AU2004242532A1 - High affinity TGF beta nucleic acid ligands and inhibitors - Google Patents

High affinity TGF beta nucleic acid ligands and inhibitors Download PDF

Info

Publication number
AU2004242532A1
AU2004242532A1 AU2004242532A AU2004242532A AU2004242532A1 AU 2004242532 A1 AU2004242532 A1 AU 2004242532A1 AU 2004242532 A AU2004242532 A AU 2004242532A AU 2004242532 A AU2004242532 A AU 2004242532A AU 2004242532 A1 AU2004242532 A1 AU 2004242532A1
Authority
AU
Australia
Prior art keywords
nucleic acid
selex
tgfp2
binding
ligand
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2004242532A
Inventor
Larry Gold
Michael Lochrie
Nikos Pagratis
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Gilead Sciences Inc
Original Assignee
Gilead Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU67487/00A external-priority patent/AU781234B2/en
Application filed by Gilead Sciences Inc filed Critical Gilead Sciences Inc
Priority to AU2004242532A priority Critical patent/AU2004242532A1/en
Publication of AU2004242532A1 publication Critical patent/AU2004242532A1/en
Abandoned legal-status Critical Current

Links

Description

AUSTRALIA
PATENTS ACT 1990 DIVISIONAL APPLICATION NAME OF APPLICANT(S): Gilead Sciences, Inc.
ADDRESS FOR SERVICE: DAVIES COLLISON CAVE Patent Attorneys 1 Nicholson Street Melbourne, 3000.
INVENTION TITLE: "High affinity TGF beta nucleic acid ligands and inhibitors" The following statement is a full description of this invention, including the best method of performing it known to us: Q.\OPER\EH\2004\Dec\12551520 357.doc 24/12/04 HIGH AFFINITY TGFP NUCLEIC ACID LIGANDS AND INHIBITORS d This application is a divisional application of Australian Application No. 67487/00 the specification and drawings of which as originally filed are incorporated herein in their entirety C1 5 by reference.
SFIELD OF THE INVENTION Described herein are methods for identifying and preparing high affinity nucleic acid C" ligands that bind human transforming growth factor 32 (TGFp2). The method utilized herein for identifying such nucleic acid ligands is called SELEX, an acronym for Systematic C Evolution of Ligands by EXponential Enrichment. This invention includes high affinity nucleic acids of human TGFp2. Further disclosed are RNA ligands to TGFp2. Also included are oligonucleotides containing nucleotide derivatives modified at the 2' position of the pyrimidines. Additionally disclosed are ligands to TGFp2 containing 2'-OCH 3 purine modifications that may have higher stability in serum and in animals. This invention also includes high affinity nucleic acid inhibitors of TGFp2. The oligonucleotide ligands of the present invention are useful in any process in which binding to TGFp2 is required. This includes, but is not limited to, their use as pharmaceuticals, diagnostics, imaging agents, and immunohistochemical reagents.
BACKGROUND OF THE INVENTION Transforming growth factor betas (TGFpS) are part of a superfamily of proteins that includes inhibins, activins, bone morphogenetic and osteogenic proteins, growth/differentiation factors, Mullerian-inhibiting substance, decapentaplegic and 60A (Drosophila), DAF-7 and unc-129 elegans), and vgl (Xenopus) (Schlunegger and Grutter (1992) Nature 358:430- 434). Three TGFP isotypes, TGFpl, TGFp2, and TGFP3, exist in mammals. There is about sequence identity between any pair of mammalian TGFps. TGFps bind to at least receptors, but only 2 or 3 of them (types I, II and possibly V) are signaling receptors. The intracellular signaling pathways activated by TGFps involve SMAD proteins and are being intensively studied (Padgett et al. (1998) Pharmacol Ther 78:47-52). The signaling receptors are found on a variety of cells. In turn, a variety of cells express TGFps.
TGFps are synthesized as precursors composed of latency-associated protein (LAP) at the amino terminus and mature TGFP at the carboxyl terminus. The precursor is cleaved and assembles as a homodimer. TGFps are secreted from cells bound to LAP and latent TGF, binding proteins (LTBPs). Latent TGFps are released from LAP and LTBP 2 I and become active by a relatively uncharacterized mechanism that may involve proteolysis by plasmin or regulation by thrombospondin (Crawford et al. (1998) Cell 93:1159-70).
The mature, released TGFP homodimer has a combined molecular weight of-25000 daltons (112 amino acids per monomer). TGFP1 and TGFP2 bind heparin and there are indications that basic amino acids at position 26 are required for heparin binding (Lyon et N al. (1997) Jour. Biol. Chem. 272:18000-18006).
x t The structure of TGFP2 has been determined using x-ray crystallography (Daopin et al. (1992) Science 257:369-373; Schlunegger and Grutter (1992) Nature 358:430-434) and is very similar to the structure of TGF3 1. TGFps belong to a structural family of proteins called the "cysteine knot" proteins that includes vascular endothelial growth factor, nerve growth factor, human chorionic gonadotropin, and platelet-derived growth factor. These proteins are structurally homologous, but have only 10-25% primary sequence homology.
The biological activities of the TGFps vary (Moses (1990) Growth Factors from Genes to Clinical Application 141-155; Wahl (1994) J. Exp. Med. 180:1587-1590). In some cases they inhibit cell proliferation (Robinson et al. (1991) Cancer Res. 113:6269- 6274) and in other cases they stimulate it (Fynan and Reiss (1993) Crit. Rev. Oncogenesis 4:493-540). They regulate extracellular matrix formation and remodeling (Koli and Arteaga (1996) Jour. Mammary Gland Biol. and Neoplasia 1:373-380). TGFps are also are very potent immunosuppressants (Letterio and Roberts (1998) Ann. Rev. Immunol.
16:137-161). TGFps are thought to play a significant role in fibrotic diseases, preventing the immune system from rejecting tumors (Fakhrai et al. (1996) Proc. Natl. Acad. USA 93:2090-2914), cancer cell growth (Koli and Arteaga (1996) J. Mammary Gland Bio. and Neoplasia 1:373-380; Reiss and Barcellos-Hoff (1997) Breast Cancer Res. and Treatment 45:81-85; Jennings and Pietenpol (1998) J. Neurooncol. 36:123-140), and tumor metastasis. They may have ancillary roles in autoimmune and infectious diseases.
Inhibition of TGFp2 by an expressed antisense RNA (Fakhrai et al. (1996) Proc. Natl.
Acad. USA 93:2090-2914) and by exogenous antisense oligonucleotides (Marzo et al.
(1997) Cancer Research 57:3200-3207) has been reported to prevent glioma formation in rats.
3 ,I The gene for mouse TGFP2 has been deleted (Sanford et al. (1997) Development 124: 2659-2670). Mice lacking TGFI2 function die near birth and have aberrant epithelial-mesencymal interactions that lead to developmental defects in the heart, eye, ear, lung, limb, craniofacial area, spinal cord, and urogenital tracts. These defects, for the most part, do not overlap abnormalities that have been observed in TGFp 1 and TGFp3
C
knockout mice. TGFps have also been overexpressed in cell lines or transgeneic mice S(Koli and Arteaga (1996) J. Mammary Gland Bio. and Neoplasia 1:373-380; Bottinger et t al. (1997) Kidney Int. 51:1355-1360; Bottinger and Kopp (1998) Miner Electrolyte Metab 24:154-160) with a variety of effects.
A method for the in vitro evolution of nucleic acid molecules with high affinity binding to target molecules has been developed. This method, Systematic Evolution of Ligands by EXponential enrichment, termed SELEX, is described in U.S. Patent Application Serial No. 07/536,428, filed June 11, 1990, entitled "Systematic Evolution of Ligands by Exponential Enrichment," now abandoned, U.S. Patent No. 5,475,096, entitled "Nucleic Acid Ligands," and U.S. Patent No. 5,270,163, entitled "Methods for Identifying Nucleic Acid Ligands" (see also W091/19813), each of which is specifically incorporated herein by reference in its entirety. Each of these applications, collectively referred to herein as the SELEX Patent Applications, describe a fundamentally novel method for making a nucleic acid ligand to any desired target molecule.
The SELEX method involves selection from a mixture of candidate oligonucleotides and step-wise iterations of binding, partitioning and amplification, using the same general selection theme, to achieve virtually any desired criterion of binding affinity and selectivity. Starting from a mixture of nucleic acids, preferably comprising a segment of randomized sequence, the SELEX method includes steps of contacting the mixture with the target under conditions favorable for binding, partitioning unbound nucleic acids from those nucleic acids which have bound to target molecules, dissociating the nucleic acid-target complexes, amplifying the nucleic acids dissociated from the nucleic acid-target complexes to yield a ligand-enriched mixture of nucleic acids, then reiterating the steps of binding, partitioning, dissociating and amplifying through as many cycles as desired to yield high affinity nucleic acid ligands to the target molecule.
The basic SELEX method may be modified to achieve specific objectives. For 0 4 example, U.S. Patent Application Serial No. 07/960,093, filed October 14, 1992, entitled S"Method for Selecting Nucleic Acids on the Basis of Structure," now abandoned, describes the use of SELEX in conjunction with gel electrophoresis to select nucleic acid molecules with specific structural characteristics, such as bent DNA (see U.S. Patent No. 5,707,796).
U.S. Patent Application Serial No. 08/123,935, filed September 17, 1993, entitled CI "Photoselection of Nucleic Acid Ligands," now abandoned, describes a SELEX based t method for selecting nucleic acid ligands containing photoreactive groups capable of binding and/or photocrosslinking to and/or photoinactivating a target molecule. U.S.
Patent Application Serial No. 08/134,028, filed October 7, 1993, entitled "High-Affinity Nucleic Acid Ligands That Discriminate Between Theophylline and Caffeine," now abandoned, describes a method for identifying highly specific nucleic acid ligands able to discriminate between closely related molecules, termed "Counter-SELEX" (see U.S.
Patent No. 5,580,737). U.S. Patent Application Serial No. 08/143,564, filed October 1993, entitled "Systematic Evolution of Ligands by EXponential Enrichment: Solution SELEX," now abandoned, (see also U.S. Patent No. 5,567,588) and U.S. Patent No.
5,861,254, entitled "Flow Cell SELEX," describe SELEX-based methods which achieve highly efficient partitioning between oligonucleotides having high and low affinity for a target molecule. U.S. Patent No 5,496,938, entitled Nucleic Acid Ligands to HIV-RT and HIV-1 Rev," describes methods for obtaining improved nucleic acid ligands after the SELEX process has been performed. U.S. Patent No. 5,705,337, entitled "Systematic Evolution of Ligands by EXponential Enrichment: Chemi-SELEX," describes methods for covalently linking a ligand to its target.
The SELEX method encompasses the identification of high-affinity nucleic acid ligands containing modified nucleotides conferring improved characteristics on the ligand, such as improved in vivo stability or delivery. Examples of such modifications include chemical substitutions at the ribose and/or phosphate and/or base positions. Specific SELEX-identified nucleic acid ligands containing modified nucleotides are described in U.S. Patent Application Serial No. 08/117,991, filed September 8, 1993, entitled "High Affinity Nucleic Acid Ligands Containing Modified Nucleotides," now abandoned, that describes oligonucleotides containing nucleotide derivatives chemically modified at the and 2'-positions of pyrimidines, as well as specific RNA ligands to thrombin containing 2'- O amino modifications (see U.S. Patent No. 5,660,985). U.S. Patent Application Serial No.
O 08/134,028, supra, describes highly specific nucleic acid ligands containing one or more nucleotides modified with 2'-amino (2'-NH 2 2'-fluoro and/or 2'-O-methyl t U.S. Patent Application Serial No. 08/264,029, filed June 22, 1994, entitled "Novel Method of Preparation of Known and Novel 2' Modified Nucleosides by C Intramolecular Nucleophilic Displacement," now abandoned, describes oligonucleotides mI containing various 2'-modified pyrimidines. PCT/US98/00589 (WO 98/18480), filed January 7, 1998, entitled "Bioconjugation of Oligonucleotides," describes a method for identifying bioconjugates to a target comprising nucleic acid ligands derivatized with a O 10 molecular entity exclusively at the 5'-position of the nucleic acid ligands.
The SELEX method encompasses combining selected oligonucleotides with other selected oligonucleotides and non-oligonucleotide functional units as described in U.S.
Patent No. 5,637,459, entitled "Systematic Evolution of Ligands by Exponential Enrichment: Chimeric SELEX," and U.S. Patent No. 5,683,867, entitled "Systematic Evolution of Ligands by Exponential Enrichment: Blended SELEX," respectively. These applications allow the combination of the broad array of shapes and other properties, and the efficient amplification and replication properties, of oligonucleotides with the desirable properties of other molecules. The full text of the above described patent applications, including but not limited to, all definitions and descriptions of the SELEX process, are specifically incorporated herein by reference in their entirety.
BRIEF SUMMARY OF THE INVENTION The present invention includes methods of identifying and producing nucleic acid ligands to transforming growth factor beta (TGFP2) and the nucleic acid ligands so identified and produced. In particular, RNA sequences are provided that are capable of binding specifically to TGFP2. Also included are oligonucleotides containing nucleotide derivatives modified at the 2' position of the pyrimidines. Specifically included in the invention are the RNA ligand sequences shown in Tables 5, 7, 8, 11, 13, 14, 16-19 and Figure 9 (SEQ ID NOS:21-108 and 128-193). Also included in this invention are RNA ligands of TGFP2 that inhibit the function of TGFp2. Also described herein are 2'OMemodified nucleic acid ligands of TGFp1.
6 O Further included in this invention is a method of identifying nucleic acid ligands 0 and nucleic acid ligand sequences to TGFp2, comprising the steps of(a) preparing a candidate mixture of nucleic acids, contacting the candidate mixture of nucleic acids with TGFp2, partitioning between members of said candidate mixture on the basis of affinity to TGFp2, and amplifying the selected molecules to yield a mixture of nucleic C acids enriched for nucleic acid sequences with a relatively higher affinity for binding to Ir TGFp2.
More specifically, the present invention includes the RNA ligands to TGFp2, identified according to the above-described method, including those ligands shown in Tables 5, 7, 8, 11, 13, 14, 16-19 and Figure 9 (SEQ ID NOS:21-108 and 128-193). Also included are nucleic acid ligands to TGFp2 that are substantially homologous to any of the given ligands and that have substantially the same ability to bind TGFp2 and inhibit the function of TGFp2. Further included in this invention are nucleic acid ligands to TGFP2 that have substantially the same structural form as the ligands presented herein and that have substantially the same ability to bind TGFp2 and inhibit the function of TGFI2.
The present invention also includes other modified nucleotide sequences based on the nucleic acid ligands identified herein and mixtures of the same.
BRIEF DESCRIPTION OF THE FIGURES Figure 1 shows a flow chart summarizing the various SELEX experiments done with TGFp2. The length of the arrowheads corresponds to the round number shown to the left. Connected arrowheads indicate branches in the SELEX experiments where a pool was used to start a new branch. Under each arrowhead the fold improvement in affinity is also shown.
Figure 2 shows activity of TGFp2 following amine coupling on a BIAcore carboxymethylcellulose (CM5) chip. A CM5 chip was loaded with TGFp2 using NHS- EDC coupling as described in the Example 1 at about 18, 718, and 1692 response units for flow cell (FC) 1,2 and 3, respectively. FC-4 was left blank as a control and was used to normalize the signals from the other FCs. The chip was then exposed to 10 nM of either latency associated peptide (LAP) (Figure 2A) or TGFp soluble receptor III (sRIII) (Figure 7 2B) at 20 PL/min in binding buffer. Data were collected for an association and a dissociation phase as shown. The signal from FC-4 was subtracted from the other FCs.
Figure 3 shows affinity improvement during the spr SELEX. A CM5 chip was loaded with TGFp2 using NHS-EDC coupling as described in Example 1 at about 18, 718, and 1692 response units for flow cell (FC) 1, 2, and 3, respectively. FC-4 was left blank
C
as a control and was used to normalize the signals from the other FCs. The chip was then l exposed to 1 [M of RNA pools from the SELEX rounds (Rd) as shown at 20 JPL/min in binding buffer. Data were collected for an association and a dissociation phase as shown.
The signal from FC-4 was subtracted from the other FCs.
Figure 4 shows nitrocellulose filter binding curves with pools from the spr SELEX. High specific activity internally labeled RNA was used from rounds as shown. Labeled RNA was incubated with various concentrations of TGFI2 in the presence of ~100,000 fold molar excess unlabeled tRNA. Bound RNA was partitioned by nitrocellulose filtration and quantitated. Data were analyzed as described in Example 1.
Figure 5 shows nitrocellulose filter binding curves with various pools. High specific activity internally labeled RNA was used from rounds as shown. Labeled RNA was incubated with various concentrations of TGFp2 (no competitor tRNA was used). Bound RNA was partitioned by nitrocellulose filtration and quantitated. Data were analyzed as described in Example 1.
Figure 6 shows specificity of the bioactivity of lead TGFI31 and TGFp2 aptamers and comparison with commercial antibody preparations. RNA was either synthesized by phosphoramidite chemistry (NX22283) (SEQ ID NO:114) or by in vitro transcription.
Indicator cells (mink lung epithelial cells) were incubated with either TGFPl, TGFp2 or TGFp3 and dilutions of RNA or antibody as described. The extent of cell proliferation was measured by 3 H-thymidine incorporation and the data were analyzed as described.
The points represent an average of n=2-6 and error bars are standard errors. Symbols designated by TGFl TGFp2 or TGFp3 indicate data obtained from cells treated with either TGF1, TGFp2 or TGFp3, respectively. MAB and pAB designate monoclonal and polyclonal antibodies, respectively. Random, NX22283, and 40-03 designate the use of random RNA, the TGFp2, or the TGFlI lead aptamer, respectively. The aptamer 40-03 8 O was described in the U.S. Patent Application Serial No. 09/046,247, filed March 23, 1998, O entitled "High-Affinity High Affinity TGFp Nucleic Acid Ligands and Inhibitors." Figure 7 shows boundaries of TGFp2 ligands 14-1 (SEQ ID NO:72), 21-21 (SEQ ID NO:87), and 21-4 (SEQ ID NO:86). RNA aptamers were end labeled at the 5' end or at the 3' end partially hydrolyzed at high pH, and partitioned for binding to CI TGFp2 by nitrocellulose filtration as described in the Example 1. The amounts of TGF32 V used for binding partitioning is as shown. Recovered RNA was analyzed on high resolution sequencing gels and visualized by autoradiography. Unselected hydrolyzed RNA was used as a marker (Alk. hydr.) to align the banding pattern to the sequence of each ligand. The observed boundary bands are shown with and their position in the sequence pattern is shown by arrowheads. No protein and input lanes show the background binding to nitrocellulose and the starting unhydrolyzed RNA. The observed boundaries for each ligand is summarized at the bottom of the figure.
Figure 8 shows the putative structures of TGFp2 aptamers. The minimal required sequences were fit into similar structures. Ligand 14i-lt5-41 (SEQ ID NO:131) and 21a- 4(ML-110) (SEQ ID NO:144) were transcribed in vitro and contained extra bases at their ends (shown in lower case) to allow efficient in vitro transcription. Bold-faced letters indicate positions that are identical to invariant positions of the biased SELEX with the 21- 21 sequence (SEQ ID NO:93).
Figure 9 shows the molecular structure of NX22323 40k PEG (SEQ ID NO: 115).
rG 2'OH G; rA 2'-OH A; fU 2'FU; fC 2'FC.
Figure 10 shows the putative structure of lead truncate ligand CD70 (SEQ ID NO:216). Lower case letters indicate positions requiring 2'OH and indicates GU base pairing.
Figure 11 shows the pharmacokinetics of TGFp aptamer in sprague dawley rats (dose 1 mg/kg).
Figure 12 shows the results of three separate PAI-Luciferase assays performed on MLEC with TGFlI and TGFpl aptamer M22 (SEQ ID NO:215). The negative control is an unrelated aptamer. The TGFp concentration was held constant at 20 pM for 16 hours.
Figure 13 shows the results of three separate PAI-Luciferase assays performed on MLEC with TGFp2 and the TGFp2 aptamer NX22421 (SEQ ID NO:186). The negative 9 O control is an unrelated aptamer. The TGFp concentration was held constant at 10 pM for 0 16 hours.
DETAILED DESCRIPTION OF THE INVENTION This application describes high-affinity nucleic acid ligands to TGFP2 identified C1 through the method known as SELEX. SELEX is described in U.S. Patent Application tI) Serial No. 07/536,428, entitled "Systematic Evolution of Ligands by EXponential Enrichment," now abandoned, U.S. Patent No. 5,475,096, entitled "Nucleic Acid Ligands," and U.S. Patent No. 5,270,163, entitled "Methods for Identifying Nucleic Acid Ligands,"(see also WO91/19813). These applications, each specifically incorporated herein by reference in its entirety, are collectively called the SELEX Patent Applications.
Nucleic acid ligands to TGFp have been identified through the SELEX method. These TGFp nucleic acid ligands are described in U. S. Patent No. 5,731,144, U.S. Patent Application Serial No. 09/046,247, filed March 23, 1998, both entitled, "High Affinity TGFp Nucleic Acid Ligands and Inhibitors," and U.S. Patent Application Serial No.
09/275,850, filed March 24, 1999, entitled "Truncation SELEX Method." These applications are specifically incorporated herein by reference in their entirety.
Certain terms used to described the invention herein are defined as follows.
"Nucleic Acid Ligand" as used herein is a non-naturally occurring nucleic acid having a desirable action on a target. Nucleic acid ligands are also referred to herein as "aptamers." A desirable action includes, but is not limited to, binding of the target, catalytically changing the target, reacting with the target in a way which modifies/alters the target or the functional activity of the target, covalently attaching to the target as in a suicide inhibitor, and facilitating the reaction between the target and another molecule. In a preferred embodiment, the desirable action is specific binding to a target molecule, such target molecule being a three dimensional chemical structure other than a polynucleotide that binds to the nucleic acid ligand through a mechanism which predominantly depends on Watson/Crick base pairing or triple helix binding, wherein the nucleic acid ligand is not a nucleic acid having the known physiological function of being bound by the target molecule. Nucleic acid ligands include nucleic acids that are identified from a candidate mixture of nucleic acids, said nucleic acid ligand being a ligand of a given target by the O o method comprising: a) contacting the candidate mixture with the target, wherein nucleic 0 acids having an increased affinity to the target relative to the candidate mixture may be partitioned from the remainder of the candidate mixture; b) partitioning the increased affinity nucleic acids from the remainder of the candidate mixture; and c) amplifying the increased affinity nucleic acids to yield a ligand-enriched mixture of nucleic acids.
CN "Candidate Mixture" is a mixture of nucleic acids of differing sequence from in which to select a desired ligand. The source of a candidate mixture can be from naturallyoccurring nucleic acids or fragments thereof, chemically synthesized nucleic acids, enzymatically synthesized nucleic acids or nucleic acids made by a combination of the foregoing techniques. In a preferred embodiment, each nucleic acid has fixed sequences ("1 surrounding a randomized region to facilitate the amplification process.
"Nucleic Acid" means either DNA, RNA, single-stranded or double-stranded and any chemical modifications thereof. Modifications include, but are not limited to, those which provide other chemical groups that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole. Such modifications include, but are not limited to, 2'-position sugar modifications, 5-position pyrimidine modifications, 8-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil, backbone modifications, methylations, unusual base-pairing combinations such as the isobases isocytidine and isoguanidine and the like.
Modifications can also include 3' and 5' modifications such as capping.
"SELEX" methodology involves the combination of selection of nucleic acid ligands which interact with a target in a desirable manner, for example binding to a protein, with amplification of those selected nucleic acids. Iterative cycling of the selection/amplification steps allows selection of one or a small number of nucleic acids which interact most strongly with the target from a pool which contains a very large number of nucleic acids. Cycling of the selection/amplification procedure is continued until a selected goal is achieved. In the present invention, the SELEX methodology is employed to obtain nucleic acid ligands to TGFp2. The SELEX methodology is described in the SELEX Patent Applications.
11 O "Target" means any compound or molecule of interest for which a ligand is Sdesired. A target can be a protein, peptide, carbohydrate, polysaccharide, glycoprotein, hormone, receptor, antigen, antibody, virus, substrate, metabolite, transition state analog, cofactor, inhibitor, drug, dye, nutrient, growth factor, etc. without limitation. In this application, the target is TGF02.
N, In its most basic form, the SELEX process may be defined by the following series Sof steps: 1) A candidate mixture of nucleic acids of differing sequence is prepared. The candidate mixture generally includes regions of fixed sequences each of the members of the candidate mixture contains the same sequences in the same location) and regions of randomized sequences. The fixed sequence regions are selected either: a) to assist in the amplification steps described below; b) to mimic a sequence known to bind to the target; or c) to enhance the concentration of a given structural arrangement of the nucleic acids in the candidate mixture. The randomized sequences can be totally randomized the probability of finding a base at any position being one in four) or only partially randomized the probability of finding a base at any location can be selected at any level between 0 and 100 percent).
