SlideShare a Scribd company logo
1 of 11
Download to read offline
WHITE PAPER:
                      “Aptamers and their applications at IceNine Bio”
                            (excerpts from various proposals)

                                                 April 2011

     Aptamer development has been limited to one-target at a time
    Aptamers are single-stranded DNA or RNA (ssDNA or ssRNA)
molecules that can bind to pre-selected targets including proteins and
peptides with high affinity and specificity. These molecules can assume a
variety of shapes due to their propensity to form helices and single-
stranded loops, explaining their versatility in binding to dverse targets.
They are used as sensors [1], and therapeutic tools [2], and to regulate
cellular processes [3], as well as to guide drugs to their specific cellular                            Amplify
                                                                                                         eluted
targets [4-7]. Contrary to the actual genetic material, their specificity and                           binders
characteristics are not directly determined by their primary sequence, but
instead by their tertiary structure [8].
    Aptamers are generated from large random libraries by an iterative
process often called Systematic Evolution of Ligands by Exponential
Enrichment (SELEX) [9, 10]. The conventional SELEX technique (Figure
1) starts with a large library of random single stranded nucleotides or
aptamers (ca. 1015 unique sequences). A typical library will contain a Figure 1. Schematic of conventional,
randomized region of ca. 40 nucleotides flanked by two constant regions single-target DNA aptamer selection.
for PCR priming. The library is exposed to a target and the bound
aptamers are partitioned and amplified for the next round. With each round the stringency of the binding
conditions is increased until the only remaining aptamers in the pool are highly specific for, and bind with high
affinity to, the target. Once multiple (typically 10-15) rounds of SELEX are completed, the DNA sequences are
usually identified by conventional cloning and sequencing.
   While in general the accepted process for selecting aptamers, so-called “Systematic Evolution of Ligands by
EXponential enrichment (SELEX)” [9, 10] is quite effective, SELEX is commonly performed against only a
single protein target at a time. Because the process is tedious and time consuming, the yield of just one or,
at best, several aptamer candidates for a single target greatly limits throughput.
   IceNine has addressed this severe limitation by successfully parallel-izing the conventional
aptamer selection process.

    Additional Documented Applications of Aptamers
    So far, aptamers are best known as ligands to proteins, rivaling antibodies in both affinity and specificity
[11-14], and the first aptamer-based therapeutics were recently FDA-approved (Macugen) [15-17]. More
recently, however, aptamers have also been developed to bind small organic molecules and cellular toxins [18-
26], viruses [27, 28], and even targets as small as heavy metal ions [29-33]. While aptamers are analogous to
antibodies in their range of target recognition and variety of applications, they possess several key advantages
over their protein counterparts [34]:
       They are self-refolding, single-chain, and redox-insensitive. They also lack the large hydrophobic
       cores of proteins and thus do not aggregate. They tolerate (or recover from) pH and temperatures
       that proteins do not.
       They are easier and more economical to produce (especially at the affinity reagent scale). In
       stark contrast to peptides, proteins and to some small chemicals, oligonucleotides ( = DNA
       aptamers) are made through chemical synthesis, a process that is well defined, highly reproducible,
       sequence independent and can be readily and predictably scaled up. Their production does not
       depend on bacteria, cell cultures or animals.
       In contrast to antibodies, toxicity and low immunogenicity of particular antigens do not interfere with
       the aptamer selection. Further, using the technology proposed, highly custom or “orphaned”
       targets can be address rapidly and cheaply.

                                                       1
They are capable of greater specificity and affinity than antibodies [35].
       They can easily be modified chemically to yield improved, custom tailored properties. For instance,
       reporter and functional groups and PEG can easily be attached to the aptamer in a deterministic
       way. In fact, they can even be combined with antibodies [36, 37]. Similarly, their ADME properties
       can be readily tuned by conjugation to other groups (PEG, etc).
       Their small size leads to a high number of moles of target bound per gram, and they may have
       improved transport properties allowing cell specific targeting and improved tissue penetration [38-
       42].
       They are much more stable at ambient temperature than antibodies yielding a much higher shelf
       life, and they can tolerate transportation without any special requirements for cooling, eliminating
       the need for a continuous cold chain.

    . The fact that, after selection, aptamers can be produced by chemical synthesis eliminates batch-to-batch
variation which complicates production of therapeutic proteins [43] and the variability of diagnostic antibody
reagents. Aptamers identified by SELEX can also be easily analyzed and manipulated to characterize the
minimum sequence requirements for aptamer:target recognition. Sequences can be inspected for primary and
secondary structure motifs [44-48], and single base perturbations of identified aptamers can be studied for the
resulting affects on binding affinity. This sort of rational/directed optimization would be much more challenging
using the antibody approach.
    In most applications, aptamers have been successfully employed as direct replacements for antibodies.
Aptamer dot-blots [49] and aptamer-westerns [50, 51] are well supported in the literature. With respect to the
integration of aptamers into biosensors, numerous publications support the feasibility of this approach. For
instance, aptamers have been incorporated into lead sensors [30-32], drug sensors [24, 26], and estrogen
measuring devices [52]. They can be used in single-target measuring devices or even in arrays [38], further
extending their versatility. Recently, aptamers have also been used as chimeric conjugates to siRNAs to
improve delivery [53].


   Ongoing aptamer development and applications at BioTex

    The principal scientist at IceNine, Dr. Bill Jackson, has considerable experience in the development and
novel application of nucleic acid aptamers, and these molecules have enjoyed recent increasing acceptance in
both diagnostic and therapeutic settings – (in 2010, Dr. Jackson authored a 200+ page market research report
on the use of aptamers in both diagnostic and therapeutic applications) [54].

    In addition to the aptamer selection services offered by IceNine, our group maintains an active research
program involving novel uses of aptamers. The PI and his colleagues have considerable experience in
developing novel molecular diagnostics using a variety of analytical techniques including mass spectrometry
[55-59] and DNA microarrays [56, 60]. More recently the PI has been involved in a variety of projects involving
aptamers [61-64]. Recent funding has included an EPA SBIR Phase I project for aptamer-based detection of
the cyanobacterial toxin, anatoxin-a, a project to develop aptamer-mimetics to proteins such as stem cell factor
(SCF) to replace peptide agonists with inexpensive aptamers, and the aforementioned project to develop a
platform for massively parallel SELEX. We have also developed recombinant techniques to express small
foreign RNA’s (aptamers and siRNA) within the ribosome of E. coli [62-65]. Currently we are developing
aptamer affinity reagents to high priority cancer biomarker proteins under Contract SBIR Phase I
funding from the National Cancer Institute.




                                                       2
Application of aptamers in flow cytometry
    In addition to validation of aptamers via SPR, we have successfully employed aptamers in
fluorescence activated cell sorting (FACS) with collaborators at nearby M.D. Anderson Cancer Center
(Houston, TX). Specifically, our collaborator, Dr. Laurence Cooper at MDACC has been using our aptamers
to label and differentiate a panel of recombinant proteins expressed on desired T-cell subsets using flow
cytometry. The availability of an inexpensive and efficacious alternative to monoclonal antibodies (mAbs) in
this application is important as a method to produce cellular reagents to be used in compliance with current
good manufacturing practice (cGMP). Obtaining monoclonal antibodies (mAbs) as reagents to validate
the manufacture of cell-based therapeutics for clinical application is tedious, labor intensive and
expensive as applied to generation of clinical-grade reagents. This is chiefly because of the concern of
adventitious virus that may accompany the production of mAbs. Aptamer technology based on in vitro DNA
synthesis will avoid this issue and greatly simplify and reduce the costs associated with producing materials for
use in compliance with cGMP.




                  Figure 2. Demonstration of the feasibility of FACS using fluorescent aptamers selected
                  against cell surface proteins. While some cross-reactivity is observed, screening and better
                  selection of recombinant aptamer targets should alleviate these problems.


     Figure 2 above shows some of the preliminary fluorescence activated cell sorting (FACS) data utilizing our
aptamers as replacements to fluorescently labeled mAb reagents. Briefly, cells were first transfected with a
defined membrane-bound interleukins (IL7, IL15, or IL21). Cells were then washed to remove serum and
culture media and incubated at 37° for 60 minutes with an aptamer developed to either IL-7, IL-15, or IL-21.
                                     C
Aptamers were synthesized with a 3’-biotin for facile labeling in situ with streptavidin-FITC conjugate.
Following binding, aptamer-bound cells were washed once with PBS and then stained with streptavidin-FITC
followed by an additional wash with PBS. Finally, cells were sorted by conventional FACS based on FITC
fluorescence; cell counts to the right of the vertical blue line represent positive binding. As expected from an
initial screen, there is some cross-reactivity of the tested aptamers. This is due to the use of a common
recombinant Fc region for presenting these three cytokines on the cell surface. Nevertheless, some degree of
specificity/orthogonality of the aptamers is seen, and the experiment demonstrates that in principal, expensive
mAbs could be replaced by aptamers for the FACS application.




