Skip to main navigation menu Skip to main content Skip to site footer

Review article: Biomedical intelligence

Vol. 143 No. 0102 (2013)

Aptamer-modified nanoparticles and their use in cancer diagnostics and treatment

  • Christine Reinemann
  • Beate Strehlitz
DOI
https://doi.org/10.4414/smw.2014.13908
Cite this as:
Swiss Med Wkly. 2013;143:w13908
Published
29.12.2013

Summary

Aptamers are single-stranded deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) oligonucleotides, which are able to bind their target with high selectivity and affinity. Owing to their multiple talents, aptamers combined with nanoparticles are nanosystems well qualified for the development of new biomedical devices for analytical, imaging, drug delivery and many other medical applications. Because of their target affinity, aptamers can direct the transport of aptamer-nanoparticle conjugates. The binding of the aptamers to the target “anchors” the nanoparticle-aptamer conjugates at their site of action. In this way, nanoparticle-based bioimaging and smart drug delivery are enabled, especially by use of systematically developed aptamers for cancer-associated biomarkers.

This review article gives a brief overview of recent relevant research into aptamers and trends in their use in cancer diagnostics and therapy. A concise description of aptamers, their development and functionalities relating to nanoparticle modification is given. The main part of the article is dedicated to current developments of aptamer-modified nanoparticles and their use in cancer diagnostics and treatment.

Summary and outlook

The huge potential of aptamer-nanoparticle-based detection of cancer cells and biomedical in-vivo imaging has been shown, with some examples. Gold nanoparticles offer unique properties that make them ideal detection materials. Specifically, the colour change caused by their aggregation or disaggregation and connected to the aptamer-target binding reaction has been used in the development of assays and tests. The colour change can be processed even with the naked eye, without any device necessary [34]. This application is especially user-friendly and, in principle, comparable to the well-known pregnancy test. Point-of-care diagnostics based on this principle soon will easily find the way to commercialisation.

Besides gold nanoparticles, other metal and magnetic nanoparticles, silica- or polymer-based nanoparticles and quantum dots are used. For combined diagnostic and medical use, the silica- or polymer-based nanoparticles have to be loaded with the drug. They can additionally include organic dyes for optical detection, just as functional groups for their conjugation with aptamers [52, 61].

Aptamer modified magnetic nanoparticles offer the possibility of magnetic resonance imaging as well as application of magnetic fluid hypothermia therapy [64].

Quantum dots extend optical detection methods in various directions. They are particular suitable for quantum dot-stimulated FRET-based (beacon) aptamer assays [61].

Aptamer-modified nanoparticles for medical applications are developed mostly for the detection of analytes, for biomedical in-vivo imaging and for in-vivo drug delivering.

Merging nanoparticles with aptamers permits additional or enhanced medical tools for many biomedical applications by combination of the complementary outstanding properties of each. Nanoparticles that are suitable for detection and loading with drugs can be directed to the site of their action by use of the affinity and specificity of aptamers, which are conjugated to the nanoparticles or fixed on their surface. This is of great relevance in cancer therapy in particular. Most anticancer pharmaceuticals have destructive effects not only on the cancer cells, but also on healthy cells. Aptamers can facilitate cell-specific drug delivery in a selective manner to the sick cancer cells because of their binding specificity. This can enhance therapeutic effects and diminish adverse effects. Moreover, simultaneous in-vivodetection and therapy for cancer cells lowers the burden for cancer patients. However, the widespread phenomenon of possible nonspecific accumulation of nanoparticles in the liver, which is not caused by aptamer modification, must be taken into account.

Methods for aptamer-based nanoparticle application have been developed, but mostly by use of a few aptamers. The development of more aptamers specific to cancer cells or marker proteins is urgently needed, especially for these applications. This is the precondition for provision of these new detection, imaging and therapy methods.

