild, R.; et al. Silvestrol and episilvestrol, potential anticancer rocaglate derivatives from Aglaia silvestris. J. Org. Chem. 2004, 69, 3350–3358. [Google Scholar] [CrossRef]
- Zhang, X.; Bi, C.; Lu, T.; Zhang, W.; Yue, T.; Wang, C.; Tian, T.; Zhang, X.; Huang, Y.; Lunning, M.; et al. Targeting translation initiation by synthetic rocaglates for treating MYC-driven lymphomas. Leukemia 2020, 34, 138–150. [Google Scholar] [CrossRef]
- Bordeleau, M.E.; Robert, F.; Gerard, B.; Lindqvist, L.; Chen, S.M.; Wendel, H.G.; Brem, B.; Greger, H.; Lowe, S.W.; Porco, J.A., Jr.; et al. Therapeutic suppression of translation initiation modulates chemosensitivity in a mouse lymphoma model. J. Clin. Investig. 2008, 118, 2651–2660. [Google Scholar] [CrossRef]
- Low, W.K.; Dang, Y.; Schneider-Poetsch, T.; Shi, Z.; Choi, N.S.; Merrick, W.C.; Romo, D.; Liu, J.O. Inhibition of eukaryotic translation initiation by the marine natural product pateamine A. Mol. Cell 2005, 20, 709–722. [Google Scholar] [CrossRef] [PubMed]
- Low, W.K.; Dang, Y.; Schneider-Poetsch, T.; Shi, Z.; Choi, N.S.; Rzasa, R.M.; Shea, H.A.; Li, S.; Park, K.; Ma, G.; et al. Isolation and identification of eukaryotic initiation factor 4A as a molecular target for the marine natural product Pateamine, A. Methods Enzym. 2007, 431, 303–324. [Google Scholar] [CrossRef]
- Low, W.K.; Li, J.; Zhu, M.; Kommaraju, S.S.; Shah-Mittal, J.; Hull, K.; Liu, J.O.; Romo, D. Second-generation derivatives of the eukaryotic translation initiation inhibitor pateamine A targeting eIF4A as potential anticancer agents. Bioorg. Med. Chem. 2014, 22, 116–125. [Google Scholar] [CrossRef] [PubMed]
- Saito, H.; Handa, Y.; Chen, M.; Schneider-Poetsch, T.; Shichino, Y.; Takahashi, M.; Romo, D.; Yoshida, M.; Furstner, A.; Ito, T.; et al. DMDA-PatA mediates RNA sequence-selective translation repression by anchoring eIF4A and DDX3 to GNG motifs. Nat. Commun. 2024, 15, 7418. [Google Scholar] [CrossRef]
- 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]
- Peron, G.; Mastinu, A.; Pena-Corona, S.I.; Hernandez-Parra, H.; Leyva-Gomez, G.; Calina, D.; Sharifi-Rad, J. Silvestrol, a potent anticancer agent with unfavourable pharmacokinetics: Current knowledge on its pharmacological properties and future directions for the development of novel drugs. Biomed. Pharmacother. 2024, 177, 117047. [Google Scholar] [CrossRef]
- Kuznetsov, G.; Xu, Q.; Rudolph-Owen, L.; Tendyke, K.; Liu, J.; Towle, M.; Zhao, N.; Marsh, J.; Agoulnik, S.; Twine, N.; et al. Potent in vitro and in vivo anticancer activities of des-methyl, des-amino pateamine A, a synthetic analogue of marine natural product pateamine A. Mol. Cancer Ther. 2009, 8, 1250–1260. [Google Scholar] [CrossRef]
- 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]
- Thompson, P.A.; Eam, B.; Young, N.P.; Fish, S.; Chen, J.; Barrera, M.; Howard, H.; Sung, E.; Parra, A.; Staunton, J.; et al. Targeting Oncogene mRNA Translation in B-Cell Malignancies with eFT226, a Potent and Selective Inhibitor of eIF4A. Mol. Cancer Ther. 2021, 20, 26–36. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Gurel, G.; Blaha, G.; Moore, P.B.; Steitz, T.A. U2504 determines the species specificity of the A-site cleft antibiotics: The structures of tiamulin, homoharringtonine, and bruceantin bound to the ribosome. J. Mol. Biol. 2009, 389, 146–156. [Google Scholar] [CrossRef] [PubMed]
