Next Article in Journal
Improving Drug Safety in Pediatric and Young Adult Patients with Hemato-Oncological Diseases: A Prospective Study of Active Pharmacovigilance
Previous Article in Journal
Recent Advances in Pyrimidine-Based Drugs
Previous Article in Special Issue
Assessment of Aptamer as a Potential Drug Targeted Delivery for Retinal Angiogenesis Inhibition
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Aptamers’ Potential to Fill Therapeutic and Diagnostic Gaps

by
Alfredo Berzal-Herranz
* and
Cristina Romero-López
*
Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas. PTS Granada, Av. del Conocimiento 17, 18016 Granada, Spain
*
Authors to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(1), 105; https://doi.org/10.3390/ph17010105
Submission received: 4 December 2023 / Revised: 22 December 2023 / Accepted: 4 January 2024 / Published: 12 January 2024
(This article belongs to the Special Issue Potential of the Aptamers to Fill Therapeutic and Diagnostic Gaps)
More than 30 years ago, in 1990, three independent research groups published several papers demonstrating that genetics could be performed in vitro in the absence of living organisms or cells [1,2,3,4]. This represented a major breakthrough in experimental Molecular Biology, defining the basis for what we now call vitro molecular selection strategies [5]. These strategies allow the selection of nucleic acid molecules, genotype, that can express a specific phenotype on their own. One phenotype that has attracted significant interest and has been widely exploited is nucleic acids’ ability to bind to a specific target molecule, to the extent that the selection of nucleic acids capable of binding specifically and with high affinity to a defined target now constitutes its own dedicated discipline, known as aptamer technology. These efficient nucleic acid binders are referred to as aptamers and the procedure used to select them is known as SELEX, terms coined by Ellington and Szostak [1] and Tuerk and Gold [2], respectively. Aptamers are short RNA or DNA oligonucleotides, and their functionality resides in their three-dimensional structure. Although different procedures have been developed to improve the aptamers’ efficiency and to extend the technical procedure of SELEX [6,7,8,9,10,11,12,13], the technology adheres to the basic principles defined by Gold and Szostak’s research groups back in 1990.
As a result of the thousands of manuscripts published in the past thirty years that describe the selection of RNA or DNA aptamers and their applications, it is widely accepted that they have a broad range of potential applications in different fields of knowledge regarding aptamers technology [14,15,16,17]. Firstly, aptamers are considered excellent candidates for the development of therapeutic molecules to fight different diseases [18,19,20,21,22,23]. Many assays have been performed to investigate their potential to combat infectious diseases caused by RNA viruses, either by targeting a variety of cellular or viral proteins, e.g., [24,25,26,27,28] or by targeting structural elements of the viral RNA genome [29,30,31,32,33,34,35,36,37,38,39]. These publications reported the achievement of a range of therapeutic activity levels in cell culture assays that highlighted the potential of this molecular antiviral strategy. Cancer, in its broadest sense, attracts a great deal of interest and many resources, and aptamer technology is no stranger to this. Cancer treatment has also been the focus of many aptamer-based studies [40,41,42,43,44,45,46]. However, other diseases such as neurodegenerative or cardiovascular diseases, among others, have also been targeted in aptamer studies [47,48,49,50,51,52]. In addition to the development of aptamers as therapeutic molecules, other notable approaches include the applications of aptamers as biosensors, through the development of aptamer-based devices and platforms for disease diagnosis [53,54,55,56,57,58,59,60,61,62,63,64], and as molecular tools to deliver therapeutic drugs to specific target cells, as well as to improve access to malignant molecules whose inactivation is desirable [65,66,67,68,69,70]. These applications have driven the significant development of the aptamers-based discipline in recent years.
In this Special Issue, we have compiled a collection of original articles and reviews focusing on the various factors that define aptamer technology and the main trends in the field. Wrenger and coworkers comprehensively summarize the recent achievements in applying aptamers to the diagnosis of infectious viral diseases, with particular focus on review work applied to RNA viruses such as flaviviruses, influenza virus and coronaviruses [71]. Their review highlights the importance of this discipline aimed at applying aptamers to pathogen detection, which is probably one of the fastest growing applications of aptamers.
Five original research articles included in this Special Issue cover a variety of issues related to using aptamers to treat and diagnose cancer. Dr. González et al. provide evidence of the potential application of selected DNA aptamers, directed against the vaccinia-related kinase 1 (VRK1), as anti-cancer drugs [72]. VRK1 is involved in cell cycle progression [73] and has been linked to the development of several types of cancer [74,75,76,77,78,79,80]. They show inhibition of the cell cycle progression and induction of apoptosis of the MCF7 breast cancer cell line treated with aptamers. Dr. Dubin’s group selects and characterizes DNA aptamers targeting the human programmed death-ligand 1 (PD-L1), whose expression level reflects the immune status of tumors [81]. The assay—both in cell culture of several human tumor cell lines and in two mouse tumor models—of the selected fluorescently labeled aptamers demonstrates their potential for the development of a non-invasive diagnostic method of tumor classification based on PD-L1 status and, therefore, their usefulness for decision-making in personalized immunotherapy. In another interesting article, Dr. Calzada’s group reports on the optimization of the delivery system of an aptamer probe that has already proven to be efficient in the imaging detection of a cancer biomarker. They have developed a device that significantly improves tumor uptake of the aptamer probes in both cell culture and BALB/c mice [82]. The last two articles within this series describe the development of aptamer-based drug delivery devices [83,84]. In their article, Qi and coworkers describe the design of a gemcitabine dendrimer linked to an aptamer specific for breast cancer cells. The aptamer provides the desired cell specificity to deliver the antitumor drug, reducing off-target cytotoxic effects and enhancing specific activity against target cells, improving the therapeutic efficacy of the drug [83]. Heus’ group also designed a smart approach that can be used to improve cell specificity and drug delivery efficiency [84]. They applied an innovative 3D-SELEX strategy against cellular spheroids of breast cancer tumor cells, using non-malignant breast cells as target for counter selection. They demonstrated the specificity of selected aptamers for the breast tumor cells. Further, they designed aptamer multimers which intercalate DNA boxes that efficiently bind and carry doxorubicin, a very potent chemotherapeutic drug. This approach can significantly reduce the undesirable side effects of the drug [84].