2) The candidate mixture is contacted with the selected target under conditions favorable for binding between the target and members of the candidate mixture. Under these circumstances, the interaction between the target and the nucleic acids of the candidate mixture can be considered as forming nucleic acid-target pairs between the target and those nucleic acids having the strongest affinity for the target.
3) The nucleic acids with the highest affinity for the target are partitioned from those nucleic acids with lesser affinity to the target. Because only an extremely small number of sequences (and possibly only one molecule of nucleic acid) corresponding to the highest affinity nucleic acids exist in the candidate mixture, it is generally desirable to set the partitioning criteria so that a significant amount of the nucleic acids in the candidate mixture (approximately 5-50%) are retained during partitioning.
4) Those nucleic acids selected during partitioning as having the relatively higher affinity to the target are then amplified to create a new candidate mixture that is enriched in nucleic acids having a relatively higher affinity for the target.
S12 O 5) By repeating the partitioning and amplifying steps above, the newly formed 0 candidate mixture contains fewer and fewer weakly binding sequences, and the average degree of affinity of the nucleic acids to the target will generally increase. Taken to its extreme, the SELEX process will yield a candidate mixture containing one or a small number of unique nucleic acids representing those nucleic acids from the original Ci candidate mixture having the highest affinity to the target molecule.
V The SELEX Patent Applications describe and elaborate on this process in great detail. Included are targets that can be used in the process, methods for partitioning nucleic acids within a candidate mixture, and methods for amplifying partitioned nucleic O 10 acids to generate enriched candidate mixture. The SELEX Patent Applications also describe ligands obtained to a number of target species, including both protein targets where the protein is and is not a nucleic acid binding protein.
The SELEX method further encompasses combining selected nucleic acid ligands with lipophilic or non-immunogenic, high molecular weight compounds in a diagnostic or therapeutic complex as described in U.S. Patent No. 6,011,020, "Nucleic Acid Ligand Complexes." VEGF nucleic acid ligands that are associated with a lipophilic compound, such as diacyl glycerol or dialkyl glycerol, in a diagnostic or therapeutic complex are described in U.S. Patent No. 5,859,228, entitled "Vascular Endothelial Growth Factor (VEGF) Nucleic Acid Ligand Complexes." VEGF nucleic acid ligands that are associated with a lipophilic compound, such as a glycerol lipid, or a non-immunogenic, high molecular weight compound, such as polyalkylene glycol, are further described in U.S. Patent Application Serial No. 08/897,351, filed July 21, 1997, entitled "Vascular Endothelial Growth Factor (VEGF) Nucleic Acid Ligand Complexes". VEGF nucleic acid ligands that are associated with a non-immunogenic, high molecular weight compound or lipophilic compound are also further described in PCT/US 97/18944 (WO 98/18480), filed October 17, 1997, entitled "Vascular Endothelial Growth Factor (VEGF) Nucleic Acid Ligand Complexes." Each of the above described patent applications which describe modifications of the basic SELEX procedure are specifically incorporated by reference herein in their entirety.
In certain embodiments of the present invention it is desirable to provide a complex comprising one or more nucleic acid ligands to TGFP2 covalently linked with a non- 13 O immunogenic, high molecular weight compound or lipophilic compound. A complex as 0 used herein describes the molecular entity formed by the covalent linking of the nucleic acid Q ligand of TGFp2 to a non-immunogenic, high molecular weight compound. A nonimmunogenic, high molecular weight compound is a compound between approximately 100 Da to 1,000,000 Da, more preferably approximately 1000 Da to 500,000 Da, and most C preferably approximately 1000 Da to 200,000 Da, that typically does not generate an to immunogenic response. For the purposes of this invention, an immunogenic response is one that causes the organism to make antibody proteins. In one preferred embodiment of the invention, the non-immunogenic, high molecular weight compound is a polyalkylene glycol.
In the most preferred embodiment, the polyalkylene glycol is polyethylene glycol (PEG).
("1 More preferably, the PEG has a molecular weight of about 10-80K. Most preferably, the PEG has a molecular weight of about 20-45K. In certain embodiments of the invention, the non-immunogenic, high molecular weight compound can also be a nucleic acid ligand.
In another embodiment of the invention it is desirable to have a complex comprised of a nucleic acid ligand to TGFp2 and a lipophilic compound. Lipophilic compounds are compounds that have the propensity to associate with or partition into lipid and/or other materials or phases with low dielectric constants, including structures that are comprised substantially of lipophilic components. Lipophilic compounds include lipids as well as nonlipid containing compounds that have the propensity to associate with lipid (and/or other materials or phases with low dielectric constants). Cholesterol, phospholipid, and glycerol lipids, such as dialkylglycerol, diacylglycerol, and glycerol amide lipids are further examples of lipophilic compounds. In a preferred embodiment, the lipophilic compound is a glycerol lipid.
The non-immunogenic, high molecular weight compound or lipophilic compound may be covalently bound to a variety of positions on the nucleic acid ligand to TGFp2, such as to an exocyclic amino group on the base, the 5-position of a pyrimidine nucleotide, the 8position of a purine nucleotide, the hydroxyl group of the phosphate, or a hydroxyl group or other group at the 5' or 3' terminus of the nucleic acid ligand to TGFp2. In embodiments where the lipophilic compound is a glycerol lipid, or the non-immunogenic, high molecular weight compound is polyalkylene glycol or polyethylene glycol, preferably the nonimmunogenic, high molecular weight compound is bonded to the 5' or 3' hydroxyl of the 14 0 phosphate group thereof. In the most preferred embodiment, the lipophilic compound or 0 non-immunogenic, high molecular weight compound is bonded to the 5' hydroxyl of the phosphate group of the nucleic acid ligand. Attachment of the non-immunogenic, high molecular weight compound or lipophilic compound to the nucleic acid ligand of TGFP can be done directly or with the utilization of linkers or spacers.
A "linker" is a molecular entity that connects two or more molecular entities through Scovalent bonds or non-covalent interactions, and can allow spatial separation of the molecular entities in a manner that preserves the functional properties of one or more of the molecular entities. A linker can also be known as a spacer.
The complex comprising a nucleic acid ligand to TGFP2 and a non-immunogenic, high molecular weight compound or lipophilic compound can be further associated with a lipid construct. Lipid constructs are structures containing lipids, phospholipids, or derivatives thereof comprising a variety of different structural arrangements which lipids are known to adopt in aqueous suspension. These structures include, but are not limited to, lipid bilayer vesicles, micelles, liposomes, emulsions, lipid ribbons or sheets, and may be complexed with a variety of drugs and components which are known to be pharmaceutically acceptable. In the preferred embodiment, the lipid construct is a liposome. The preferred liposome is unilamellar and has a relative size less than 200 nm. Common additional components in lipid constructs include cholesterol and alpha-tocopherol, among others. The lipid constructs may be used alone or in any combination which one skilled in the art would appreciate to provide the characteristics desired for a particular application. In addition, the technical aspects of lipid constructs and liposome formation are well known in the art and any of the methods commonly practiced in the field may be used for the present invention.
The SELEX method further comprises identifying bioconjugates to a target.
Copending PCT Patent Application No. US98/00589 (WO 98/30720), filed January 7, 1998, entitled "Bioconjugation of Oligonucleotides," describes a method for enzymatically synthesizing bioconjugates comprising RNA derivatized exclusively at the 5'-position with a molecular entity, and a method for identifying bioconjugates to a target comprising nucleic acid ligands derivatized with a molecular entity exclusively at the 5'-position of the nucleic acid ligands. A "bioconjugate" as used herein refers to any oligonucleotide which has been derivatized with another molecular entity. In the preferred embodiment, the 0 molecular entity is a macromolecule. As used herein, a "macromolecule" refers to a large O organic molecule. Examples of macromolecules include, but are not limited to nucleic acids, oligonucleotides, proteins, peptides, carbohydrates, polysaccharides, glycoproteins, lipophilic compounds, such as cholesterol, phospholipids, diacyl glycerols and dialkyl glycerols, hormones, drugs, non-immunogenic high molecular weight compounds, C fluorescent, chemiluminescent and bioluminescent marker compounds, antibodies and t biotin, etc. without limitation. In certain embodiments, the molecular entity may provide certain desirable characteristics to the nucleic acid ligand, such as increasing RNA hydrophobicity and enhancing binding, membrane partitioning and/or permeability.
Additionally, reporter molecules, such as biotin, fluorescein or peptidyl metal chelates for incorporation of diagnostic radionuclides may be added, thus providing a bioconjugate which may be used as a diagnostic agent.
The methods described herein and the nucleic acid ligands identified by such methods are useful for both therapeutic and diagnostic purposes. Therapeutic uses include the treatment or prevention of diseases or medical conditions in human patients.
Therapeutic uses may also include veterinary applications.
Diagnostic utilization may include both in vivo or in vitro diagnostic applications.
The SELEX method generally, and the specific adaptations of the SELEX method taught and claimed herein specifically, are particularly suited for diagnostic applications. SELEX identifies nucleic acid ligands that are able to bind targets with high affinity and with surprising specificity. These characteristics are, of course, the desired properties one skilled in the art would seek in a diagnostic ligand.
The nucleic acid ligands of the present invention may be routinely adapted for diagnostic purposes according to any number of techniques employed by those skilled in the art or by the methods described in PCT/US98/00589 (WO 98/30720). Diagnostic agents need only be able to allow the user to identify the presence of a given target at a particular locale or concentration. Simply the ability to form binding pairs with the target may be sufficient to trigger a positive signal for diagnostic purposes. Those skilled in the art would also be able to adapt any nucleic acid ligand by procedures known in the art to incorporate a labeling tag in order to track the presence of such ligand. Such a tag could O 16 0 be used in a number of diagnostic procedures. The nucleic acid ligands to TGF32 O described herein may specifically be used for identification of the TGFI2 protein.
SELEX provides high affinity ligands of a target molecule. This represents a singular achievement that is unprecedented in the field of nucleic acids research. The present invention applies the SELEX procedure to the specific target ofTGFP1. In the (C7 Example section below, the experimental parameters used to isolate and identify the in nucleic acid ligands to TGFP2 are described.
SIn order to produce nucleic acids desirable for use as a pharmaceutical, it is preferred that the nucleic acid ligand: 1) binds to the target in a manner capable of achieving the desired effect on the target; 2) be as small as possible to obtain the desired effect; 3) be as stable as possible; and 4) be a specific ligand to the chosen target. In most situations, it is preferred that the nucleic acid ligand have the highest possible affinity to the target.
In co-pending and commonly assigned U.S. Patent No. 5,496,938, ('938 patent), methods are described for obtaining improved nucleic acid ligands after SELEX has been performed. The '938 patent, entitled "Nucleic Acid Ligands to HIV-RT and HIV-1 Rev," is specifically incorporated herein by reference in its entirety.
In the present invention, SELEX experiments were performed in order to identify RNA ligands with specific high affinity for TGFp2 from degenerate libraries containing 33, 34 or 40 random positions (33N, 34N or 40N) (Table This invention includes the specific RNA ligands to TGFP2 shown in Tables 5, 7, 8, 11, 13, 14, 16-19 and Figure 9 (SEQ ID NOS:21-108 and 128-193), identified by the methods described in Example 1.
This invention further includes RNA ligands to TGFI2 which inhibit TGFI2 function, presumably by inhibiting the interaction ofTGFp2 with its receptor. The scope of the ligands covered by this invention extends to all nucleic acid ligands ofTGFp2, modified and unmodified, identified according to the SELEX procedure. More specifically, this invention includes nucleic acid sequences that are substantially homologous to the ligands shown in Tables 5, 7, 8, 11, 13, 14, 16-19 and Figure 9 (SEQ ID NOS:21-108 and 128- 193). By substantially homologous it is meant a degree of primary sequence homology in excess of 70%, most preferably in excess of 80%, and even more preferably in excess of 95% or 99%. The percentage of homology as described herein is calculated as the 17 Spercentage of nucleotides found in the smaller of the two sequences which align with 0 identical nucleotide residues in the sequence being compared when 1 gap in a length of nucleotides may be introduced to assist in that alignment. A review of the sequence homologies of the ligands of TGFI2, shown in Tables 5, 7, 8, 11, 13, 14, 16-19 and Figure 9 (SEQ ID NOS:21-108 and 128-193) shows that some sequences with little or no C primary homology may have substantially the same ability to bind TGFp2. For these in reasons, this invention also includes nucleic acid ligands that have substantially the same structure and ability to bind TGFp2 as the nucleic acid ligands shown in Tables 5, 7, 8, 11, 13, 14, 16-19 and Figure 9 (SEQ ID NOS:21-108 and 128-193). Substantially the same
O
S 10 ability to bind TGFp2 means that the affinity is within one or two orders of magnitude of the affinity of the ligands described herein. It is well within the skill of those of ordinary skill in the art to determine whether a given sequence substantially homologous to those specifically described herein-- has substantially the same ability to bind TGFp.
This invention also includes nucleic acid ligands that have substantially the same postulated structure or structural motifs. Substantially the same structure or structural motifs can be postulated by sequence alignment using the Zukerfold program (see Zuker (1989) Science 244:48-52). As would be known in the art, other computer programs can be used for predicting secondary structure and structural motifs. Substantially the same structure or structural motif of nucleic acid ligands in solution or as a bound structure can also be postulated using NMR or other techniques as would be known in the art.
One potential problem encountered in the therapeutic, prophylactic and in vivo diagnostic use of nucleic acids is that oligonucleotides in their phosphodiester form may be quickly degraded in body fluids by intracellular and extracellular enzymes such as endonucleases and exonucleases before the desired effect is manifest. Certain chemical modifications of the nucleic acid ligand can be made to increase the in vivo stability of the nucleic acid ligand or to enhance or to mediate the delivery of the nucleic acid ligand.
See, U.S. Patent Application Serial No. 08/117,991, filed September 8, 1993, entitled "High Affinity Nucleic Acid Ligands Containing Modified Nucleotides," now abandoned and U.S. Patent No. 6,011,020, entitled "Nucleic Acid Ligand Complexes," which are specifically incorporated herein by reference in their entirety. Modifications of the nucleic acid ligands contemplated in this invention include, but are not limited to, those which 0 18 Sprovide other chemical groups that incorporate additional charge, polarizability, o hydrophobicity, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole. Such modifications include, but are not limited to, 2'-position sugar modifications, 5-position pyrimidine modifications, 8position purine modifications, modifications at exocyclic amines, substitution of 4- N thiouridine, substitution of 5-bromo or 5-iodo-uracil, backbone modifications, phosphorothioate or alkyl phosphate modifications, methylations, unusual base-pairing combinations such as the isobases isocytidine and isoguanidine and the like.
Modifications can also include 3' and 5' modifications such as capping.
O 10 Where the nucleic acid ligands are derived by the SELEX method, the modifications can be pre- or post- SELEX modifications. Pre-SELEX modifications yield nucleic acid ligands with both specificity for their SELEX target and improved in vivo stability. Post-SELEX modifications made to 2'-OH nucleic acid ligands can result in improved in vivo stability without adversely affecting the binding capacity of the nucleic acid ligand. The preferred modifications of the nucleic acid ligands of the subject invention are 5' and 3' phosphorothioate capping and/or inverted phosphodiester linkage at the 3' end. In one preferred embodiment, the preferred modification of the nucleic acid ligand is a inverted phosphodiester linkage at the 3' end. Additional 2'-fluoro and/or 2'amino (2'-NH 2 and/or 2'-O methyl (2'-OMe and/or 2'-OCH 3 modification of some or all of the nucleotides is preferred. Described herein are nucleic acid ligands that were 2'-F modified and incorporated into the SELEX process. Also described herein are nucleic acid ligands that were 2'-OCH 3 modified after the SELEX process was performed.
Other modifications are known to one of ordinary skill in the art. Such modifications may be made post-SELEX (modification of previously identified unmodified ligands) or by incorporation into the SELEX process.
As described above, because of their ability to selectively bind TGFp2, the nucleic acid ligands to TGFp2 described herein are useful as pharmaceuticals. This invention, therefore, also includes a method for treating TGFp2-mediated pathological conditions by administration of a nucleic acid ligand capable of binding to TGFp2.
Therapeutic compositions of the nucleic acid ligands may be administered parenterally by injection, although other effective administration forms, such as 19 0 intraarticular injection, inhalant mists, orally active formulations, transdermal O iontophoresis or suppositories, are also envisioned. One preferred carrier is physiological saline solution, but it is contemplated that other pharmaceutically acceptable carriers may also be used. In one preferred embodiment, it is envisioned that the carrier and the ligand constitute a physiologically-compatible, slow release formulation. The primary solvent in C such a carrier may be either aqueous or non-aqueous in nature. In addition, the carrier may c, contain other pharmacologically-acceptable excipients for modifying or maintaining the 4 pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of
(N
Sthe formulation. Similarly, the carrier may contain still other pharmacologically- 0 S 10 acceptable excipients for modifying or maintaining the stability, rate of dissolution, release, or absorption of the ligand. Such excipients are those substances usually and customarily employed to formulate dosages for parental administration in either unit dose or multi-dose form.
Once the therapeutic composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder.
Such formulations may be stored either in a ready to use form or requiring reconstitution immediately prior to administration. The manner of administering formulations containing nucleic acid ligands for systemic delivery may be via subcutaneous, intramuscular, intravenous, intranasal or vaginal or rectal suppository.
The following Examples are provided to explain and illustrate the present invention and are not intended to be limiting of the invention. Example 1 describes the various materials and experimental procedures used in Examples 2-5. Example 2 describes the isolation and characteristics of nucleic acid ligands that bind human TGFp2. Example 3 describes the nucleic acid ligands isolated by the SELEX method using a biased round O library, the sequences of TGFI2 nucleic acid ligands isolated from the biased SELEX process, and the binding of nucleic acid ligands isolated from the biased SELEX process. Example 4 describes substitutions of 2'-OH purines with 2'-OCH 3 purines in NX22284 (SEQ ID NO:115) and NX22385 (SEQ ID NO:189). Example 5 describes the pharmacokinetic properties of NX22323 (SEQ ID NO:121). Example 6 describes 2'-OMe modification of lead TGFpl truncate ligand CD70 (SEQ ID NO:216). Example 7 describes a Mink Lung Epithelial Cell (MLEC) PAI Luciferase Assay ofTGFI aptamers to TGFp1 and TGFp2.
(N
0 EXAMPLES Example 1. Experimental Procedures Materials Monoclonal and polyclonal antibodies that recognize human TGFl1, TGFp2 or CI TGFp3 were purchased from R&D Systems, Inc. (Minneapolis, MN). DNA tr) oligonucleotides were purchased from Operon, Inc. (Alameda, CA) or Oligos, Etc.
(Redding Center, CT). The BIAcore 2000 and IAsys plus instruments are products of Biacore, Inc. (Paramus, NJ) and Affinity Sensors, Inc. (Cambridge, U. respectively.
O
Nitrocellulose filters and filtering manifolds were obtained from Millipore (Bedford, MA).
Mink lung epithelial cells (#CCL64) were purchased from the American Type Culture Collection (Rockville, MD). The cloning vectors pCR-Script and pUC9 were obtained inhouse or from Stratagene, Inc. (La Jolla, CA) or Life Technologies, Inc. (Gaithersburg, MD), respectively. E coli strains were obtained from Stratagene. The QIAprep spin miniprep kit was from QIAgen, Inc. (Chatsworth, CA). The Big Dye sequencing kit and model 377 sequencer can be purchased from Applied Biosystems (Foster City, CA). T7 RNA polymerase and Thermus aquaticus DNA polymerase were purchased from Enzyco, Inc. (Denver, CO) and Perkin Elmer (Norwalk, CT), respectively. All restriction enzymes were purchased from New England Biolabs. E. coli RNase H was obtained from Boehringer Mannheim. All synthetic nucleic acids with a name that begins with "NX" were synthesized at NeXstar Pharmaceuticals, Inc. (Boulder, CO) using an ABI model 394 DNA/RNA synthesizer (Applied Biosystems). Yeast tRNA (type X-SA) and porcine intestinal mucosca-derived heparin (molecular weight 5000), were purchased from Sigma (St. Louis, MO) and Calbiochem (La Jolla, CA), respectively.
Preparation of round 0 nucleic acid library The initial (round 0) library ofribonucleic acid molecules that was used to isolate TGFp2 nucleic acid ligands was generated as follows. Two DNA oligonucleotides (40 N7 round 0 (RO) DNA template and 5'N7 primer) were annealed and filled in with Klenow to produce a 40 N7 RO DNA transcription template (Table This template was transcribed using T7 RNA polymerase, 3 mM 2-F uridine and cytosine, 1 mM 2'-OH guanosine and adenine, and at
P
-ATP as described in (Fitzwater and Polisky (1996) S21 0 Meth. Enz. 267:275-301). This resulted in a RO 40N7 nucleic acid pool with the 0 following sequence which has 5' and 3' "fixed" regions and a 40 base long random sequence region: 5'-GGGAGGACGAUGCGG-40N-CAGACGACUCGCCCGA-3') RO 40N7 nucleic acid fixed region random region 3' fixed region (SEQ ID NO:6) CI A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U (Table 1) C-i Spot SELEX Spot SELEX was performed as described in U.S. Patent No. 5,972,599, entitled S"High Affinity Nucleic Acid Ligands ofCytokines," which is hereby incorporated by reference in its entirety, using nucleic acid that was internally labeled using aX- P
ATP.
The conditions and progress of this SELEX experiment are summarized in Table 3.
Briefly, human TGFP2 (or no protein) was applied to a 13 mm diameter nitrocellulose filter and allowed to absorb but not completely dry. The filter was incubated with RNA in Dulbecco's phosphate-buffered saline, 1 mM MgCl, and then washed as summarized in Table 3. Filter-bound and protein-bound nucleic acid was visualized and quantitated on an Instant Imager (Packard Instrument Co., Downers Grove, IL) and the protein-bound nucleic acid was eluted in 50% phenol, 4 M urea for 45 minutes at 65 0 C. Eluted nucleic acid was ethanol precipitated and then reverse transcribed using avian myeloblastosis virus reverse transcriptase and subjected to the polymerase chain reaction (PCR) using 5'N7 and 3'N7 primers for 15 cycles. This resulting transcription template was transcribed with T7 RNA polymerase in the presence of 2'-F pyrimidine nucleotides, 2'-OH purine ribonucleotides and a 32 P-ATP, and carried to the next spot round. The pool from the first spot round was also transcribed as above in the absence of cta-P-ATP for use in round 2 of the surface plasmon resonance biosensor SELEX.