                                                             3
Development of Aptamers and Sensing Chemistry for the Hormone, Thyroxine (T4) and the Protein, Insulin
     Researchers at BioTex/IceNine have also employed aptamers in competitive sensing chemistries
for detection of environmental and clinical analytes. For instance, a sensor for the thyroid hormone (small
molecule), thyroxine (so-called “free-T4”) was developed.           Figure 3 below depicts the modular,
aptamer/quantum-dot-based sensing scheme employed.
     To select a DNA aptamer to the molecule, thyroxine was covalently immobilized via its primary amine to a
solid phase gel by standard chemistries. After 10 rounds of conventional (e.g. single-target) selection,
individual aptamers were cloned and sequenced. Evolution of a unique sequence, characterized by a high GC
content, as typical for known thyroxine aptamers [66] was clearly observed (sequence not shown).
     Figure 4 shows the performance of the FRET-aptamer sensor which has been described in detail
elsewhere [67]. The sensor was found non-responsive to several structurally similar chemicals and was thus,
specific for the analyte. BioTex has considerable fluorescent biosensor expertise [67-70] especially through
co-investigator, Dr. Ralph Ballerstadt. The fluorescence emitted from the sensor was measured in a portable,
inexpensive Qubit™ fluorometer (Invitrogen). Thus, using a sensing cocktail that can be readily lyophilized
and reconstituted by the sample, such an assay could be taken to the field or bedside for environmental or
clinical use.
     This sort of sensing scheme can be readily devised for proteins as well. We have developed an
analogous sensing chemistry using aptamers specific for insulin evolved at BioTex. Figure 5 (next page)
shows that result. These data demonstrate not only the ability of BioTex researchers to select DNA
aptamers to novel targets, but also the broad, modular applicability of DNA aptamers in numerous
applications.
                A                                QD Nanoshell                B

                                                         Immobilized
                                                         analyte-                                             Fluorescence Emission
                                                         analog


                                                     Aptamer with
                                                     quencher dye
                                                                                                                          analyte
                 EX. hν                                                          EX. hν
                          ~ 10 nm                PEG coating

               Figure 3. Schematic of Modular, Aptamer-based QD-FRET sensing chemistry (approximate
               scale). In a competitive-binding fluorescence resonance energy transfer (FRET) assay, quantum
               dots (QDs) are conjugated to an immobilized version of the intended analyte (T4 in the text
               example). Aptamers which bind T4 with high specificity are identified by in vitro selection or
               "SELEX". Panel (A): Aptamers synthesized with a terminal fluorophore for quenching of the QD
               are bound to immobilized T4. Panel (B) When free T4 in the sample is exposed to this reagent
               mixture, QD-quenching aptamers are released from the QDs resulting in a fluorescence signal
               proportional to the T4 concentration.

                                                                Qubit Reading (5 min values)
                                           200
                                           180
                                           160
                                           140
                                           120
                                    AFUs




                                           100
                                           80
                                           60
                                           40
                                           20
                                            0
                                                 0       0.05    0.1        0.15     0.2         0.25   0.3       0.35
                                                                       Thyroxine Conc. [ug/ul]


                              Figure 4. Quantitative detection of the small molecule thyroxine
                              (T4) via FRET-aptamer detection. Insets show structure of T4 and
                              portable (4.5 x 6.5 x 1.8 inch) Qubit™ fluorometer (Invitrogen) used
                              to acquire data.

                                                                             4
B
                                                                           A




                Figure 5. Response curve of quantum-dot-based sensor (Figure 4) to insulin as formulated with
                the insulin-specific aptamer #43 selected at BioTex. (Panel A) The increase in fluorescence with
                increasing insulin concentrations is due to FRET. The putative secondary structure of insulin-
                aptamer #43 is shown in panel B.



Whole cell SELEX for aptamers competing with ‘Stem Cell Factor’ (SCF)
    In addition to selection of aptamers against purified targets, we also have significant experience in
selection of aptamers against the surface of whole cells. Specifically, under NSF funding, we collaborated
with our literal neighbor, Synthecon Inc. (Houston, TX) in an attempt to develop an inexpensive DNA aptamer
mimetic for so-called stem cell factor (SCF or “kit ligand”). Recombinant SCF is a rather expensive, but
commonly used reagent in stem cell culture. To achieve this goal, we enriched our randomized DNA library for
aptamers binding whole stem cells and then selectively displaced the desired aptamers from the c-Kit receptor
using SCF itself. Although we were unable to evolve an agonist mimetic (to effect stem cell proliferation in
culture), did successfully develop a number of candidate c-Kit specific antagonists. We are currently
quantifying their affinity.




                                                            5
Additional Aptamer Applications:

Aptamer-Western blots of 2D gels and Affinity Depletion of Abundant Proteins
    It has already been demonstrated that aptamers can perform well in a Western-blot application [50, 51].
Similarly they have functioned as direct replacements for antibody reporters in ELISAs and dot blots [49]. A
number of aptamer conjugation/functionalization schemes are likely accepted for this purpose. Most simply,
aptamers can synthesized to contain either a 5’- or 3’-fluorphore of choice. Alternatively, as described in
Preliminary Work, we have considerable experience in conjugating aptamers to quantum dots with the
advantage of higher quantum yield and larger Stokes’ shifts of fluorescence. Finally, for additional signal,
aptamers can be synthesized with a 5’- or 3’-biotin and detected by a streptavidin/horseradish peroxidase
conjugate (strep-HRP). Numerous commercial sources for both fluorescent and chromagenic HRP substrates
are available. Such an approach is often referred to as an “ELONA” [71, 72] or aptamer-ELISA (“ELASA”) [73].

Aptamer-bead precipitation or enrichment
     As mentioned elsewhere, one of the attractions/advantages of aptamers is that they are readily
synthesized with modifications for conjugation, covalent coupling, fluorescent reporting, etc. Specifically,
aptamers can be readily coupled to beads or nanoparticles by synthesizing them with a terminal biotin or
amino-group. Such functionalized particles have been used to concentrate transcription factors [74], or, in the
form of magnetic beads, have been employed to enrich bacterial pathogens [75] or cancer cells [76]. Similarly,
it should be feasible to develop highly multiplexed bead-based assays such as Luminex/Magpix.

Immunohistochemistry
    Employing aptamers in conventional histology and molecular imaging is another logical extension for their
use in lieu of antibodies. In recent years, directly, or indirectly labeled aptamers have, for instance, been used
to detect individual receptors [77] or to differentiate cancerous from non-cancerous cells [78].




   SUMMARY:

   Aptamers represent a promising form of synthetic, inexpensive affinity reagent which can be
employed in numerous applications. Despite their promise, however, they have generally only been
developed or ‘evolved’ against a single target at a time. High costs for aptamer selection (as well as
now-expiring intellectual property constraints) have until now hindered their more widespread use.
IceNine’s novel approach to multiplex aptamer selection can inexpensively provide aptamer affinity
reagents to a revolutionary breadth of targets. Finally, our team has the expertise to demonstrate and
support our customers in a variety of novel aptamer-based application areas.




                                                        6
REFERENCES CITED:
1. Holthoff EL, Bright FV: Molecularly templated materials in chemical sensing. Anal. Chim. Acta 2007,
594:147-161.

2. Kaur G, Roy I: Therapeutic applications of aptamers. Expert opinion on investigational drugs 2008, 17:43-
60.

3. Toulme JJ, Di Primo C, Boucard D: Regulating eukaryotic gene expression with aptamers. FEBS letters
2004, 567:55-62.

4. Chu TC, Marks 3rd JW, Lavery LA, Faulkner S, Rosenblum MG, Ellington AD, Levy M: Aptamer:toxin
conjugates that specifically target prostate tumor cells. Cancer Res. 2006, 66:5989-92.

5. Chu TC, Twu KY, Ellington AD, Levy M: Aptamer mediated siRNA delivery. Nucl. Acids Res. 2006,
34:e73-.

6. Chu TC, Shieh F, Lavery LA, Levy M, Richards-Kortum R, Korgel BA, Ellington AD: Labeling tumor cells
with fluorescent nanocrystal-aptamer bioconjugates. Biosens Bioelectron. 2006, Feb 20.

7. Cao Z, Tong R, Mishra A, Xu W, Wong GCL, Cheng J, Lu Y: Reversible Cell-Specific Drug Delivery with
Aptamer-Functionalized Liposomes. Angew. Chem. Int. Ed. 2009, 48:6494-6498.

8. Breaker RR: Natural and engineered nucleic acids as tools to explore biology. Nature 2004, 432:838-
45.

9. Tuerk C, Gold L: Systematic evolution of ligands by exponential enrichment: RNA ligands to
bacteriophage T4 DNA polymerase. Science 1990, 249:505-10.

10. Ellington AD, Szostak JW: In vitro selection of RNA molecules that bind specific ligands. Nature 1990,
346:818-22.

11. Tuerk C, MacDougal S, Gold L: RNA pseudoknots that inhibit human immunodeficiency virus type 1
reverse transcriptase. Proceedings of the National Academy of Sciences 1992, 89:6988-92.

12. Lee JF, Hesselberth JR, Meyers LA, Ellington AD: Aptamer Database. Nucleic Acids Res 2004, 32:D95-
100.

13. Ruckman J, Green LS, Beeson J, Waugh S, Gillette WL, Henninger DD, Claesson-Welsh L, Janjic N: 2’-
Fluoropyrimidine RNA-based Aptamers to the 165-Amino Acid Form of Vascular Endothelial Growth
Factor (VEGF165), Inhibition Of Receptor Binding And Vegf-Induced Vascular Permeability Through
Interactions Requiring The Exon 7-Encoded Domain. J Biol Chem 1998, 273:20556-7.

14. Jayasena SD: Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin.
Chem. 1999, 45:1628-1650.

15. Lee J-H, Canny MD, De Erkenez A, Krilleke D, Ng Y-S, Shima DT, Pardi A, Jucker F: A therapeutic
aptamer inhibits angiogenesis by specifically targeting the heparin binding domain of VEGF165. Proc.
Natl. Acad. Sci. 2005, 102:18902-18907.

16. Ng EWM, Shima DT, Calias P, Cunningham ET, Guyer DR, Adamis AP: Pegaptanib, a targeted anti-
VEGF aptamer for ocular vascular disease. Nature Reviews Drug Discovery 2006, 5:123-132.