- Alvandi, F.; Kwitkowski, V.E.; Ko, C.W.; Rothmann, M.D.; Ricci, S.; Saber, H.; Ghosh, D.; Brown, J.; Pfeiler, E.; Chikhale, E.; et al. U.S. Food and Drug Administration approval summary: Omacetaxine mepesuccinate as treatment for chronic myeloid leukemia. Oncologist 2014, 19, 94–99. [Google Scholar] [CrossRef]
- 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]
- Walker, Z.J.; Idler, B.M.; Davis, L.N.; Stevens, B.M.; VanWyngarden, M.J.; Ohlstrom, D.; Bearrows, S.C.; Hammes, A.; Smith, C.A.; Jordan, C.T.; et al. Exploiting Protein Translation Dependence in Multiple Myeloma with Omacetaxine-Based Therapy. Clin. Cancer Res. 2021, 27, 819–830. [Google Scholar] [CrossRef]
- Janes, M.R.; Fruman, D.A. Targeting TOR dependence in cancer. Oncotarget 2010, 1, 69–76. [Google Scholar] [CrossRef]
- Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One drug, many effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef]
- Kwitkowski, V.E.; Prowell, T.M.; Ibrahim, A.; Farrell, A.T.; Justice, R.; Mitchell, S.S.; Sridhara, R.; Pazdur, R. FDA approval summary: Temsirolimus as treatment for advanced renal cell carcinoma. Oncologist 2010, 15, 428–435. [Google Scholar] [CrossRef] [PubMed]
- Fruman, D.A.; Rommel, C. PI3K and cancer: Lessons, challenges and opportunities. Nat. Rev. Drug Discov. 2014, 13, 140–156. [Google Scholar] [CrossRef] [PubMed]
- Benjamin, D.; Colombi, M.; Moroni, C.; Hall, M.N. Rapamycin passes the torch: A new generation of mTOR inhibitors. Nat. Rev. Drug Discov. 2011, 10, 868–880. [Google Scholar] [CrossRef] [PubMed]
- Choo, A.Y.; Blenis, J. Not all substrates are treated equally: Implications for mTOR, rapamycin-resistance and cancer therapy. Cell Cycle 2009, 8, 567–572. [Google Scholar] [CrossRef]
- Yun, S.; Vincelette, N.D.; Knorr, K.L.; Almada, L.L.; Schneider, P.A.; Peterson, K.L.; Flatten, K.S.; Dai, H.; Pratz, K.W.; Hess, A.D.; et al. 4EBP1/c-MYC/PUMA and NF-kappaB/EGR1/BIM pathways underlie cytotoxicity of mTOR dual inhibitors in malignant lymphoid cells. Blood 2016, 127, 2711–2722. [Google Scholar] [CrossRef]
- Demosthenous, C.; Han, J.J.; Stenson, M.J.; Maurer, M.J.; Wellik, L.E.; Link, B.; Hege, K.; Dogan, A.; Sotomayor, E.; Witzig, T.; et al. Translation initiation complex eIF4F is a therapeutic target for dual mTOR kinase inhibitors in non-Hodgkin lymphoma. Oncotarget 2015, 6, 9488–9501. [Google Scholar] [CrossRef]
- Ghobrial, I.M.; Siegel, D.S.; Vij, R.; Berdeja, J.G.; Richardson, P.G.; Neuwirth, R.; Patel, C.G.; Zohren, F.; Wolf, J.L. TAK-228 (formerly MLN0128), an investigational oral dual TORC1/2 inhibitor: A phase I dose escalation study in patients with relapsed or refractory multiple myeloma, non-Hodgkin lymphoma, or Waldenstrom’s macroglobulinemia. Am. J. Hematol. 2016, 91, 400–405. [Google Scholar] [CrossRef]
- Wu, X.; Xu, Y.; Liang, Q.; Yang, X.; Huang, J.; Wang, J.; Zhang, H.; Shi, J. Recent Advances in Dual PI3K/mTOR Inhibitors for Tumour Treatment. Front. Pharmacol. 2022, 13, 875372. [Google Scholar] [CrossRef]
- Fan, Q.W.; Knight, Z.A.; Goldenberg, D.D.; Yu, W.; Mostov, K.E.; Stokoe, D.; Shokat, K.M.; Weiss, W.A. A dual PI3 kinase/mTOR inhibitor reveals emergent efficacy in glioma. Cancer Cell 2006, 9, 341–349. [Google Scholar] [CrossRef]
- Yu, Y.; Yoon, S.O.; Poulogiannis, G.; Yang, Q.; Ma, X.M.; Villen, J.; Kubica, N.; Hoffman, G.R.; Cantley, L.C.; Gygi, S.P.; et al. Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling. Science 2011, 332, 1322–1326. [Google Scholar] [CrossRef]