Surface plasmon resonance biosensor SELEX Rounds 2-spr through 9-spr were done using surface Ilasmon resonance biosensor technology on a BIAcore model 2000 instrument. For this experiment 1XDPBS, 1 mM MgCI z 0.005% P20 surfactant (cat#BR-1000-54, Biacore, Inc., Piscataway, NJ) was used as the running buffer. TGFP2 was amine coupled onto a CM5 BIACORE chip (Biacore, Inc., Piscataway, NJ) using the Biacore amine coupling kit (cat#BR-1000-50, Biacore, 22 O Inc., Piscataway, NJ) per manufacturer's instructions. Briefly, TGFp2 aliquots (3 4L, in 4 0 mM HCI at 100 pg/mL) were diluted in 30 uL of 10 mM CH 3 COONa, pH 5.0 and injected on an EDC-NHS activated chip at 25*C, 5 uL /min, in different volumes to achieve different loading levels, as measured in response units Following coupling, the chip was washed with 3M NaCI for about 1.5 minutes at 10 p.L/min. Under these experimental C- conditions, TGFp2 loading of 15RU/IL could be achieved. TGFp2 was loaded in flow ¢c€ tr cells 1, 2, and 3 while flow cell 4 was kept blank for control and background subtractions. Before use, the chip was tested for activity by testing binding of LAP and or soluble receptor III (R&D Systems, Minneapolis, MN) at 37 0 C. At the end of each test injections the chip was regenerated using a 1 minute wash with 10 mM NaOH. For C"1 SELEX rounds, RNA pools, generated by in vitro transcription without any labeled nucleotides, were in running buffer and were injected over the TGFp2 loaded CM5 chips at 5 pL/min at 37 0 C. The concentration and volume of the RNA pools used at each round are as shown in Table 4. At each round the RNA pools were applied in 40 pL injections and each injection cycle was followed by a dissociation phase where the chip was washed with DPBS, ImM MgCl, at 20 pL/min while three 100 pL fractions (5 minutes each) were collected. Following the last injection-dissociation cycle, the chip was treated with 0.25% SDS and the eluted RNA was collected as the final fraction. The third fractions of each injection cycle and the SDS elution were pooled and amplified by RT/PCR to generate the template pool for the next SELEX round.
Resonant Mirror Optical Biosensor SELEX Rounds 10-rm through 13-rm were done using an IASYS plus resonant mirror optical biosensor instrument. Round 9-spr from the surface plasmon resonance SELEX was used as the starting material. For this experiment, 1XDPBS, 1 mM MgCl 2 0.005% P20 surfactant (cat#BR-1000-54, Biacore, Inc., Piscataway, NJ) were used as the running buffer. TGFp2 was amine coupled onto a CMD IASYS cuvette (Affinity Sensors, Cambridge, UK) according to the manufacturer's protocol. Briefly, the CMD cuvette was activated with 0.2 M EDC, 0.05 M NHS for 10 minutes, and TGFp2 was coupled by injection 35 tL of 0.4 pM TGFB2, 10 mM CH 3 COONa, pH 5.5 in 35 pL of
CH
3 COONa, pH 5.5. The coupling reaction was at 25C for about 10 minutes and resulted in about 2,000 Arcsec of signal. Unreacted sites were capped by exposing the cuvette in 23 O 1M ethanolamine for 1-2 minutes. Following coupling and capping the cuvette was Sexposed to 3M NaCI for 1-2 minutes and was ready for use. The cuvettes were routinely tested for activity by measuring binding of LAP and or soluble receptor III (R&D Systems, Minneapolis, MN) at 37 0 C. At the end of each test injection the chip was regenerated using 1 minute wash with 50 mM NaCO3. For SELEX rounds, RNA pools, CI generated by in vitro transcription without any labeled nucleotides, were in running buffer.
¢c€ tIn They were injected in the TGFP2 loaded CMD cuvette and incubated for 27-60 minutes (Table 6) under 100% steering at 37 0 C. Following binding, the RNA was replaced with buffer and bound RNA was observed to dissociate from the cuvette surface. Dissociation was allowed for 30-150 minutes (Table 6) at 37 0 C while the buffer was exchanged several times to avoid evaporation. Following dissociation, the remaining RNA was eluded with
H
2 0 or 0.25% SDS and the RNA was amplified as above and carried to the next SELEX round.
SELEX using filter partitioning and polyanion competition For rounds 9b through 22a, SELEX using filter partitioning was performed essentially as described in (Fitzwater and Polisky (1996) Meth. Enz. 267:275-301) except that 1) heparin or yeast tRNA was included to compete off ligands that bound nonspecifically, 2) the binding buffer was HBSMCK (50 mM HEPES, pH 7.4, 140 mM NaCI, 1 mM MgCI1, 1 mM CaCl, 3 mM KCI), 3) extensive efforts were undertaken to reduce filter binding sequences (preadsorbtion of nucleic acid onto filters after elution and transcription, blocking of filters with tRNA and bovine serum albumin prior to partitioning, addition of 0.5 M urea to the wash buffer) and 4) the transcripts were initiated with a 5:1 molar mixture of guanosine:2'-fluoro-nucleotides. Initiation with guanosine allows nucleic acids to be used in SELEX or bioactivity assays without radiolabeling and alleviates a phosphatase step if the nucleic acid is to be 5'-end radiolabeled for binding studies.
Round 8-spr from the surface plasmon resonance SELEX was used as the starting material. From rounds 9b to 14i, the SELEX process was performed using protein-excess conditions. The concentrations of nucleic acid and protein were equimolar in round Nucleic acid-excess conditions were used from rounds 16a to 22a (Table Competitors (yeast tRNA and heparin) were used from rounds 9b to 14i. Filters were washed with 24 O 15 mL HBSMCK buffer from rounds 9b to 12d and increasing amounts (5-50 mL) of 0 HBSMCK, 0.5 M urea from rounds 13i to 22a.
Sequencing of nucleic acid ligand pools Nucleic acid pools were sequenced as described in (Fitzwater and Polisky (1996) Meth. Enz. 267:275-301).
SScreening nucleic acid ligand pools using ligand-specific reverse transcription-PCR SNucleic acids from the various pools was reverse transcribed with clone-"specific" primers (ML-85 (SEQ ID NO:16) for ligand 14i-1 and ML-81 (SEQ ID NO: 18) for ligand 21a-21) for 12, 15 or 18 cycles. Mixtures of pure nucleic acid ligands and round 0 40N7 nucleic acid that contained 10%, 0.3% or 0.1% ligand were processed in the same manner and served to quantitate signals from RT-PCR of the nucleic acid pools.
Cloning, screening and sequencing of nucleic acid ligands Nucleic acid ligands were cloned using two methods. In one method the ligands were directly cloned into pCR-Script according to the manufacturer's instructions and transformed into E. coli strain XL-1 Blue MRF' Kan. In the other method the doublestranded DNA transcription template was amplified by PCR using primers ML-34 (SEQ ID NO: 11) and ML-78 (SEQ ID NO:12), digested with BamHI and EcoRI restriction enzymes, and cloned into BamHI and EcoRI-digested pUC9. The ligation was transformed into E. coli strain DH5at. Colonies were selected on ampicillin plates and screened for inserts by PCR using vector-specific primers (RSP1 (SEQ ID NO: 13) and FSP2 (SEQ ID NO: Typically 90%-100% of the clones had inserts. Some colonies or nucleic acid pools were also screened using 14i-1, 21a-4, or 21a-21 ligand-specific primers (ML-79 (SEQ ID NO:17), ML-81 (SEQ ID NO:18), and ML-85 (SEQ ID NO:16), respectively) in an attempt to identify clones that were different from those already isolated.
Plasmid minipreps from the transformants were prepared using the QIAprep spin miniprep kit (QIAGEN, Inc., Valencia, CA) or PERFECT prep plasmid DNA kit Inc., Boulder, CO). Sequencing reactions were performed with the Big Dye kit and a sequencing primer (RSP2). The sequencing products were analyzed on an ABI model 377 sequencer.
O Nucleic acid ligand boundaries 0 The boundaries and 3' end) of the smallest ligand that can bind TGFi2 was determined essentially as described in (Fitzwater and Polisky (1996) Meth. Enz. 267:275- 301). The protein concentrations used were 0, 1 nM, and 10 nM and the nucleic acid /protein ratio was 1. The binding buffer used in this experiment was HBSMC, 0.01% C HSA. Binding reactions were incubated at 37 0 C for 30 minutes, filtered through 0.45 jm, itr nitrocellulose filters (15 mm),and then washed with 15 mL HBSMC. The RNA was recovered by phenol-urea extraction, eluted RNA was ethanol precipitated in the presence of glycogen, resuspended in H 2 O, supplemented with equal volume 2X formamide dye, and analyzed on 8% acrylamide, 8M urea sequencing gels. Truncated RNAs that were bound to TGFI2 were visualized and developed on a FUJIX BAS1000 phosporimager (FUJI Medical Systems, USA).
Nucleic acid ligand truncation Truncated versions of full length nucleic acid ligands were generated in three ways.
In one method, E. coli RNase H and hybrid 2'-OCH 3 RNA/DNA oligonucleotides (5'N7 cleave, 3'N7 cleave; Table 1) were used to cleave nucleic acids at a specific site.
Truncation SELEX is described in U.S. Patent Application Serial No. 09/275,850, filed March 24, 1999, entitled "Truncation SELEX Method," which is hereby incorporated by reference in its entirety. In a second method, overlapping DNA oligonucleotides encoding the desired ligand sequence were annealed, extended by Klenow DNA polymerase, and then transcribed. In a third method, ligands were chemically synthesized with the desired sequence.
Binding of nucleic acid ligands to human TGF3's The binding activity of individual ligands was determined by measuring the equilibrium dissociation constants using nitrocellulose partitioning of labeled RNA as a function of protein concentration. RNA was body-labeled or guanylated and then labeled with y- 32 P ATP and T4 polynucleotide kinase. Binding reactions were set at various protein concentrations (typically varied in either 3-fold or 10-fold increments) while maintaining the labeled RNA concentration constant at less than 0.1 nM, and incubated at 37 0 C for 10 minutes. Protein-RNA complexes were partitioned away from uncomplexed RNA, by filtering the binding reactions through a nitrocellulose/cellulose O 26 o acetated mixed matrix (0.45 im pore size filter disks, type HA; Millipore, Co., Bedford, O MA). For filtration, the filters were placed onto a vacuum manifold (12-well, Millipore, or 96-well BRL) and wetted by aspirating 1-5 mL of binding buffer. The binding reactions were aspirated through the filters, washed with 1-5 mL of binding buffer and counted in a scintillation counter (Beckmann).
CN To obtain the monophasic equilibrium dissociation constants of RNA ligands to i hTGFp2 the binding reaction:
KD
C, R:P R+P 0 wherein R=RNA; P=Protein and KD=dissociation constant is converted into an equation for the fraction of RNA bound at equilibrium: q=(f/2RT)(PT+RT+KD-((PT RT KD) 2 -4 P, RT) m wherein q=fraction of RNA bound; P- total protein concentration; RT -total RNA concentration and f=retention efficiency of RNA-protein complexes. The average retention efficiency for RNA-TGFp2 complexes on nitrocellulose filters is 0.3-0.8. Kd values were obtained by least square fitting of the data points using the software Kaleidagraph (Synergy Software, Reading, PA).
Competition between ligands "P-labeled test ligands at a concentration of 1 nM were mixed with increasing concentrations of unlabeled NX22283 (SEQ ID NO: 114). Then, an amount of TGFp2 estimated to be near the Kd of the test ligands was added (1 nM for NX22283, 1 nM for 21a-21, 3 nM for 21a-4, and 10 nM for 14i-1). The reactions were incubated, filtered, washed, and counted as for a binding reaction.
Off-rate of NX22283 1 nM "P-labeled NX22283 was mixed with 10 nM TGFP2, incubated for minutes to allow the protein to bind to the nucleic acid, and then a 1000-fold excess (1 pM) of unlabeled NX22283 was added. At various time points the reactions were filtered and washed to measure the amount of "P-labeled NX22283 that remained bound.
Biased SELEX A library of sequences was constructed based on the sequence of the 34-mer truncate (NX22284 (SEQ ID NO:1 15)) of nucleic acid ligand 21a-21. The sequence of the S27 SDNA template (34N7.2 a-21 (SEQ ID NO:7)) is shown in Table 1. The randomized 0 region is 34 bases long. At each position the randomized region consists of 62.5% of the NX22284 sequence and 12.5% of the other 3 nucleotides. Thus the randomized region is Smutagenized at each position but at the same time is biased toward the sequence of NX22284. The fixed regions (5'N7, 3'N7) were the same as used for the primary CN SELEX.
V To generate 34N7.21a-21 round 0 nucleic acid, the DNA template was amplified by PCR using the 5'N7 (SEQ ID NO:2) and 3'N7 (SEQ ID NO:3) primers (Table 1).
This PCR product was transcribed as described above in the filter partitioning SELEX O 10 section. This resulted in a 34N7.21a-21 round 0 nucleic acid pool with the sequence shown in Table 1 (SEQ ID NO: Filter partitioning as described above and in Fitzwater and Polisky (1996) (Meth.
Enz. 267:275-301) with no competitors was used to enrich nucleic acids ligands that bound to human TGFp2 the best. The protein concentration was reduced from -150-300 nM to 50 pM. The nucleic acid concentration was reduced from 1 p.M to 1 nM. The nucleic acid/protein ratio ranged from 0.25 to 125. The round 5a pool of ligands was cloned into pUC9 and sequenced as described above.
Bioactivity of TGFp2 nucleic acid ligands The bioactivity of TGFp2 nucleic acid ligands was measured with mink lung epithelial cells. Proliferation of these cells is inhibited by TGFp2. Human TGFP2 was titrated on the cells and 'H-thymidine incorporation was measured. The point at which 3
H-
thymidine incorporation by the cells was inhibited by 90-100 was determined (typically 1-4 pM). This inhibitory amount of TGFp2 along with varying amounts of nucleic acid ligand (typically 0.3 or 1 nM to 1 or 3 pM, in 3 fold increments) was used. Typically, cells were plated at 10E5/mL in 96-well plates in 100 IiL MEM, 10mM HEPES pH 7.4, 0.2% FBS. Following a 4 hour incubation at 37 0 C, when cells were well attached to the well surface, TGFp2 was added at 1-4 pM with or without nucleic acid ligands as follows: the ligands were diluted across the 96 well plate in 3-fold dilution steps and then TGF32 was added at 1-4 pM to all wells except controls. The cells were incubated for 16-18 hours prior to addition of 3 H-thymidine, and then incubation was continued for additional hours following 3 H-thymidine addition at 0.25 iCi per well. After incubation, O 28 0 the cells were lysed with 1% Triton X-100 and harvested onto GF/B filter plates in a 0 Packard 96 well plate harvester, and 'H-thymidine incorporation in cellular DNA was quantitated by scintillation counting in MicroScint (Packard, Mariden, CT) in a Packard STop-Count. Data were plotted as of maximum 3 H-thymidine incorporation vs RNA concentration, and were fitted by the software Kaleidagraph (Synergy Software, Reading, N PA) to the equation m3*(m0+ml+(m2)-((m0+m where mO is the concentration of competitor RNA; ml is the m2 is the concentration of TGFp2, and m3 is the plateau value of the fraction of maximum 'H-thymidine incorporation. K, values were determined from IC 5 0 values according to the equation where is the molar concentration of TGFp2 present in the assay and Kdr is the concentration of TGFp2 causing 50% inhibition of MLEC proliferation as determined by TGFp2 titration experiments. This assay was also used to determine the isotype specificity of RNA ligands where the three TGFp isotypes were independently used as inhibitors of MLEC replication.
Pharmacokinetic properties of NX22323 (SEQ ID NO:121) The pharmacokinetic properties ofTGFp2 ligand NX22323 were determined in Sprague-Dawley rats. NX22323 was suspended in sterile PBS and stored at Prior to animal dosing NX22323 was diluted with sterile PBS, to a final concentration of 0.925 mg/mL (18 1 M, based on the oligonucleotide molecular weight and the ultraviolet absorption at 260 nm with an extinction coefficient of 0.037 mg of oligo/mL). Sprague- Dawley rats (n 2) were administered a single dose of NX22323 by intravenous bolus injection through the tail vein. Blood samples (approximately 400 gL) were obtained by venipuncture under isofluorane anesthesia and placed in EDTA-containing tubes. The EDTA-treated blood samples were immediately processed by centrifugation to obtain plasma and stored frozen 5-20 0 C. Time points for blood sample collection ranged from to 2880 minutes.
Standards and quality control samples prepared in blank rat plasma and plasma samples were analyzed by a double hybridization assay. To prepare plasma samples for hybridization analysis, 25 gL of plasma sample (or a dilution in plasma of the sample) was added to 100 L of 4 x SSC, 0.5 sarkosyl. A 25 L aliquot was then mixed with 25 tL of 4 x SSC, 0.5 sarkosyl containing 24 gM capture oligonucleotide conjugated to 29 O magnetic beads and 28 gM detect oligonucleotide conjugated to biotin in a covered 96- Swell microtiter plates. The mixture was allowed to incubate at 45 0 C. for 1 hour. Unbound oligonucleotide was removed and 0. 1 ng streptavidin alkaline phosphatase/pL NTT Buffer (0.8 M NaC1, 20 mM Tris pH 7.5, 0.5% Tween 20) added to each well followed by a 30 minute incubation at room temperature. The streptavidin alkaline phosphatase was CI removed and the plate was washed twice with 200 piL NTT Buffer. The NTT Buffer was tn removed and replaced with 50 pL DEA buffer (0.02% NaN 3 1 mM MgCI,, 1% diethanolamine (Tropix, Inc., Bedford, MA), pH 10) and 34 pL/mL 25 mM chemiluminescent substrate for alkaline phosphatase (Tropix, Inc., Bedford, MA), and S 10 20% Sapphire chemiluminescence amplifier (Tropix, Inc., Bedford, MA) in DEA buffer .L/well) was added. The plate was incubated for 20 minutes at room temperature and read on a luminometer. A standard curve of NX22323 was fit using a variable slope sigmoidal dose response non linear regression equation (PRISM, version 2.00, GraphPad, San Diego, CA). Sample and quality control concentrations were extrapolated from the standard curve and corrected for dilution.
The average plasma concentration at each time point was calculated and utilized in the pharmacokinetic analysis. Both noncompartmental and compartmental pharmacokinetic analysis were carried out using WinNonlin version 1.5 (Scientific Consulting, Inc.). In the noncompartmental analysis, the following parameters were calculated; the maximum concentration extrapolated at zero time (Cmax), the area under the curve from zero to the last time point (AUClast), the area under the curve from zero to infinite time (AUCINF), the terminal phase half life (Beta tl/2), the clearance rate the mean residence time calculated from zero to infinite time (MRTINF), the volume of distribution at steady state (Vss), and the volume of distribution during elimination (Vz).
In the case of compartmental analysis, the following parameters were calculated based on the minimum number of monoexponential equations to adequately fit the data: the maximum concentration extrapolated at zero time (Cmax), the area under the curve from zero to infinite time (AUCINF), the distribution phase half life (Alpha tl/2), terminal phase half life (Beta tl/2), the exponential constant for the distribution phase the exponential constant for the terminal phase the clearance rate the mean residence 0 time calculated from zero to infinite time (MRTINF), and the volume of distribution at O steady state (Vss).
Example 2. Isolation of Nucleic Acid Ligands that Bind Human TGF32 Several SELEX experiments on TGFp2 have been attempted as summarized in Figure 1. Several partitioning methods were applied at various stages of the SELEX progress including standard filtration through nitrocellulose, spot, surface plasmon resonance biosensor (BIAcore), resonant mirror biosensor (lasys), polystyrene beads, and polyacrylamide gel shift. The combination of spot SELEX, surface plasmon resonance biosensor SELEX, and filter partitioning SELEX (with competitors) described here had the best overall improvement in affinity (->1000 fold) and thus is described in detail. In addition, a branch of this SELEX that utilized resonant mirror biosensor technology is also described.
Spot SELEX conditions Spot SELEX was chosen to initiate the SELEX process on human TGFp2 because it would allow a large amount of protein and nucleic acid to interact. The conditions used are shown in Table 3. The results of round 1 were acceptable. The background was very low and the signal to noise ratio was 5. At this point the population from the first round was used for the surface plasmon SELEX in addition to continuing with the spot partition method. Ten rounds of spot SELEX were completed as summarized in Table 3 and a modest improvement in the affinity of the pool of about 30 fold was observed. These pools were not analyzed further.
Surface plasmon resonance biosensor (spr) SELEX Surface plasmon resonance biosensors were chosen as a partitioning medium because they provide very low background nucleic acid binding to the sensor, so that higher degrees of enrichment can be obtained. In addition binding and elution of nucleic acid can be monitored and quantitated in real time.
TGFp2 was coupled to a BIAcore biosensor using amine coupling chemistry.
TGFp2 coupled in this manner binds latency-associated protein, and recombinant soluble TGFp receptors. Figures 2A and 2B show typical sensograms obtained with LAP and recombinant sRIII where ko, and kff rates indicative of avid binding were observed.
During this SELEX experiment as summarized in Table 4, a binding signal was first S31 O observed on the biosensor in round (Rd) 6, increased up to round 9, and then decreased in O rounds 10 and 11. Figure 3 shows sensograms with 0, 4-spr, 6-spr, 8-spr, and 9.-spr.
Figure 4 shows typical filter binding curves, in the presence and absence of competitor tRNA, with representative pools from the spr SELEX and from these data it seems that round 9 binds in a biphasic manner with a high affinity and low affinity Kd of 30 and 160 C'1 nM, respectively.
In In bioactivity assays the K, of the round 0 pool was about 2.6 pM and the K, of the round 9-spr pool was about 711 nM (see below). Sequence analysis of representative pools indicated that such pools maintained significant complexity up to round 8 while after O 10 round 9 such pools were strongly biased towards a single sequence.
Round 8-spr was cloned and sequenced. A total of 69 clones representing 51 different sequences were analyzed. Four sequences (Nos 8.2 (SEQ ID NO:22), 8.3 (SEQ ID NO:23), 8.9 (SEQ ID NO:27), and 8.48 (SEQ ID NO:54); see Table 5) were represented more than once and accounted for 21 of the 69 clones. All four of these sequences bound TGFP2 and were 2 or 3 base variants of a clone (14i-1) isolated from the filter SELEX (see below). Twenty three other sequences were nonbinding or filterbinding sequences (see Table 5) and 25 clones were not tested for binding.
Resonant mirror (rm) optical biosensor SELEX Since the affinity of nucleic acid pools selected on the surface plasmon resonance biosensor peaked at round 9-spr, resonant mirror (rm) optical biosensor technology was tested to determine if it could advance the affinity of nucleic acid ligands any further.
Resonant mirror optical biosensor technology offers many of the same advantage as surface plasmon biosensor technology, but in addition the binding is done in a cuvette under equilibrium conditions rather than over the surface of a chip under flow conditions.
Within the cuvette the binding can be extended for long time periods. Therefore, the nucleic acid/protein binding reaction can be more stringent and selective.
For resonant mirror SELEX, TGFP2 was coupled to two biosensor cuvettes using amine coupling chemistry. In one cuvette the TGFP2 was inactivated by SDS denaturation and this cuvette served to assess background. The other cuvette containing active TGFp2 was used for the SELEX. Beginning with round 9-spr, five rounds were 32 O done using resonant mirror optical biosensor technology. The conditions used for and the o results of the resonant mirror SELEX are shown in Table 6.
Biosensor signals were observed for each round. The binding of the nucleic acid pools from rounds 10-rm to 12-rm was assessed (Table 6; Figure The pool K, improved modestly up to round 12-rm with no further improvement in the subsequent C rounds. The round 12-rm pool binds biphasically with high and low affinity Kd of-2nM t and -150nM, respectively.
In bioactivity assays the K, of the round 13-rm pool was about 505 nM. Round 13rm was chosen for subcloning and sequence analysis. Of 15 clones that were sequenced, 0 10 all 15 (Table 7) were 1 to 5 base variants of a clone (14i-1), which was originally isolated from the filter SELEX (see below).
Filter partitioning SELEX Round 8-spr was used as the starting material for a filter SELEX. The properties of round 8-spr were studied and it was found that 1) a significant fraction bound to a nitrocellulose filter 2) significant nucleic acid binding (defined here as signal/noise>2) to TGFp2 was not detectable using nucleic acid-excess conditions, and 3) in the presence or absence of polyanionic competitors there was a significant decrease in the binding of round 0, but not round 8-spr to TGFp2. These findings had implications that are addressed below.
Use of a competitor The binding of round 8-spr nucleic acid to TGFp2 in the presence of a polyanionic competitor (yeast tRNA) was studied at various ratios of competitor to nucleic acid. It was found that a 75,000 fold excess of tRNA over round 8-spr nucleic acid resulted in inhibition of binding, whereas a 6,000 fold excess of tRNA over the round 0 nucleic acid pool resulted in 50% inhibition of binding. Heparin also competed with RNA for binding to TGFp2, but about 10-fold more heparin was needed to inhibit RNA binding to TGFp2 to the same degree as that observed using tRNA. By including a 100,000-fold excess of yeast tRNA over RNA in a TGFp2/RNA binding experiment, a 100-fold difference in binding between round 0 and round 8-spr was detected, whereas a 3-fold difference was observed in the absence of any competitor. Thus, in the presence of an appropriate amount of competitor, the binding of selected nucleic acid pools is unaffected, whereas the binding 33 O of round 0 nucleic acid is reduced substantially. When competitors are not included in Sstudying the binding of TGF32 to nucleic acid the affinity of nucleic acids selected using the SELEX process can be grossly underestimated. In this regard TGF02 is similar to other "professional" nucleic acid binding proteins restriction enzymes, polymerases, transcription factors, etc.), in that it possesses both a low affinity, nonspecific and a high I affinity, specific nucleic acid binding activity. The difference between these 2 binding I modes can be revealed in the presence of competitors. Competitors are often used in the study of transcription factors. For example, it can be difficult to detect specific binding of a crude extract containing a transcription factor to oligonucleotides representing their cognate site in gel-shift experiments, unless a competitor, such as poly [dI-dC] 0 poly [dIdC], is included in the binding reaction.
Nonspecific binding can involve the binding of multiple proteins per nucleic acid, often at low affinity sites, giving a false appearance of high affinity. A protein can bind at multiple sites on a nucleic acid or protein aggregates may form on a single protein bound to a nucleic acid. TGFP2 is well known to be "sticky". In the absence of a competitor of nonspecific interactions, TGFI32 may form large networks and complexes of nucleic acid and protein involving primarily nonspecific interactions. Gel shift analysis of TGF32, in the absence of competitor, supports these ideas because TGF32 does not form distinct (one to one) complexes with nucleic acid in gels, but instead either remains in the well at the top of a gel or forms smears that may represent large heterogeneous nucleic acid/protein complexes.