17. Siddiqui MAA, Keating GM: Pegaptanib: in exudative age-related macular degeneration. Drugs 2005,
65:1571-1577.

18. Sazani PL, Larralde R, Szostak JW: A Small Aptamer with Strong and Specific Recognition of the
Triphosphate of ATP. J Am Chem Soc 2004, 126:8371.
                                                     7
19. Tang J, Breaker RR: Mechanism for allosteric inhibition of an ATP-sensitive ribozyme. Nucleic acids
research 1998, 26:4214-21.

20. Yang Q, Goldstein IJ, Mei H-Y, Engelke DR: DNA ligands that bind tightly and selectively to
cellobiose. Proc Natl Acad Sci U S A 1998, 95:5462-5467.

21. Babendure JR, Adams SR, Tsien RY: Aptamers Switch on Fluorescence of Triphenylmethane Dyes. J
Am Chem Soc 2003, 125:14716-7.

22. Hirao I, Yoshinari S, Yokoyama S, Endo Y, Ellington AD: In vitro selection of aptamers that bind to
ribosome-inactivating toxins. Nucleic Acids Symp Ser 1997, 37:283-4.

23. Hirao I, Harada Y, Nojima T, Osawa Y, Masaki H, Yokoyama S: In Vitro Selection of RNA Aptamers that
Bind to Colicin E3 and Structurally Resemble the Decoding Site of 16S Ribosomal RNA†. Biochemistry
2004, 43:3214-3221.

24. Liu J, Lu Y: Fast Colorimetric Sensing of Adenosine and Cocaine Based on a General Sensor Design
Involving Aptamers and Nanoparticles. Angew. Chem., Int. Ed. 2006, 117:90-94.

25. Stojanovic MN, de Prada P, Landry DW: Aptamer-Based Folding Fluorescent Sensor for Cocaine. J
Am Chem Soc 2001, 123:4928-4931.

26. Stojanovic MN, Landry DW: Aptamer-Based Colorimetric Probe for Cocaine. J Am Chem Soc 2002,
124:9678-9679.

27. Gopinath SCB, Misono TS, Kawasaki K, Mizuno T, Imai M, Odagiri T, Kumar PKR: An RNA aptamer that
distinguishes between closely related human influenza viruses and inhibits haemagglutinin-mediated
membrane fusion. J Gen Virol 2006, 87:479-87.

28. Gopinath SCB, Sakamaki Y, Kawasaki K, Kumar PKR: An Efficient RNA Aptamer against Human
Influenza B Virus Hemagglutinin. Journal of Biochemistry 2006 139(5):837-846 2006, 139:837-46.

29. Liu J, Lu Y: A Colorimetric Lead Biosensor Using DNAzyme-Directed Assembly of Gold
Nanoparticles. Journal of the American Chemical Society 2003, 125:6642-6643.

30. Chang I-H, Tulock J, Liu J, Kim W-S, Cannon Jr. D, Lu Y, Bohn P, Sweedler J, Cropek D: Miniaturized
Lead Sensor Based on Lead-Specific DNAzyme in a Nanocapillary Interconnected Microfluidic Device.
Environ. Sci. Technol. 2005, 39:3756.

31. Swearingen CB, Wernette DP, Cropek DM, Lu Y, Sweedler JV, Bohn PW: Immobilization of a Catalytic
DNA Molecular Beacon on Au for Pb(II) Detection. Anal. Chem. 2005, 77:442-8.

32. Wernette DP, Kim H-K, Liu J, Swearingen CB, Yue Z, Zavareh M, Ingram CW, Economy J, Shannon MA,
Bohn PW, Lua Y: New Catalytic DNA Biosensors for Trace Contaminants in Water.

33. Wrzesinski J, Ciesiolka J: Characterization of structure and metal ions specificity of Co2+-binding
RNA aptamers. Biochemistry 2005, 44:6257-68.

34. Stoltenburg R, Reinemann C, Strehlitz B: SELEX--a (r)evolutionary method to generate high-affinity
nucleic acid ligands. Biomolecular engineering 2007, 24:381-403.

35. Jayasena SD: Aptamers: An Emerging Class of Molecules That Rival Antibodies in Diagnostics.
Clinical Chemistry 1999, 45:1628–1650.

36. Ferreira CS, Papamichael K, Guilbault G, Schwarzacher T, Gariepy J, Missailidis S: DNA aptamers
against the MUC1 tumour marker: design of aptamer-antibody sandwich ELISA for the early diagnosis
of epithelial tumours. Analytical and bioanalytical chemistry 2008, 390:1039-50.

                                                  8
37. Burbulis I, Yamaguchi K, Yu R, Resnekov O, Brent R: Quantifying small numbers of antibodies with a
“near-universal” protein-DNA chimera. Nature Methods 2007, 4:1011-3.

38. McCauley TG, Hamaguchi N, Stanton M: Aptamer-based biosensor arrays for detection and
quantification of biological macromolecules. Analytical biochemistry 2003, 319:244-50.

39. Cao Z, Tong R, Mishra A, Xu W, Wong GCL, Cheng J, Lu Y: Reversible Cell-Specific Drug Delivery with
Aptamer-Functionalized Liposomes. Angew. Chem. Int. Ed. 2009, 48:6494-6498.

40. De Rosa G, La Rotonda MI: Nano and Microtechnologies for the Delivery of Oligonucleotides with
Gene Silencing Properties. Molecules 2009, 14:2801-2823.

41. Ferreira CSM, Cheung MC, Missailidis S, Bisland S, Gariepy J: Phototoxic aptamers selectively enter
and kill epithelial cancer cells. Nucl. Acids Res. 2009, 37:866-876.

42. Yan AC, Levy M: Aptamers and aptamer targeted delivery. rnabiology 2009, 6:316-320.

43. Cload S, McCauley T, Keefe A, Healy J, Wilson C: Properties of Therapeutic Aptamers. In Aptamer
Handbook, The. Wiley-VCH Verlag GmbH & Co. KGaA; 2006.

44. Mathews DH, Sabina J, Zuker M, Turner DH: Expanded Sequence Dependence of Thermodynamic
Parameters Improves Prediction of RNA Secondary Structure. J Mol Biol 1999, 288:911-40.

45. Markham NR, Zuker M: UNAFold: software for nucleic acid folding and hybridization. Methods Mol.
Biol 2008, 453:3-31.

46. Zuker M: Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res
2003, 31:3406-15.

47. Zuker M and S: Optimal computer folding of large RNA sequences using thermodymanics and
auxiliary information. Nucleic Acids Res. 1981, 9:133-148.

48. Hofacker IL: Vienna RNA secondary structure server. Nucleic Acids Res 2003, 31:3429-3431.

49. Zhu J, Li T, Hu J, Wang E: A novel dot-blot DNAzyme-linked aptamer assay for protein detection.
Anal Bioanal Chem 2010, 397:2923-2927.

50. Tombelli S, Mascini M: Aptamers as molecular tools for bioanalytical methods. Curr. Opin. Mol. Ther
2009, 11:179-188.

51. Shin S, Kim I-H, Kang W, Yang JK, Hah SS: An alternative to Western blot analysis using RNA
aptamer-functionalized quantum dots. Bioorg. Med. Chem. Lett 2010, 20:3322-3325.

52. Kim YS, Jung HS, Matsuura T, Lee HY, Kawai T, Gu MB: Electrochemical detection of 17beta-estradiol
using DNA aptamer immobilized gold electrode chip. Biosensors & bioelectronics 2007, 22:2525-31.

53. Neff CP, Zhou J, Remling L, Kuruvilla J, Zhang J, Li H, Smith DD, Swiderski P, Rossi JJ, Akkina R: An
Aptamer-siRNA Chimera Suppresses HIV-1 Viral Loads and Protects from Helper CD4+ T Cell Decline
in Humanized Mice. Sci Transl Med 2011, 3:66ra6.

54. Jackson GW, Strych U: Report #BIO071A - Nucleic Acid Aptamers for Diagnostics and Therapeutics:
Global Markets. BCC Research; 2010.

55. Jackson GW: Microbial Identification by MALDI-TOF Mass Spectrometry of Ribosomal RNA. 2006.




                                                   9
56. Jackson GW, Karouia F, Putonti C, Fofanov Y, Fox GE, Willson RC: Microarray Designs and MALDI-
TOF Mass Spectrometry for Rapid Microbial Identification. In 229th National Meeting of the American
Chemical Society. San Diego, CA: 2005.

57. Jackson GW, McNichols RJ, Fox GE, Willson RC: Bacterial genotyping by 16S rRNA mass cataloging.
BMC bioinformatics 2006, 7:321.

58. Jackson GW, McNichols RJ, Fox GE, Willson RC: Universal Bacterial Identification by Mass
Spectrometry of 16S Ribosomal RNA Cleavage Products. Intl J Mass Spectrometry 2007, 261:218-26.

59. Jackson GW, McNichols RJ, Fox GE, Willson RC: Toward universal flavivirus identification by mass
cataloging. J Mol Diagn 2008, 10:135-41.

60. Jackson GW, Fox GE, Willson RC: Novel Microarray Designs With Real-Time Hybridization
Monitoring For Microbial Identification. In 22nd Annual Meeting of the Houston Society for Engineering in
Medicine and Biology. Houston, TX: 2005.

61. Potty AS, Kourentzi K, Fang H, Jackson GW, Zhang X, Legge GB, Willson RC: Biophysical
characterization of DNA aptamer interactions with vascular endothelial growth factor. Biopolymers
2009, 91:145-56.