This Special Issue also includes two original research articles focusing on the applications of aptamers as anticoagulant tools, with demonstrable applications in the fight against ischemic stroke. In the first of these articles, Shea and coworkers characterize the biological activity of a previously described RNA aptamer, BB-031, targeted against the Von Willebrand Factor (VWF), which plays a critical role in thrombosis [85]. Their study uses a microfluidic model of arterial occlusion. They show that BB-031 induces thrombolysis in this model, providing evidence of dose-dependent inhibition of VWF by the aptamer that leads to a reduction in thrombus surface area and recanalization. On the other hand, Dr. Pasternak’s group has addressed the optimization of antithrombin DNA aptamers through the introduction of chemical modifications. In particular, they analyze the effect of pyrrolo-2′-deoxycytidine (Py-dC) and its derivatives modifications. They demonstrate that while all variants tested exhibited anticoagulant activity, a variant with a decyl derivative of Py-dC containing a long, linear aliphatic side chain exhibited greater thrombin inhibition activity [86]. As indicated above, the activity of the aptamers relies on their 3D structure. The antithrombin aptamers optimized by Pasternak and co-workers adopt a G-quadruplex structure, whose thermodynamic stability is preserved with little variation in the chemically modified variants tested. Interestingly, this G-quadruplex structure is shared by other aptamers isolated in independent experiments targeting very different molecules. This is the case for AT11-L0, a DNA aptamer targeted against the nucleolin (NCL), a nuclear protein that plays an important role in angiogenesis in retinal neovascular diseases, since its inhibition promotes an antiangiogenic effect [87]. This aptamer has been used by Moreira and co-workers to functionalize liposomes that could be loaded with antiangiogenetic drugs [88]. The authors show that the complexes allow efficient targeting of NCL, making liposomes functionalized with this aptamer a promising tool to enable effective anti-angiogenic drug delivery. Recent advances in G-quadruplex-mediated cancer therapy are described in [89].
Finally, Dr. Maher III’s group aimed to understand the biological mechanism of a previously described DNA aptamer conjugate with streptavidine, also capable of adopting G-quadruplex structure, shown to promote remyelination in a mouse model of chronic spinal cord demyelination [90]. In their most recent paper, the authors provide evidence that this macromolecular aptamer-based tool binds to the cell membrane of human and adult rat oligodendrocytes, suggesting that binding to a membrane molecule triggers the remyelination pathway [91].
This set of original research contributions providing evidence of the versatility of applying aptamers for the diagnosis or treatment of complex diseases is completed by two very interesting review articles by the groups of Drs. Ilgu and Marty, respectively [92,93]. Dr. Ilgu’s group nicely summarizes the main achievements in the application of aptamers to the treatment or diagnosis of neurological diseases, highlighting the potential future development of the field [92]. Meanwhile, Dr. Marty et al. present a comprehensive review of the current state of the art in biomarker detection using electrochemical aptasensors [93]. The authors provide a very careful summary of disease diagnostic strategies by detecting a variety of biomarkers using an electrochemical aptasensor. They provide examples of biomarkers for the early detection of cancer, heart diseases, degenerative diseases (such as Alzheimer’s or multiple sclerosis), diabetes or infectious diseases. The review also includes very interesting information on the functioning mechanisms and properties of the electrochemical aptasensors. The advantages and future perspectives of aptasensors are also discussed [93].
Aptamers, as specific and efficient binders, have inevitably been compared to antibodies in terms of their potential analytical and diagnostic applications. In relation to this, Dr. Bruno has provided a very interesting article in which he discusses situations in which aptamers can fill the gap left by antibodies, indicating that there is no need to compete with antibodies and it is necessary to decide which molecule may be most appropriate for each specific problem [94]. He offers a very interesting reflection on the futility of competing with antibodies and the need to look and identify (using his same words) “niches were aptamers are truly needed or wanted”. Readers can learn a lot from this interesting review article [94]. Continuing this line of investigation, Marty and co-workers provide another review article summarizing the applications of aptamers in the lateral flow assay procedure for rapid detection of different analytes, biomarkers or specific molecules in point-of-care diagnostics. The aptamers are used to substitute the antibodies in this widely used in vitro detection platform, in which several limitations derived from the use of antibodies have been identified. At the same time, the use of aptamers also has some drawbacks that the authors highlight in this article.
The influence of two physical properties of protein ligands, molecular weight and isoelectric point, on aptamers’ affinity has been studied by Fischer and co-workers. Their study including nearly 300 target proteins and peptides spanning a wide range of isoelectric points and molecular weight concludes that there is a significant inverse correlation between the isoelectric point of protein ligands and aptamers’ affinity. In contrast, there appears to be no correlation with molecular mass [95].
In a comprehensive review article, Tickner and Farzan discuss the utility of riboswitches for optimizing adeno-associated viruses (AAV) as transgene vehicles in gene therapy [96]. Riboswitches are structural RNA elements that change their structure as a result of their specific binding of a small molecule, leading to a change in the function of the RNA molecules in which they are contained. Riboswitches constitute an aptamer included in an RNA regulatory element, whose structure is sensitive to aptamer–ligand binding. Original natural riboswitches were identified as regulators of gene expression in bacteria [97,98,99]. Riboswitches have been artificially engineered to develop ligand-responsive regulator devices [100,101,102,103,104,105,106]. The review by Tickner and co-workers provides a detailed description of different types of artificial riboswitches described to date that function in human cells, explaining their mechanisms and their applications in the context of AAV-mediated transgenes delivery [96].