Besides the implications for doing binding curves, the nucleic acid -binding properties of TGF32 may have implications for SELEX. For example the high level of nonspecific binding of nucleic acid by TGF32 may have interfered with previous SELEXs by obscuring specific interactions or preventing the isolation of nucleic acid /protein complexes that involved only specific binding interactions. That is, if mixtures of specific and nonspecific nucleic acid interactions exist in nucleic acid/TGFI32 complexes that form, then the selection for specific interactions may be difficult, if the nonspecific interactions are not eliminated. Lack of progress in some previous SELEX experiments may have been due to efficient competition by the large excess (>1012 10 1 4) of low affinity nucleic acids that contain nonspecific binding sites with a smaller number (-10-1000) of high 34 affinity nucleic acids that contain specific binding sites, especially in early rounds of o SELEX.
Use of protein-excess or nucleic acid-excess conditions Given the discussion above, a question arises as to which SELEX conditions are better for a protein, such as TGFp2, protein-excess or nucleic acid-excess. Protein-excess C conditions may tend to encourage nonspecific interactions. However as long as the In competitor/nucleic acid ratio is high enough to eliminate enough nonspecific interactions, but retain specific interactions, this may not be an issue. One advantage to using protein excess is the bound to background ratios are better and background is lower, which would result in better levels of enrichment.
Nucleic acid-excess conditions may not discourage nonspecific interactions because within nucleic acid pools used for SELEX the ratio of nonspecific to specific binding nucleic acids (which is what is most important) would be the same no matter what the nucleic acid concentration is. In addition, nucleic acid-excess would reduce the competitor/nucleic acid ratio which would tend to increase nonspecific interactions. As discussed above the ratio of tRNA to nucleic acid must be at least 100,000 in early rounds of the filter SELEX in order for affinity enrichment to be efficient. This can be technically difficult in early rounds of SELEX when the nucleic acid concentration is typically higher.
One advantage to using excess nucleic acid is that more members of a given sequence would be represented in a pool. However if there had been enough enrichment using a method such as surface plasmon resonance SELEX) prior to filter SELEX there will probably be multiple representatives of a given sequence and this would not be an issue.
Filter SELEX conditions The conditions used at each round of the filter SELEX are shown in Table Multiple conditions (up to 12) were used in each round varying nucleic acid/protein ratios, competitor/nucleic acid ratios, filter washing buffers, and filter washing volumes.
Typically conditions that resulted in the lowest background and a significant bound/background ratio were processed for the next round. Only data for SELEX rounds that were used in the next round are shown in Table The SELEX began by using an amount of tRNA competitor (100,000-fold excess) that was determined in the SELEXreaction to inhibit binding of round 8-spr to TGFP2 by 0 about 60%. SELEX reactions with competitor were done for round 9b through 14i. The Sinclusion of tRNA in round 9 also dramatically reduced binding of round 8 -spr nucleic acid to nitrocellulose filters from -10-15% to The higher the "background" binding is in a SELEX reaction, the lower the maximum possible enrichment. Thus, inclusion of tRNA in the early rounds of the filter SELEX may have had a dual benefit. It not only CI may have eliminated nonspecific binding of TGFp2 to nucleic acid, but also allowed more in enrichment by reducing background. At round 15c lower affinity competitors were no longer effective at reducing binding of nucleic acid and were not used. This is presumably because the nucleic acid pool bound TGFp2 with adequate specificity and affinity.
O 10 Therefore from round 16 to 22, the presumed specific nucleic acids were allowed to compete with each other by using more traditional nucleic acid -excess conditions.
The background increased to unacceptable levels in rounds 15c and 16a. Gel shift partitioning was investigated as an alternative partitioning procedure at this point but did not work. By modifying the washing conditions the background was reduced to 0.2% in round 17a. After round 18b it was possible to do SELEX rounds at protein concentrations below 1 nM and under nucleic acid-excess conditions. It was also found that nucleic acid concentrations above 1-5 nM helped to reduce background in some rounds.
In summary, during the filter SELEX, the concentration of the protein was reduced 30,000-fold, from 300 nM in round 9b to 10 pM in round 22a. The background binding to filters was reduced from 10% to Nucleic acid pools that bound to TGFp2 only when protein-excess conditions (-100 protein/1 nucleic acid) were used were selected to bind under high nucleic acid/protein (>100/1) or competitor/nucleic acid (>10 7 /1)-excess conditions.
Binding of nucleic acid pools from filter SELEX The binding of TGFp2 to selected nucleic acid pools improved steadily, but slowly and erratically. There was an improvement in the binding of round 10b (Kd--100 nM) compared to the starting pool (round 8-spr; Kd=-500 nM). The affinity of round 1 la was the same as 10b and that of round 12d improved modestly to -40 nM. Rounds 13i and bound TGFp2 approximately the same (Kd-30), while round 14i may have bound worse Round 16a nucleic acid (Kd=-10 nM) bound slightly better than round There was -2-fold improvement in affinity of the nucleic acid pool from rounds 16a 36 O to 17a (Kd=-5 nM). The K, of round 18a nucleic acid nM) was equivalent to round o 17a nucleic acid. There was another slight increase in affinity from round 18b to 19a 2 3 nM). The affinity of rounds 20a, 21a, and 22a plateaued at about 1 nM. The SELEX was stopped at round 22a because the bound to background ratio was below 2 and it would have been technically difficult to reduce the protein concentration to 1-3 pM in C round 23.
ti In summary the Kd improved from -500 nM in round 9b to -1 nM in round 21a, resulting in an overall improvement of -500-fold in the filter SELEX and >10,000-fold in the entire SELEX. The average improvement per round was about 1.6-fold. This rate of improvement is slow compared to an average SELEX experiment, which may take rounds using only surface plasmon resonance technology or -10 rounds using only filter partitioning.
Inhibition of bioactivity by nucleic acid pools Rounds 0, 9-spr, 13-rm, 14i, 18b, 19a, and latency-associated protein (LAP) were tested on mink lung epithelial cells for their ability to reverse TGFp2-mediated inhibition of 3 H-thymidine incorporation. The results are that the K, of the round 9-spr pool was about 711 nM. The K,.s of the round 14i, 18b, 19a and 21a pools were about 231 nM, 309 nM, 154 nM and 10 nM, respectively. The of LAP was about 0.5 nM.
From these results it can be concluded that inhibitors of TGFp2 were enriched in the later rounds of the TGFP2 SELEX. In addition, there is a continuous correlation between the affinity measured in vitro and the inhibitory activity measured in vivo: LAP round 19A round 14i round 13-rm round 8-spr round 0.
Sequencing of nucleic acid ligand clones isolated from filter SELEX Based on several criteria (pool Kd, filter-binding background, bound to noise background, inhibitory activity in cell assay, and absence of aberrant products during the RT-PCR steps of SELEX) round 21a was subcloned for sequence analysis. Forty eight clones were sequenced from round 21a. Two unique sequences represented by clones 21a- 4 (SEQ ID NO:86) and 21a-21 (SEQ ID NO:87) (the first number refers to the SELEX round a clone was initially isolated from and the second number is a clone number) were identified (Table Several clones were minor variants (1-6 bases different) of clones 21a-4 and 21a-21. One hundred more clones were screened by PCR using primers specific 37 0 for clones 21a-4 and 21a-21. Of these, 90 were clone 21a-21-like, 9 were clone 21a-4-like, O and 1 was a third unique sequence (21a-48), which was shown to be a nitrocellulose filterbinding sequence. In conclusion, round 21 a consists almost entirely of two sequences and variations of those sequences. This was not surprising because round 21 a was the second to last round and the bulk affinity of the nucleic acid pools had not improved much from C, round 19a to 21a.
In Since the sequence diversity of round 21a was restricted, 3 other rounds (14i, 16a and 18a) were also sequenced. Only one more novel sequence (14i-1 (SEQ ID NO:72) and variants) was isolated. Two filter-binding sequences were also isolated (16a-l and a variant of 21a-48). Therefore, as with rounds 8-spr, 13-rm, and 21a, these 3 rounds also did not contain diverse TGFp2-binding nucleic acid ligands.
The sequences of 14i-1, 21a-4, and 21a-21 are shown in Table 8. The affinity of the sequences for human TGFp2 is about 10 nM, 3 nM and 1 nM respectively. Therefore, these 3 sequences are ligands that bind human TGFp2 with high affinity.
The ligands were tested for inhibitory bioactivity. The K, of 14i-1, 21a-4, and 21a- 21 are about 200 nM, 30 nM and 10 nM respectively. Thus these ligands are also inhibitory ligands. As for the pools the binding affinity correlates well with the inhibitory activity. This is not surprising since it is likely the TGFp2 ligands bind near the heparin binding site which is very close to the TGFI receptor binding region. The inhibitory activity of ligand 21a-21 was also compared to that of antibodies.
Clones were isolated and sequenced from six rounds (8-spr, 13-rm, 14i, 16a, 18a, and 21a). The number of each type of sequence is summarized in Table 9. Out of 264 clones analyzed by sequencing and 100 clones analyzed by a PCR-based analysis using ligand-specific primers (Table 10), only 3 different TGFp2 ligand sequences (and minor variants) were obtained. Fifteen sequences were filter binding sequences and 36 were nucleic acids that do not bind well to filters or TGFp2. The degree of restriction in sequence diversity observed in this SELEX is very unusual. Generally one can isolate dozens of different nucleic acid ligands and usually it is possible to find high affinity rounds were one ligand represents <10% of the population.
Since sequencing and screening of 6 rounds of SELEX that are as much as 13 rounds apart did not result in a diverse set of sequences the properties of the pools were 38 investigated further to determine where the sequence diversity was restricted. Selected Snucleic acid pools were sequenced and semi-quantitative RT-PCR on nucleic acid pools using ligand-specific primers was done. The results are shown in (Table 10). Taken together with the sequencing results, it appears that a restriction in sequence diversity during the SELEX process may have occurred near rounds 6-spr or 7-spr.
Clone 14i-1 is first detectable in round 6-spr, becomes most frequent near round 14i, and decreases in frequency in later rounds. Clone 21a-4 is first detectable by sequencing in round 14i, is most abundant in round 16a, and decreases in frequency by round 21a. However 21a-4 may exist in prior rounds. (RT-PCR analysis of pools using a primer specific for clone 21a-4 was not done.) Clone 21a-21 was rare in round 14i (<1/104 clones by sequencing; estimate <1/200-500 clones by RT-PCR), became more frequent in round 16a, and composes most of round 18b and 21a.
It appears the surface plasmon resonance biosensor SELEX resulted in a high degree of diversity restriction, which has been observed before using this technology. The reason why various later rounds would consist of virtually one sequence is not clear.
Perhaps only a very small number of sequences bind TGFP2 under the selection conditions used. Perhaps a change in selection conditions such as the inclusion of competitors at round 9 or the switch from protein-excess binding reactions to nucleic acid-excess binding reactions at round 16 resulted in the emergence of clone 21a-21 as the predominant clone by round 21a. It seems as though the selection pressures were significant because the predominant ligand in a pool changed in as few as 2 rounds.
The pattern of changes in the population of nucleic acid ligands can be explained by analogy to the theory of natural selection. In an early SELEX round, a variety of sequences will exist. Strong selective pressure may narrow the sequence variation considerably, to the point that a single sequence is predominant. However rare ligands still exist that can be selected in future rounds or during significant changes in selective pressure. This is true in any SELEX experiment, but the TGFP2 SELEX experiment described here may be an extreme example. In spite of the restriction on sequence diversity, better binding ligands could eventually be isolated. Note that ligand 21a-21 was first identified by sequencing in round 16a. Thus rare, high affinity nucleic acid ligands may exist even in round 22 that would only become predominant under the correct 39 O selection conditions. One approach for isolating such rare sequences might be to Sspecifically deplete late rounds of SELEX of known sequences by hybrid selection, restriction enzyme digestion of PCR products, site-directed RNase H cleavage of nucleic acid), an approach that this TGFp SELEX is well suited for since essentially only different sequences (3 ligands and 2 filter binding sequences) were present in later rounds.
C Isolating a sequence that is present in <1/1000 clones might be easy using depletion tV methods, but would be tedious using sequencing or PCR screening methods.
These results raise questions about when a SELEX is done and how to judge whether it is done. In this SELEX, standard criteria for judging when a SELEX is done such as Kd improvement, and sequencing of clones or bulk nucleic acid pools may not be good criteria for judging if the SELEX had proceeded as far as it could. Often there are technical limitations (background, reaction volumes, loss of low amounts of protein to large surfaces) that determine when a SELEX must be terminated and these are artificially limiting. Perhaps a "depletion SELEX" round should be done at the end of every SELEX to attempt enrichment of ligands that would be difficult to isolate by currently used methods.
Specificity of human TGFp2 ligands For nucleic acid ligands to be most useful in the applications claimed herein they should be highly specific for a particular subtype of TGFp. The specificity of human TGFp2 ligands was investigated by in several ways as discussed below.
The specificity of TGFp2 ligands was examined using the cell culture bioactivity assay where the specificity of the TGFp2 (described here) and TGFP 1 (see U.S. Patent Application Serial No. 09/046,247, filed March 23, 1998, entitled "High Affinity TGFp Nucleic Acid Ligands and Inhibitors," which is incorporated herein by reference in its entirety) aptamers was compared to the specificity of antibodies. Two types of antibodies were used namely, monoclonal antibodies and immunopurified polyclonal antibodies. It was found (Figure 6) that the TGFp2 ligand NX22283 (SEQ ID NO: 114) inhibited TGFp2 protein bioactivity (KI.=10 nM), but not TGFl1 (K,.>1000 nM) or TGFp3 bioactivity (K,.>1000 nM). The TGFp2 ligand NX22283 inhibits the TGFp2 bioactivity with a potency equivalent to that of a monoclonal antibody while the most o potent inhibitor of TGF2 bioactivity in this experiment was an affinity-purified O polyclonal antibody.
The specificity of a TGFp2 ligand for TGFp2 compared to TGFp3 was also analyzed in nucleic acid binding assays. The affinity of round 0 40N7 nucleic acid or the full-length TGFp2 ligand 21a-21 to human TGFp2 protein was >10 .M or 1 nM, rC respectively. The affinity of round 0 nucleic acid or ligand 21a-21 to human TGFp3 to protein was >10 pM or >30 pM, respectively. Therefore, the TGFp2 ligand does not bind significantly to TGFp3.
It was found that the TGFpl ligand 40-03 (1 (see U.S. Patent Application Serial No. 09/046,247, filed March 23, 1998, entitled "High Affinity TGFp Nucleic Acid Ligands and Inhibitors") bound to TGFp3 although 1000-fold worse. These results indicate there may be one or more amino acids in common between TGFp1 and TGF33 that are not found in TGFp2 so that a TGFP1 ligand can bind TGFpl and TGFp3, but not TGFp2 and so that the TGFp2 ligand 21a-21 binds TGFp2 but not TGFl1 or TGFp3.
Indeed, as shown in Table 12, there are 19 amino acids out of 122 that are found in TGFp2, but not in TGFI1 or TGFp3. Three of these differences (Lys-25, Arg-26, and Lys-94 in TGFp2) are within a putative heparin binding region and may be important for determining the binding specificity ofTGFP ligands.
Truncation of nucleic acid ligands It is desirable to obtain the smallest "truncate" of a full length nucleic acid ligand so that it can be synthesized efficiently at the least cost. The goal of this study was to obtain ligands that are less than half their original length (<35 bases), yet retain about the same affinity as the full length ligand. Several approaches were used to identify truncates of the three TGFp2 ligands.
RNase H and hybrid 2'-OCH 3 RNA/DNA oligonucleotides (5'N7 cleave (SEQ ID NO:19), 3'N7 cleave (SEQ ID NO:20), Table 1) were used to remove the 5' and 3' fixed sequences from 2'-F pyrimidine, 2'-OH purine nucleic acid ligands as described in U.S.
Patent Application Serial No. 09/275,850, filed March 24, 1999, entitled "Truncation SELEX Method," which is hereby incorporated by reference in its entirety.
Second, the "boundaries" of the ligands were identified using a previously described method (Fitzwater and Polisky (1996) Meth in Enzymol 267:275-301).
41 O Boundaries define the 5' and 3 ends of the smallest truncate. However boundary 0 determination does not identify internal deletions that can be made. Also because of the nature of the boundary determination method, if a boundary falls within a run of pyrimidines or is too close to either end, then which nucleotide is the boundary must be determined by other methods generation of ligands beginning or ending with each C1, candidate boundary position followed by analysis of their binding to TGFp2).
A third method used relied on plausible structural motifs to define hypothetical sequence boundaries. Synthetic oligonucleotides corresponding to these boundaries were synthesized and were tested for binding to TGFp2.
A fourth approach for identifying TGFp2 ligand truncates was to look at the location of sequence variations in each ligand. In ligands 21 a-4 and 21a-21 the changes that occurred in sequence variants were distributed randomly throughout their sequences.
However in ligand 14i-1, the sequence changes in variants were highly localized. This implied that the variable region of ligand 14i-1 could tolerate changes without affecting binding and that the whole variable region may be dispensable.
A fifth approach was to make internal deletions based on predicted structures.
Portions of putative bulges, loops, or base pair(s) within predicted stems can be deleted.
The success of this method depends critically on how close the structural model is to the real structure. For 21a-21 the most stable structure was found to be incorrect. Only when a structure closer to the real structure was identified (by using the biased SELEX method) could internal deletions of 21 a-21 successfully be made.
Truncation of ligand 14i-1 Using the RNase H truncation method it was determined that ligand 14i-1 requires the but not the 3' fixed sequence (Table 13). Consistent with this result, when both the 5' and 3' fixed sequences were removed, ligand 14i-1 did not bind TGFp2.
Conventional boundary experiments defined the 3' end of ligand 14i-1 (Figure 7) to be within positions 39-45. In the same experiment we failed to observe a clear boundary at the 5' end of this ligand. Of the 60 sequence variants of ligand 14i-1, 54 have nucleotide changes that occur within the last 16 bases at the 3' end of the selected sequence region. Most of the variants have single base changes, but a few have as many as 6 bases changed. Such changes may or may not affect binding. If they affect binding then that 42 O region is important for binding. More likely, since there are so many changes in so many O clones within that region, those bases are probably not important. It was surmised they may be able to be deleted, possibly along with the adjacent 3' fixed region, without affecting binding. This idea was confirmed by the following two experiments: 1) The binding of 8 sequences that varied within the 16 base region and had different sequence changes was tested. They all bound as well to TGFP2 as the 14i-1 ligand.
t 2) A 38 base long truncate of 14i-1 (14i-lt5-41 (SEQ ID NO:131); Table 13 that lacked the 3' fixed region and the 16 base variable region bound to TGFi2 as well as the full length (71 base long) ligand.
Four sequences that removed additional bases from the ends of 14i-1 beyond those removed in 14i-lt5-41 were made [(14i-lt5-38 (SEQ ID NO:132), 14i-lt5-35 (SEQ ID NO:133), 14i-1(ML-87) (SEQ ID NO:135), and 14i-1(ML-89) (SEQ ID NO:136)]. Also one internal deletion of 14i-lt5-41 was made [14i-1(ML-86) (SEQ ID NO:134)]. None of these bound to TGFP2 (Table 13). Taken as a whole these experiments showed that the boundaries of ligand 14i-1 fall within positions 5-7 at the 5' end and 39-41 at the 3' end.
All these results defined a truncate for ligand 14i-1 that is 38 bases long (Table 13).
Truncation of ligand 21a-4 (SEQ ID NO:86) Using the RNase H truncation method it was determined that ligand 21a-4 requires the but not the 3' fixed sequence (Table 14). When both the 5' and 3' fixed sequences were removed ligand 21 a-4 did not bind TGFP2.
The boundaries of ligand 21a-4 (Figure 7) are at positions U11 in the 5' fixed region and within positions 52-56 on the 3' end, defining a truncate that is between 42-46 bases long (Table This is consistent with RNase H truncation results which show 21a- 4 requires the 5' end, but not the 3' end to bind TGFp2.
By examining hypothetical structures, the boundaries for 21a-4 were predicted to occur at position G12 at the 5' end and position C48 at the 3' end. These positions agree well with the region defined by the boundary method. A 37 base long truncate of 21a-4 (excluding sequences required to initiate transcription), beginning at position 12 and ending at position 48 [(21a-4(ML-110) (SEQ ID NO: 144); Table bound as well to TGFB2 as the full length 21a-4 ligand.
43 C" One sequence that removes 4 additional bases from the 3 end of 21 a-4(ML 110) was made that is called 21a-4(ML- 111) (SEQ ID NO:145). The binding of 21a-4(ML- 111) was reduced 30-fold compared to 21a-4(ML-110). Also three internal deletions of 21a-4(ML-110) were made [21a-4(ML-92 (SEQ ID NO:141), ML-108 (SEQ ID NO:142) and ML-109) (SEQ ID NO:143)]. None of these bound well to TGFP2 (Table 14). A c sequence [21a-4(ML-91) (SEQ ID NO:140)] that added 2 bases to the 3' end of21a- 4(ML 111) did not have any improved binding compared to 21a-4(ML-110). Thus, the smallest truncate ofligand 21a-4 identified, that retains binding, is 42 bases long.
Truncation of ligand 21a-21 (SEQ ID NO:72) Using the RNase H truncation method it was concluded that ligand 21a-21 requires the but not the 5' fixed sequence (Table 11). However, when both the 5' and 3' fixed sequences were removed, ligand 21a-21 bound TGFp2. This seems paradoxical since removal of the 3' end alone eliminates binding. However, the data can be interpreted to mean that the 3' deletion folds in a structure that does not bind to TGFp2, while the truncate that lacks both ends does not fold into a dead end structure. Indeed Mfold structure predictions indicate this may be the case.
The boundaries of ligand 21a-21 (Figure 7) are at position G21 on the 5' end and within positions 50-55 on the 3' end, defining a truncate that is 30-35 bases long. The results are consistent with RNAse H truncation data which shows that 21a-21 requires neither the 5' nor the 3' end to bind TGFp2. The truncate identified by boundaries falls completely within that defined by RNase H truncation.
Synthetic sequences based on putative structures were also tested as summarized in Table 11. Results from these experiments are in agreement with the RNAse H and conventional boundary experiments.
Several additional end truncates and internal deletions of 21a-21(ML-95) were made (Table 11). The 9 end truncates included 21a-21(ML-96), 21a-21(ML-97), 21a- 21(ML-103) 21a-21(ML-104) 21a-21(ML-105), NX22286, NX22301, NX22302 and NX22303. Of these only NX22301, which removes one base at the 5' end, binds as well as 21a-21(ML-95). Internal deletions included 21a-21(ML-99), 21a-21(ML-101), 21a- 21(ML-102), 21a-21(ML-114), 21a-21(ML-115), 21a-21(ML-116), 21a-21(ML-118), 21a- 21(ML-120), 21a-21(ML-122), 21a-21(ML-128), 21a-21(ML-132), 21a-21(ML-134), 21a- 44 21(ML-136) and 21a-21(ML-138). Of these 14 internal deletions, only 21a-21(ML-130) bound about as well as 21 a-21 Three sequences [21a-21(ML-94), NX22283 and NX22285), were made that are longer than 21a-21(ML-95). Of these only NX22285 may have bound (marginally) better than 21a-21(ML-95). Thus, the shortest ligand identified that binds TGFp2 is the 34-mer NX22284.
t NX22283 and NX22284, which are synthetic analogs of the transcribed ligands j 21a-21(ML-94) and 21a-21(ML-95), respectively, bound with identical affinity to TGFp2 (Table 11). The synthetic nucleic acids also have the same inhibitory bioactivity as their S 10 transcribed analogs; on the other hand, the short, 30-base long NX22286 and its transcribed analog 21a-21 (ML-96) do not bind TGFp2 and they do not inhibit TGFp2 bioactivity. Therefore synthetic nucleic acids have the same properties as their transcribed counterparts.
To summarize, truncated 14i-1, 21a-4, and 21a-21 ligands were identified that bind TGFI2 as well as the full length ligands and are 38, 37 (excluding 5 bases added to improve transcription yield), and 32 bases long, respectively. Twenty four sequences were made in an attempt to shorten the NX22284 truncated ligand (8 single base deletions and 16 multiple base deletions). Only 2 of them, (21a-21(ML-130) and NX22301, bind to TGFp2. Therefore, it appears that the sequence and spacing of structural elements in NX22284 must be maintained for binding to occur.
Competition between ligands for binding TGF32 Examination of the ability of different ligands to compete with each other for binding to a protein can indicate whether the ligands bind to a similar (or overlapping) or distinctly different regions on the protein.