62. Zhang X, Potty AS, Jackson GW, Stepanov V, Tang A, Liu Y, Kourentzi K, Strych U, Fox GE, Willson RC:
Engineered 5S ribosomal RNAs displaying aptamers recognizing vascular endothelial growth factor
and malachite green. J Mol Recognit 2009, 22:154-161.

63. Zhang X, Potty ASR, Jackson GW, Fox GE, Willson RC: Molecular Recognition of Foreign Sequences
in Engineered 5S Ribosomal RNA. In AFFINITY 2007, The 17th Biennial Meeting of the International Society
for Molecular Recognition. New York University, New York, NY: 2007.

64. Zhang X, Potty ASR, Jackson GW, Fox GE, Willson RC: Engineered 5S Ribosomal RNAs displaying
Anti-VEGF and Malachite Green Aptamers. 12th Annual Structural Biology Symposium, University of Texas
Medical Branch at Galveston, May 18-19, 2007. 2007.

65. Liu Y, Stepanov V, Strych U, Willson R, Jackson G, Fox G: DNAzyme-mediated recovery of small
recombinant RNAs from a 5S rRNA-derived chimera expressed in Escherichia coli. BMC Biotechnology
2010, 10:85.

66. Lavesque D, Beaudoin J-D, Roy S, Perreault J-P: In vitro selection and characterization of RNA
aptamers binding thyroxine hormone. Biochem. J. 2007, 403:129-138.

67. Jackson GW, Strych U, Frank E, Willson RC, Ballerstadt R, McNichols RJ: Portable FRET Sensing of
Proteins, Hormones, and Toxins Using DNA Aptamers and Quantum Dots. In Technical Proceedings of
the 2009 Nanotechnology Conference and Trade Show, Nanotech 2009. Houston, TX: 2009.

68. Ballerstadt R, Gowda A, McNichols RJ: In Vivo Performance Evaluation of a Transdermal Near-
Infrared FRET Affinity Sensor for Continuous Glucose Monitoring. Diabetes Technology & Therapeutics
2006, 8:296-311.

69. Ballerstadt R, Polak A, Beuhler A, Frye J: In-vitro long-term performance study of a near-infrared
fluorescence affinity sensor for glucose monitoring. Biosensors & Bioelectronics 2004, 19:905-14.

70. Ballerstadt R, Gowda A, McNichols RJ: Fluorescence Resonance Energy Transfer-Based Near-
Infrared Fluorescence Sensor for Glucose Monitoring. Diabetes Technology & Therapeutics 2004, 6:191-
200.

71. Ramos E, Piñeiro D, Soto M, Abanades DR, Martín ME, Salinas M, González VM: A DNA aptamer
population specifically detects Leishmania infantum H2A antigen. Lab. Invest 2007, 87:409-416.

                                                   10
72. Yan XR, Gao XW, Yao LH, Zhang ZQ: [Novel methods to detect cytokines by enzyme-linked
oligonucleotide assay]. Sheng wu gong cheng xue bao = Chinese journal of biotechnology 2004, 20:679-82.

73. Bruno JG, Carrillo MP, Cadieux CL, Lenz DE, Cerasoli DM, Phillips T: DNA aptamers developed against
a soman derivative cross-react with the methylphosphonic acid core but not with flanking hydrophobic
groups. J. Mol. Recognit 2009, 22:197-204.

74. Oguro A, Ohtsu T, Nakamura Y: An aptamer-based biosensor for mammalian initiation factor
eukaryotic initiation factor 4A. Anal. Biochem 2009, 388:102-107.

75. Joshi R, Janagama H, Dwivedi HP, Senthil Kumar TMA, Jaykus L-A, Schefers J, Sreevatsan S: Selection,
characterization, and application of DNA aptamers for the capture and detection of Salmonella enterica
serovars. Mol. Cell. Probes 2009, 23:20-28.

76. Smith JE, Medley CD, Tang Z, Shangguan D, Lofton C, Tan W: Aptamer-conjugated nanoparticles for
the collection and detection of multiple cancer cells. Anal. Chem 2007, 79:3075-3082.

77. Chen Y, Munteanu AC, Huang Y-F, Phillips J, Zhu Z, Mavros M, Tan W: Mapping receptor density on
live cells by using fluorescence correlation spectroscopy. Chemistry 2009, 15:5327-5336.

78. Chen HW, Medley CD, Sefah K, Shangguan D, Tang Z, Meng L, Smith JE, Tan W: Molecular recognition
of small-cell lung cancer cells using aptamers. ChemMedChem 2008, 3:991-1001.




                                                  11

More Related Content

What's hot

Aptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic Use
Aptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic UseAptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic Use
Aptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic Useajithnandanam
 
Antibody and Aptamer, Choose Which One?
Antibody and Aptamer, Choose Which One?Antibody and Aptamer, Choose Which One?
Antibody and Aptamer, Choose Which One?creative biogene
 
Aptamers as targeted therapeutics
Aptamers as targeted therapeuticsAptamers as targeted therapeutics
Aptamers as targeted therapeuticsDibya Sundar
 
Bobs Aptamer Assay Slide Presentation_R
Bobs Aptamer Assay Slide Presentation_RBobs Aptamer Assay Slide Presentation_R
Bobs Aptamer Assay Slide Presentation_RRobert Bruce
 
Aptamer Mediated siRNA Delivery
Aptamer Mediated siRNA DeliveryAptamer Mediated siRNA Delivery
Aptamer Mediated siRNA DeliverySaw Yi
 
Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...
Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...
Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...ponenciasexpoquim11
 
Biopharmaceutics of antisense molecule and aptamers
Biopharmaceutics of antisense molecule and aptamersBiopharmaceutics of antisense molecule and aptamers
Biopharmaceutics of antisense molecule and aptamersSUJITHA MARY
 
BIOPHARMACEUTICS OF ANTISENSE MOLECULE AND APTAMERS
BIOPHARMACEUTICS  OF  ANTISENSE MOLECULE AND  APTAMERSBIOPHARMACEUTICS  OF  ANTISENSE MOLECULE AND  APTAMERS
BIOPHARMACEUTICS OF ANTISENSE MOLECULE AND APTAMERSMUSTAFIZUR RAHMAN
 
2013-09-03 Radboudumc NCMLS Technical Forum
2013-09-03 Radboudumc NCMLS Technical Forum2013-09-03 Radboudumc NCMLS Technical Forum
2013-09-03 Radboudumc NCMLS Technical ForumAlain van Gool
 
Proteomics course 1
Proteomics course 1Proteomics course 1
Proteomics course 1utpaltatu
 
Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015
Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015
Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015Alain van Gool
 
Aptamers and antisense oligonucleotides
Aptamers and antisense oligonucleotidesAptamers and antisense oligonucleotides
Aptamers and antisense oligonucleotidesSagugowda
 
Peptide Mass Fingerprinting (PMF) and Isotope Coded Affinity Tags (ICAT)
Peptide Mass Fingerprinting  (PMF) and Isotope Coded Affinity Tags (ICAT)Peptide Mass Fingerprinting  (PMF) and Isotope Coded Affinity Tags (ICAT)
Peptide Mass Fingerprinting (PMF) and Isotope Coded Affinity Tags (ICAT)Suresh Antre
 
Functional proteomics, and tools
Functional proteomics, and toolsFunctional proteomics, and tools
Functional proteomics, and toolsKAUSHAL SAHU
 

What's hot (20)

Aptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic Use
Aptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic UseAptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic Use
Aptamers - New Class of Oligonucleotide for Therapeutic and Diagnostic Use
 
Aptamer presentation version 4
Aptamer presentation version 4Aptamer presentation version 4
Aptamer presentation version 4
 
Antibody and Aptamer, Choose Which One?
Antibody and Aptamer, Choose Which One?Antibody and Aptamer, Choose Which One?
Antibody and Aptamer, Choose Which One?
 
Aptamers as targeted therapeutics
Aptamers as targeted therapeuticsAptamers as targeted therapeutics
Aptamers as targeted therapeutics
 
Bobs Aptamer Assay Slide Presentation_R
Bobs Aptamer Assay Slide Presentation_RBobs Aptamer Assay Slide Presentation_R
Bobs Aptamer Assay Slide Presentation_R
 
Aptamers as Drug of future
Aptamers as Drug of futureAptamers as Drug of future
Aptamers as Drug of future
 
Aptamer Mediated siRNA Delivery
Aptamer Mediated siRNA DeliveryAptamer Mediated siRNA Delivery
Aptamer Mediated siRNA Delivery
 
Session 5.3: Levy
Session 5.3: LevySession 5.3: Levy
Session 5.3: Levy
 
Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...
Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...
Víctor M. González - Introducción a la tecnología de aptámeros y posibles apl...
 
Biopharmaceutics of antisense molecule and aptamers
Biopharmaceutics of antisense molecule and aptamersBiopharmaceutics of antisense molecule and aptamers
Biopharmaceutics of antisense molecule and aptamers
 
BIOPHARMACEUTICS OF ANTISENSE MOLECULE AND APTAMERS
BIOPHARMACEUTICS  OF  ANTISENSE MOLECULE AND  APTAMERSBIOPHARMACEUTICS  OF  ANTISENSE MOLECULE AND  APTAMERS
BIOPHARMACEUTICS OF ANTISENSE MOLECULE AND APTAMERS
 
2013-09-03 Radboudumc NCMLS Technical Forum
2013-09-03 Radboudumc NCMLS Technical Forum2013-09-03 Radboudumc NCMLS Technical Forum
2013-09-03 Radboudumc NCMLS Technical Forum
 
Proteomics course 1
Proteomics course 1Proteomics course 1
Proteomics course 1
 
Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015
Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015
Overview Radboudumc Center for Proteomics, Glycomics and Metabolomics april 2015
 
Salisha ppt (1) (1)
Salisha ppt (1) (1)Salisha ppt (1) (1)
Salisha ppt (1) (1)
 
Proteomics
ProteomicsProteomics
Proteomics
 
Aptamers and antisense oligonucleotides
Aptamers and antisense oligonucleotidesAptamers and antisense oligonucleotides
Aptamers and antisense oligonucleotides
 
Proteomics
ProteomicsProteomics
Proteomics
 
Peptide Mass Fingerprinting (PMF) and Isotope Coded Affinity Tags (ICAT)
Peptide Mass Fingerprinting  (PMF) and Isotope Coded Affinity Tags (ICAT)Peptide Mass Fingerprinting  (PMF) and Isotope Coded Affinity Tags (ICAT)
Peptide Mass Fingerprinting (PMF) and Isotope Coded Affinity Tags (ICAT)
 
Functional proteomics, and tools
Functional proteomics, and toolsFunctional proteomics, and tools
Functional proteomics, and tools
 

Similar to White Paper Aptamer Applications

Target identification and validation in drug discovery
Target identification and validation in drug discoveryTarget identification and validation in drug discovery
Target identification and validation in drug discoverySpringer
 
5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral Flow5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral FlowLisa Thurston
 
Shah Presentation1[1811].pptx
Shah Presentation1[1811].pptxShah Presentation1[1811].pptx
Shah Presentation1[1811].pptxShahnawaz Rayeen
 
protein microarray.pptx
protein microarray.pptxprotein microarray.pptx
protein microarray.pptxAtulSingh77625
 
protein microarray-types and approaches.pptx
protein microarray-types and approaches.pptxprotein microarray-types and approaches.pptx
protein microarray-types and approaches.pptxSachin Teotia
 
Viral Protein Structure Predictions - Consensus Strategy
Viral Protein Structure Predictions - Consensus StrategyViral Protein Structure Predictions - Consensus Strategy
Viral Protein Structure Predictions - Consensus StrategyKeiji Takamoto
 
Aptamer-Drug Conjugate (ApDC) Current Research Progress.pdf
Aptamer-Drug Conjugate (ApDC) Current Research Progress.pdfAptamer-Drug Conjugate (ApDC) Current Research Progress.pdf
Aptamer-Drug Conjugate (ApDC) Current Research Progress.pdfDoriaFang
 
Caracterizacion de anticuerpos
Caracterizacion de anticuerposCaracterizacion de anticuerpos
Caracterizacion de anticuerposCeciliaParrado
 
New Progress of Targeted Degradation Based On Nucleic Acid.pdf
New Progress of Targeted Degradation Based On Nucleic Acid.pdfNew Progress of Targeted Degradation Based On Nucleic Acid.pdf
New Progress of Targeted Degradation Based On Nucleic Acid.pdfDoriaFang
 
Genotyping methods of nosocomial infections pathogen
Genotyping methods of nosocomial infections pathogenGenotyping methods of nosocomial infections pathogen
Genotyping methods of nosocomial infections pathogenimprovemed
 
Proteomics 2009 V9p1696
Proteomics 2009 V9p1696Proteomics 2009 V9p1696
Proteomics 2009 V9p1696jcruzsilva
 
JBEI Research Highlights - August 2017
JBEI Research Highlights - August 2017 JBEI Research Highlights - August 2017
JBEI Research Highlights - August 2017 Irina Silva
 
Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...
Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...
Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...SynEnthu
 

Similar to White Paper Aptamer Applications (20)

Target identification and validation in drug discovery
Target identification and validation in drug discoveryTarget identification and validation in drug discovery
Target identification and validation in drug discovery
 
Aptamers based drug delivery
Aptamers based drug deliveryAptamers based drug delivery
Aptamers based drug delivery
 
5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral Flow5 Reasons Aptamers for Lateral Flow
5 Reasons Aptamers for Lateral Flow
 
Shah Presentation1[1811].pptx
Shah Presentation1[1811].pptxShah Presentation1[1811].pptx
Shah Presentation1[1811].pptx
 
protein microarray.pptx
protein microarray.pptxprotein microarray.pptx
protein microarray.pptx
 
protein microarray-types and approaches.pptx
protein microarray-types and approaches.pptxprotein microarray-types and approaches.pptx
protein microarray-types and approaches.pptx
 
Viral Protein Structure Predictions - Consensus Strategy
Viral Protein Structure Predictions - Consensus StrategyViral Protein Structure Predictions - Consensus Strategy
Viral Protein Structure Predictions - Consensus Strategy
 
Aptamer-Drug Conjugate (ApDC) Current Research Progress.pdf
Aptamer-Drug Conjugate (ApDC) Current Research Progress.pdfAptamer-Drug Conjugate (ApDC) Current Research Progress.pdf
Aptamer-Drug Conjugate (ApDC) Current Research Progress.pdf
 
Caracterizacion de anticuerpos
Caracterizacion de anticuerposCaracterizacion de anticuerpos
Caracterizacion de anticuerpos
 
3targeting
3targeting3targeting
3targeting
 
Proteomics
ProteomicsProteomics
Proteomics
 
targeting
targetingtargeting
targeting
 
New Progress of Targeted Degradation Based On Nucleic Acid.pdf
New Progress of Targeted Degradation Based On Nucleic Acid.pdfNew Progress of Targeted Degradation Based On Nucleic Acid.pdf
New Progress of Targeted Degradation Based On Nucleic Acid.pdf
 
Vector Engineering.pptx
Vector Engineering.pptxVector Engineering.pptx
Vector Engineering.pptx
 
Genotyping methods of nosocomial infections pathogen
Genotyping methods of nosocomial infections pathogenGenotyping methods of nosocomial infections pathogen
Genotyping methods of nosocomial infections pathogen
 
3targeting
3targeting3targeting
3targeting
 
Proteomics 2009 V9p1696
Proteomics 2009 V9p1696Proteomics 2009 V9p1696
Proteomics 2009 V9p1696
 
PROTEIN MICROARRAYS
PROTEIN MICROARRAYSPROTEIN MICROARRAYS
PROTEIN MICROARRAYS
 
JBEI Research Highlights - August 2017
JBEI Research Highlights - August 2017 JBEI Research Highlights - August 2017
JBEI Research Highlights - August 2017
 
Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...
Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...
Conrado et al. 2011 NAR DNA-guided assembly of biosynthetic pathways promotes...
 

White Paper Aptamer Applications

  • 1. WHITE PAPER: “Aptamers and their applications at IceNine Bio” (excerpts from various proposals) April 2011 Aptamer development has been limited to one-target at a time Aptamers are single-stranded DNA or RNA (ssDNA or ssRNA) molecules that can bind to pre-selected targets including proteins and peptides with high affinity and specificity. These molecules can assume a variety of shapes due to their propensity to form helices and single- stranded loops, explaining their versatility in binding to dverse targets. They are used as sensors [1], and therapeutic tools [2], and to regulate cellular processes [3], as well as to guide drugs to their specific cellular Amplify eluted targets [4-7]. Contrary to the actual genetic material, their specificity and binders characteristics are not directly determined by their primary sequence, but instead by their tertiary structure [8]. Aptamers are generated from large random libraries by an iterative process often called Systematic Evolution of Ligands by Exponential Enrichment (SELEX) [9, 10]. The conventional SELEX technique (Figure 1) starts with a large library of random single stranded nucleotides or aptamers (ca. 1015 unique sequences). A typical library will contain a Figure 1. Schematic of conventional, randomized region of ca. 40 nucleotides flanked by two constant regions single-target DNA aptamer selection. for PCR priming. The library is exposed to a target and the bound aptamers are partitioned and amplified for the next round. With each round the stringency of the binding conditions is increased until the only remaining aptamers in the pool are highly specific for, and bind with high affinity to, the target. Once multiple (typically 10-15) rounds of SELEX are completed, the DNA sequences are usually identified by conventional cloning and sequencing. While in general the accepted process for selecting aptamers, so-called “Systematic Evolution of Ligands by EXponential enrichment (SELEX)” [9, 10] is quite effective, SELEX is commonly performed against only a single protein target at a time. Because the process is tedious and time consuming, the yield of just one or, at best, several aptamer candidates for a single target greatly limits throughput. IceNine has addressed this severe limitation by successfully parallel-izing the conventional aptamer selection process. Additional Documented Applications of Aptamers So far, aptamers are best known as ligands to proteins, rivaling antibodies in both affinity and specificity [11-14], and the first aptamer-based therapeutics were recently FDA-approved (Macugen) [15-17]. More recently, however, aptamers have also been developed to bind small organic molecules and cellular toxins [18- 26], viruses [27, 28], and even targets as small as heavy metal ions [29-33]. While aptamers are analogous to antibodies in their range of target recognition and variety of applications, they possess several key advantages over their protein counterparts [34]: They are self-refolding, single-chain, and redox-insensitive. They also lack the large hydrophobic cores of proteins and thus do not aggregate. They tolerate (or recover from) pH and temperatures that proteins do not. They are easier and more economical to produce (especially at the affinity reagent scale). In stark contrast to peptides, proteins and to some small chemicals, oligonucleotides ( = DNA aptamers) are made through chemical synthesis, a process that is well defined, highly reproducible, sequence independent and can be readily and predictably scaled up. Their production does not depend on bacteria, cell cultures or animals. In contrast to antibodies, toxicity and low immunogenicity of particular antigens do not interfere with the aptamer selection. Further, using the technology proposed, highly custom or “orphaned” targets can be address rapidly and cheaply. 1
  • 2. They are capable of greater specificity and affinity than antibodies [35]. They can easily be modified chemically to yield improved, custom tailored properties. For instance, reporter and functional groups and PEG can easily be attached to the aptamer in a deterministic way. In fact, they can even be combined with antibodies [36, 37]. Similarly, their ADME properties can be readily tuned by conjugation to other groups (PEG, etc). Their small size leads to a high number of moles of target bound per gram, and they may have improved transport properties allowing cell specific targeting and improved tissue penetration [38- 42]. They are much more stable at ambient temperature than antibodies yielding a much higher shelf life, and they can tolerate transportation without any special requirements for cooling, eliminating the need for a continuous cold chain. . The fact that, after selection, aptamers can be produced by chemical synthesis eliminates batch-to-batch variation which complicates production of therapeutic proteins [43] and the variability of diagnostic antibody reagents. Aptamers identified by SELEX can also be easily analyzed and manipulated to characterize the minimum sequence requirements for aptamer:target recognition. Sequences can be inspected for primary and secondary structure motifs [44-48], and single base perturbations of identified aptamers can be studied for the resulting affects on binding affinity. This sort of rational/directed optimization would be much more challenging using the antibody approach. In most applications, aptamers have been successfully employed as direct replacements for antibodies. Aptamer dot-blots [49] and aptamer-westerns [50, 51] are well supported in the literature. With respect to the integration of aptamers into biosensors, numerous publications support the feasibility of this approach. For instance, aptamers have been incorporated into lead sensors [30-32], drug sensors [24, 26], and estrogen measuring devices [52]. They can be used in single-target measuring devices or even in arrays [38], further extending their versatility. Recently, aptamers have also been used as chimeric conjugates to siRNAs to improve delivery [53]. Ongoing aptamer development and applications at BioTex The principal scientist at IceNine, Dr. Bill Jackson, has considerable experience in the development and novel application of nucleic acid aptamers, and these molecules have enjoyed recent increasing acceptance in both diagnostic and therapeutic settings – (in 2010, Dr. Jackson authored a 200+ page market research report on the use of aptamers in both diagnostic and therapeutic applications) [54]. In addition to the aptamer selection services offered by IceNine, our group maintains an active research program involving novel uses of aptamers. The PI and his colleagues have considerable experience in developing novel molecular diagnostics using a variety of analytical techniques including mass spectrometry [55-59] and DNA microarrays [56, 60]. More recently the PI has been involved in a variety of projects involving aptamers [61-64]. Recent funding has included an EPA SBIR Phase I project for aptamer-based detection of the cyanobacterial toxin, anatoxin-a, a project to develop aptamer-mimetics to proteins such as stem cell factor (SCF) to replace peptide agonists with inexpensive aptamers, and the aforementioned project to develop a platform for massively parallel SELEX. We have also developed recombinant techniques to express small foreign RNA’s (aptamers and siRNA) within the ribosome of E. coli [62-65]. Currently we are developing aptamer affinity reagents to high priority cancer biomarker proteins under Contract SBIR Phase I funding from the National Cancer Institute. 2
  • 3. Application of aptamers in flow cytometry In addition to validation of aptamers via SPR, we have successfully employed aptamers in fluorescence activated cell sorting (FACS) with collaborators at nearby M.D. Anderson Cancer Center (Houston, TX). Specifically, our collaborator, Dr. Laurence Cooper at MDACC has been using our aptamers to label and differentiate a panel of recombinant proteins expressed on desired T-cell subsets using flow cytometry. The availability of an inexpensive and efficacious alternative to monoclonal antibodies (mAbs) in this application is important as a method to produce cellular reagents to be used in compliance with current good manufacturing practice (cGMP). Obtaining monoclonal antibodies (mAbs) as reagents to validate the manufacture of cell-based therapeutics for clinical application is tedious, labor intensive and expensive as applied to generation of clinical-grade reagents. This is chiefly because of the concern of adventitious virus that may accompany the production of mAbs. Aptamer technology based on in vitro DNA synthesis will avoid this issue and greatly simplify and reduce the costs associated with producing materials for use in compliance with cGMP. Figure 2. Demonstration of the feasibility of FACS using fluorescent aptamers selected against cell surface proteins. While some cross-reactivity is observed, screening and better selection of recombinant aptamer targets should alleviate these problems. Figure 2 above shows some of the preliminary fluorescence activated cell sorting (FACS) data utilizing our aptamers as replacements to fluorescently labeled mAb reagents. Briefly, cells were first transfected with a defined membrane-bound interleukins (IL7, IL15, or IL21). Cells were then washed to remove serum and culture media and incubated at 37° for 60 minutes with an aptamer developed to either IL-7, IL-15, or IL-21. C Aptamers were synthesized with a 3’-biotin for facile labeling in situ with streptavidin-FITC conjugate. Following binding, aptamer-bound cells were washed once with PBS and then stained with streptavidin-FITC followed by an additional wash with PBS. Finally, cells were sorted by conventional FACS based on FITC fluorescence; cell counts to the right of the vertical blue line represent positive binding. As expected from an initial screen, there is some cross-reactivity of the tested aptamers. This is due to the use of a common recombinant Fc region for presenting these three cytokines on the cell surface. Nevertheless, some degree of specificity/orthogonality of the aptamers is seen, and the experiment demonstrates that in principal, expensive mAbs could be replaced by aptamers for the FACS application. 3