The Special Issue concludes with a very interesting article by Andrianova and Kuznetsov, who review a topic of great interest: Biocomputing based on the use of DNA aptamers and their most important feature—their excellent binding abilities—for the creation of logic gates applied to medicine and analytical chemistry [107]. A logic gate performs a Boolean function, a basic logical function that converts the input signals into a logic output. Briefly, an aptamer-based biosensor device constitutes a logic gate that, in the presence of the ligand, generates an output signal. In this review, the authors summarize different biocomputing approaches involving aptamers, describing the various methodologies applied to detect an output signal. They distinguish between optical and electrochemical methods for obtaining an output signal, which are also explained in detail. This represents an innovative and growing application of aptamers, with medicine and analytical chemistry currently being the fields in which it could have the greatest impact. Its development is supported by the continuous identification of new aptamers against their specific ligands.
This Special Issue constitutes a representative sample of reviews and original research articles, providing a snapshot of the current state of the art of aptamers technology. By reading these papers, readers can get a broad idea of the possibilities of use of aptamers, of different strategies for developing tools based on these molecules, and ways to potentially improve their efficacy. This reinforces their potential use in the clinic, which may currently be the most attractive and desired application for aptamer technology.

Acknowledgments

The Guest Editors would like to thank all the contributors to this special issue.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ellington, A.D.; Szostak, J.W. In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346, 818–822. [Google Scholar] [CrossRef]
  2. Tuerk, C.; Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249, 505–510. [Google Scholar] [CrossRef]
  3. Robertson, D.L.; Joyce, G.F. Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA. Nature 1990, 344, 467–468. [Google Scholar] [CrossRef]
  4. Green, R.; Ellington, A.D.; Szostak, J.W. In vitro genetic analysis of the tetrahymena self-splicing intron. Nature 1990, 347, 406–408. [Google Scholar] [CrossRef]
  5. Burke, J.M.; Berzal-Herranz, A. In vitro selection and evolution of RNA: Applications for catalytic RNA, molecular recognition, and drug discovery. FASEB J. 1993, 7, 106–112. [Google Scholar] [CrossRef]
  6. Blind, M.; Blank, M. Aptamer selection technology and recent advances. Mol. Ther. Nucleic Acids 2015, 4, e223. [Google Scholar] [CrossRef]
  7. Urak, K.T.; Shore, S.; Rockey, W.M.; Chen, S.J.; McCaffrey, A.P.; Giangrande, P.H. In vitro RNA SELEX for the generation of chemically-optimized therapeutic rna drugs. Methods 2016, 103, 167–174. [Google Scholar] [CrossRef]
  8. Sharma, T.K.; Bruno, J.G.; Dhiman, A. ABCs of DNA aptamer and related assay development. Biotechnol. Adv. 2017, 35, 275–301. [Google Scholar] [CrossRef]
  9. Kratschmer, C.; Levy, M. Effect of chemical modifications on aptamer stability in serum. Nucleic Acid. Ther. 2017, 27, 335–344. [Google Scholar] [CrossRef]
  10. Shen, R.; Tan, J.; Yuan, Q. Chemically modified aptamers in biological analysis. ACS Appl. Bio Mater. 2020, 3, 2816–2826. [Google Scholar] [CrossRef]
  11. Oliveira, R.; Pinho, E.; Sousa, A.L.; DeStefano, J.J.; Azevedo, N.F.; Almeida, C. Improving aptamer performance with nucleic acid mimics: De novo and post-SELEX approaches. Trends Biotechnol. 2022, 40, 549–563. [Google Scholar] [CrossRef]
  12. Ji, D.; Feng, H.; Liew, S.W.; Kwok, C.K. Modified nucleic acid aptamers: Development, characterization, and biological applications. Trends Biotechnol. 2023, 41, 1360–1384. [Google Scholar] [CrossRef] [PubMed]
  13. Zhu, C.; Feng, Z.; Qin, H.; Chen, L.; Yan, M.; Li, L.; Qu, F. Recent progress of SELEX methods for screening nucleic acid aptamers. Talanta 2024, 266, 124998. [Google Scholar] [CrossRef]
  14. Nimjee, S.M.; Rusconi, C.P.; Sullenger, B.A. Aptamers: An emerging class of therapeutics. Annu. Rev. Med. 2005, 56, 555–583. [Google Scholar] [CrossRef]
  15. Marton, S.; Reyes-Darias, J.A.; Sanchez-Luque, F.J.; Romero-Lopez, C.; Berzal-Herranz, A. In vitro and ex vivo selection procedures for identifying potentially therapeutic DNA and RNA molecules. Molecules 2010, 15, 4610–4638. [Google Scholar] [CrossRef]