The ability of NX22283 (SEQ ID NO:114), a truncate of ligand 21a-21 (SEQ ID NO:87) (Table 11), to compete with 4 ligands (14i-1 (SEQ ID NO:72), 21a-4 (SEQ ID NO:86), 21a-21, and NX22283) was tested. The results were that NX22283 competed best with itself, then with 21a-21, 21a-4, and 14i-1, in decreasing effectiveness. Thus, the ability of NX22283 to compete correlates with how related its sequence is to the sequence of the test ligand. NX22283 is most closely related to itself and competes best with itself.
NX22283 is a truncate of 21a-21, and competes with 21a-21 second best. The sequence of 2, 21a-4 may be distantly related to 21a-21 and NX22283 competes with 21a-4 third best.
The sequence ofligand 14i-1 is not related to NX22283, and NX22283 is least capable of competing with 14i-1.
The concentration range of NX22283 required to inhibit 50 of the binding of the other ligands was 10-fold. Since these differences in the amount of NX22283 it took to C compete off the other ligands can be attributed to differences in their affinity there is Sprobably only one type of binding region for these ligands on TGFp2. However, there may be one or more similar sites per homodimer of TGFp2. If there were two distinct types of nucleic acid binding sites on TGFp2 (as is the case for the HIV-1 gag protein; Lochrie et al. (1997) Nuc. Acids Res. 25:2902-2910) it should take >1000 times as much competitor the difference between the Kd of round 0 nucleic acid and the Kd of NX22283) to compete off a ligand binding at a second distinct site, because presumably a ligand that has high affinity at one site would have low affinity for a distinct site. This was not observed.
Off-rate of NX22283 The half-life for NX22283-TGFP2 complex was measured in 2 experiments to be or 3 minutes. Almost all of the ligand dissociated from TGFp2 in 60-75 minutes.
Although these times may seem short, they are typical of in vitro off-rate measurements for nucleic acid ligands that have been isolated by filter partitioning SELEX.
Example 3. Nucleic acid ligands isolated by the SELEX method using a biased round 0 library A biased SELEX is one in which the sequences in a nucleic acid pool are altered to bias the result toward a certain outcome. The primary goals of a "biased" SELEX are to obtain ligands that have a higher affinity and to determine what the putative secondary structure of a ligand may be. The starting, round 0 nucleic acid library (called 34N7.21a- 21) used for the TGFp2 biased SELEX had the same 5' and 3' fixed regions (5'N7 and 3'N7) as the prior TGFp2 SELEX (Table It was made as a 2'-F pyrimidine, 2'-OH purine nucleic acid. However, as described in Example 1, the random region was 34 bases long. Within the randomized region 62.5% of the nucleotides at each position correspond to the NX22284 (SEQ ID NO:115) sequence. The remaining 37.5% correspond to the O 46 other three nucleotides. Thus each position is mutagenized and the sequence of the pool is 0 biased toward the NX22284 sequence. Selection for ligands that bind to TGFp2 using such a pool should allow variants of NX22284 to be isolated, some of which may not have been present in the original 30N7 round O pool.
The bulk Kd of the round 0 34N7.21a-21 pool was about 870 nM (Table 15) using protein-excess binding conditions. This is at least 10-fold better than for the unbiased round 0 40N7 pool, as would be expected. This round 0 nucleic acid pool also bound under nucleic acid-excess conditions in small scale SELEX type reactions, although poorer than in protein-excess reactions, as would be expected. The progress of the biased SELEX S 10 is shown in Table The conditions used in the biased SELEX and the results are shown in Table A total of 9 rounds were done. Attempts were made to obtain higher affinity ligands by using competitors, starting at round 4. Both yeast tRNA (low affinity) and NX22284 (high affinity) were used as competitors. Both are nonamplifiable during the PCR step of SELEX. The series was done without competitors while the series was done with competitors.
The binding of the nucleic acid pools to TGFP2 was measured for rounds 0 to 8 (Table 15) and found to improve from -870 nM for the round 0 nucleic acid library to -1 nM for the round 5a nucleic acid pool. Competition seemed to have little consistent effect on affinity improvement in this SELEX experiment. Probably competition should have been initiated with NX22284 at round 1. Peak improvement in the pool affinity plateaued in rounds 5, 6 and 7, and 8. Therefore round 5a, the earliest round with the best affinity, was subcloned and sequenced.
Sequences of TGF32 nucleic acid ligands obtained from a biased SELEX.
As shown in Table 16, 25 unique sequences were obtained. One to nine changes from the starting sequence were found. All of the clones were 34 bases long within the selected sequence, consistent with studies (see "Truncation of ligand 21a-21" above) where it was difficult to delete any internal bases.
Covariance between pairs of positions was analyzed by eye and by using the consensus structure matrix program (Davis et al. (1995) Nucleic Acids Research 23:4471- 4479). Covariance was observed between 2 different areas implying the existence of 2 47 1 stems in the structure. The pattern ofcovariance suggests the structural model shown in Figure 8 or a similar variant of that structure some base pairing could occur within the loop). This predicted structure is the third most stable structure predicted by the Mfold program (Zuker (1989) Science 244:48-52). A curious example of possible covariance is observed at positions 15 and 25 in the loop region. A 15 and G25 were observed to covary
C
I to C15 and U25 in 2 clones (#18 and Ligand 21a-4 also has the C/U combination at ¢c€ t the bottom of its putative loop.
Of 34 bases, 11 are "invariant" among these 25 clones (Table 16 All of the invariant positions are predicted to occur in the loop and bulge regions except C34, the last nucleotide. The last base of all 3 truncated TGFI2 ligands (Figure 8) is a C. Removal of this C results in loss of binding. If invariant positions indicate regions where TGF32 binds the NX22284 ligand, then binding may occur primarily in the bulge and stem loop regions. The stems must be base paired, but can vary in sequence implying that the structure of the stems may be more important than their sequence. The stem may be a structure used to present the bulge, loop and C34 nucleotides in the proper orientation to bind TGFp2.
Clone 5a-l 1 from the biased SELEX is similar to clone 21-4 from the primary SELEX, particularly at positions that are invariant in clones from the biased SELEX, thus reinforcing the new structural model and the importance of the invariant positions. It has not been possible to fit ligand 14i-1 into a similar structure. Perhaps it represents a second sequence motif capable of binding TGFp2.
Binding of nucleic acid ligands isolated from the biased SELEX The binding of clones from the biased SELEX was compared to the binding of full length ligand 21a-21. The majority of the clones bound as well to TGFp2 as 21a-21 (Table 16). One clone (#20 (SEQ ID NO:160)) bound about 6-fold worse and one clone (#13 (SEQ ID NO:154)) bound about 5-fold better than full length ligand 21a-21. The average Kd of the clones (weighting clones found more than once) is 1.2 nM, which agrees with the round 5a pool Kd of-1 nM. Thus, the ligands that were isolated in this manner were not vastly different in affinity from the starting sequence.
One would expect there to be an optimal number of changes that results in higher affinity ligands. Clones with only a few changes might be expected to bind about the 1 48 same as the starting sequence, clones with a threshold number of changes may bind better, and clones with too many changes may bind worse. Indeed there may be a correlation between the number of changes and the affinity. Clones with I to 4 changes tend to bind F, the same or worse than ligand 21a-21. Clones with 5-8 changes tend to bind better than ligand 21a-21. The worst binder was the one with the most changes The ligand N, that bound to TGFp2 the best (clone #13) had 7 changes relative to the starting sequence.
l t When the clones that bound better and those that bound worse are aligned (Table 17) it appears that an A at position 5 may be important for higher affinity binding since the ligands that bind to TGFp2 best all have an A at position 5 and all clones with an A at position 5 bind at least as well as 21 a-21. In contrast clones with a U, C or G at position tend to bind worse than 21 a-21. With regard to the pattern of base pair changes in the putative stems there is no single change that correlates with better binding. In addition, the better binders do not consistently have GC-rich stems. However the pattern of changes in the stems of the poor binders does not overlap with that seen in the stems of the better binders. Thus, various stem sequences may result in better binding for subtle reasons.
A point mutant that eliminated binding of the full length 21-21 transcript (21 a- 21(ML-107); Table 11) changes U at position 6 to G. A G was found at position 6 in three clones from the biased SELEX 9 and 35), one of which (clone has only one other base change while the others had additional changes. All three clones from the biased SELEX that have a G at position 6 bind TGFp2. Thus it would seem that the U6G change alone eliminates binding, but this binding defect can be reversed when combined with other sequence changes.
To summarize, some changes (such as A at position 5) may act independently and be able to confer better binding alone, while others changes at position 6 and in the stems) may influence binding in a more unpredictable way that depends on what other changes are also present.
Presumably sequences that lack an "invariant" nucleotide would not bind to TGFP2. Some of the invariant bases have been deleted and others have been changed (Table 11). None of these 10 altered sequences [21a-4(ML-111); 21a-21(ML-96, 97, 101, 102, 103, 104, 105, 120, NX22286] bind to TGFp2.
49 S Example 4. Substitutions of 2'-OH purines with 2'-OCH 3 purines in NX22284 O Substitutions of 2'-OH purines with 2'-OCH, purines sometimes results in nucleic acid ligands that have a longer half life in serum and in animals. Since the nucleic acid S ligands described here are ultimately intended for use as diagnostics, therapeutics, imaging, or histochemical reagents the maximum number of 2'-OH purines that could be CI substituted with 2'-OCH 3 purines in ligand NX22284 was determined. NX22284 is a 34i mer truncate of the 70 base long 21a-21TGFp2 ligand (Table 18). NX22284 has 17 2'-OH S purines and binds about 2-fold worse than ligand 21 a-21.
Initially an all 2'-OCH 3 purine substituted sequence was synthesized (NX22304).
O Another sequence has all 2'-OH purines substituted with 2'-OCH 3 purines except six purines at its 5' end. Neither bound to TGFp2 or had measurable bioactivity (Table 18).
Therefore a set of sequences was synthesized (NX22356-NX22360; Table 18) such that groups of 3 or 4 2'-OH purines were substituted with 2'-OCH 3 purines. The binding of NX22357 was reduced about 2-fold and the bioactivity was reduced The binding and bioactivity ofNX22356, NX22258 and NX22360 were unaffected. In contrast the binding of NX22359 was reduced over 100-fold and its bioactivity was reduced over 30-fold. Therefore, the sequence ofNX22359 was "deconvoluted" one base at a time in order to determine which individual purines in NX22359 cannot be 2'-OCH 3 purines. NX22374, NX22375 and NX22376 are deconvolutions ofNX22359. All three of these sequences had greatly reduced binding and bioactivity. This suggests that G20, A22 and A24 cannot be 2'-OCH 3 purines.
NX22377 was designed to determine if a sequence with an intermediate number of 2'-OCH 3 purines could bind TGFp2 and retain bioactivity. NX22377 has 10 2'-OCH 3 purines out of 17 (representing the 2'-OCH 3 purines in NX22356, NX22357 and NX22360). The binding and bioactivity of NX22377 are identical to NX22284.
NX22417 was designed to test the possibility that G20, A22 and A24 must be 2'-OH purines in order to retain binding and bioactivity. In NX22417 G20, A22, and A24 are 2'-OH purines while the other 14 purines are 2'-OCH 3 NX22417 binds to TGFp2 as well as NX22284, but its bioactivity is reduced about 10 fold. Since substitution of G20 (NX22374) or A24 (NX22376) alone had a less severe effect than substitution of A22 (NX22375), nucleic acids were synthesized that had all 2'-OCH 3 purines except position A22 (see NX22384) or G20 and A22 (see O
C
I NX22383). NX22383 and NX22384 did not bind or inhibit TGF32, again suggesting that at least S3 purines at positions 20, 22, and 24 must be 2'OH to retain binding and bioactivity.
NX22384 was analyzed by mass spectroscopy to ensure its lack of binding and inhibitory CK, activity was not due to incomplete deprotection or an incorrect sequence. The results are that NX22383 may be 0.5- 0.9 daltons more than the predicted molecular weight and therefore is very C likely to be what it should be.
CN Since NX22357 bound to TGFP2 slightly worse than NX22284, but had a
C
reduced bioactivity, it was possible that one or more of the three 2'-OCH 3 purines in O NX22357 (G5, A8 or Al 1) may also be required for bioactivity. This notion was tested by C 0 synthesizing NX22420 and NX22421. NX22421 has all three of these bases (G5, A8, and All) as 2'-OH purines (along with G20, A22 and A24, which require 2'-OH groups).
NX22420 has A8 (along with G20, A22 and A24) as 2'-OH purines. NX22421 has A8 and Al (along with G20, A22 and A24) as 2'-OH purines. A8 was retained as a 2'- OH purine in both NX22420 and NX22421 because it was invariant among the clones from the biased SELEX and therefore it was inferred that A8 might be less tolerant to change at the 2' ribose position (as was the case for G20, A22 and A24). Indeed both NX22420 and NX22421 had approximately the same binding and inhibitory activity as NX22284. In summary, the NX22284 sequence can retain maximal binding and inhibitory activity when four purines (AS, G20, A22 and A24) are 2'-OH and the other purines are 2'- :0 OCH 3 Note that all four of these positions were invariant among the clones isolated using the biased SELEX method.
While studies were being done on substituting the 2'-OH purines of NX22284, two shorter versions of NX22284 (21a-21[ML-130] and 21a-21[ML-134]; Table 11) were discovered that bound well to TGFP2 as transcripts. The 2'-OCH 3 purine substitution pattern of NX22420 was transferred to these sequences. NX22426 is the 2'-OCH 3 purine analog of 21a-21 (ML-134) and NX22427 is the 2'-OCH 3 purine analog of 21a-21(ML- 130). NX22426 bound well to TGFp2, but had 25-fold reduced bioactivity. NX22427 may have slightly better binding and inhibitory activity than NX22284.
In summary, the human TGFP2 ligand isolated by using combined spot, spr, and filter SELEX methods which have the best combination of affinity, short length, and O 51 0 inhibitory activity is NX22427, a 32-mer with 12 2'-OCH 3 purines out of a total of 16 O purines.
Substitutions of 2'-OH purines with 2'-OCH 3 purines in NX22385 Some of the ligands that were isolated using the biased SELEX method clone 13) bound better to TGFp2.
(C7 To compare the properties of a truncated clone 13 to truncated 21 a-21, NX22385 was ¢c€ In synthesized. NX22385 (Table 19) is a 34 base long, 2'-F pyrimidine, 2'-OH purine version of biased SELEX clone #13. It binds about 2.5-fold better than NX22284, the corresponding 34 base long truncate of 21 a-21, but its inhibitory activity is about 4-fold worse.
0 10 For reasons mentioned in the previous section it was of interest to determine if the properties of a truncated clone 13 t when synthesized as a 2'-F pyrimidine, 2'-OCH 3 purine nucleic acid. Two 2'-OCH 3 purine versions of NX22385 (NX22424 and NX22425; Table 19) were synthesized based on the 2'-OCH 3 pattern of NX22420, a truncate of 21a-21. In both nucleic acids A8, G20, A22 and A24 were retained as 2'-OH purines, as in NX22420. In NX22424, the purines that are unique to clone 13 (A5, A6 and G12) are 2'-OH purines. In NX22425, those purines are 2'-OCH 3 purines. Analogs of NX22424 and NX22425 were also synthesized in which A24 (NX22386) or G20 and A24 (NX22387) are 2'-OCH 3 purines. NX22386 and NX22387 were expected to serve as negative controls since 2'-OCH 3 G20 or A24 version of NX22284 were inactive. As expected NX22386 and NX22387 did not bind or inhibit TGFp2. NX22424 and NX22425 bound to TGFp2 as well as NX22284, but were reduced >100-fold in bioactivity Table 19). Therefore, while other sequences that bind as well as NX22284 were isolated, no other sequence was identified that have better bioactivity.
Example 5. Pharmacokinetic properties ofNX22323 NX22323 is a 5'-polyethylene glycol-modified version of NX22284 (see Table 11; Figure The plasma concentrations of NX22323 were measured in rats over a 48 hour time period and are shown in Figure 11 with the corresponding pharmacokinetic parameters in Tables 20 and 21. These data demonstrate biphasic clearance ofNX22323 from plasma with an initial clearance half life (a of 1 hour and a terminal clearance half life (p of 8 hours. The volume of distribution at steady state was approximately 140 mL/kg suggesting only minor distribution of the aptamer with the majority remaining O 52 in plasma and extracellular water. The clearance rate determined by compartmental 0 analysis was 0.40 mL/(min*kg). This value was consistent with other aptamers with similar chemical composition (5'-PEG 40K, dT, 2'F pyrimidine, 2'-OH purine nucleic acid). These data support daily administration of NX22323 for efficacy evaluation.
Example 6. 2' OMe Modification of Lead Truncate Ligand STGFl1 nucleic acid ligands are disclosed in U.S. Patent Application Serial No.
09/275,850, filed March 24, 1999, entitled "Truncation SELEX Method," which is incorporated herein by reference in its entirety. A lead aptamer was generated by truncation SELEX by hybridization (see Table 11, Family 4, Ligand #70 in U.S. Patent Application Serial No. 09/275,850), herein called CD70. CD70 derivative oligonucleotides were synthesized containing 2' OMe modifications at various positions as summarized in Table 22. The results suggest that 13 out of 16 purines can be substituted with their 2'OMe counterparts without any loss of activity. The molecule with the maximum 2'OMe modifications (CD70-ml3) is also bioactive (Table 22). Figure shows a putative structure of CD70-m 13 (SEQ ID NO:206) and the positions of that require the presence of 2'OH nucleotides. Of interest is the A position at the 3' end of the molecule which according to the proposed structure does not participate in a secondary structure. Deletion of this single stranded A affects somewhat the binding activity of the molecule but it completely eliminates its bioactivity (Table 22). The 2'OH bases and the 3' final A are in close proximity in the proposed structure. This suggests a domain of the molecule responsible for target binding. Under these circumstances, it is expected that the loop shown at the top of the proposed structure (Figure 10) may not be necessary for binding. This was confirmed by replacing such a loop with a PEG linker and showing that such modified molecules retain binding (Table 22). The PEG linker was conjugated to the aptamer as shown in U.S. Patent Application Serial No. 08/991,743, filed December 16, 1997, entitled "Platelet Derived Growth Factor (PDGF) Nucleic Acid Ligand Complexes," which is hereby incorporated by reference in its entirety. The shortest binding aptamer identified from these experiments is CD70-m22 (SEQ ID NO:215), a 34-mer (including the PEG linker).
53 O Example 7. Mink Lung Epithelial Cell (MLEC) PAI Luciferase Assay 0 The MLEC PAI Luciferase Assay was performed in order to test the ability of Q TGFp aptamers to interfere with the biological activity of TGFP in vitro. Mink Lung Epithelial Cells were transfected with a PAI/Luciferase construct that allows for the direct measurement of PAI promoter upregulation by TGFp. Both TGFpl and TGFi2 can C upregulate the PAI promoter. Stimulation of PAI/Luciferase expression results in a it quantifiable light emission when the Luciferase substrate Luciferin is present. MLEC- PAI-Luc cells between P6,, were plated at 3x10 4 cells per well in MEM supplemented with 10 mM HEPES and 0.2% FBS (MEM-S), and allowed to adhere for 4-5 hours. Serial dilutions of inhibitors (antibodies or aptamers) were prepared in MEM-S for incubation with MLEC. Two columns of wells were maintained for each TGFp and untreated control groups. TGFI(1 or 2) was added to each well, except for the untreated control cells, to 10 pM or 20 pM and the cells were incubated for 15-16 hours. Following TGFp stimulation, MEM-S was replaced with DPBS supplemented with Ca 2 and Mg 2 at 1 mM each. Cells were processed with the Luc-lite kit (Packard Instruments) per manufacturer's instructions. Relative luciferase activity was determined by chemiluminescence detection (Top Count, Packard Instruments) after a 10 minute dark adaptation. Data were generated as CPS (counts per second). The results are set forth in Figures 12 and 13. It is clear from these figures that both TGFp aptamers interfere with the biological activity of their respective cytokines. The apparent Ki in these experiments is about 200 nM.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in Australia.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
54 TABLE 1. Sequences used during SELEX.
(all are shown in a 5' to 3' direction, and separated by a blank every 10 bases) Sequences involved in SELEX process: C1 DNA template for round 0 of spot SELEX) TCGGGCGAGT CGTCTGNNNN NNNNNNNNNN NNt4NNNNNNN NNNNNNfNNNN NNNNNNCCGC ATCGTCCTCC C 71 (SEQ ID NO: 1) A=dA; C=dC; G=dG; T=dT; N+25% each of dA, dC, dG, or dT (5'N7; primer used in PCR steps of SELEX) c-i TAATACQACT CACTATAGGG AGQACGATQjC GG 32 (SEQ ID NO: 2) A=dA; C=dC; G=dG; T=dT c-i (3'N7; primer used in RT and PCR steps of SELEX) TCGGGCQAGT CGTCTG 16 (SEQ ID NO: 3) A=dA; C=dC; G=dG; T=dT (Transcription template for round 0 of spot SELEX) (SEQ ID NO:4) (SEQ ID NO: AdA; CdC; G=dG; T=dT; N=25% each of dA, dC, dG, or dT (RO 40N7; nucleic acid library for round 0 of spot SELEX) GGGAGGACGA UGCGGNNNN NNNNNNNNNN NNNNNNNNN NNNrNNNNNNN NNNNNCAGAC GACUCGCCCG A 71 (SEQ ID NO: 6) A=2'-OH A; C=2'-F C; G=2'-OH G; N=25 each of 2'-OH A, 2'-F C, 2'-OH G, and 2'-F U; U=2'-F U (34N7.21a-21 DNA template for round 0 of biased GGGAGGACGA TGCGGNNNNN NNNNNNNNNN NNNNNNNNNN NNNNNNNNNC AGACGACTCG CCCGA 65 (SEQ ID NO: 7) A=dA; C=dC; G=dG; T=dT, N=62.5 NX22284 sequence as DNA and 12.5% of the other 4 nucleotides (dA, dC, dG, or dT) at each position (Transcription template for round 0 of biased SELEX) TAATACGACTCACTATAGGGAGGACGATGCGG-34N-CAGACGACTCGCCCGA (SEQ ID NO: 8) ATTATGCTGAGTGATATCCCTCCTGCTACGCC-34N-GTCTGCTGAGCGGGCT (SEQ ID NO: 9) A~dA; C=dC; G=dG; T=dT, N=62.5 NX22284 sequence as DNA and 12.5% of the other 4 nucleotides (dA, dC, dG, or dT) at each position TABLE 1. (Conutinued) (34N7.21a-21 nucleic acid library for round 0, biased GGGAGGACGA UGCGGNNNNN NNNNNN[NP'JN NNNNNNNNNN NNNNNNNNNC AGACGACUCG CCCGA 65 (SEQ ID NO: A=2'-OH A; C=2'-F C; G=2'-OH G; N=62.5 NX22284 sequence and 12.5% of other 4 nucleotides (2'-OH A, 2'-F C, 2'-OH G, or 2'-F U) at each position; U=2'-F U Sequences used for subcloning. screening. sequencing ligand ri (ML-34; used for subcloning) CGCAGGATCC TAATACGACT CACTATA 27 (SEQ ID NO: 11) AdA; C=dC; GdG; T=dT (ML-78; used for subcloning) GGCAGAATTC TCATCTACTT AGTCGGGCGA GTCGTCTG (SEQ ID NO: 12) A=dA; C=dC; G=dG; T=dT (RSP1 vector-specific primer used to screen transformants for ligand inserts) AGCGGATAAC AATTTCACAC AGG 23 (SEQ ID NO: 13) A=dA; C=dC; G=dG; TdT (FSP2; vector-specific primer used to screen transformants for ligand inserts) GTGCTGCAAG GCGATTfAAGT TGG 23 (SEQ ID NO: 14) A=dA; C=dC; G=dG; TdT (RSP2; primer for sequencing ligands) ACTTTATGCT TCCGGCTCG 19 (SEQ ID NO: A=dA; C=dC; G=dG; T=dT Sequences used to detect specific ligands (ligand 14i-1 specific primer; GCCAAATGCC GAGAGAACG 19 (SEQ ID NO: 16) A=dA; C=dC; GdG; TdT (ligand 21a-4 specific primer; ML-79) GGGGACAAGC GGACTTAG 18 (SEQ ID NO: 17) A=dA; C=dC; GdG; TdT (ligand 21a-21 specific primer; ML-81) GGGAGTACAG CTATACAG 18 (SEQ ID NO: 18) A=dA; C=dC; GdG; TdT 56 TABLE 1. (Continued) Seciuences used for RNAse H cleavage (5'N7 cleave) C1 CCGCaugcuc cuccc 15 (SEQ ID NO: 19) a=2'-OCH 3 A; c=2'-OCH 3 C; C=dC; g=2'-OCH 3 G; G=dG; u=2'-OCH 3
U
(3'N7 cleave) In ucgggcgagu cgTCTG 16 (SEQ ID NO: a=2'-OCH 3 A; c=2-OCH 3 C; CdC; g=2-OCH 3 G; G=dG; u=2'-OCH 3 U; T=dT 2004242532 24 Dec 2004 Table 2. Conditions and results of filter SELEX Rounda 9b 11a 1 2d 13i 14i 1 6a 1 7a 1 8b 1 9a 21a 22a rRNAlb. nM I nM 1 nM I nM 0.2 nM 0.4 nM 0.1 nM 10 nM 55 nM 30OnM 15 nM 7 nM 0.33 nM 0.63 nM 0.07 nM rTGFQ2], nM 100 nM 30 nM 30 nM 20 nM 10 nM 10 nM 10 nM 10 nM 3 nM 3 nM 0.1 nM 0.03 nM 0.03 nM 0.01 nM RNAb/rotein 0.01 0.03 0.03 0.01 0.04 0.01 1.0 5.5 10 5 70 11 21 7 [Competitor] 100 iM tRNA 100 g~M tRNA 100 g.M tRNA 250 pM tRNA 10jiM tRNA 10 iM heparin 0 0 0 0 0 0 0 0 Bound 4.2 4.3 1.5 2.2 2.6 14.5 8.8 9.6 1.9 2.3 0.17 0.1 0.3 0.12 Background 1.1 0.13 0.2 0.3 0.16 0.55 2.2 2.1 0.17 0.6 0.05 0.04 0.1 0.09 Bound/Background 4 33 8 7 16 20 4 5 11 4 3 3 3 Kd (nM) nd 100 2 'Number designates the round of SELEX and letter designates the condition used for that round.
'NA, nucleic acid library Only those rounds that were carried to the next round are shown Table 3. Conditions and results of Spot SELEX Rd Protein RNA (pmoles) (pmoles) 1 *200 2000 2 *200 1500 3 *200 1500 4 200 1000 *67 1000 22 1000 67 100 *22 100 7.3 100 6 22 50 *7.3 50 2.4 50 7 22 7 *7.3 7 2.4 7 8 *7.3 3 2.4 3 0.7 3 9 *7.3 1 2.4 1 0.7 1 7.3 <1I(no tRNA) 7.3 <1 (10' tR.NA)' 7.3 <1 (10' tRNA) 7.3 <1 (10' tRNA) 2 2 2 2 2 2 2 2 2 2 2 3 2 Washes' Signal/ (g.l/m in) Noise Z~ (500/10) 4.90 (1000/10) 1.80 (1000/10) 5.50 (1000/10) 11.20 (1000/10) 3.70 (1000/10) 1.58 (1000/20) 26.00 (1000/20) 11.00 (1000/20) 2.70 (1000/20) 20.70 (1000/20) 4.00 ~(1000/20) 1.20 (1000/50) 24.00 (1000/50) 7.50 (1000/50) 1.50 (1000/60) 77.00 ~(1000/60) 8.50 ~(1000/60) 1.00 ~(1000/20) 87.00 ~(1000/20) 4.00 ~(1000/20) 2.50 (1000/20) 13.70 Input Incubation
ND'
ND
ND
0.18 0.06 0.03 1.30 0.56 0.10 1.00 0.20 0.06 1.30 0.40 0.07 0.41 0.04
ND
0.23 0.01 0.006
ND
4 hrs, 20'C 0.5 his, 37 0
C
I hi, 37-C 1 hi, 37-C 1 hi, 37TC 1 hi, 37 0
C
I hi, 37-C 1 hr, 37-C 1 hr, 37-C I hi, 37 0
C
I hi, 37-C 1 hi, 37-C 1 hi, 37-C I hi, 37-C I hi, 37-C 0.75 hi, 37-C 0.75 hi, 37 0
C
0.75 hi, 37-C 1 hi, 37-C 1 hi, 37 0
C
1 hi, 37-C 0.5 hr, 37 0
C