- Bhagwat, S.V.; Gokhale, P.C.; Crew, A.P.; Cooke, A.; Yao, Y.; Mantis, C.; Kahler, J.; Workman, J.; Bittner, M.; Dudkin, L.; et al. Preclinical characterization of OSI-027, a potent and selective inhibitor of mTORC1 and mTORC2: Distinct from rapamycin. Mol. Cancer Ther. 2011, 10, 1394–1406. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Kwiatkowski, D.J. Equivalent benefit of rapamycin and a potent mTOR ATP-competitive inhibitor, MLN0128 (INK128), in a mouse model of tuberous sclerosis. Mol. Cancer Res. 2013, 11, 467–473. [Google Scholar] [CrossRef] [PubMed]
- Massard, C.; Sandhu, S.; Blanco, M.; Papadatos-Pastos, D.; Carden, C.; De Bono, J.; Saran, F.; Molife, L.R.; Kaye, S.B.; Soria, J.C.; et al. Toward a better dialogue between neuro-oncologists and phase I investigators. J. Clin. Oncol. 2012, 30, 562–563. [Google Scholar] [CrossRef] [PubMed]
- Pike, K.G.; Malagu, K.; Hummersone, M.G.; Menear, K.A.; Duggan, H.M.; Gomez, S.; Martin, N.M.; Ruston, L.; Pass, S.L.; Pass, M. Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: The discovery of AZD8055 and AZD2014. Bioorg. Med. Chem. Lett. 2013, 23, 1212–1216. [Google Scholar] [CrossRef]
- Mateo, J.; Olmos, D.; Dumez, H.; Poondru, S.; Samberg, N.L.; Barr, S.; Van Tornout, J.M.; Jie, F.; Sandhu, S.; Tan, D.S.; et al. A first in man, dose-finding study of the mTORC1/mTORC2 inhibitor OSI-027 in patients with advanced solid malignancies. Br. J. Cancer 2016, 114, 889–896. [Google Scholar] [CrossRef]
- Al-Kali, A.; Aldoss, I.; Atherton, P.J.; Strand, C.A.; Shah, B.; Webster, J.; Bhatnagar, B.; Flatten, K.S.; Peterson, K.L.; Schneider, P.A.; et al. A phase 2 and pharmacological study of sapanisertib in patients with relapsed and/or refractory acute lymphoblastic leukemia. Cancer Med. 2023, 12, 21229–21239. [Google Scholar] [CrossRef]
- Rodrik-Outmezguine, V.S.; Okaniwa, M.; Yao, Z.; Novotny, C.J.; McWhirter, C.; Banaji, A.; Won, H.; Wong, W.; Berger, M.; de Stanchina, E.; et al. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor. Nature 2016, 534, 272–276. [Google Scholar] [CrossRef]
- Janku, F.; Choong, G.M.; Opyrchal, M.; Dowlati, A.; Hierro, C.; Rodon, J.; Wicki, A.; Forster, M.D.; Blagden, S.P.; Yin, J.; et al. A Phase I Study of the Oral Dual-Acting Pan-PI3K/mTOR Inhibitor Bimiralisib in Patients with Advanced Solid Tumors. Cancers 2024, 16, 1137. [Google Scholar] [CrossRef]
- Brown, J.R.; Hamadani, M.; Hayslip, J.; Janssens, A.; Wagner-Johnston, N.; Ottmann, O.; Arnason, J.; Tilly, H.; Millenson, M.; Offner, F.; et al. Voxtalisib (XL765) in patients with relapsed or refractory non-Hodgkin lymphoma or chronic lymphocytic leukaemia: An open-label, phase 2 trial. Lancet Haematol. 2018, 5, e170–e180. [Google Scholar] [CrossRef]
- Dolly, S.O.; Wagner, A.J.; Bendell, J.C.; Kindler, H.L.; Krug, L.M.; Seiwert, T.Y.; Zauderer, M.G.; Lolkema, M.P.; Apt, D.; Yeh, R.F.; et al. Phase I Study of Apitolisib (GDC-0980), Dual Phosphatidylinositol-3-Kinase and Mammalian Target of Rapamycin Kinase Inhibitor, in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2016, 22, 2874–2884. [Google Scholar] [CrossRef]
- Carracedo, A.; Ma, L.; Teruya-Feldstein, J.; Rojo, F.; Salmena, L.; Alimonti, A.; Egia, A.; Sasaki, A.T.; Thomas, G.; Kozma, S.C.; et al. Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer. J. Clin. Investig. 2008, 118, 3065–3074. [Google Scholar] [CrossRef]