  16. Nimjee, S.M.; White, R.R.; Becker, R.C.; Sullenger, B.A. Aptamers as therapeutics. Annu. Rev. Pharmacol. Toxicol. 2017, 57, 61–79. [Google Scholar] [CrossRef]
  17. Askari, A.; Kota, S.; Ferrell, H.; Swamy, S.; Goodman, K.S.; Okoro, C.C.; Spruell Crenshaw, I.C.; Hernandez, D.K.; Oliphant, T.E.; Badrayani, A.A.; et al. UTexas aptamer database: The collection and long-term preservation of aptamer sequence information. Nucleic Acids Res. 2023, 52, gkad959. [Google Scholar] [CrossRef]
  18. Dausse, E.; Da Rocha Gomes, S.; Toulme, J.J. Aptamers: A new class of oligonucleotides in the drug discovery pipeline? Curr. Opin. Pharmacol. 2009, 9, 602–607. [Google Scholar] [CrossRef]
  19. Zhou, J.; Rossi, J. Aptamers as targeted therapeutics: Current potential and challenges. Nat. Rev. Drug Discov. 2017, 16, 181–202. [Google Scholar] [CrossRef]
  20. Zhu, G.; Chen, X. Aptamer-based targeted therapy. Adv. Drug Deliv. Rev. 2018, 134, 65–78. [Google Scholar] [CrossRef]
  21. Ismail, S.I.; Alshaer, W. Therapeutic aptamers in discovery, preclinical and clinical stages. Adv. Drug Deliv. Rev. 2018, 134, 51–64. [Google Scholar] [CrossRef] [PubMed]
  22. Allemailem, K.S.; Almatroudi, A.; Alsahli, M.A.; Basfar, G.T.; Alrumaihi, F.; Rahmani, A.H.; Khan, A.A. Recent advances in understanding oligonucleotide aptamers and their applications as therapeutic agents. 3 Biotech 2020, 10, 551. [Google Scholar] [CrossRef]
  23. Ni, S.; Zhuo, Z.; Pan, Y.; Yu, Y.; Li, F.; Liu, J.; Wang, L.; Wu, X.; Li, D.; Wan, Y.; et al. Recent progress in aptamer discoveries and modifications for therapeutic applications. ACS Appl. Mater. Interfaces 2021, 13, 9500–9519. [Google Scholar] [CrossRef]
  24. Gopinath, S.C. Methods developed for SELEX. Anal. Bioanal. Chem. 2007, 387, 171–182. [Google Scholar] [CrossRef]
  25. Chen, H.L.; Hsiao, W.H.; Lee, H.C.; Wu, S.C.; Cheng, J.W. Selection and characterization of DNA aptamers targeting all four serotypes of dengue viruses. PLoS ONE 2015, 10, e0131240. [Google Scholar] [CrossRef]
  26. Torres-Vazquez, B.; de Lucas, A.M.; Garcia-Crespo, C.; Garcia-Martin, J.A.; Fragoso, A.; Fernandez-Algar, M.; Perales, C.; Domingo, E.; Moreno, M.; Briones, C. In vitro selection of high affinity DNA and RNA aptamers that detect hepatitis C virus core protein of genotypes 1 to 4 and inhibit virus production in cell culture. J. Mol. Biol. 2022, 434, 167501. [Google Scholar] [CrossRef]
  27. Zhang, W.; Xiao, L.; Luo, J.; Wu, M.; Zhu, Y.; Cong, F. Application of aptamer-based viral detection in animals. Pol. J. Vet. Sci. 2023, 26, 521–529. [Google Scholar] [CrossRef]
  28. Chang, Z.Y.; Alhamami, F.; Chin, K.L. Aptamer-based strategies to address challenges in COVID-19 diagnosis and treatments. Interdiscip. Perspect. Infect. Dis. 2023, 2023, 9224815. [Google Scholar] [CrossRef]
  29. Romero-López, C.; Díaz-González, R.; Berzal-Herranz, A. Inhibition of hepatitis C virus internal ribosome entry site-mediated translation by an RNA targeting the conserved IIIf domain. Cell Mol. Life Sci. 2007, 64, 2994–3006. [Google Scholar] [CrossRef]
  30. Romero-López, C.; Díaz-González, R.; Barroso-delJesus, A.; Berzal-Herranz, A. Inhibition of hepatitis C virus replication and internal ribosome entry site-dependent translation by an RNA molecule. J. Gen. Virol. 2009, 90, 1659–1669. [Google Scholar] [CrossRef] [PubMed]
  31. Watrin, M.; Dausse, E.; Lebars, I.; Rayner, B.; Bugaut, A.; Toulme, J.J. Aptamers targeting RNA molecules. Methods Mol. Biol. 2009, 535, 79–105. [Google Scholar] [CrossRef]
  32. Romero-López, C.; Berzal-Herranz, B.; Gómez, J.; Berzal-Herranz, A. An engineered inhibitor RNA that efficiently interferes with hepatitis C virus translation and replication. Antivir. Res. 2012, 94, 131–138. [Google Scholar] [CrossRef]
  33. Marton, S.; Romero-Lopez, C.; Berzal-Herranz, A. RNA aptamer-mediated interference of HCV replication by targeting the CRE-5BSL3.2 domain. J. Viral Hepat. 2013, 20, 103–112. [Google Scholar] [CrossRef] [PubMed]
  34. Sánchez-Luque, F.J.; Stich, M.; Manrubia, S.; Briones, C.; Berzal-Herranz, A. Efficient HIV-1 inhibition by a 16 nt-long RNA aptamer designed by combining in vitro selection and in silico optimisation strategies. Sci. Rep. 2014, 4, 6242. [Google Scholar] [CrossRef] [PubMed]