Pre-adsorb 2 No 5 layers, 0.75his 5 layers, 1 hr 5 layers, 2.5 hrs 5 layers, 2.5 his 5 layers, 2.5 his 10 layers, 0.75 his 10 layers, 0.75hrs 10 layers, 0.75 his 10 layers, 0.75 his 10 layers, 0.75his 10 layers, 0.75his 10 layers, 10 layers, 10 layers, 1. 10 layers, 10 layers, 10 layers, 10 layers, 10 layers, 1 10 layers, 10 layers, 2 (1000/20) 2 (1000/20) 2 (1000/20) 10.50 5.00 1.80 ND 0.5 hi, 37'C 10 layers, ND 0.5 hi, 37'C 10 layers, 1.Shrs ND 0.5 hi, 37'C 10 layers, pool carried to next round 'Number of washes, volumes and duration 2 Number of filters and duration of incubation during the background counterselection step 'ND, not determined 'Fold excess tRNA over the aptamer pool STable 4. Conditions and results surface plasmon resonance biosensor (spr) SELEX.
1- Progress of BIA SELEX with TGFp32 C' Rd TGFp2, RU' [RNA], Injections Fractions Fraction RU after uM 2 (vol, pL) 3 (min FW 5
SDS
6 each) 4 SFCI FC2 FC3 FC4 S2 1293 874 294 0 4 4(40) 3(5) 3rd SDS -100 S3 1176 1178 1181 0 15 4(40) 3(5) 3rd SDS -50-100 S4 3010 2037 1767 0 10 6(40) 3(5) 3rd SDS 5520 5334 4265 0 5 6(40) 3(5) 3rd& SDS -100-150 6 4075 3143 298 0 5 6(40) 3 3rd SDS -75-100 7 3773 2616 2364 0 2 6(40) 3(5) 3rd SDS -330-220 8 2574 1842 1461 0 5 4(40) 3(5) 3rd& SDS -60-105 9 3180 2029 1688 0 3 4(40) 3(5) 3rd SDS -77-114 344 718 1692 0 1 4(40) 6(10) 6th& SDS 11 217 675 386 0 5 2(40) 6(10) 6th SDS -50-62 'Amount of TGFp2 immobilized expressed in resonance units where 1RU corresponds to I pg of protein per mm 2 The protein is immobilized in an area of 1.2 mm 2 2 concentration of RNA pools 3 Number of injections and volume of each injection 4 Number and length in min (in parentheses) of each fraction 'Fractions carried to the next round 6 Amount of RNA eluted after SDS treatment expressed in response units FC1, FC2, FC3, and FC4 designate the four flowcells of the BIA chip.
2004242532 24 Dec 2004 Table 5. Sequences isolated from round 8 of surface plasmon resonance SELEX NAMEa SEQ
ID
NO:
8.1(1) 21 8.2(1) 22 8.3(14) 23 8.5(1) 24 8.6(1) 25 8.8 26 8-9(4) 27 8.11(1) 28 8.12(1) 29 8.13(1) 30 8.15(2) 31 8.18 32 8.20(1) 33 8.21(1) 34 8.22(1) 35 8.23(1) 36 8.24(1) 37 8.25(1) 38 8.26(1) 39 8.28(1) 40 8.29(1) 41 8.31(1) 42 8.33(1) 43 8.34 44 8.35(1) 45 8.36(1) 46 8.38 47 SEQUENCEb GGGAGGACGAtJGCGG UCCUCAAUG-AtJCUU
UCCUGUUUAUGCUCCC
GGGAGGACGAtJGCGG
AAGUAACGUUUAAGUAAAAUUCGUUCUCUCGGUAUUUGGC
GGGAGGACGAtJGCGG
AAGUAAGGUUGAAGUAAAAUUCGUUCUCUCGGCAUUUGGC
GGGAGGACGAUGCGG
UCCUAACCAUCACAAEJCUCAAUUCUUAUAUUUUCCCGCCC
GGGAGGACGAUGCGG
AAACCAAAAGACCACAUCUCCAUACUCACGCUCUGCCC
GGGAGGACGAUGCGG
AUAGAUCGGUCCGAUAAGUCUUUCAUCUUUACCUGGCCCC
GGGAGGACGAUGCGG
AAGUAACGUUGAAGUAAAAUUCGUUCUCUCGGUAUUUGGC
GGGAGGACGAUGCGG ACGAUCCUUtJCCUUAACAUUUCAUCAUUUCUCGUGUGCCC GGGAGGACGAUGCGG
UCCAUCAACAAUCUUAUGAUUAUGUUUUUCCUUCCCGCCC
GGGAGGACGAUGCGG
UCCUCUGAGCCGAUCUUGUUCACUACUUCUUUUUCUGCCC
GGGAGGACGAUGCGG
UUCCUCAAUUCUUCCAUCUUCAUAAUGUUUCCCUUUGCCC
GGGAGGACGAUGCGG UCUACCCUUUAGCAGtJAUUUGUUUCCAUCGUUGUUUGCCC GGGAGGACGAUGCGG UCUCAACGAAGAACAtJCGUUGGAUACUGUUUGUCCCGCCC GGGAGGACGAUGCGG
UUCAGUUUGCUUCAGUUUUCGUUUCUAAUUCUUGUGUCGC
GGGAGGACGAUGCGG------------
AGCGGAUUAAUUAGUCJGACEJUCUUGUCCC
GGGAGGACGAUGCGG
AGACAUCUUUGUCUCGAUUAGUCAUGUUCCUUACGUGCCC
GGGAGGACGAUGGGG
UCCUCUAGCAAGCAGCUUCUCAUCUUAUUUUUCCGCCC
GGGAGGACGAUGCGG UGCACAGUGAtJGGAUGACAUUGUAUAACGGUAUGCGUCCC GGGAGGACGAUGCGG
-ACCUAUCUUUCUEJCCAAGUCAUAGUUUUACUUCCCGCCC
GGGAGGACGAUGCGG
AUGAGACCUAAUCAUCGAUCCGCUAUCUAMAACCUCACCC
GGGAGGACGAUGCGG UCCUCAGACAAAUCUUrJCUUGAAUCUUUCCUUAACUGCCC GGGAGGACGAUGCGG
-ACCGAEUCUCCAACUUGACAUUUAUUCCUCUUUCUGCCC
GGGAGGACGAEJGCGG UCCUCUGAGCCAAUC.JtCUtCGCUACUUCUUUUUCUGCCC GGGAGGACGAUGCGG AUUGUUUCUCCAACGCUUUtJCACUACCUACAUUUCUGCCC GGGAGGACGAUGCGG
AUCCUAUCCU~CUGAAUAUCAUUAAAUCAUCUUCUCCGCCC
GGGAGGACGAUGCGG
UUCAAUCAUCUUCACUCU-CAUUUCCUUUUUCCUACUCCC
GGGAGGACGAU.GCGG
CGAUAGAAUCUAGUCGUUCUAGAUGAUCUGGUACGUGCCC
BINDrNGe CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA TGFP32 CAGACGACUCGCCCGA TGFP2 CAGACGACUCGCCCGA
NONE
CAGACGACUCGCCCGA
NONE
CAGACGACUCGCCCGA
NONE
CAGACGAGUCGCCCGA TGFP32 CAGACGACUGGCCCGG
FILTER
CAGAGGACtJCGCCCGA
NONE
CAGACGACUGGCCCGA
FILTER
CAGACGACEJCGCCCGA
FILTER
CAGACGACUCGCCCGG NONE a CAGACGACtJCGCCCGA
NONE
CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
NONE
CAGACGACUCGCCCGA
CAGACGACEJCGCCCGA
CAGACGACEJCGCCCGA FI LTER CAGACGACUCGCCCGA
NONE
CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA
NONE
CAGACGACUCGCCCGA
FILTER
CAGACGACUCGCCCGA
2004242532 24 Dec 2004 Table 5. (Continued) 8.39(1 8.40(1 8.41 (1) 8.45(1) 8.46(1 8.47 (1) 8.48 (2) 8.49 (1 8.51 (1) 8.52 (1) 8.56(1) 8.57 (1) 8.61 (1) 8.62(1) 8.64(1) 8.65 (1) 8.69M1 8.71M(1 8.72M1 8.74 (1) 8.75(1) 8.76(1) 8.79(1) 8.80(1) 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
UAGUAAU CC UUGUCUU CCAtJUUUC UCU UUACCCU tJUUGCCC CCCAUUAGUCCUCAUUAGU CCCCUGUGCCC
CAUCUUAUCCUCCAUCAGUUACUCUUCGUUAUUCCCGCCC
UCC-AAAUCCUCUUCCCAUGUUAGCAUUCAGCCUUGUCCC
UUCCGACAAUUUCCUCCACCAUUAGAUUUCUUGCU4GCCC
UCUUGAUCCUCCUUUGLJG(CUUUCUUUGUCUUCCCUGCCC
AAGUAACGUUGAAGUAAAAUUCGUUCUCUCGGUAUU-GGC
-UCCGAUCAGUUCCUUCGAUUAAUCUUCUUUCCUGCCCCC
AAUCCUUCUCCCUGAUGAAUAUGACCUUUUUCUUGCUCCC
AUGAUCUUUAAUGUCUGGUUUGAGGUCAAUGCGGGUGCCC
AGAUGGUACUCCAUCUCCUUUAUGUGCCCAUCGCUGUCCC
UCCUC-GAUUCU AAUUUACUCCUUUUUCCCC
UCUACCCUUUAGCAGUAUUUGUUUCCAUCGUUGUUUGCCC
-CACAAUAUUCUCCUCUACUUCCACGUAUUUUCCUGUCCC
UCCUCAACCUUAGACUUUCAUUUCUUCAGUUCUUCUGCCC
UAGUGGUCUGUCAAAGGAAUAGCUAGUAGUGUUUGGUCCC
CAUCUUCCUUAGCAUACCAGUUUAUUCCUUUCCCUGUCCC
AGCGACAGUAUAGUUAGUACUCUAGCUC UAGUGCUGUCCC
ACCUCUCAUGAUCAGCAUCUCGCGUAAUCACGGUUCACCC
UCCGUACUCCAEJUUCCUAUUUGAUUCCUUUUCCUCUGCCC
AACCCACGACCUUACCUUAAUCAUGUAUUUCUCUCUGCCC
AGAUAAUGAGUGACGGUGAUUAUAGAUGCUGCCC
UUCCUCAAUUCUUCCAUCUUCAUAAUGUUUCCCUUUGCCC
UUCCU UCCAACGUUAUCUACUUUCU GCCC CAGAGGACUC GCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
FILTER
NONE
TGFP2 'am~es are given in the form Round 8.clone number followed by the number of clones of that sequence that were isolated in parentheses.
gaps introduced to designate sequences with selected regions that are shorter than 40 bases. An attempt was made to align such sequences with other sequences but the alignment is not necessarily optimal.
Underlined bases are those that differ from the ligand 1 4i- 1 (Table A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U.
'FILTER, filter-binding sequence; NONE, no binding to TGFI32 or filters, TGFP32, binds to TGFP2 as well as ligand 14i-1I STable 6. Conditions and results of resonant mirror (rm) optical biosensor SELEX.
Progress of IASYS SELEX with TGFp2 Rd TGFp2, Arcsec' [RNA], Vol, pL 3 Binding Dissociation Elution 6 gM 2 (min) 4 (min) CI C2 V 10 1777 0 1 50 27 29 water N 11 1777 0 10 50 30 60 water 12 1777 0 10 50 60 150 water 13 1893 0 0.05 50 37 73 water&SDS O 14 1721 0 3.5 50 30 35 water&SDS 'Amount of TGFp2 immobilized expressed in Arcsec where 1 Arcsec is 5 pg/mm 2 protein.
The protein is immobilized in an area of 4 mm 2 in cell 1 (C1).
2 Concentration of RNA pools 'Volume of RNA solution used 4 Length of binding phase in min Length of dissociation phase in min 6 Elution used 2004242532 24 Dec 2004 Table 7. Sequences isolated from round 13 of resonant mirror SELEX
NAME"
14i-1 13.20(1) 13.22 (2) 13.24(2) 13.30(1) 13.32(1) 13.34(1) 13.36(2) 13.40(1) 13.42(1) 13.44(1) 13.48(1) 13.50(1) 13.54(1) SEQ ID NO.
72 73 74 75 76 77 78 79 80 81 82 SEQUENCE b
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGGUGCGG
GGGAGGACGAUGCGG
GGGAGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
AAGUAACGUtJGUAGUAAAAUUCGtJUCUCUCGG -CAUUUGGC AAGUAACGUUAUAGUAAAAUtJCGUEJCUCUCGG
-UAUUGGC
AAGUAACGUUGUAGUAAAAUUCGUUCUCUCGG
-CGUULJGGC
AAGUAACGUEJGUAGUAAAAUUCGUUtCUCUCGG -CGUtJUGGU AAGUAACGUUGUAGUAAAAUUCGUUCUCUCGG
-CAUUUGGC
AAGUAACGUUGAAGUAAAAUUCGUUCUCUCUG -CgUUUGGU AAGUAACGUUGAAGUAAAAUtJCGUUCUCCUGG -UA-tUtGGC AAGUAACGUUGAAGUAAAAUUCGUUCUCUCGG
-CAUUUGGC
AAGUAACGUUGUAGUAAAAUUCGUUCUCUUGG
-CAUUUGC
AAGUAACGtJUAAAGUAAAAUUCGUUCUCUCGG
-CGUTJUGGC
AAGUAACGUtJGAAGUAAAAUUCGtJUCtJCUCGG
-CGUUUGGC
CAGACGACU -CGCCCGA CAGACGACU -CGCCCGA CAGACGACU -CGCCCGA CAGACGACU- CGCCCGA CAGACGACU -CGCCCGA
CAGACGACU-CGCCCGA
CAGACGACU-CGCCCGA
CAGACGACU- CGCCCGA CAGACGACU -CGCCCGA CAGACGACU -CGCCCGA CAGACGACU -CGCCCGA a, 83 GGGAGGACGAUGCGG AAGUAACGUUGUAGUAAAAUUCGUUCUCUCGG UAUUUGGC CAGACGACU -CGCCCGA
GGGAGGACG;AUGCGG
-GGAGGACGAUGCG_
AAGUAACGUUGUAGUAAAAUUCGUUCUCUUGG -UCUU_GGC CAGACGACU -CGCCCGA AAGUAACGUUGUAGUAAAAUUCGUUCUCUCGGGCAUG_
CAGACGACUUCGCCCGA
a Names are given in the form Round 13.clone number followed by the number of clones of that sequence that were isolated.
b Underlined bases are those that differ from ligand 14i-1I from the filter SELEX The sequence of 14i-1I is shown at the top for comparison. A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U.
2004242532 24 Dec 2004 Table 8. Sequences and boundaries of TGFP2 ligands isolated from rounds 14 and 21 of filter SELEX.
Name a SEQ
ID
NO:
SEQUENCE b Kd (nM) Ki (nM) 14i-1 72 21a-4 86 21a-21 87 region:
GGGAGGACGAUGCGGAAGUAACGUUGUAGUAAAAUUCGUUCUCUCGGCAUUEJGGCCAGACGACUCGCCCGA
GGGAGGACGAUGCGGCGUUGUUUAGUCGUAUGUAUAUACUAAGUCCGCUTJGUCCCCAGACGACUCGCCCGA
GGGAGGACGAUGCGG -tUCAGGAGGUTJAUUACAGAGUCUGUAUAGCUGUACUCCCCAGACGACUCGCCCGA 5' fixed selected 3' fixed 230 Names are in the form: round sequence was isolated-clone number.
b Boundaries are underlined. Fixed regions are in bold-faced type. Selected sequences are in plain type.
A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U 2004242532 24 Dec 2004 Table 9. Number of sequences isolated using the SELEX process.
SELEX round 8-spr 13-rm 14i l6a l8b 21a TOTAL Sequence 14i- 1 0 0 75 2 0 0 77 14i-I variants 21 15 22 2 0 0 21 a-4 0 0 0 0 0 3 3 7 2la-4 variants 0 0 4 7 0 2 13 2 1a-21 0 0 0 1 11 38 2la-2l variants 0 0 0 2 4 4 unidentified 36 0 0 0 0 0 36 filter-binding 12 0 1 1 0 1 TOTAL 69 15 102 15 15 48 264 2004242532 24 Dec 2004 Table 10. Characteristics of nucleic acid pools isolated using the SELEX method.
Round" 0 6-spr 8-spr 9-spr 9-rm I1I-rn 12-rm 1 3-rm Seguence of pgo 0 1 b random random slightly nonrandom of Mool 14i-1: <0.03 14i-1: -1 14i-1: -5 of transforrnantsd of clones' 14i- 1: .other: nonrandom can read sequence of ligand 1 4i- 1 can read sequence of ligand 14i-1I can read sequence of ligand 14i-1I can read variants of ligand 14i-1I sequence can read variants of ligand 14i-1 sequence 14i-1: 10-100 2la-21: <0.1 2 1a-21: 0.2-0.5 14i-1: 100 14i-1: 93 2 1a-4: 4 2 1a-2 1: 0 other: 3 14i-1: 27 21 a-4: 47 21a-21: other: 6 2 1a-2 1: 100 21a-21: 3-100 21a-21: 3-100 2la-21: 3-100 2 1 a-4: 21 a-2 1: other: 2 1 a-4: 2 1 a-2 1: other: 2004242532 24 Dec 2004 Table 10. (Continued) a spr, from surface plasmon resonance biosensor SELEX; rm, from resonant mirror optical biosensor SELEX.
b Determined by primer extension of bulk nucleic acid pools with 3'N7 primer.
c Determined by RT-PCR of bulk nucleic acid pools with a ligand-specific primer.
d Determined by PCR of individual transformants with a ligand-specific primer.
c Determined by sequencing of clones. Includes sequence variants of ligands.
2004242532 24 Dec 2004 Table 11. Truncates of human TGFP2 nucleic acid ligand 2 1a-2 1.
NAME
21a-21 2la-21 (U6G) 21a-21A5' 21a-21A3' 21a-21A5 3, 2la-21 (ML-94) 21a-21 (ML-95) 21a-21 (ML-96) 21a- 21 (ML- 97) 21a-21 (ML-99) 2la-21 (ML-1O1) 21a'-21 (ML-102) 2la-21 (ML-103) SEQUENCEa SEQ ID
BIO-
NO: BINDINGb LENGTHC ACTIVITYd
GGGAGGACGAUGCGGUUCAGGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCCCCAGACGACUCGCCCGA
87 0.5 70 1
GGGAGGACGAUGCGGUUCAGGAGGGUAUUACAGAGUCUGUAUAGCUGUACUCCCCAGACGACUCGCCCGA
88 250 34 GGtJUCAGGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCCCCAGACGAUCGCCCGA 89 0.5 56
GGGAGGACGAUGCGGUUCAGGAGGUUAUUACAGAGUCUGUAUAGCUGUAUCCCCA
100 56 GGUUtCAGGAGGUUAUUACAGAGUCUGUAUAGCUGUAUCCCCA 91 0.5 42 1
GGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCCCC
92 0.5 36
GGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCC
93 1 34
GGAGGUUAUUACAGAGUCUGUAUAGCUGUA
94 1000
GGAGGUUALTJUACAGAGUCUGUAUAGC
1000 26 GGAGGUUAUUACAGAGUCUGUAUAGC
CUCC
96 1000 GGAGGUUAUU AGAGUCU AUAGCUGUACUCC 97 1000 GGAGGUUAUU AGAGUCU AUAGC CUCC 98 1000 26
GGAGGUUAUUACAGAGUCUGUAUAGCUGUACUC
99 50 33 2004242532 24 Dec 2004 Table 11. (Continued)
NAME
21a-21 (ML-104) 21a-21 (ML-105) 21a-21 (ML-114) 21a-21 (ML-115) 21a-21 (ML-116) 21a-21 (ML-118) 21a-21 (ML-120) 21a-21 (ML-122) 21a-21 (ML-128) 21a-21 (ML-130) 21a-21 (ML-132) 21a-21 (ML-134) 21a-21 (ML-136) 21a-21 (ML-138) SEQUENCEa
SEQ
ID BIO- ACTIVITyd NO: BIN DINGb
GGAGGIJUAUUACAGAGUCUGUAUAGCUGUACU
100 70 GGAGGutJAuuACAGAGUCUGUAUAGCUGUAC 101 1000 GGAGGUUAuuACAGAGUCUGUAUAGC
GUACUCC
102 1000 GGAGGuuAUUACAGAGUCUGUAUAGCUGU
CUCC
103 1000 GGAGGUUAUUACAGAGUCUGUAUAGCU
ACUCC
104 1000 ,GGAGGUUAU ACAGAGUCUGUAUAGCUGUACUCC 105 1000 GGAGGuuAUUACAGA UCUGUAUAGCUGUACUCC 106 1000 GGAGGUUAUUACA AGU UGUAUAGCUGUACUCC 107 1000 GGAGGuuAUUACAGAGU UGUAUAGCUGUACUCC 108 1000 GG GGuUAUUACAGAGUCUGUAUAGCUGUAC
CC
109 2 GGAGGUUAtJUAC GAGUCUGUAUAGC GUACUCC 110 1000 GGAGA UAUUACAGAGUCUGUAUAGCUGUACUCC ill 10 GG GGUUAUU CAGAGUCUGUAUAGCUG AC CC 112 10000 GG GGtJUAUUA AGAGUCUGUAUAGCU UAC CC 113 10000
LENGTHC
32 31 33 33 32 33 33 32 33 32 32 33 2004242532 24 Dec 2004 Table 11. (Continued)
NAME
SEQUENCEa
SEQ
ID
NX22283 NX22284 NX22285 NX22286 NX22301 NX22302 NX22303 NX22323 NO: BINDINGb LENGTHC GGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCCCC[3'TI 114 0.6 36 GGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCC [3'TI 115 1 34 GGAGGUtJAUUACAGAGUCUGUAUAGCUGUACUCCCCA 116 2 37
GGAGGUUAUUACAGAGUCUGUAUAGCUGUA
117 130 30 GAGGUUAUUACAGAGUCUGUAUAGCUGUACUCC
TI
118 1 33 AGGUUAUUACAGAGUCUGUAUAGCUGUACUCC [3 'TI 119 100 32 GGUUAUUACAGAGUCUGUAUAGCUGUACUCC [3'TI 120 >100 31 PEG-GGAGGUUAUUACAGAGUCUGUAUAGCUGUACUCC [3'T] 121 nt 34
BIO-
ACTIVITYd 1 2 >100 3 8 The fixed regions are indicated by bold-faced letters. The point mutant in 21a-21(U6G) is underlined and in bold type. A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U The italicized G at the 5' end of the 5' RNase H cleavage products indicates that -50% of the time cleavage leaves 2 G's and 50% of the time one G is left. The boundaries in 21a-21 are underlined b Binding is expressed as the ratio of the Kd of ligand /Kd of NX22284. The K 4 of NX22284 is -2 nM.
Length is given in bases.
d Bioactivity is expressed as the ratio of the Ki of ligand /Ki of NX22284. The K, of NX22284 is -10 nM.
2004242532 24 Dec 2004 Table 12. Alignment of human transforming growth factor 03 amino acid sequences.
SEQ
ID
NO.
122 123 124 TGF91: ALDTNYCFSS TEKNCCVRQL YIDFRKDLGW KWIHEPKGYH ANFCLGPCPY
IWSLDTQYSK
TGFf92: AIJDAAYCFRN VQDNCCLRPL YIDFKRDLGW KWIHEPKGYN ANFCAGACPY LWSSDTQHSR TGF9g3: ALDTNYCFRN LEENCCVRPL YIDFRQDLGW KWVHEPKGYY ANFCSGPCPY
LRSADTTIHST
TGF92 specific: AA VQD L KR N A A S R 60 60 60 TGFI91: VLALYNQHNP GASAAPCCVP QALEPLPIVY YVGRKPKVEQ LSNMIVRSCK
CS
TGF9?2: VLSLYNTINP EASASPCCVS QDLEPLTILY YIGKTPKIEQ LSNNIVKSCK
CS
TGFI93: VLGLYNThNP EASASPCCVP QDLEPLTILY YVGRTPKVEQ LSNMVVKSCK
CS
TGF92 specific: S I S I K I 112 112 112 125 126 127 2004242532 24 Dec 2004 77 Table 13. Truncates of human TGFP2 nucleic acid ligand 14i-1.
NAME
SEOUENCEa SEQ ID NO. BINDINGb LENGTHC 14i-1 1 4 i- 1 ASd 14i-1A3 d 14i-1A5,3-d 14i-1t5-41 14i-1t5-38 14i-1t5-35 14i-1(ML-86) 14i-1(ML-87) 14i-1(ML-89) GGGAGGACGAUGCGGAAGUAACGIJEGUAGUAAAUUCGUUCUCUCGGCA
CCGACGACCGCCCGA
72 1 71
GGAAGUAACGUUGUAGUAAAAUUCGUUCUCUCGGCAUGGCCAGACGAUCGCCCGA
128 >100 56
GGGAGGACGAUGCGGAAGUAACGUUGUAGUAAAAUUCGUUCUCUCGGCAUUUGGCCA
129 3 57
GGAAGUAACGUUGUAGUAAAAUUCGUUCUCUCGGCAUGGCCA
gGGAgGAUGCGGAAGUAACGUUGUAGUAAAAUUCcUUC gGGAgGAUGCGGAAGUAACGUUGUAGUAAAAUUCc gGGAgGAUGCGGAAGUAACGUUGUAGUAAAAU gGGAgGAUGCGGAAGUAACGUUGUAGU UCctUC gGGAgGAUGCGGAAGUAACGUUGUAGU gGgaGgAGUAACGUUGUAGU 130 131 132 133 134 135 136 >100 1 >100 >100 >100 >100 >100 Lowercase letters indicate bases not found at that position in the full length ligand that were added or changed to maintain transcriptional efficiency. Boundaries are underlined. The fixed regions are in bold-faced type. The italicized G at the 5' end of the 5' RNase H cleavage products indicates that -50% of the time cleavage leaves 2 G's and 50% of the time one G is left.
A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F.
Binding is expressed as the ratio of Kd (ligand)/Kd (14i-1). The Kd of 14i-l is -10 nM.
Length is in bases.
Produced by RNase H digestion.
2004242532 24 Dec 2004 Table 14. Truncates of human TGFP2 nucleic acid ligand 21a-4.
Name Sequencea SEQ ID NO. Bindinjib Lenth 21a-4
GGGAGGACGAUGCGGCGUUGUUAGUCGUAUGUAUAUACUAGUCCGCUGUCCCCAGAG
86 1 71 2la-4A5' d GGCGUUGUUUAGUCGUAUGUAUAUA
CUAAGUCCGCGUCCCCAGACGACUCGCCCGA
137 >100 56 21a-4A3 d GGGAGGACGAUGCGGCGUUGUUUAGUCGUAUGUAUAUAIAGUCCGCUUGUCCCCA 138 1 57 2la-4A5', 3 'd
GGCGUUGUUAGUCGUAUGUAUAUACUAAGUCCGCUUGUCCCCA
139 >100 42 2la-4(ML-91) ggGgaGCGGCGUUGUUAGUCGUAUGUAUAUACUAAGUCCGC1 140 1 44 21a-4(ML-92) ggGgaGCGGCGUUGUU gaaa
AGUCCGCUU
141 >100 27 21a-4 (ML-108) ggGgaGCGGCGUUGUUU CGUAUGUAUAU
AAGUCCGCUJ
142 >100 38 21a-4(ML-109) ggGgaGCGGCGUUGUUU AUGUAU
AAGUCCGCUU
143 >100 33 21a-4(ML-110) ggGgaGCGGCGUUGUUUAGUCGUAUGUAUAUACUAAGUCCGC 144 1 42 2la-4(ML-111) ggGgaGCGGCGUUGUUUAGUCGUAUGUAUAUAAGU 145 30 38 a Lowercase letters indicate bases not found at that position in the full length ligand. Underlining indicates boundary positions.
The fixed region sequences are indicated in bold-faced lettering. The italicized G at the 5' end of the 5' RNase H cleavage products indicates that -50% of the time cleavage leaves 2 Gs and 50% of the time one G is left. A=2'-OH A; C=2'-F C; G=2'-OH G; U=2-F U b Binding is expressed as the ratio of Kd (ligand)/Kd (21a-4). The K. of 21a-4 is -3 nM.
c Length is expressed in bases.
d These ligands were generated by RNAse H digestion of 21a-4.
2004242532 24 Dec 2004 Table 15. Biased SELEX conditions and results.
Round" [RNA]' Nm [TGFfP2], rim RNAb/ protein Bound/ background [Competitor] Bound Background 34N7. 2la-21 la 1000 2a 450 3a 10 4a 50 4b 50 8 8 6a 4 6b 6 7a 5 7b 5 round 0 150 300 50 10 10 1 1 0.5 0.5 0 .25 0.25 0.05 0 .05 1 0.01 nucleic acid 7 1.5 0.2 5 5 8 8 8 12 20 20 20 20 125 90 0 0 0 0 333 riM NX22284 0 100 riM NX22284 0 100 nM NX22284 0 200 riM NX22284 mM tRNA 0 100 nM NX22284 mM tRNA 1.4 1.7 17 .5 11.0 2.2 1.4 0.8 2.9 1.8 0.5 0.15 1.4 1.0 1.0 0.9 1.3 0.9 0.7 2.9 1.3 0. 14 0.1 1.0 1.7 17.5 12 .3 1.7 1.5 1. 1 1.0 1.4 3.4 1.5 0.9 1.2 1.2 1.0 Kd (nM)c 870 395 186 17 8 1 17 1 1 1 1 3 nd rid 125 0.9 1.05 1. 1 0.6 0.5 0.6 0.15 0.5 0.14 a a series, without competitor; b series, with competitors b nucleic acid ligand library cnd, not determined 2004242532 24 Dec 2004 Table 16. Nucleic acid ligands isolated from round 5a of a human TGFj32 biased SELEX.
NAMEO
5' FIXED SELECTED b 3' FIXED A CAGACC3ACUCGCCCGA SEQ ID NO: CHANGESc B INDING d A B C 72 0 putative structural element: Si B S2 L S2 Si 21 a-21: GGGAGGACGAUGCGGUUCAA~ACAffgagU~gUgAAQGUACUCCC 1: (2) 2: (1) 4: (1) 6 (2) 7: (1) 9: (1) (1) 11: (2) 13: (1) 14: (1) (1) 17: (1) 18: (1) 19: (2) (1) 21: (2) 22: (4) (1) 26: (1) 28: (1) 29: (1)
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAIJGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
GGUGAUUAUUACAGAGUAUGUAUAGCUGUACCCC
AGGCGUUAUUAGAGAGUCUGUAUAGCUCUAGCCC
GGAGGGUAUUACAGAGUAUGUAUAGCUGUACUCC
GGAGGUUAUtJAUAGAGUCUGUAUAGCUAUACCCC
GAGGGUUAUUAUAGAGUCUGCAUAGCUAUACCCC
UGAGAGUAUUACGGAGUAUGUAUAGCCGUACCCC
GGGCAUUAUUUCAGAGUCUGUAUAGCUGUAGCCC
GCGGAUUAUCACAGAGUAUGUAUAGCUGUGCCGC
UGUGAAUAUUAGAGAGUCUGUAUAGCUCUACCCC
CGGGAUUAUUACUGAGUCUGUAUAGCAGUACCCC
GUGGAAUAUUACGGAGUCUGUAUAGCCGUACUCC
GGGGACUAUUAGUGAGUCUGUAUAGCACUACCCC
GUGGAUUAUUACAGCGUCUGUAUAUCUGUACCCC
GCAGGUUAUUACAGAGUCUGUAUAGCUGUACUGC
GGUAGAUAUCACUGAGUCUGUAUAGCAGUGUCCC
AGGGAUUAUtJACAGAGUCUGUAUAGCUGUACCCC
GUGGAUUTAUTUACAGAGUCUGUAUAGCUGUACCCC
GGGCGUUAUUACAGAGUCUGUAUAGCUGUAGCCC
GGUGGUUAUUACACAGUAUGUAUAGGUGUACCCC
AGGGAAUAUUACAGAGUAUGUAUAGCUGUACCCC
GGAGUUUAUUACAGCGUCUGUAUAUCUGUAGCCC
CAGACGACUCGCCCGA
CAGACGACUCGCC
-GA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAI3ACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACtCGCCCGA
ICAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 1,63 164 1,65 166 0.8 0.6 1.4 1.6 0.3 0.3 0.3 0.4 0.2 0.4 0.4 0.8 5.7 0.7 1. 1 3.1 2004242532 24 Dec 2004 Table 16. (Continued) NAMEa 5' FIXED
GGGAGGACGAUGCGG
34: GGGAGGACGAUGCGG
GGGAGGACGAUGCGG
36: GGGAGGACGAUGCGG invariant: SELECTED b 31 FIXED UGAGGUUAUUtACAGAGUCUGUAUAGCUGUACUCC
GGUGGUUAUUAGAGAGUCUGUAUAGCUCUACGCC
GGGGAGUAUUAAAGAGUCUGUAUAGCUUUACCCC
GGAGGAUAUUAUAGAGUCUGUAUAGCUAUACCCC
UAU GU UG AUA C
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
CAGACGACUCGCCCGA
SEQ ID NO: CHANGE Sc B IND ING d A 'B C 167 1 2.4 168 4 1.7 169 6 0.8 170 4 1.9 aNumber of clones isolated for each sequence is indicated in parentheses.
bNucleotides that differ from the starting sequence are shown in bold-faced lettering. A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U Putative structural elements: Si1, stem 1; B, bulge; S2, stem 2; L, loop. The sequence of ligand 2 1a-21 is shown at the top for comparison.
cNumber of changes from starting sequence.
d Binding is expressed as Kd (ligand)/ Kd (21 a-2 The Kd of ligand 2 1a-21 is about 1 nM.
2004242532 24 Dec 2004 Table 17. Highest and lowest affinity TGFP12 nucleic acid ligands from biased SELEX
NAME
5' FIXED SELECTED a 3' FIXED BINDING' CHANGESC SEO ID NO.
HIGHEST AFFINITY LIGANDS: 13: GGGAGGACGAUGCGG UGUGAAUAUUAGAGAGUCUGUAUAGCUCUAC
CCC
154 14: GGGAGGACGAUGCGG CGGGAUUtAUUACUGAGUCUGUAUAGCAGUACCCC 155 21: GGGAGGACGAUGCGG
AGGGAUUAUTUACAGAGUCUGUAUAGCUGUACCCC
161 GGGAGGACGAUGCGG
GGGGAGUAUUAAAGAGUCUGUAUAGCUUUACCCC
169 putative structural elements: S1 B S2 L S2 Si 21 a-21: .AUUGCGCAGGGGUUCUGGQ jA9UGUACUCCC GGGAGACAUGCGUUAGGAG~J72 LOWEST AFFINITY LIGANDS: 36: GGGAGGACGAUGCGG
GGAGGAUALUAUAGAGUCUGUAUAGCUAUACCCC
170 GGGAGGACGAUGCGG UGAGGUUAUtACAGAGUCUGUAUAGCUGUACUCC 167 26: GGGAGGACGAUGCGG
GGUGGUUAUUACACAGUAUGUAUAGGUGUACCCC
164 6: GGGAGGACGAUG COG GGAGGUUAUUAUAGAGUCUGUAUAGCUAUACCCC 149 GGGAGGACGAUGCGG
GGUAGAUAUCACUGAGUCUGUAUAGCAGUGUCCC
160
CAGACGACUCGCCCGA
0.2 7
CAGACGACUCGCCCGA
0.4 6
CAGACGACUCGCCCGA
0.7 4
CAGACGACUCGCCCGA
0.8 6
CAGACGACUCGCCCGA
1.0 0
CAGACGACUCGCCCGA
2.0 4
CAGACGACUCGCCCGA
2.4 1
CAGACGACUCGCCCGA
3.1 4
CAGACGACUCGCCCGA
3.3 3
CAGACGACUCGCCCGA
5.7 9 invariant: UAU GU3 UG AUA
C
aNucleotides that differ from the starting sequence are shown in bold-faced lettering. A=2'-OH A; C=2'-F C; G=2'-OH G; U=2'-F U Putative structural elements: S1, stemi; B, bulge; S2, stem2; L, loop.
bBinding is expressed as K. (ligand)! Kd (21 a-2l1). The Kd of 21 a-21 is 1 nM Number of changes from starting sequence.
2004242532 24 Dec 2004 Table 18. Substitution of 2'-OH purines with 2'-OCH 3 purines in NX22284 ligand.
NAME
NX2 2284 NX2 2304 NX2 2355 NX22356 NX22357 NX22358 NX22359 NX 22360 NX2 2374 NX 22375 NX2 2376 NX2 2377 NX2 2383 NX2 2384 NX2 2417 NX2 2420 NX2 2421 NX2 2426 NX2 2427 aSBQ UEN CEa SEO ID NO: GGAGGUUAUTUACAGAGUCUGUAUAGCUGUACUCC [3 'TI 115 ggaggUUaUUaCagagUCUgUaUagCUgUaCUCC TI 171 GGGUATa~ggCga g~~CC[3 'TI 172 ggagGUUAUUACAGAGUCUGtJAUAGCUGUAUCC [3 'TI 173 GGAGgUUaUUaCAGAGUCUGUAUAGCUGUAUCC [3 'TI 174 GGGUAU~ggCUUUGUU C[3 'TI 175 GGGUAUCGGC ~~GUUCC[3 'TI 176 GGGLUUUCGGCUUU[~ga C3 'TI 177 GGGUAUCGGC UUGUUCC[3 'TI 178 GGAGGUUAUJEACAGAGUCUGUaUAGCUGUAUCC TI 179 GGAGGUTUALUACAGAGUCUGUAUaGCUGUACUCC [3 'TI 180 ggaggtUtaUUaCAGAGUCUGUAUAgCUgUaUCC TI 181 ggaggUUaUlUaCagagUCUGUAUagCUgUaCUcCC[3' TI 182 ggaggUTUaLUaCagagUCUgUAUagCUgUaCUCC 3' TI 183 ggaggUUaUUaCagagUCUGUAUAgCUgUaCUCC TI 184 ggaggUUAUtaCagagUCUGUAUAgCUgUaUCC [3 'TI 185 ggagGUUAUUACagagUCUGUAUAgCUgUaUCC [3 'TI 186 ggaga-UAUTUaCagagUCUGUAUAgCUgUaCUCC TI 187 gg-ggtJUAUJEaCagagUCtGUAUAgCUgUaCCC[3 'TI 188 B INDING' 1 >100 >100 1 2 1 >100 1 25 >100 50 1 500 10000 1 1 2 1 0.3
LENGTHC
34 34 34 34 34 34 34 34 34 34 34 34 34 34 34 34 34 33 32
BIOACTIVITY'
1 >100 >100 1 1 1 >100 >300 >100 1 >100 >100 1 1 0.7 A, 2'-OH A; C, 2'-F C; G, 2'-OH G; U, 2'-F U; a, 2'-OCH 3 A; g, 2'-OCH 3 G. [37T] signifies a 3T, 3'dT cap.
bBinding is expressed as the ratio of the Kd of ligand /Kd of NX22284. The K-d of NX22284 is -1 nM.
Length is given in bases. X 2 8 .T eK f N 2 2 4i 1 M b Bioactivity is expressed as the ratio of the K, of ligand of N228.TeKofN 284i-1nM 2004242532 24 Dec 2004 Table 19. Truncates and 2'-OCH 3 purine modifications of nucleic acid ligand 13 from a biased SELEX
NAME
NX2 2385 NX22386 NX22387 NX2 2424 NX2 2425 SEQUENCEa SEQ ID NO: UGUGAAUAUUEAGAGAGUCUGUAUAGCtJCUACCCC[13 'TI 189 UgUgaAUaUUaGagagUCUGUAUagCUCUaCCCC [3 'TI 190 UgUgaaUaUt~agagagtCUgUAUagCUCUaCCCC T] 191 UgUgAAUAUTUaGagagtJCUGUAUAgCUCUaCCCC [3 'TI 192 UgUgaatJAUUagagagUCUGUAUAgCUCt~aCCCC [3 'TI 193 BINDINGb 0.4 3000 3000 0.6 1.5 LENGTHC BIQACTIVITyd 34 4 34 >100 34 34 >100 34 >100 a A, 2'-OH A; C, 2'-F C; G, 2'-OH G; U, 2'-F U; a, 2'-OCH 3 A; g, 2'-OCH 3 G. [37T] signifies a 3'dT cap.
b Binding is expressed as the ratio of the Kd of IigandfKd of NX22284. The Kd of NX22284 is 2 nM.
c Length is given in bases.
d Bioactivity is expressed as the ratio of the K, of 1igand/K, of NX22284. The Ki of NX22284 is 10 nM.
Table 20. Pharmacokinetic properties of NX22323 in rats using a noncompartmental analysis.
Parameter Units Estimate Cmax (pig/mL) 27.1 AUClast ((Iig*min)/mL) 3028.0 AUCINF ((jjig*mjn)/mL) 3058.0 Beta t112 (min) 630.9 Cl (mL/(min*kg)) 0.33 MRTINF (min) 350.4 Vss (mL/kg) 115.0 Vz (mL/kg) 298.0 Table 21. Pharmacokinetic properties of NX22323 in rats using a compartmental analysis.
Parameter Units Estiate tdError I% Error Cmax (p.g/mL) 16.3 3.3 20.2 AUCfNF ((p.g*min)/mL) 2486 274 11.0 Alpha-tl/2 (min) 63.5 19.1 30.2 Beta-t1/2 (min) 467.2 83.2 17.8 A (p.g/mL) 14.63 3.21 21.9 B (pg/mL) 1.70 0.84 49.1 Cl (mL/(min*kg) 0.402 0.044 11.0 MRTINF (min) 360.3 35.69.
Vss (mL/kg) 144.9 23.1 1.
2004242532 24 Dec 2004 TABLE 22. Binding and inhibitory activity of 2'-Omethyl- and Pegyl-modifications of lead TGFR1 truncate ligand SEQ ID NO: Binding Bic ChD7O GGGUGCCUUUUGCCUaGGUUGUGaUUUGUaACCUUCUGCCCa 216..
ChD7O-ml gggUgCCUUUUGCCUAGGUUGUGAUUUGUAACCUJCUGCCCA 194 ChD7 0-m2 GGGUGCCUUUt~gCCUaggUUGUGAUUUGUAACCUUCUGCCCA 195 ChD7O-m3 GGGUGCCUtUUGCGUAGGUUgUgaUUUgUAACCUUCUGCCCA 196..
ChD7O-m4 GGUCUUGCAGUUGUU~aCU~C~ 197 gGGUGCCUUUUGCCUAGGUUgUgaUUUgUAACCUUCUGCCCA 198..
ChD7O-m6 GgGUGCCUUUUGCCUAGGUUgUgaUUUgUAACCUUCUGCCCA 199..
ChD7O-m7 GGgUGCCUUUUGCCUAGGUUgt~gaUUUgUAACCUUCUGCCCA 200..
ChD7O-m8 GGGUgCCUUUUGCCUAGGUUgUgaUUUgUAACCUUCUGCCCA 201 ChD7O-m9 GGGUGCCUUUUgCCUAGGUUgUgaUUUgUAACCUUCUGCCCA 202 ChD7Q-mlO GGGUGCCt.UUUGCCUaGGUtgtgaUUUgUAACCUUCUGCCCA 203..
ChD7Q-rnll GGGUGCCUUUUGCCUAgGUUgUgaUUUgUAACCUUCUGCCCA 204..
ChD7O-m12 GGGUGCCUUUUGCCUAGgUUgUgaUUUgUAACCUUCUGCCCA 205 activity ChD7O-m13 GGGUGCCUUUUGCCUAGGUUgUgaUUUgUaACCUUCUGCCCA ChD7O-m14 GGGUGCCUUUUGCCUAGGUUgUgaUUUgUAaCCUUCUGCCCA ChD7 0-miS GGUGCCUUUtJGCCUAGGUUgUgaUUUgUAACCUUCUgCCGA ChD7 0-mi 6 GGGUGCCUUUUGCCUAGGUUgUgaUUUgUAACCUUCUGCCCa ChD7O-m17 gggUGCCUUUUGCCUaggUUgUgaUUUgUaaCCUUCUGCCCa 3
U
ChD7O-m18 gggUGCCUUUUGCCUaggUUgUgaUUUgUaACCUUCUGCCCa 3
U
ChD7O-m19 gggUGCCUUUUGCCUaggUUgUgaUUUgUaaCCUUCUGCCC 3
U
ChD7O-m20 gggUGCCUUUUGCCUaggUUgU gUaaCCUUCUGCCCa3'
U
ChD7O-m2i gggUGCCUUUUGCCUaggUUg UaaCCUUCUGCCCa3'
U
ChD7O-m22 gggUGCCUUUUGCCUaggUU aaCCUUCUGCCCa3'
U
206 207 208 209 210 211 212 213 214 215