- Jin, X.; Yu, R.; Wang, X.; Proud, C.G.; Jiang, T. Progress in developing MNK inhibitors. Eur. J. Med. Chem. 2021, 219, 113420. [Google Scholar] [CrossRef]
- Konicek, B.W.; Stephens, J.R.; McNulty, A.M.; Robichaud, N.; Peery, R.B.; Dumstorf, C.A.; Dowless, M.S.; Iversen, P.W.; Parsons, S.; Ellis, K.E.; et al. Therapeutic inhibition of MAP kinase interacting kinase blocks eukaryotic initiation factor 4E phosphorylation and suppresses outgrowth of experimental lung metastases. Cancer Res. 2011, 71, 1849–1857. [Google Scholar] [CrossRef]
- Santag, S.; Siegel, F.; Wengner, A.M.; Lange, C.; Bomer, U.; Eis, K.; Puhler, F.; Lienau, P.; Bergemann, L.; Michels, M.; et al. BAY 1143269, a novel MNK1 inhibitor, targets oncogenic protein expression and shows potent anti-tumor activity. Cancer Lett. 2017, 390, 21–29. [Google Scholar] [CrossRef]
- Webster, K.R.; Goel, V.K.; Hung, I.N.; Parker, G.S.; Staunton, J.; Neal, M.; Molter, J.; Chiang, G.G.; Jessen, K.A.; Wegerski, C.J.; et al. eFT508, a Potent and Selective Mitogen-Activated Protein Kinase Interacting Kinase (MNK) 1 and 2 Inhibitor, Is Efficacious in Preclinical Models of Diffuse Large B-Cell Lymphoma (DLBCL). Blood 2015, 126, 1554. [Google Scholar] [CrossRef]
- Yang, H.; Chennamaneni, L.R.; Ho, M.W.T.; Ang, S.H.; Tan, E.S.W.; Jeyaraj, D.A.; Yeap, Y.S.; Liu, B.; Ong, E.H.; Joy, J.K.; et al. Optimization of Selective Mitogen-Activated Protein Kinase Interacting Kinases 1 and 2 Inhibitors for the Treatment of Blast Crisis Leukemia. J. Med. Chem. 2018, 61, 4348–4369. [Google Scholar] [CrossRef]
- Wan, W.; Zhang, X.; Huang, C.; Chen, L.; Yang, X.; Bao, K.; Peng, T. Preclinical anti-angiogenic and anti-cancer activities of BAY1143269 in glioblastoma via targeting oncogenic protein expression. Pharmacol. Res. Perspect. 2022, 10, e00981. [Google Scholar] [CrossRef] [PubMed]
- Reich, S.H.; Sprengeler, P.A.; Chiang, G.G.; Appleman, J.R.; Chen, J.; Clarine, J.; Eam, B.; Ernst, J.T.; Han, Q.; Goel, V.K.; et al. Structure-based Design of Pyridone-Aminal eFT508 Targeting Dysregulated Translation by Selective Mitogen-activated Protein Kinase Interacting Kinases 1 and 2 (MNK1/2) Inhibition. J. Med. Chem. 2018, 61, 3516–3540. [Google Scholar] [CrossRef] [PubMed]
- Falchook, G.S.; Infante, J.R.; Meric-Bernstam, F.; Miller, L.L.; Morison, K.; Vallner, D.; Sperry, S.; Goel, V.; Chiang, G.G.; Webster, K.; et al. A phase 1 dose escalation study of eFT508, an inhibitor of mitogen-activated protein kinase-interacting serine/threonine kinase-1 (MNK-1) and MNK-2 in patients with advanced solid tumors. J. Clin. Oncol. 2017, 35, 2579. [Google Scholar] [CrossRef]
- Ferrario, C.; Mackey, J.; Gelmon, K.A.; Levasseur, N.; Sorensen, P.H.; Oo, H.Z.; Negri, G.L.; Tse, V.W.L.; Spencer, S.E.; Cheng, G.; et al. Phase Ib Pharmacodynamic Study of the MNK Inhibitor Tomivosertib (eFT508) Combined With Paclitaxel in Patients With Refractory Metastatic Breast Cancer. Clin. Cancer Res. 2025, 31, 491–502. [Google Scholar] [CrossRef]
- Xu, Y.; Poggio, M.; Jin, H.Y.; Shi, Z.; Forester, C.M.; Wang, Y.; Stumpf, C.R.; Xue, L.; Devericks, E.; So, L.; et al. Translation control of the immune checkpoint in cancer and its therapeutic targeting. Nat. Med. 2019, 25, 301–311. [Google Scholar] [CrossRef] [PubMed]
- El-Khoueiry, A.B.; Tchekmedyian, N.; Sanborn, R.E.; Peguero, J.A.; Pulver, K.; Dembla, V.; Thomas, J.S.; Chiang, G.G.; Densel, M.; Sankar, N.; et al. A phase II, open-label study of tomivosertib (eFT508) added on to continued checkpoint inhibitor therapy in patients (pts) with insufficient response to single-agent treatment. J. Clin. Oncol. 2020, 38, 3112. [Google Scholar] [CrossRef]