  35. Fernández-Sanlés, A.; Berzal-Herranz, B.; González-Matamala, R.; Ríos-Marco, P.; Romero-López, C.; Berzal-Herranz, A. RNA aptamers as molecular tools to study the functionality of the hepatitis C virus CRE region. Molecules 2015, 20, 16030–16047. [Google Scholar] [CrossRef] [PubMed]
  36. Durand, G.; Dausse, E.; Goux, E.; Fiore, E.; Peyrin, E.; Ravelet, C.; Toulme, J.J. A combinatorial approach to the repertoire of RNA kissing motifs; towards multiplex detection by switching hairpin aptamers. Nucleic Acids Res. 2016, 44, 4450–4459. [Google Scholar] [CrossRef]
  37. Toulme, J.J.; Peyrin, E.; Duconge, F. Nucleic acid aptamers. Methods 2016, 97, 1–2. [Google Scholar] [CrossRef]
  38. Sett, A.; Zara, L.; Dausse, E.; Toulme, J.J. Engineering light-up aptamers for the detection of rna hairpins through kissing interaction. Anal. Chem. 2020, 92, 9113–9117. [Google Scholar] [CrossRef]
  39. Panda, K.; Alagarasu, K.; Parashar, D. Oligonucleotide-based approaches to inhibit dengue virus replication. Molecules 2021, 26, 956. [Google Scholar] [CrossRef]
  40. Santulli-Marotto, S.; Nair, S.K.; Rusconi, C.; Sullenger, B.; Gilboa, E. Multivalent RNA aptamers that inhibit CTLA-4 and enhance tumor immunity. Cancer Res. 2003, 63, 7483–7489. [Google Scholar]
  41. Tabatabaee, A.; Nafari, B.; Farhang, A.; Hariri, A.; Khosravi, A.; Zarrabi, A.; Mirian, M. Targeting vimentin: A multifaceted approach to combatting cancer metastasis and drug resistance. Cancer Metastasis Rev. 2023; ahead of print. [Google Scholar] [CrossRef]
  42. Szymanowski, W.; Szymanowska, A.; Bielawska, A.; Lopez-Berestein, G.; Rodriguez-Aguayo, C.; Amero, P. Aptamers as potential therapeutic tools for ovarian cancer: Advancements and challenges. Cancers 2023, 15, 5300. [Google Scholar] [CrossRef]
  43. Venkatesan, S.; Chanda, K.; Balamurali, M.M. Recent advancements of aptamers in cancer therapy. ACS Omega 2023, 8, 32231–32243. [Google Scholar] [CrossRef] [PubMed]
  44. Razlansari, M.; Jafarinejad, S.; Rahdar, A.; Shirvaliloo, M.; Arshad, R.; Fathi-Karkan, S.; Mirinejad, S.; Sargazi, S.; Sheervalilou, R.; Ajalli, N.; et al. Development and classification of RNA aptamers for therapeutic purposes: An updated review with emphasis on cancer. Mol. Cell Biochem. 2023, 478, 1573–1598. [Google Scholar] [CrossRef] [PubMed]
  45. Goh, K.W.; Stephen, A.; Wu, Y.S.; Sim, M.S.; Batumalaie, K.; Gopinath, S.C.B.; Guad, R.M.; Kumar, A.; Sekar, M.; Subramaniyan, V.; et al. Molecular targets of aptamers in gastrointestinal cancers: Cancer detection, therapeutic applications, and associated mechanisms. J. Cancer 2023, 14, 2491–2516. [Google Scholar] [CrossRef]
  46. Ciccone, G.; Ibba, M.L.; Coppola, G.; Catuogno, S.; Esposito, C.L. The small RNA landscape in NSCLC: Current therapeutic applications and progresses. Int. J. Mol. Sci. 2023, 24, 6121. [Google Scholar] [CrossRef]
  47. Woodruff, R.S.; Sullenger, B.A. Modulation of the coagulation cascade using aptamers. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 2083–2091. [Google Scholar] [CrossRef]
  48. Qu, J.; Yu, S.; Zheng, Y.; Zheng, Y.; Yang, H.; Zhang, J. Aptamer and its applications in neurodegenerative diseases. Cell Mol. Life Sci. 2017, 74, 683–695. [Google Scholar] [CrossRef] [PubMed]
  49. Chabata, C.V.; Frederiksen, J.W.; Sullenger, B.A.; Gunaratne, R. Emerging applications of aptamers for anticoagulation and hemostasis. Curr. Opin. Hematol. 2018, 25, 382–388. [Google Scholar] [CrossRef]
  50. Zheng, Y.; Zhang, L.; Zhao, J.; Li, L.; Wang, M.; Gao, P.; Wang, Q.; Zhang, X.; Wang, W. Advances in aptamers against Ab and applications in Ab detection and regulation for alzheimer’s disease. Theranostics 2022, 12, 2095–2114. [Google Scholar] [CrossRef]
  51. Kong, A.H.; Wu, A.J.; Ho, O.K.; Leung, M.M.; Huang, A.S.; Yu, Y.; Zhang, G.; Lyu, A.; Li, M.; Cheung, K.H. Exploring the potential of aptamers in targeting neuroinflammation and neurodegenerative disorders: Opportunities and challenges. Int. J. Mol. Sci. 2023, 24, 11780. [Google Scholar] [CrossRef]
  52. Nourizad, A.; Golmohammadi, S.; Aghanejad, A.; Tohidkia, M.R. Recent trends in aptamer-based nanobiosensors for detection of vascular endothelial growth factors (VEGFs) biomarker: A review. Environ. Res. 2023, 236, 116726. [Google Scholar] [CrossRef] [PubMed]
  53. Davydova, A.; Vorobyeva, M. Aptamer-based biosensors for the colorimetric detection of blood biomarkers: Paving the way to clinical laboratory testing. Biomedicines 2022, 10, 1606. [Google Scholar] [CrossRef]