Claims (4)

1. A complex comprised of a TGFP2 nucleic acid ligand and a non- I immunogenic, high molecular weight compound or lipophilic compound. t 2. The complex of claim 1 further comprising a linker between said ligand and said non-immunogenic, high molecular weight compound or lipophilic compound.
3. The complex of claim 2 wherein said non-immunogenic, high molecular weight compound is a polyalkylene glycol.
4. The complex of claim 3 wherein said polyalkylene glycol is polyethylene glycol (PEG). The complex of claim 4 wherein said PEG has a molecular weight of about between 10-80 K.
6. The complex of claim 5 wherein said PEG has a molecular weight of about K. DATED this 24 th day of December, 2004 GILEAD SCIENCES, INC. by DAVIES COLLISON CAVE Patent Attorneys for the Applicant(s)
AU2004242532A 1999-07-29 2004-12-24 High affinity TGF beta nucleic acid ligands and inhibitors Abandoned AU2004242532A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2004242532A AU2004242532A1 (en) 1999-07-29 2004-12-24 High affinity TGF beta nucleic acid ligands and inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/363939 1999-07-29
AU67487/00A AU781234B2 (en) 1999-07-29 2000-07-26 High affinity TGF beta nucleic acid ligands and inhibitors
AU2004242532A AU2004242532A1 (en) 1999-07-29 2004-12-24 High affinity TGF beta nucleic acid ligands and inhibitors