References

  1. Andy Ellington’s Blog [Internet]. Austin: Andrew Ellington. 2010 July –. On Aptamers; 2011 March 6 [cited 2012 Dec 20]; [about three screens]. Available from: http://ellingtonlab.org/blog/2011/03/06/on-aptamers/
  2. Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346(6287):818–22.
  3. Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990;249(4968):505–10.
  4. Hermann T, Patel DJ. Adaptive recognition by nucleic acid aptamers. Science. 2000;287(5454):820–5.
  5. Ozalp VC, Eyidogan F, Oktem HA. Aptamer-gated nanoparticles for smart drug delivery. Pharmaceuticals. 2011;4(2011):1137–57.
  6. Jenison RD, Gill SC, Pardi A, Polisky B. High-resolution molecular discrimination by RNA. Science. 1994;263(5152):1425–9.
  7. Stoltenburg R, Reinemann C, Strehlitz B. SELEX-A (r)evolutionary method to generate high-affinity nucleic acid ligands. Biomol Eng. 2007;24(4):381–403.
  8. James W. Aptamers. In: Meyers RA, editor. Encyclopedia of Analytical Chemistry. Chichester: John Wiley & Sons, Ltd.; 2000. P. 4848–71.
  9. Daniels DA, Chen H, Hicke BJ, Swiderek KM, Gold L. A tenascin-C aptamer identified by tumor cell SELEX: Systematic evolution of ligands by exponential enrichment. Proc Natl Acad Sci U S A. 2003;100(26):15416–21.
  10. Sefah K, Shangguan D, Xiong XL, O’Donoghue MB, Tan WH. Development of DNA aptamers using Cell-SELEX. Nature Protoc. 2010;5(6):1169–85.
  11. Tucker CE, Chen LS, Judkins MB, Farmer JA, Gill SC, Drolet DW. Detection and plasma pharmacokinetics of an anti-vascular endothelial growth factor oligonucleotide-aptamer (NX1838) in rhesus monkeys. J Chromatogr B. 1999;732(1):203–12.
  12. Ni X, Castanares M, Mukherjee A, Lupold SE. Nucleic Acid Aptamers: Clinical Applications and Promising New Horizons. Curr Med Chem. 2011;18(27):4206–14.
  13. Carlson B. Aptamers: The New Frontier In Drug Development? Biotechnol Healthc. 2007;(April):31–5.
  14. Vallian S, Khazaei MR. Medical applications of aptamers. Res Pharm Sci. 2007;2(2):59–66.
  15. Lee JF, Stovall GM, Ellington AD. Aptamer therapeutics advance. Curr Opin Chem Biol. 2006;10(3):282–9.
  16. Green LS, Jellinek D, Bell C, Beebe LA, Feistner BD, Gill SC, et al. Nuclease-Resistant Nucleic-Acid Ligands to Vascular-Permeability Factor Vascular Endothelial Growth-Factor. Chem Biol. 1995;2(10):683–95.
  17. Rhodes A, Deakin A, Spaull J, Coomber B, Aitken A, Life P, et al. The generation and characterization of antagonist RNA aptamers to human oncostatin M. J Biol Chem. 2000;275(37):28555–61.
  18. Dougan H, Lyster DM, Vo CV, Stafford A, Weitz JI, Hobbs JB. Extending the lifetime of anticoagulant oligodeoxynucleotide aptamers in blood. Nucl Med Biol. 2000;27(3):289–97.
  19. Klussmann S. The Aptamer Handbook. Functional Oligonucleotides and Their Applications. Weinheim: WILEY-VCH Verlag GmbH & Co. KGaA; 2006.
  20. Petersen M, Wengel J. LNA: a versatile tool for therapeutics and genomics. Trends Biotechnol. 2003;21(2):74–81.
  21. Schmidt KS, Borkowski S, Kurreck J, Stephens AW, Bald R, Hecht M, et al. Application of locked nucleic acids to improve aptamer in vivo stability and targeting function. Nucleic Acids Res. 2004;32(19):5757–65.
  22. Burke DH, Scates L, Andrews K, Gold L. Bent pseudoknots and novel RNA inhibitors of type 1 human immunodeficiency virus (HIV-1) reverse transcriptase. J Mol Biol. 1996;264(4):650–66.
  23. Wilson C, Nix J, Szostak J. Functional requirements for specific ligand recognition by a biotin-binding RNA pseudoknot. Biochemistry. 1998;37(41):14410–9.
  24. Xiao Z, Farokhzad OC. Aptamer-functionalized nanoparticles for medical applications: challenges and opportunities. ACS Nano. 2012;6(5):3670–6.
  25. Phillips JA, Lopez-Colon D, Zhu Z, Xu Y, Tan W. Applications of aptamers in cancer cell biology. Anal Chim Acta. 2008;621(2):101–8.
  26. Fang X, Tan W. Aptamers generated from cell-SELEX for molecular medicine: a chemical biology approach. Acc Chem Res. 2010;43(1):48–57.
  27. Shangguan DH, Cao ZHC, Li Y, Tan WH. Aptamers evolved from cultured cancer cells reveal molecular differences of cancer cells in patient samples. Clin Chem. 2007;53(6):1153–5.
  28. Shangguan D, Li Y, Tang ZW, Cao ZHC, Chen HW, Mallikaratchy P, et al. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc Natl Acad Sci U S A. 2006;103(32):11838–43.
  29. Mallikaratchy P, Tang ZW, Kwame S, Meng L, Shangguan DH, Tan WH. Aptamer directly evolved from live cells recognizes membrane bound immunoglobin heavy mu chain in Burkitt’s lymphoma cells. Mol Cell Proteomics. 2007;6(12):2230–8.
  30. Tang ZW, Shangguan D, Wang KM, Shi H, Sefah K, Mallikratchy P, et al. Selection of aptamers for molecular recognition and characterization of cancer cells. Anal Chem. 2007;79(13):4900–7.
  31. Zhang K, Tan T, Fu JJ, Zheng T, Zhu JJ. A novel aptamer-based competition strategy for ultrasensitive electrochemical detection of leukemia cells. Analyst. 2013;138(21):6323–30.
  32. Sheng W, Chen T, Tan W, Fan ZH. Multivalent DNA Nanospheres for Enhanced Capture of Cancer Cells in Microfluidic Devices. ACS Nano. 2013;7(8):7067–76.
  33. Medley CD, Smith JE, Tang Z, Wu Y, Bamrungsap S, Tan W. Gold nanoparticle-based colorimetric assay for the direct detection of cancerous cells. Anal Chem. 2008;80(4):1067–72.
  34. Liu G, Mao X, Phillips JA, Xu H, Tan W, Zeng L. Aptamer-nanoparticle strip biosensor for sensitive detection of cancer cells. Anal Chem. 2009;81(24):10013–8.
  35. Chen X, Estevez MC, Zhu Z, Huang YF, Chen Y, Wang L, et al. Using aptamer-conjugated fluorescence resonance energy transfer nanoparticles for multiplexed cancer cell monitoring. Anal Chem. 2009;81(16):7009–14.
  36. Herr JK, Smith JE, Medley CD, Shangguan D, Tan W. Aptamer-conjugated nanoparticles for selective collection and detection of cancer cells. Anal Chem. 2006;78(9):2918–24.
  37. Lu W, Arumugam SR, Senapati D, Singh AK, Arbneshi T, Khan SA, et al. Multifunctional oval-shaped gold-nanoparticle-based selective detection of breast cancer cells using simple colorimetric and highly sensitive two-photon scattering assay. ACS Nano. 2010;4(3):1739–49.
  38. Lupold SE, Hicke BJ, Lin Y, Coffey DS. Identification and characterization of nuclease-stabilized RNA molecules that bind human prostate cancer cells via the prostate-specific membrane antigen. Cancer Res. 2002;62(14):4029–33.
  39. Walter JG, Petersen S, Stahl F, Scheper T, Barcikowski S. Laser ablation-based one-step generation and bio-functionalization of gold nanoparticles conjugated with aptamers. J Nanobiotechnology. 2010;8:21.
  40. Ferreira CS, Matthews CS, Missailidis S. DNA aptamers that bind to MUC1 tumour marker: design and characterization of MUC1-binding single-stranded DNA aptamers. Tumor Biol. 2006;27(6):289–301.
  41. Cheng AK, Su H, Wang YA, Yu HZ. Aptamer-based detection of epithelial tumor marker mucin 1 with quantum dot-based fluorescence readout. Anal Chem. 2009;81(15):6130–9.
  42. Ireson CR, Kelland LR. Discovery and development of anticancer aptamers. Mol Cancer Ther. 2006;5(12):2957–62.
  43. Watanabe T, Hirano K, Takahashi A, Yamaguchi K, Beppu M, Fujiki H, et al. Nucleolin on the cell surface as a new molecular target for gastric cancer treatment. Biol Pharm Bull. 2010;33(5):796–803.
  44. Ko MH, Kim S, Kang WJ, Lee JH, Kang H, Moon SH, et al. In vitro derby imaging of cancer biomarkers using quantum dots. Small. 2009;5(10):1207–12.
  45. Hua X, Zhou Z, Yuan L, Liu S. Selective collection and detection of MCF-7 breast cancer cells using aptamer-functionalized magnetic beads and quantum dots based nano-bio-probes. Anal Chim Acta. 2013;788:135–40.
  46. Charlton J, Sennello J, Smith D. In vivo imaging of inflammation using an aptamer inhibitor of human neutrophil elastase. Chem Biol. 1997;4(11):809–16.
  47. Javier DJ, Nitin N, Levy M, Ellington A, Richards-Kortum R. Aptamer-targeted gold nanoparticles as molecular-specific contrast agents for reflectance imaging. Bioconjug Chem. 2008;19(6):1309–12.
  48. Kim D, Jeong YY, Jon S. A drug-loaded aptamer-gold nanoparticle bioconjugate for combined CT imaging and therapy of prostate cancer. ACS Nano. 2010;4(7):3689–96.
  49. Zhou J, Soontornworajit B, Martin J, Sullenger BA, Gilboa E, Wang Y. A hybrid DNA aptamer-dendrimer nanomaterial for targeted cell labeling. Macromol Biosci. 2009;9(9):831–5.
  50. Hwang do W, Ko HY, Lee JH, Kang H, Ryu SH, Song IC, et al. A nucleolin-targeted multimodal nanoparticle imaging probe for tracking cancer cells using an aptamer. J Nucl Med. 2010;51(1):98–105.
  51. Albanese A, Tang PS, Chan WC. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu Rev Biomed Eng. 2012;14:1–16.
  52. Chen T, Shukoor MI, Chen Y, Yuan Q, Zhu Z, Zhao Z, et al. Aptamer-conjugated nanomaterials for bioanalysis and biotechnology applications. Nanoscale. 2011;3(2):546–56.
  53. Farokhzad OC, Jon S, Khademhosseini A, Tran TN, Lavan DA, Langer R. Nanoparticle-aptamer bioconjugates: a new approach for targeting prostate cancer cells. Cancer Res. 2004;64(21):7668–72.
  54. Farokhzad OC, Cheng J, Teply BA, Sherifi I, Jon S, Kantoff PW, et al. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc Natl Acad Sci U S A. 2006;103(16):6315–20.
  55. Cheng J, Teply BA, Sherifi I, Sung J, Luther G, Gu FX, et al. Formulation of functionalized PLGA-PEG nanoparticles for in vivo targeted drug delivery. Biomaterials. 2007;28(5):869–76.
  56. Gu F, Zhang L, Teply BA, Mann N, Wang A, Radovic-Moreno AF, et al. Precise engineering of targeted nanoparticles by using self-assembled biointegrated block copolymers. Proc Natl Acad Sci U S A. 2008;105(7):2586–91.
  57. Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA-PEG nanoparticles. Proc Natl Acad Sci U S A. 2008;105(45):17356–61.
  58. Kolishetti N, Dhar S, Valencia PM, Lin LQ, Karnik R, Lippard SJ, et al. Engineering of self-assembled nanoparticle platform for precisely controlled combination drug therapy. Proc Natl Acad Sci U S A. 2010;107(42):17939–44.
  59. Wang AZ, Bagalkot V, Vasilliou CC, Gu F, Alexis F, Zhang L, et al. Superparamagnetic iron oxide nanoparticle-aptamer bioconjugates for combined prostate cancer imaging and therapy. ChemMedChem. 2008;3(9):1311–5.
  60. Aravind A, Jeyamohan P, Nair R, Veeranarayanan S, Nagaoka Y, Yoshida Y, et al. AS1411 aptamer tagged PLGA-lecithin-PEG nanoparticles for tumor cell targeting and drug delivery. Biotechnol Bioeng. 2012;109(11):2920–31.
  61. Lopez-Colon D, Jimenez E, You M, Gulbakan B, Tan W. Aptamers: turning the spotlight on cells. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2011;3(3):328–40.
  62. Michalet X, Pinaud FF, Bentolila LA, Tsay JM, Doose S, Li JJ, et al. Quantum dots for live cells, in vivo imaging, and diagnostics. Science. 2005;307(5709):538–44.
  63. Savla R, Taratula O, Garbuzenko O, Minko T. Tumor targeted quantum dot-mucin 1 aptamer-doxorubicin conjugate for imaging and treatment of cancer. J Control Release. 2011;153(1):16–22.
  64. McCarthy JR, Weissleder R. Multifunctional magnetic nanoparticles for targeted imaging and therapy. Adv Drug Deliv Rev. 2008;60(11):1241–51.