  54. Lafi, Z.; Gharaibeh, L.; Nsairat, H.; Asha, N.; Alshaer, W. Aptasensors: Employing molecular probes for precise medical diagnostics and drug monitoring. Bioanalysis 2023, 15, 1439–1460. [Google Scholar] [CrossRef]
  55. Lee, M.; Shin, S.; Kim, S.; Park, N. Recent advances in biological applications of aptamer-based fluorescent biosensors. Molecules 2023, 28, 7327. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, J.; Chen, D.; Huang, W.; Yang, N.; Yuan, Q.; Yang, Y. Aptamer-functionalized field-effect transistor biosensors for disease diagnosis and environmental monitoring. Exploration 2023, 3, 20210027. [Google Scholar] [CrossRef] [PubMed]
  57. Niazi, S.; Khan, I.M.; Akhtar, W.; Ul Haq, F.; Pasha, I.; Khan, M.K.I.; Mohsin, A.; Ahmad, S.; Zhang, Y.; Wang, Z. Aptamer functionalized gold nanoclusters as an emerging nanoprobe in biosensing, diagnostic, catalysis and bioimaging. Talanta 2023, 268, 125270. [Google Scholar] [CrossRef] [PubMed]
  58. Park, S.; Cho, E.; Chueng, S.D.; Yoon, J.S.; Lee, T.; Lee, J.H. Aptameric fluorescent biosensors for liver cancer diagnosis. Biosensors 2023, 13, 617. [Google Scholar] [CrossRef] [PubMed]
  59. Kara, N.; Ayoub, N.; Ilgu, H.; Fotiadis, D.; Ilgu, M. Aptamers targeting membrane proteins for sensor and diagnostic applications. Molecules 2023, 28, 3728. [Google Scholar] [CrossRef]
  60. Li, Y.; Tam, W.W.; Yu, Y.; Zhuo, Z.; Xue, Z.; Tsang, C.; Qiao, X.; Wang, X.; Wang, W.; Li, Y.; et al. The application of aptamer in biomarker discovery. Biomark. Res. 2023, 11, 70. [Google Scholar] [CrossRef]
  61. Kizilkurtlu, A.A.; Demirbas, E.; Agel, H.E. Electrochemical aptasensors for pathogenic detection toward point-of-care diagnostics. Biotechnol. Appl. Biochem. 2023, 70, 1460–1479. [Google Scholar] [CrossRef]
  62. Zhang, T.; Liu, J.; Zhang, L.; Irfan, M.; Su, X. Recent advances in aptamer-based biosensors for potassium detection. Analyst 2023, 148, 5340–5354. [Google Scholar] [CrossRef]
  63. Liang, G.; Song, L.; Gao, Y.; Wu, K.; Guo, R.; Chen, R.; Zhen, J.; Pan, L. Aptamer sensors for the detection of antibiotic residues- a mini-review. Toxics 2023, 11, 513. [Google Scholar] [CrossRef]
  64. Yang, L.F.; Ling, M.; Kacherovsky, N.; Pun, S.H. Aptamers 101: Aptamer discovery and in vitro applications in biosensors and separations. Chem. Sci. 2023, 14, 4961–4978. [Google Scholar] [CrossRef] [PubMed]
  65. Romero-López, C.; Barroso-delJesus, A.; Puerta-Fernández, E.; Berzal-Herranz, A. Interfering with hepatitis C virus IRES activity using RNA molecules identified by a novel in vitro selection method. Biol. Chem. 2005, 386, 183–190. [Google Scholar] [CrossRef]
  66. Zhang, T.; Jin, X.; Zhang, N.; Jiao, X.; Ma, Y.; Liu, R.; Liu, B.; Li, Z. Targeted drug delivery vehicles mediated by nanocarriers and aptamers for posterior eye disease therapeutics: Barriers, recent advances and potential opportunities. Nanotechnology 2022, 33, 162001. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, G.; Mao, D.; Wang, X.; Chen, J.; Gu, C.; Huang, S.; Yang, Y.; Zhang, F.; Tan, W. Aptamer-based self-assembled nanomicelle enables efficient and targeted drug delivery. J. Nanobiotechnol. 2023, 21, 415. [Google Scholar] [CrossRef]
  68. Jabbari, A.; Sameiyan, E.; Yaghoobi, E.; Ramezani, M.; Alibolandi, M.; Abnous, K.; Taghdisi, S.M. Aptamer-based targeted delivery systems for cancer treatment using DNA origami and DNA nanostructures. Int. J. Pharm. 2023, 646, 123448. [Google Scholar] [CrossRef] [PubMed]
  69. Giles, B.; Nakhjavani, M.; Wiesa, A.; Knight, T.; Shigdar, S.; Samarasinghe, R.M. Unravelling the glioblastoma tumour microenvironment: Can aptamer targeted delivery become successful in treating brain cancers? Cancers 2023, 15, 4376. [Google Scholar] [CrossRef]
  70. He, S.; Du, Y.; Tao, H.; Duan, H. Advances in aptamer-mediated targeted delivery system for cancer treatment. Int. J. Biol. Macromol. 2023, 238, 124173. [Google Scholar] [CrossRef]
  71. Kruger, A.; de Jesus Santos, A.P.; de Sa, V.; Ulrich, H.; Wrenger, C. Aptamer applications in emerging viral diseases. Pharmaceuticals 2021, 14, 622. [Google Scholar] [CrossRef]
  72. Carrion-Marchante, R.; Frezza, V.; Salgado-Figueroa, A.; Perez-Morgado, M.I.; Martin, M.E.; Gonzalez, V.M. DNA aptamers against vaccinia-related kinase (VRK) 1 block proliferation in MCF7 breast cancer cells. Pharmaceuticals 2021, 14, 473. [Google Scholar] [CrossRef] [PubMed]
  73. Valbuena, A.; Sanz-Garcia, M.; Lopez-Sanchez, I.; Vega, F.M.; Lazo, P.A. Roles of VRK1 as a new player in the control of biological processes required for cell division. Cell Signal 2011, 23, 1267–1272. [Google Scholar] [CrossRef] [PubMed]
  74. Santos, C.R.; Rodriguez-Pinilla, M.; Vega, F.M.; Rodriguez-Peralto, J.L.; Blanco, S.; Sevilla, A.; Valbuena, A.; Hernandez, T.; van Wijnen, A.J.; Li, F.; et al. VRK1 signaling pathway in the context of the proliferation phenotype in head and neck squamous cell carcinoma. Mol. Cancer Res. 2006, 4, 177–185. [Google Scholar] [CrossRef] [PubMed]
  75. Fournier, M.V.; Martin, K.J.; Kenny, P.A.; Xhaja, K.; Bosch, I.; Yaswen, P.; Bissell, M.J. Gene expression signature in organized and growth-arrested mammary acini predicts good outcome in breast cancer. Cancer Res. 2006, 66, 7095–7102. [Google Scholar] [CrossRef] [PubMed]
  76. Valbuena, A.; Suarez-Gauthier, A.; Lopez-Rios, F.; Lopez-Encuentra, A.; Blanco, S.; Fernandez, P.L.; Sanchez-Cespedes, M.; Lazo, P.A. Alteration of the VRK1-p53 autoregulatory loop in human lung carcinomas. Lung Cancer 2007, 58, 303–309. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, N.; Kwon, J.H.; Kim, Y.B.; Kim, S.H.; Park, S.J.; Xu, W.; Jung, H.Y.; Kim, K.T.; Wang, H.J.; Choi, K.Y. Vaccinia-related kinase 1 promotes hepatocellular carcinoma by controlling the levels of cell cycle regulators associated with G1/S transition. Oncotarget 2015, 6, 30130–30148. [Google Scholar] [CrossRef]
  78. Huang, W.; Cui, X.; Chen, Y.; Shao, M.; Shao, X.; Shen, Y.; Liu, Q.; Wu, M.; Liu, J.; Ni, W.; et al. High VRK1 expression contributes to cell proliferation and survival in hepatocellular carcinoma. Pathol. Res. Pract. 2016, 212, 171–178. [Google Scholar] [CrossRef] [PubMed]
  79. Li, J.; Wang, T.; Pei, L.; Jing, J.; Hu, W.; Sun, T.; Liu, H. Expression of VRK1 and the downstream gene BANF1 in esophageal cancer. Biomed. Pharmacother. 2017, 89, 1086–1091. [Google Scholar] [CrossRef]
  80. Ben, Z.; Gong, L.; Qiu, Y. High expression of VRK1 is related to poor prognosis in glioma. Pathol. Res. Pract. 2018, 214, 112–118. [Google Scholar] [CrossRef]
  81. Malicki, S.; Pucelik, B.; Zyla, E.; Benedyk-Machaczka, M.; Galan, W.; Golda, A.; Sochaj-Gregorczyk, A.; Kaminska, M.; Encarnacao, J.C.; Chruscicka, B.; et al. Imaging of clear cell renal carcinoma with immune checkpoint targeting aptamer-based probe. Pharmaceuticals 2022, 15, 697. [Google Scholar] [CrossRef]
  82. Castelli, R.; Ibarra, M.; Faccio, R.; Miraballes, I.; Fernandez, M.; Moglioni, A.; Cabral, P.; Cerecetto, H.; Glisoni, R.J.; Calzada, V. T908 polymeric micelles improved the uptake of Sgc8-c aptamer probe in tumor-bearing mice: A co-association study between the probe and preformed nanostructures. Pharmaceuticals 2021, 15, 15. [Google Scholar] [CrossRef] [PubMed]
  83. Qi, J.; Zeng, Z.; Chen, Z.; Nipper, C.; Liu, X.; Wan, Q.; Chen, J.; Tung, C.H.; Zu, Y. Aptamer-gemcitabine conjugates with enzymatically cleavable linker for targeted delivery and intracellular drug release in cancer cells. Pharmaceuticals 2022, 15, 558. [Google Scholar] [CrossRef] [PubMed]
  84. Nelissen, F.H.T.; Peeters, W.J.M.; Roelofs, T.P.; Nagelkerke, A.; Span, P.N.; Heus, H.A. Improving breast cancer treatment specificity using aptamers obtained by 3d cell-SELEX. Pharmaceuticals 2021, 14, 349. [Google Scholar] [CrossRef]
  85. Shea, S.M.; Thomas, K.A.; Rassam, R.M.G.; Mihalko, E.P.; Daniel, C.; Sullenger, B.A.; Spinella, P.C.; Nimjee, S.M. Dose-dependent Von Willebrand factor inhibition by aptamer BB-031 correlates with thrombolysis in a microfluidic model of arterial occlusion. Pharmaceuticals 2022, 15, 1450. [Google Scholar] [CrossRef]
  86. Kotkowiak, W.; Jahnz-Wechmann, Z.; Pasternak, A. A comprehensive analysis of the thrombin binding aptamer containing functionalized pyrrolo-2′-deoxycytidines. Pharmaceuticals 2021, 14, 1326. [Google Scholar] [CrossRef] [PubMed]
  87. Leaderer, D.; Cashman, S.M.; Kumar-Singh, R. Topical application of a G-quartet aptamer targeting nucleolin attenuates choroidal neovascularization in a model of age-related macular degeneration. Exp. Eye Res. 2015, 140, 171–178. [Google Scholar] [CrossRef] [PubMed]
  88. Moreira, D.; Lopes-Nunes, J.; Santos, F.M.; Campello, M.P.C.; Oliveira, M.C.; Paulo, A.; Tomaz, C.; Cruz, C. Assessment of aptamer as a potential drug targeted delivery for retinal angiogenesis inhibition. Pharmaceuticals 2023, 16, 751. [Google Scholar] [CrossRef]