- Yang, H.; Li, Z.; Wu, Z.; Chen, X.; Bo, L.; Patel, H.; Zhang, B.; Xiong, W.; Wang, W.; Chen, Z.S. Reversal of ABCG2-mediated drug resistance by tinodasertib (ETC-206). Front. Pharmacol. 2025, 16, 1606857. [Google Scholar] [CrossRef] [PubMed]
- Matos, M.; Kannourakis, G.; Aghmesheh, M.; Mendis, S.R.; Mahon, K.; Dave, H.P.; Barde, P.J.; Schwedat, A.; Patava, J. Safety, tolerability, and preliminary efficacy of tinodasertib as a monotherapy or in combination with pembrolizumab or irinotecan in metastatic colorectal cancer: Interim results from a phase II open-label, dose-finding, run-in and cohort expansion study. J. Clin. Oncol. 2025, 43, 183. [Google Scholar] [CrossRef]
- Fu, S.C.; Huang, H.C.; Horton, P.; Juan, H.F. ValidNESs: A database of validated leucine-rich nuclear export signals. Nucleic Acids Res. 2013, 41, D338–D343. [Google Scholar] [CrossRef]
- Azizian, N.G.; Li, Y. XPO1-dependent nuclear export as a target for cancer therapy. J. Hematol. Oncol. 2020, 13, 61. [Google Scholar] [CrossRef]
- Nakayama, R.; Zhang, Y.X.; Czaplinski, J.T.; Anatone, A.J.; Sicinska, E.T.; Fletcher, J.A.; Demetri, G.D.; Wagner, A.J. Preclinical activity of selinexor, an inhibitor of XPO1, in sarcoma. Oncotarget 2016, 7, 16581–16592. [Google Scholar] [CrossRef]
- Tabe, Y.; Kojima, K.; Yamamoto, S.; Sekihara, K.; Matsushita, H.; Davis, R.E.; Wang, Z.; Ma, W.; Ishizawa, J.; Kazuno, S.; et al. Ribosomal Biogenesis and Translational Flux Inhibition by the Selective Inhibitor of Nuclear Export (SINE) XPO1 Antagonist KPT-185. PLoS ONE 2015, 10, e0137210. [Google Scholar] [CrossRef]
- Aladhraei, M.; Kassem Al-Thobhani, A.; Poungvarin, N.; Suwannalert, P. Association of XPO1 Overexpression with NF-kappaB and Ki67 in Colorectal Cancer. Asian Pac. J. Cancer Prev. 2019, 20, 3747–3754. [Google Scholar] [CrossRef]
- Gravina, G.L.; Mancini, A.; Sanita, P.; Vitale, F.; Marampon, F.; Ventura, L.; Landesman, Y.; McCauley, D.; Kauffman, M.; Shacham, S.; et al. Erratum to: KPT-330, a potent and selective exportin-1 (XPO-1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin in prostate cancer models. BMC Cancer 2016, 16, 8. [Google Scholar] [CrossRef]
- Taagepera, S.; McDonald, D.; Loeb, J.E.; Whitaker, L.L.; McElroy, A.K.; Wang, J.Y.; Hope, T.J. Nuclear-cytoplasmic shuttling of C-ABL tyrosine kinase. Proc. Natl. Acad. Sci. USA 1998, 95, 7457–7462. [Google Scholar] [CrossRef]
- Subhash, V.V.; Yeo, M.S.; Wang, L.; Tan, S.H.; Wong, F.Y.; Thuya, W.L.; Tan, W.L.; Peethala, P.C.; Soe, M.Y.; Tan, D.S.P.; et al. Anti-tumor efficacy of Selinexor (KPT-330) in gastric cancer is dependent on nuclear accumulation of p53 tumor suppressor. Sci. Rep. 2018, 8, 12248. [Google Scholar] [CrossRef] [PubMed]
- Volpon, L.; Culjkovic-Kraljacic, B.; Sohn, H.S.; Blanchet-Cohen, A.; Osborne, M.J.; Borden, K.L.B. A biochemical framework for eIF4E-dependent mRNA export and nuclear recycling of the export machinery. RNA 2017, 23, 927–937. [Google Scholar] [CrossRef] [PubMed]
- Topisirovic, I.; Siddiqui, N.; Lapointe, V.L.; Trost, M.; Thibault, P.; Bangeranye, C.; Pinol-Roma, S.; Borden, K.L. Molecular dissection of the eukaryotic initiation factor 4E (eIF4E) export-competent RNP. EMBO J. 2009, 28, 1087–1098. [Google Scholar] [CrossRef] [PubMed]