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
AU58839/96A Addition AU732820B2 (en) 1991-06-10 1996-05-30 High-affinity oligonucleotide ligands to growth factors
AU67487/00A Division AU781234B2 (en) 1999-07-29 2000-07-26 High affinity TGF beta nucleic acid ligands and inhibitors

Publications (1)

Publication Number Publication Date
AU2004242532A1 true AU2004242532A1 (en) 2005-01-27

Family

ID=34427360

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2004242532A Abandoned AU2004242532A1 (en) 1999-07-29 2004-12-24 High affinity TGF beta nucleic acid ligands and inhibitors

Country Status (1)

Country Link
AU (1) AU2004242532A1 (en)

Similar Documents

Publication Publication Date Title
AU781234B2 (en) High affinity TGF beta nucleic acid ligands and inhibitors
US6124449A (en) High affinity TGFβ nucleic acid ligands and inhibitors
AU692469B2 (en) Nucleic acid ligands and improved methods for producing the same
US6344321B1 (en) Nucleic acid ligands which bind to hepatocyte growth factor/scatter factor (HGF/SF) or its receptor c-met
US6127119A (en) Nucleic acid ligands of tissue target
AU777043B2 (en) Nucleic acid ligands to CD40ligand
JP4531132B2 (en) High affinity oligonucleotide ligands for growth factors
US5723594A (en) High affinity PDGF nucleic acid ligands
AU767501B2 (en) Tenascin-C nucleic acid ligands
US20090075922A1 (en) Nucleic Acid Ligands Which Bind to Hepatocyte Growth Factor/Scatter Factor (HGF/SF) or its Receptor c-met
EP1204672A1 (en) High affinity vascular endothelial growth factor (vegf) receptor nucleic acid ligands and inhibitors
US20060084797A1 (en) High affinity TGFbeta nucleic acid ligands and inhibitors
AU2004242532A1 (en) High affinity TGF beta nucleic acid ligands and inhibitors
AU782620B2 (en) High affinity vascular endothelial growth factor (VEGF) receptor nucleic acid ligands and inhibitors
AU6234900A (en) Nucleic acid ligands to hepatocyte growth factor/scatter factor (hgf/sf) or its receptor c-met and to integrins

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted