CA2325013A1 - Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation - Google Patents

Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation Download PDF

Info

Publication number
CA2325013A1
CA2325013A1 CA002325013A CA2325013A CA2325013A1 CA 2325013 A1 CA2325013 A1 CA 2325013A1 CA 002325013 A CA002325013 A CA 002325013A CA 2325013 A CA2325013 A CA 2325013A CA 2325013 A1 CA2325013 A1 CA 2325013A1
Authority
CA
Canada
Prior art keywords
oligonucleotides
nucleic acid
compounds
oligonucleotide
silico
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
CA002325013A
Other languages
French (fr)
Inventor
Lex M. Cowsert
Brenda F. Baker
John Mcneil
Susan M. Freier
Henri M. Sasmor
Douglas G. Brooks
Cara Ohasi
Jacqueline R. Wyatt
Alexander H. Borchers
Timothy A. Vickers
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.)
Ionis Pharmaceuticals Inc
Original Assignee
Individual
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 US09/067,638 external-priority patent/US7321828B2/en
Application filed by Individual filed Critical Individual
Publication of CA2325013A1 publication Critical patent/CA2325013A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids
    • G16B30/10Sequence alignment; Homology search
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids
    • G16B30/20Sequence assembly
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B35/00ICT specially adapted for in silico combinatorial libraries of nucleic acids, proteins or peptides
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B35/00ICT specially adapted for in silico combinatorial libraries of nucleic acids, proteins or peptides
    • G16B35/20Screening of libraries
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/60In silico combinatorial chemistry
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/0068Means for controlling the apparatus of the process
    • B01J2219/007Simulation or vitual synthesis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B30/00ICT specially adapted for sequence analysis involving nucleotides or amino acids

Abstract

Iterative, preferably computer based iterative processes for generating synthetic compounds with desired physical, chemical and/or bioactive properties, i.e., active compounds, are provided. During iterations of the processes, a target nucleic acid sequence is provided or selected, and a library of candidate nucleobase sequences is generated in silico according to defined criteria. A "virtual" oligonucleotide chemistry is chosen and a library of virtual oligonucleotide compounds having the selected nucleobase sequences is generated. These virtual compounds are reviewed and compounds predicted to have particular properties are selected. The selected compounds are robotically synthesized and are preferably robotically assayed for a desired physical, chemical or biological activity. Active compounds are thus generated and, at the same time, preferred sequences and regions of the target nucleic acid that are amenable to oligonucleotide or sequence-based modulation are identified.