- Kirli, K.; Karaca, S.; Dehne, H.J.; Samwer, M.; Pan, K.T.; Lenz, C.; Urlaub, H.; Gorlich, D. A deep proteomics perspective on CRM1-mediated nuclear export and nucleocytoplasmic partitioning. Elife 2015, 4, e11466. [Google Scholar] [CrossRef]
- Saulino, D.M.; Younes, P.S.; Bailey, J.M.; Younes, M. CRM1/XPO1 expression in pancreatic adenocarcinoma correlates with survivin expression and the proliferative activity. Oncotarget 2018, 9, 21289–21295. [Google Scholar] [CrossRef]
- Cosson, A.; Chapiro, E.; Bougacha, N.; Lambert, J.; Herbi, L.; Cung, H.A.; Algrin, C.; Keren, B.; Damm, F.; Gabillaud, C.; et al. Gain in the short arm of chromosome 2 (2p+) induces gene overexpression and drug resistance in chronic lymphocytic leukemia: Analysis of the central role of XPO1. Leukemia 2017, 31, 1625–1629. [Google Scholar] [CrossRef]
- Shen, A.; Wang, Y.; Zhao, Y.; Zou, L.; Sun, L.; Cheng, C. Expression of CRM1 in human gliomas and its significance in p27 expression and clinical prognosis. Neurosurgery 2009, 65, 153–159. [Google Scholar] [CrossRef]
- Etchin, J.; Sanda, T.; Mansour, M.R.; Kentsis, A.; Montero, J.; Le, B.T.; Christie, A.L.; McCauley, D.; Rodig, S.J.; Kauffman, M.; et al. KPT-330 inhibitor of CRM1 (XPO1)-mediated nuclear export has selective anti-leukaemic activity in preclinical models of T-cell acute lymphoblastic leukaemia and acute myeloid leukaemia. Br. J. Haematol. 2013, 161, 117–127. [Google Scholar] [CrossRef]
- Landes, J.R.; Moore, S.A.; Bartley, B.R.; Doan, H.Q.; Rady, P.L.; Tyring, S.K. The efficacy of selinexor (KPT-330), an XPO1 inhibitor, on non-hematologic cancers: A comprehensive review. J. Cancer Res. Clin. Oncol. 2023, 149, 2139–2155. [Google Scholar] [CrossRef]
- Abdul Razak, A.R.; Mau-Soerensen, M.; Gabrail, N.Y.; Gerecitano, J.F.; Shields, A.F.; Unger, T.J.; Saint-Martin, J.R.; Carlson, R.; Landesman, Y.; McCauley, D.; et al. First-in-Class, First-in-Human Phase I Study of Selinexor, a Selective Inhibitor of Nuclear Export, in Patients With Advanced Solid Tumors. J. Clin. Oncol. 2016, 34, 4142–4150. [Google Scholar] [CrossRef]
- Chari, A.; Vogl, D.T.; Gavriatopoulou, M.; Nooka, A.K.; Yee, A.J.; Huff, C.A.; Moreau, P.; Dingli, D.; Cole, C.; Lonial, S.; et al. Oral Selinexor-Dexamethasone for Triple-Class Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 381, 727–738. [Google Scholar] [CrossRef] [PubMed]
- Grosicki, S.; Simonova, M.; Spicka, I.; Pour, L.; Kriachok, I.; Gavriatopoulou, M.; Pylypenko, H.; Auner, H.W.; Leleu, X.; Doronin, V.; et al. Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): A randomised, open-label, phase 3 trial. Lancet 2020, 396, 1563–1573. [Google Scholar] [CrossRef] [PubMed]
- Gasparetto, C.; Schiller, G.J.; Tuchman, S.A.; Callander, N.S.; Baljevic, M.; Lentzsch, S.; Rossi, A.C.; Kotb, R.; White, D.; Bahlis, N.J.; et al. Once weekly selinexor, carfilzomib and dexamethasone in carfilzomib non-refractory multiple myeloma patients. Br. J. Cancer 2022, 126, 718–725. [Google Scholar] [CrossRef] [PubMed]
- Gasparetto, C.; Lentzsch, S.; Schiller, G.; Callander, N.; Tuchman, S.; Chen, C.; White, D.; Kotb, R.; Sutherland, H.; Sebag, M.; et al. Selinexor, daratumumab, and dexamethasone in patients with relapsed or refractory multiple myeloma. EJHaem 2021, 2, 56–65. [Google Scholar] [CrossRef]
- Gonzalez-Calle, V.; Rodriguez-Otero, P.; Sureda, A.; De Arriba, F.; Reinoso, M.; Ribas, P.; Gonzalez-Rodriguez, A.P.; Gonzalez, Y.; Oriol, A.; Martinez-Lopez, J.; et al. Selinexor, daratumumab, bortezomib and dexamethasone for the treatment of patients with relapsed or refractory multiple myeloma: Results of the phase II, nonrandomized, multicenter GEM-SELIBORDARA study. Haematologica 2024, 109, 2219–2228. [Google Scholar] [CrossRef]
- White, D.; Schiller, G.J.; Madan, S.; Lentzsch, S.; Chubar, E.; Lavi, N.; Van Domelen, D.R.; Bentur, O.S.; Baljevic, M. Efficacy and safety of once weekly selinexor 40 mg versus 60 mg with pomalidomide and dexamethasone in relapsed and/or refractory multiple myeloma. Front. Oncol. 2024, 14, 1352281. [Google Scholar] [CrossRef]
- Wang, A.Y.; Weiner, H.; Green, M.; Chang, H.; Fulton, N.; Larson, R.A.; Odenike, O.; Artz, A.S.; Bishop, M.R.; Godley, L.A.; et al. A phase I study of selinexor in combination with high-dose cytarabine and mitoxantrone for remission induction in patients with acute myeloid leukemia. J. Hematol. Oncol. 2018, 11, 4. [Google Scholar] [CrossRef]
- Garzon, R.; Savona, M.; Baz, R.; Andreeff, M.; Gabrail, N.; Gutierrez, M.; Savoie, L.; Mau-Sorensen, P.M.; Wagner-Johnston, N.; Yee, K.; et al. A phase 1 clinical trial of single-agent selinexor in acute myeloid leukemia. Blood 2017, 129, 3165–3174. [Google Scholar] [CrossRef]
- Kuruvilla, J.; Savona, M.; Baz, R.; Mau-Sorensen, P.M.; Gabrail, N.; Garzon, R.; Stone, R.; Wang, M.; Savoie, L.; Martin, P.; et al. Selective inhibition of nuclear export with selinexor in patients with non-Hodgkin lymphoma. Blood 2017, 129, 3175–3183. [Google Scholar] [CrossRef]
- Kalakonda, N.; Maerevoet, M.; Cavallo, F.; Follows, G.; Goy, A.; Vermaat, J.S.P.; Casasnovas, O.; Hamad, N.; Zijlstra, J.M.; Bakhshi, S.; et al. Selinexor in patients with relapsed or refractory diffuse large B-cell lymphoma (SADAL): A single-arm, multinational, multicentre, open-label, phase 2 trial. Lancet Haematol. 2020, 7, e511–e522. [Google Scholar] [CrossRef] [PubMed]
- Kasamon, Y.L.; Price, L.S.L.; Okusanya, O.O.; Richardson, N.C.; Li, R.J.; Ma, L.; Wu, Y.T.; Theoret, M.; Pazdur, R.; Gormley, N.J. FDA Approval Summary: Selinexor for Relapsed or Refractory Diffuse Large B-Cell Lymphoma. Oncologist 2021, 26, 879–886. [Google Scholar] [CrossRef] [PubMed]
- Lassman, A.B.; Wen, P.Y.; van den Bent, M.J.; Plotkin, S.R.; Walenkamp, A.M.E.; Green, A.L.; Li, K.; Walker, C.J.; Chang, H.; Tamir, S.; et al. A Phase II Study of the Efficacy and Safety of Oral Selinexor in Recurrent Glioblastoma. Clin. Cancer Res. 2022, 28, 452–460. [Google Scholar] [CrossRef] [PubMed]
- Gavriatopoulou, M.; Chari, A.; Chen, C.; Bahlis, N.; Vogl, D.T.; Jakubowiak, A.; Dingli, D.; Cornell, R.F.; Hofmeister, C.C.; Siegel, D.; et al. Integrated safety profile of selinexor in multiple myeloma: Experience from 437 patients enrolled in clinical trials. Leukemia 2020, 34, 2430–2440. [Google Scholar] [CrossRef]
- Hashmi, H.; Green, K. The 'comeback' of Selinexor: From toxic to tolerable. Curr. Probl. Cancer 2022, 46, 100789. [Google Scholar] [CrossRef]
- Emdal, K.B.; Palacio-Escat, N.; Wigerup, C.; Eguchi, A.; Nilsson, H.; Bekker-Jensen, D.B.; Ronnstrand, L.; Kazi, J.U.; Puissant, A.; Itzykson, R.; et al. Phosphoproteomics of primary AML patient samples reveals rationale for AKT combination therapy and p53 context to overcome selinexor resistance. Cell Rep. 2022, 40, 111177. [Google Scholar] [CrossRef]