  • 4. Development of Aptamers and Sensing Chemistry for the Hormone, Thyroxine (T4) and the Protein, Insulin Researchers at BioTex/IceNine have also employed aptamers in competitive sensing chemistries for detection of environmental and clinical analytes. For instance, a sensor for the thyroid hormone (small molecule), thyroxine (so-called “free-T4”) was developed. Figure 3 below depicts the modular, aptamer/quantum-dot-based sensing scheme employed. To select a DNA aptamer to the molecule, thyroxine was covalently immobilized via its primary amine to a solid phase gel by standard chemistries. After 10 rounds of conventional (e.g. single-target) selection, individual aptamers were cloned and sequenced. Evolution of a unique sequence, characterized by a high GC content, as typical for known thyroxine aptamers [66] was clearly observed (sequence not shown). Figure 4 shows the performance of the FRET-aptamer sensor which has been described in detail elsewhere [67]. The sensor was found non-responsive to several structurally similar chemicals and was thus, specific for the analyte. BioTex has considerable fluorescent biosensor expertise [67-70] especially through co-investigator, Dr. Ralph Ballerstadt. The fluorescence emitted from the sensor was measured in a portable, inexpensive Qubit™ fluorometer (Invitrogen). Thus, using a sensing cocktail that can be readily lyophilized and reconstituted by the sample, such an assay could be taken to the field or bedside for environmental or clinical use. This sort of sensing scheme can be readily devised for proteins as well. We have developed an analogous sensing chemistry using aptamers specific for insulin evolved at BioTex. Figure 5 (next page) shows that result. These data demonstrate not only the ability of BioTex researchers to select DNA aptamers to novel targets, but also the broad, modular applicability of DNA aptamers in numerous applications. A QD Nanoshell B Immobilized analyte- Fluorescence Emission analog Aptamer with quencher dye analyte EX. hν EX. hν ~ 10 nm PEG coating Figure 3. Schematic of Modular, Aptamer-based QD-FRET sensing chemistry (approximate scale). In a competitive-binding fluorescence resonance energy transfer (FRET) assay, quantum dots (QDs) are conjugated to an immobilized version of the intended analyte (T4 in the text example). Aptamers which bind T4 with high specificity are identified by in vitro selection or "SELEX". Panel (A): Aptamers synthesized with a terminal fluorophore for quenching of the QD are bound to immobilized T4. Panel (B) When free T4 in the sample is exposed to this reagent mixture, QD-quenching aptamers are released from the QDs resulting in a fluorescence signal proportional to the T4 concentration. Qubit Reading (5 min values) 200 180 160 140 120 AFUs 100 80 60 40 20 0 0 0.05 0.1 0.15 0.2 0.25 0.3 0.35 Thyroxine Conc. [ug/ul] Figure 4. Quantitative detection of the small molecule thyroxine (T4) via FRET-aptamer detection. Insets show structure of T4 and portable (4.5 x 6.5 x 1.8 inch) Qubit™ fluorometer (Invitrogen) used to acquire data. 4
  • 5. B A Figure 5. Response curve of quantum-dot-based sensor (Figure 4) to insulin as formulated with the insulin-specific aptamer #43 selected at BioTex. (Panel A) The increase in fluorescence with increasing insulin concentrations is due to FRET. The putative secondary structure of insulin- aptamer #43 is shown in panel B. Whole cell SELEX for aptamers competing with ‘Stem Cell Factor’ (SCF) In addition to selection of aptamers against purified targets, we also have significant experience in selection of aptamers against the surface of whole cells. Specifically, under NSF funding, we collaborated with our literal neighbor, Synthecon Inc. (Houston, TX) in an attempt to develop an inexpensive DNA aptamer mimetic for so-called stem cell factor (SCF or “kit ligand”). Recombinant SCF is a rather expensive, but commonly used reagent in stem cell culture. To achieve this goal, we enriched our randomized DNA library for aptamers binding whole stem cells and then selectively displaced the desired aptamers from the c-Kit receptor using SCF itself. Although we were unable to evolve an agonist mimetic (to effect stem cell proliferation in culture), did successfully develop a number of candidate c-Kit specific antagonists. We are currently quantifying their affinity. 5
  • 6. Additional Aptamer Applications: Aptamer-Western blots of 2D gels and Affinity Depletion of Abundant Proteins It has already been demonstrated that aptamers can perform well in a Western-blot application [50, 51]. Similarly they have functioned as direct replacements for antibody reporters in ELISAs and dot blots [49]. A number of aptamer conjugation/functionalization schemes are likely accepted for this purpose. Most simply, aptamers can synthesized to contain either a 5’- or 3’-fluorphore of choice. Alternatively, as described in Preliminary Work, we have considerable experience in conjugating aptamers to quantum dots with the advantage of higher quantum yield and larger Stokes’ shifts of fluorescence. Finally, for additional signal, aptamers can be synthesized with a 5’- or 3’-biotin and detected by a streptavidin/horseradish peroxidase conjugate (strep-HRP). Numerous commercial sources for both fluorescent and chromagenic HRP substrates are available. Such an approach is often referred to as an “ELONA” [71, 72] or aptamer-ELISA (“ELASA”) [73]. Aptamer-bead precipitation or enrichment As mentioned elsewhere, one of the attractions/advantages of aptamers is that they are readily synthesized with modifications for conjugation, covalent coupling, fluorescent reporting, etc. Specifically, aptamers can be readily coupled to beads or nanoparticles by synthesizing them with a terminal biotin or amino-group. Such functionalized particles have been used to concentrate transcription factors [74], or, in the form of magnetic beads, have been employed to enrich bacterial pathogens [75] or cancer cells [76]. Similarly, it should be feasible to develop highly multiplexed bead-based assays such as Luminex/Magpix. Immunohistochemistry Employing aptamers in conventional histology and molecular imaging is another logical extension for their use in lieu of antibodies. In recent years, directly, or indirectly labeled aptamers have, for instance, been used to detect individual receptors [77] or to differentiate cancerous from non-cancerous cells [78]. SUMMARY: Aptamers represent a promising form of synthetic, inexpensive affinity reagent which can be employed in numerous applications. Despite their promise, however, they have generally only been developed or ‘evolved’ against a single target at a time. High costs for aptamer selection (as well as now-expiring intellectual property constraints) have until now hindered their more widespread use. IceNine’s novel approach to multiplex aptamer selection can inexpensively provide aptamer affinity reagents to a revolutionary breadth of targets. Finally, our team has the expertise to demonstrate and support our customers in a variety of novel aptamer-based application areas. 6
  • 7. REFERENCES CITED: 1. Holthoff EL, Bright FV: Molecularly templated materials in chemical sensing. Anal. Chim. Acta 2007, 594:147-161. 2. Kaur G, Roy I: Therapeutic applications of aptamers. Expert opinion on investigational drugs 2008, 17:43- 60. 3. Toulme JJ, Di Primo C, Boucard D: Regulating eukaryotic gene expression with aptamers. FEBS letters 2004, 567:55-62. 4. Chu TC, Marks 3rd JW, Lavery LA, Faulkner S, Rosenblum MG, Ellington AD, Levy M: Aptamer:toxin conjugates that specifically target prostate tumor cells. Cancer Res. 2006, 66:5989-92. 5. Chu TC, Twu KY, Ellington AD, Levy M: Aptamer mediated siRNA delivery. Nucl. Acids Res. 2006, 34:e73-. 6. Chu TC, Shieh F, Lavery LA, Levy M, Richards-Kortum R, Korgel BA, Ellington AD: Labeling tumor cells with fluorescent nanocrystal-aptamer bioconjugates. Biosens Bioelectron. 2006, Feb 20. 7. Cao Z, Tong R, Mishra A, Xu W, Wong GCL, Cheng J, Lu Y: Reversible Cell-Specific Drug Delivery with Aptamer-Functionalized Liposomes. Angew. Chem. Int. Ed. 2009, 48:6494-6498. 8. Breaker RR: Natural and engineered nucleic acids as tools to explore biology. Nature 2004, 432:838- 45. 9. Tuerk C, Gold L: Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 1990, 249:505-10. 10. Ellington AD, Szostak JW: In vitro selection of RNA molecules that bind specific ligands. Nature 1990, 346:818-22. 11. Tuerk C, MacDougal S, Gold L: RNA pseudoknots that inhibit human immunodeficiency virus type 1 reverse transcriptase. Proceedings of the National Academy of Sciences 1992, 89:6988-92. 12. Lee JF, Hesselberth JR, Meyers LA, Ellington AD: Aptamer Database. Nucleic Acids Res 2004, 32:D95- 100. 13. Ruckman J, Green LS, Beeson J, Waugh S, Gillette WL, Henninger DD, Claesson-Welsh L, Janjic N: 2’- Fluoropyrimidine RNA-based Aptamers to the 165-Amino Acid Form of Vascular Endothelial Growth Factor (VEGF165), Inhibition Of Receptor Binding And Vegf-Induced Vascular Permeability Through Interactions Requiring The Exon 7-Encoded Domain. J Biol Chem 1998, 273:20556-7. 14. Jayasena SD: Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin. Chem. 1999, 45:1628-1650. 15. Lee J-H, Canny MD, De Erkenez A, Krilleke D, Ng Y-S, Shima DT, Pardi A, Jucker F: A therapeutic aptamer inhibits angiogenesis by specifically targeting the heparin binding domain of VEGF165. Proc. Natl. Acad. Sci. 2005, 102:18902-18907. 16. Ng EWM, Shima DT, Calias P, Cunningham ET, Guyer DR, Adamis AP: Pegaptanib, a targeted anti- VEGF aptamer for ocular vascular disease. Nature Reviews Drug Discovery 2006, 5:123-132. 17. Siddiqui MAA, Keating GM: Pegaptanib: in exudative age-related macular degeneration. Drugs 2005, 65:1571-1577. 18. Sazani PL, Larralde R, Szostak JW: A Small Aptamer with Strong and Specific Recognition of the Triphosphate of ATP. J Am Chem Soc 2004, 126:8371. 7
  • 8. 19. Tang J, Breaker RR: Mechanism for allosteric inhibition of an ATP-sensitive ribozyme. Nucleic acids research 1998, 26:4214-21. 20. Yang Q, Goldstein IJ, Mei H-Y, Engelke DR: DNA ligands that bind tightly and selectively to cellobiose. Proc Natl Acad Sci U S A 1998, 95:5462-5467. 21. Babendure JR, Adams SR, Tsien RY: Aptamers Switch on Fluorescence of Triphenylmethane Dyes. J Am Chem Soc 2003, 125:14716-7. 22. Hirao I, Yoshinari S, Yokoyama S, Endo Y, Ellington AD: In vitro selection of aptamers that bind to ribosome-inactivating toxins. Nucleic Acids Symp Ser 1997, 37:283-4. 23. Hirao I, Harada Y, Nojima T, Osawa Y, Masaki H, Yokoyama S: In Vitro Selection of RNA Aptamers that Bind to Colicin E3 and Structurally Resemble the Decoding Site of 16S Ribosomal RNA†. Biochemistry 2004, 43:3214-3221. 24. Liu J, Lu Y: Fast Colorimetric Sensing of Adenosine and Cocaine Based on a General Sensor Design Involving Aptamers and Nanoparticles. Angew. Chem., Int. Ed. 2006, 117:90-94. 25. Stojanovic MN, de Prada P, Landry DW: Aptamer-Based Folding Fluorescent Sensor for Cocaine. J Am Chem Soc 2001, 123:4928-4931. 26. Stojanovic MN, Landry DW: Aptamer-Based Colorimetric Probe for Cocaine. J Am Chem Soc 2002, 124:9678-9679. 27. Gopinath SCB, Misono TS, Kawasaki K, Mizuno T, Imai M, Odagiri T, Kumar PKR: An RNA aptamer that distinguishes between closely related human influenza viruses and inhibits haemagglutinin-mediated membrane fusion. J Gen Virol 2006, 87:479-87. 28. Gopinath SCB, Sakamaki Y, Kawasaki K, Kumar PKR: An Efficient RNA Aptamer against Human Influenza B Virus Hemagglutinin. Journal of Biochemistry 2006 139(5):837-846 2006, 139:837-46. 29. Liu J, Lu Y: A Colorimetric Lead Biosensor Using DNAzyme-Directed Assembly of Gold Nanoparticles. Journal of the American Chemical Society 2003, 125:6642-6643. 30. Chang I-H, Tulock J, Liu J, Kim W-S, Cannon Jr. D, Lu Y, Bohn P, Sweedler J, Cropek D: Miniaturized Lead Sensor Based on Lead-Specific DNAzyme in a Nanocapillary Interconnected Microfluidic Device. Environ. Sci. Technol. 2005, 39:3756. 31. Swearingen CB, Wernette DP, Cropek DM, Lu Y, Sweedler JV, Bohn PW: Immobilization of a Catalytic DNA Molecular Beacon on Au for Pb(II) Detection. Anal. Chem. 2005, 77:442-8. 32. Wernette DP, Kim H-K, Liu J, Swearingen CB, Yue Z, Zavareh M, Ingram CW, Economy J, Shannon MA, Bohn PW, Lua Y: New Catalytic DNA Biosensors for Trace Contaminants in Water. 33. Wrzesinski J, Ciesiolka J: Characterization of structure and metal ions specificity of Co2+-binding RNA aptamers. Biochemistry 2005, 44:6257-68. 34. Stoltenburg R, Reinemann C, Strehlitz B: SELEX--a (r)evolutionary method to generate high-affinity nucleic acid ligands. Biomolecular engineering 2007, 24:381-403. 35. Jayasena SD: Aptamers: An Emerging Class of Molecules That Rival Antibodies in Diagnostics. Clinical Chemistry 1999, 45:1628–1650. 36. Ferreira CS, Papamichael K, Guilbault G, Schwarzacher T, Gariepy J, Missailidis S: DNA aptamers against the MUC1 tumour marker: design of aptamer-antibody sandwich ELISA for the early diagnosis of epithelial tumours. Analytical and bioanalytical chemistry 2008, 390:1039-50. 8
  • 9. 37. Burbulis I, Yamaguchi K, Yu R, Resnekov O, Brent R: Quantifying small numbers of antibodies with a “near-universal” protein-DNA chimera. Nature Methods 2007, 4:1011-3. 38. McCauley TG, Hamaguchi N, Stanton M: Aptamer-based biosensor arrays for detection and quantification of biological macromolecules. Analytical biochemistry 2003, 319:244-50. 39. Cao Z, Tong R, Mishra A, Xu W, Wong GCL, Cheng J, Lu Y: Reversible Cell-Specific Drug Delivery with Aptamer-Functionalized Liposomes. Angew. Chem. Int. Ed. 2009, 48:6494-6498. 40. De Rosa G, La Rotonda MI: Nano and Microtechnologies for the Delivery of Oligonucleotides with Gene Silencing Properties. Molecules 2009, 14:2801-2823. 41. Ferreira CSM, Cheung MC, Missailidis S, Bisland S, Gariepy J: Phototoxic aptamers selectively enter and kill epithelial cancer cells. Nucl. Acids Res. 2009, 37:866-876. 42. Yan AC, Levy M: Aptamers and aptamer targeted delivery. rnabiology 2009, 6:316-320. 43. Cload S, McCauley T, Keefe A, Healy J, Wilson C: Properties of Therapeutic Aptamers. In Aptamer Handbook, The. Wiley-VCH Verlag GmbH & Co. KGaA; 2006. 44. Mathews DH, Sabina J, Zuker M, Turner DH: Expanded Sequence Dependence of Thermodynamic Parameters Improves Prediction of RNA Secondary Structure. J Mol Biol 1999, 288:911-40. 45. Markham NR, Zuker M: UNAFold: software for nucleic acid folding and hybridization. Methods Mol. Biol 2008, 453:3-31. 46. Zuker M: Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res 2003, 31:3406-15. 47. Zuker M and S: Optimal computer folding of large RNA sequences using thermodymanics and auxiliary information. Nucleic Acids Res. 1981, 9:133-148. 48. Hofacker IL: Vienna RNA secondary structure server. Nucleic Acids Res 2003, 31:3429-3431. 49. Zhu J, Li T, Hu J, Wang E: A novel dot-blot DNAzyme-linked aptamer assay for protein detection. Anal Bioanal Chem 2010, 397:2923-2927. 50. Tombelli S, Mascini M: Aptamers as molecular tools for bioanalytical methods. Curr. Opin. Mol. Ther 2009, 11:179-188. 51. Shin S, Kim I-H, Kang W, Yang JK, Hah SS: An alternative to Western blot analysis using RNA aptamer-functionalized quantum dots. Bioorg. Med. Chem. Lett 2010, 20:3322-3325. 52. Kim YS, Jung HS, Matsuura T, Lee HY, Kawai T, Gu MB: Electrochemical detection of 17beta-estradiol using DNA aptamer immobilized gold electrode chip. Biosensors & bioelectronics 2007, 22:2525-31. 53. Neff CP, Zhou J, Remling L, Kuruvilla J, Zhang J, Li H, Smith DD, Swiderski P, Rossi JJ, Akkina R: An Aptamer-siRNA Chimera Suppresses HIV-1 Viral Loads and Protects from Helper CD4+ T Cell Decline in Humanized Mice. Sci Transl Med 2011, 3:66ra6. 54. Jackson GW, Strych U: Report #BIO071A - Nucleic Acid Aptamers for Diagnostics and Therapeutics: Global Markets. BCC Research; 2010. 55. Jackson GW: Microbial Identification by MALDI-TOF Mass Spectrometry of Ribosomal RNA. 2006. 9
  • 10. 56. Jackson GW, Karouia F, Putonti C, Fofanov Y, Fox GE, Willson RC: Microarray Designs and MALDI- TOF Mass Spectrometry for Rapid Microbial Identification. In 229th National Meeting of the American Chemical Society. San Diego, CA: 2005. 57. Jackson GW, McNichols RJ, Fox GE, Willson RC: Bacterial genotyping by 16S rRNA mass cataloging. BMC bioinformatics 2006, 7:321. 58. Jackson GW, McNichols RJ, Fox GE, Willson RC: Universal Bacterial Identification by Mass Spectrometry of 16S Ribosomal RNA Cleavage Products. Intl J Mass Spectrometry 2007, 261:218-26. 59. Jackson GW, McNichols RJ, Fox GE, Willson RC: Toward universal flavivirus identification by mass cataloging. J Mol Diagn 2008, 10:135-41. 60. Jackson GW, Fox GE, Willson RC: Novel Microarray Designs With Real-Time Hybridization Monitoring For Microbial Identification. In 22nd Annual Meeting of the Houston Society for Engineering in Medicine and Biology. Houston, TX: 2005. 61. Potty AS, Kourentzi K, Fang H, Jackson GW, Zhang X, Legge GB, Willson RC: Biophysical characterization of DNA aptamer interactions with vascular endothelial growth factor. Biopolymers 2009, 91:145-56. 62. Zhang X, Potty AS, Jackson GW, Stepanov V, Tang A, Liu Y, Kourentzi K, Strych U, Fox GE, Willson RC: Engineered 5S ribosomal RNAs displaying aptamers recognizing vascular endothelial growth factor and malachite green. J Mol Recognit 2009, 22:154-161. 63. Zhang X, Potty ASR, Jackson GW, Fox GE, Willson RC: Molecular Recognition of Foreign Sequences in Engineered 5S Ribosomal RNA. In AFFINITY 2007, The 17th Biennial Meeting of the International Society for Molecular Recognition. New York University, New York, NY: 2007. 64. Zhang X, Potty ASR, Jackson GW, Fox GE, Willson RC: Engineered 5S Ribosomal RNAs displaying Anti-VEGF and Malachite Green Aptamers. 12th Annual Structural Biology Symposium, University of Texas Medical Branch at Galveston, May 18-19, 2007. 2007. 65. Liu Y, Stepanov V, Strych U, Willson R, Jackson G, Fox G: DNAzyme-mediated recovery of small recombinant RNAs from a 5S rRNA-derived chimera expressed in Escherichia coli. BMC Biotechnology 2010, 10:85. 66. Lavesque D, Beaudoin J-D, Roy S, Perreault J-P: In vitro selection and characterization of RNA aptamers binding thyroxine hormone. Biochem. J. 2007, 403:129-138. 67. Jackson GW, Strych U, Frank E, Willson RC, Ballerstadt R, McNichols RJ: Portable FRET Sensing of Proteins, Hormones, and Toxins Using DNA Aptamers and Quantum Dots. In Technical Proceedings of the 2009 Nanotechnology Conference and Trade Show, Nanotech 2009. Houston, TX: 2009. 68. Ballerstadt R, Gowda A, McNichols RJ: In Vivo Performance Evaluation of a Transdermal Near- Infrared FRET Affinity Sensor for Continuous Glucose Monitoring. Diabetes Technology & Therapeutics 2006, 8:296-311. 69. Ballerstadt R, Polak A, Beuhler A, Frye J: In-vitro long-term performance study of a near-infrared fluorescence affinity sensor for glucose monitoring. Biosensors & Bioelectronics 2004, 19:905-14. 70. Ballerstadt R, Gowda A, McNichols RJ: Fluorescence Resonance Energy Transfer-Based Near- Infrared Fluorescence Sensor for Glucose Monitoring. Diabetes Technology & Therapeutics 2004, 6:191- 200. 71. Ramos E, Piñeiro D, Soto M, Abanades DR, Martín ME, Salinas M, González VM: A DNA aptamer population specifically detects Leishmania infantum H2A antigen. Lab. Invest 2007, 87:409-416. 10
  • 11. 72. Yan XR, Gao XW, Yao LH, Zhang ZQ: [Novel methods to detect cytokines by enzyme-linked oligonucleotide assay]. Sheng wu gong cheng xue bao = Chinese journal of biotechnology 2004, 20:679-82. 73. Bruno JG, Carrillo MP, Cadieux CL, Lenz DE, Cerasoli DM, Phillips T: DNA aptamers developed against a soman derivative cross-react with the methylphosphonic acid core but not with flanking hydrophobic groups. J. Mol. Recognit 2009, 22:197-204. 74. Oguro A, Ohtsu T, Nakamura Y: An aptamer-based biosensor for mammalian initiation factor eukaryotic initiation factor 4A. Anal. Biochem 2009, 388:102-107. 75. Joshi R, Janagama H, Dwivedi HP, Senthil Kumar TMA, Jaykus L-A, Schefers J, Sreevatsan S: Selection, characterization, and application of DNA aptamers for the capture and detection of Salmonella enterica serovars. Mol. Cell. Probes 2009, 23:20-28. 76. Smith JE, Medley CD, Tang Z, Shangguan D, Lofton C, Tan W: Aptamer-conjugated nanoparticles for the collection and detection of multiple cancer cells. Anal. Chem 2007, 79:3075-3082. 77. Chen Y, Munteanu AC, Huang Y-F, Phillips J, Zhu Z, Mavros M, Tan W: Mapping receptor density on live cells by using fluorescence correlation spectroscopy. Chemistry 2009, 15:5327-5336. 78. Chen HW, Medley CD, Sefah K, Shangguan D, Tang Z, Meng L, Smith JE, Tan W: Molecular recognition of small-cell lung cancer cells using aptamers. ChemMedChem 2008, 3:991-1001. 11