  89. Carvalho, J.; Mergny, J.L.; Salgado, G.F.; Queiroz, J.A.; Cruz, C. G-quadruplex, friend or foe: The role of the G-quartet in anticancer strategies. Trends Mol. Med. 2020, 26, 848–861. [Google Scholar] [CrossRef]
  90. Nastasijevic, B.; Wright, B.R.; Smestad, J.; Warrington, A.E.; Rodriguez, M.; Maher, L.J., 3rd. Remyelination induced by a DNA aptamer in a mouse model of multiple sclerosis. PLoS ONE 2012, 7, e39595. [Google Scholar] [CrossRef]
  91. Fereidan-Esfahani, M.; Yue, W.Y.; Wilbanks, B.; Johnson, A.J.; Warrington, A.E.; Howe, C.L.; Rodriguez, M.; Maher, L.J., 3rd. Remyelination-promoting DNA aptamer conjugate myaptavin-3064 binds to adult oligodendrocytes in vitro. Pharmaceuticals 2020, 13, 403. [Google Scholar] [CrossRef]
  92. Ozturk, M.; Nilsen-Hamilton, M.; Ilgu, M. Aptamer applications in neuroscience. Pharmaceuticals 2021, 14, 1260. [Google Scholar] [CrossRef] [PubMed]
  93. Majdinasab, M.; Marty, J.L. Recent advances in electrochemical aptasensors for detection of biomarkers. Pharmaceuticals 2022, 15, 995. [Google Scholar] [CrossRef] [PubMed]
  94. Bruno, J.G. Applications in which aptamers are needed or wanted in diagnostics and therapeutics. Pharmaceuticals 2022, 15, 693. [Google Scholar] [CrossRef] [PubMed]
  95. Drees, A.; Trinh, T.L.; Fischer, M. The influence of protein charge and molecular weight on the affinity of aptamers. Pharmaceuticals 2023, 16, 457. [Google Scholar] [CrossRef]
  96. Tickner, Z.J.; Farzan, M. Riboswitches for controlled expression of therapeutic transgenes delivered by adeno-associated viral vectors. Pharmaceuticals 2021, 14, 554. [Google Scholar] [CrossRef]
  97. Nahvi, A.; Sudarsan, N.; Ebert, M.S.; Zou, X.; Brown, K.L.; Breaker, R.R. Genetic control by a metabolite binding mRNA. Chem. Biol. 2002, 9, 1043. [Google Scholar] [CrossRef] [PubMed]
  98. Winkler, W.; Nahvi, A.; Breaker, R.R. Thiamine derivatives bind messenger RNAs directly to regulate bacterial gene expression. Nature 2002, 419, 952–956. [Google Scholar] [CrossRef]
  99. Winkler, W.C.; Cohen-Chalamish, S.; Breaker, R.R. An mRNA structure that controls gene expression by binding FMN. Proc. Natl. Acad. Sci. USA 2002, 99, 15908–15913. [Google Scholar] [CrossRef]
  100. Lee, C.H.; Han, S.R.; Lee, S.W. Therapeutic applications of aptamer-based riboswitches. Nucleic Acid. Ther. 2016, 26, 44–51. [Google Scholar] [CrossRef]
  101. Findeiss, S.; Etzel, M.; Will, S.; Morl, M.; Stadler, P.F. Design of artificial riboswitches as biosensors. Sensors 2017, 17, 1990. [Google Scholar] [CrossRef]
  102. Harbaugh, S.V.; Martin, J.A.; Weinstein, J.; Ingram, G.; Kelley-Loughnane, N. Screening and selection of artificial riboswitches. Methods 2018, 143, 77–89. [Google Scholar] [CrossRef] [PubMed]
  103. Stifel, J.; Sporing, M.; Hartig, J.S. Expanding the toolbox of synthetic riboswitches with guanine-dependent aptazymes. Synth. Biol. 2019, 4, ysy022. [Google Scholar] [CrossRef]
  104. Ogawa, A.; Itoh, Y. In vitro selection of RNA aptamers binding to nanosized DNA for constructing artificial riboswitches. ACS Synth. Biol. 2020, 9, 2648–2655. [Google Scholar] [CrossRef] [PubMed]
  105. Shanidze, N.; Lenkeit, F.; Hartig, J.S.; Funck, D. A theophylline-responsive riboswitch regulates expression of nuclear-encoded genes. Plant Physiol. 2020, 182, 123–135. [Google Scholar] [CrossRef]
  106. Ogawa, A.; Inoue, H.; Itoh, Y.; Takahashi, H. Facile expansion of the variety of orthogonal ligand/aptamer pairs for artificial riboswitches. ACS Synth. Biol. 2023, 12, 35–42. [Google Scholar] [CrossRef]
  107. Andrianova, M.; Kuznetsov, A. Logic gates based on DNA aptamers. Pharmaceuticals 2020, 13, 417. [Google Scholar] [CrossRef]
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

Berzal-Herranz, A.; Romero-López, C. Aptamers’ Potential to Fill Therapeutic and Diagnostic Gaps. Pharmaceuticals 2024, 17, 105. https://doi.org/10.3390/ph17010105

AMA Style

Berzal-Herranz A, Romero-López C. Aptamers’ Potential to Fill Therapeutic and Diagnostic Gaps. Pharmaceuticals. 2024; 17(1):105. https://doi.org/10.3390/ph17010105

Chicago/Turabian Style

Berzal-Herranz, Alfredo, and Cristina Romero-López. 2024. "Aptamers’ Potential to Fill Therapeutic and Diagnostic Gaps" Pharmaceuticals 17, no. 1: 105. https://doi.org/10.3390/ph17010105

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