- Kwanten, B.; Deconick, T.; Walker, C.; Wang, F.; Landesman, Y.; Daelemans, D. E3 ubiquitin ligase ASB8 promotes selinexor-induced proteasomal degradation of XPO1. Biomed. Pharmacother. 2023, 160, 114305. [Google Scholar] [CrossRef]
- Neggers, J.E.; Vanstreels, E.; Baloglu, E.; Shacham, S.; Landesman, Y.; Daelemans, D. Heterozygous mutation of cysteine528 in XPO1 is sufficient for resistance to selective inhibitors of nuclear export. Oncotarget 2016, 7, 68842–68850. [Google Scholar] [CrossRef]
- Restrepo, P.; Bhalla, S.; Ghodke-Puranik, Y.; Aleman, A.; Leshchenko, V.; Melnekoff, D.T.; Agte, S.; Jiang, J.; Madduri, D.; Richter, J.; et al. A Three-Gene Signature Predicts Response to Selinexor in Multiple Myeloma. JCO Precis. Oncol. 2022, 6, e2200147. [Google Scholar] [CrossRef]
- Totiger, T.M.; Chaudhry, S.; Musi, E.; Afaghani, J.; Montoya, S.; Owusu-Ansah, F.; Lee, S.; Schwartz, G.; Klimek, V.; Taylor, J. Protein biomarkers for response to XPO1 inhibition in haematologic malignancies. J. Cell. Mol. Med. 2023, 27, 587–590. [Google Scholar] [CrossRef]
- Jimenez, I.; Carabia, J.; Bobillo, S.; Palacio, C.; Abrisqueta, P.; Pages, C.; Nieto, J.C.; Castellvi, J.; Martinez-Ricarte, F.; Escoda, L.; et al. Repolarization of tumor infiltrating macrophages and increased survival in mouse primary CNS lymphomas after XPO1 and BTK inhibition. J. Neurooncol. 2020, 149, 13–25. [Google Scholar] [CrossRef]
- Tyler, P.M.; Servos, M.M.; de Vries, R.C.; Klebanov, B.; Kashyap, T.; Sacham, S.; Landesman, Y.; Dougan, M.; Dougan, S.K. Clinical Dosing Regimen of Selinexor Maintains Normal Immune Homeostasis and T-cell Effector Function in Mice: Implications for Combination with Immunotherapy. Mol. Cancer Ther. 2017, 16, 428–439. [Google Scholar] [CrossRef]
- Fisher, J.G.; Doyle, A.D.P.; Graham, L.V.; Sonar, S.; Sale, B.; Henderson, I.; Del Rio, L.; Johnson, P.W.M.; Landesman, Y.; Cragg, M.S.; et al. XPO1 inhibition sensitises CLL cells to NK cell mediated cytotoxicity and overcomes HLA-E expression. Leukemia 2023, 37, 2036–2049. [Google Scholar] [CrossRef]
- Fisher, J.G.; Bartlett, L.G.; Kashyap, T.; Walker, C.J.; Khakoo, S.I.; Blunt, M.D. Modulation of anti-tumour immunity by XPO1 inhibitors. Explor. Target. Anti-Tumor Ther. 2025, 6, 1002310. [Google Scholar] [CrossRef]
- Wang, D.; Fu, H.; Que, Y.; Ruan, H.; Xu, M.; Long, X.; Yu, Q.; Li, C.; Li, Z.; Cai, S.; et al. A novel two-step administration of XPO-1 inhibitor may enhance the effect of anti-BCMA CAR-T in relapsed/refractory extramedullary multiple myeloma. J. Transl. Med. 2023, 21, 812. [Google Scholar] [CrossRef] [PubMed]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gitareja, K.; Chelliah, S.S.; Sanij, E.; Sandhu, S.; Kang, J.; Khot, A. Ribosome Biogenesis and Function in Cancer: From Mechanisms to Therapy. Cancers 2025, 17, 2534. https://doi.org/10.3390/cancers17152534
Gitareja K, Chelliah SS, Sanij E, Sandhu S, Kang J, Khot A. Ribosome Biogenesis and Function in Cancer: From Mechanisms to Therapy. Cancers. 2025; 17(15):2534. https://doi.org/10.3390/cancers17152534
Chicago/Turabian StyleGitareja, Kezia, Shalini S. Chelliah, Elaine Sanij, Shahneen Sandhu, Jian Kang, and Amit Khot. 2025. "Ribosome Biogenesis and Function in Cancer: From Mechanisms to Therapy" Cancers 17, no. 15: 2534. https://doi.org/10.3390/cancers17152534
APA StyleGitareja, K., Chelliah, S. S., Sanij, E., Sandhu, S., Kang, J., & Khot, A. (2025). Ribosome Biogenesis and Function in Cancer: From Mechanisms to Therapy. Cancers, 17(15), 2534. https://doi.org/10.3390/cancers17152534