Description

IDENTIFICATION OF GENETIC TARGETS FOR MODULATION
BY OLIGONUCLEOTIDES AND GENERATION OF
OLIGONUCLEOTIDES FOR GENE MODULATION
CROSS REFERENCE TO RELATED APPLICATIONS
The present application is a continuation-in-part of U.S. Serial No.
09/067,638 filed April 28, 1998, which claims priority to provisional application Serial No.
60/081,483 filed April 13, 1998, each of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The present invention relates generally to the generation and identification of synthetic compounds having defined physical, chemical or bioactive properties.
More particularly, the present invention relates to the automated generation of oligonucleotide compounds targeted to a given nucleic acid sequence via computer-based, iterative robotic synthesis of synthetic oligonucleotide compounds and robotic or robot-assisted analysis of the activities of such compounds. Information gathered from assays of such compounds is used to identify nucleic acid sequences that are tractable to a variety of nucleotide sequence-based technologies, for example, antisense drug discovery and target validation.
2 BACKGROUND OF THE INVENTION
1. Oligonucleotide Technology Synthetic oligonucleotides of complementarity to targets are known to hybridize with particular, target nucleic acids in a sequence-specific manner. In one example, S compounds complementary to the "sense" strand of nucleic acids that encode polypeptides, are referred to as "antisense oligonucleotides." A subset of such compounds may be capable of modulating the expression of a target nucleic acid; such synthetic compounds are described herein as "active oligonucleotide compounds."
Oligonucleotide compounds are commonly used in vitro as research reagents and diagnostic aids, and in vivo as therapeutic and bioactive agents.
Oligonucleotide compounds can exert their effect by a variety of means. One such means takes advantage of an endogenous nuclease, such as RNase H in eukaryotes or ltNase P in prokaryotes, to degrade the DNA/RNA hybrid formed between the oligonucleotide sequence and mRNA
(Chiang et al., J. Biol. Chem., 1991, 266, 18162; Forster et al., Science, 1990, 249, 783).
Another means involves covalently linking of a synthetic moiety having nuclease activity to an oligonucleotide having an antisense sequence. This does not rely upon recruitment of an endogenous nuclease to modulate target activity. Synthetic moieties having nuclease activity include, but are not limited to, enzymatic lZNAs, lanthanide ion complexes, and other reactive species. (Haseloff et al., Nature, 1988, 334, 585; Baker et al., J. Am. Chem.
Soc., 1997, 119, 8749).
Despite the advances made in utilizing antisense technology to date, it is still common to identify target sequences amenable to antisense technologies through an empirical approach (Szoka, Nature Biotechnology, 1997, I5, 509). Accordingly, the need exists for systems and methods for efficiently and effectively identifying target nucleotide sequences that are suitable for antisense modulation. The present disclosure answers this need by providing systems and methods for automatically identifying such sequences via in silico, robotic or other automated means.
2. Identification of Active Oligonucleotide Compounds Traditionally, new chemical entities with useful properties are generated by (1) identifying a chemical compound (called a "lead compound") with some desirable property or activity, (2) creating variants of the lead compound, and (3) evaluating the
3 property and activity of such variant compounds. The process has been called "SAR," i.e., structure activity relationship. Although "SAR" and its handmaiden, rational drug design, has been utilized with some degree of success, there are a number of limitations to these approaches to lead compound generation, particularly as it pertains to the discovery of bioactive oligonucleotide compounds. In attempting to use SAR with oligonucleotides, it has been recognized that RNA structure can inhibit duplex formation with antisense compounds, so much so that "moving" the target nucleotide sequence even a few bases can drastically decrease the activity of such compounds (Lima et al., Biochemistry, 1992, 31, 12055).
Heretofore, the preferred method of searching for lead antisense compounds has been the manual synthesis and analysis of such compounds. Consequently, a fundamental limitation of the conventional approach is its dependence upon the availability, number and cost of antisense compounds produced by manual, or at best semi-automated, means.
Moreover, the assaying of such compounds has traditionally been performed by tedious manual techniques. Thus, the traditional approach to generating active antisense compounds is limited by the relatively high cost and long time required to synthesize and screen a relatively small number of candidate antisense compounds.
Accordingly, the need exists for systems and methods for efficiently and effectively generating new active antisense and other oligonucleotide compounds targeted to specific nucleic acid sequences. The present disclosure answers this need by providing systems and methods for automatically generating and screening active antisense compounds via robotic and other automated means.
3. Gene Function Analysis Efforts such as the Human Genome Project are making an enormous amount of nucleotide sequence information available in a variety of forms, e.g., genomic sequences, cDNAs, expressed sequence tags (ESTs) and the like. This explosion of information has led one commentator to state that "genome scientists are producing more genes than they can put a function to" (Kahn, Science, 1995, 270, 369). Although some approaches to this problem have been suggested, no solution has yet emerged. For example, methods of looking at gene expression in different disease states or stages of development only provide, at best, an association between a gene and a disease or stage of development
4 (Nowak, Science, 1995, 270, 368). Another approach, looking at the proteins encoded by genes, is developing but "this approach is more complex and big obstacles remain" (Kahn, Science, 1995, 270, 369). Furthermore, neither of these approaches allows one to directly utilize nucleotide sequence information to perform gene function analysis.
In contrast, antisense technology does allow for the direct utilization of nucleotide sequence information for gene function analysis. Once a target nucleic acid sequence has been selected, antisense sequences hybridizable to the sequence can be generated using techniques known in the art. Typically, a large number of candidate antisense oligonucleotides (ASOs) are synthesized having sequences that are more-or-less randomly spaced across the length of the target nucleic acid sequence (e.g., a "gene walk") and their ability to modulate the expression of the target nucleic acid is assayed.
Cells or animals can then be treated with one or more active antisense oligonucleotides, and the resulting effects determined in order to determine the functions) of the target gene.
Although the practicality and value of this empirical approach to determining gene function has been acknowledged in the art, it has also been stated that this approach "is beyond the means of most laboratories and is not feasible when a new gene sequence is identified, but whose function and therapeutic potential are unknown" (Szoka, Nature Biotechnology, 1997, 15, 509).
Accordingly, the need exists for systems and methods for efficiently and effectively determining the function of a gene that is uncharacterized except that its nucleotide sequence, or a portion thereof, is known. The present disclosure answers this need by providing systems and methods for automatically generating active antisense compounds to a target nucleotide sequence via robotic means. Such active antisense compounds are contacted with cells, cell-free extracts, tissues or animals capable of expressing the gene of interest and subsequent biochemical or biological parameters are measured. The results are compared to those obtained from a control cell culture, cell-free extract, tissue or animal which has not been contacted with an active antisense compound in order to determine the function of the gene of interest.
4. Target Validation Determining the nucleotide sequence of a gene is no longer an end unto itself;
rather, it is "merely a means to an end. The critical next step is to validate the gene and its [gene] product as a potential drug target" (Glasser, Genetic Engineering News, 1997, 17, 1 ). This process, i.e., confirming that modulation of a gene that is suspected of being involved in a disease or disorder actually results in an effect that is consistent with a causal relationship between the gene and the disease or disorder, is known as target validation.
5 Efforts such as the Human Genome Project are yielding a vast number of complete or partial nucleotide sequences, many of which might correspond to or encode targets useful for new drug discovery efforts. The challenge represented by this plethora of information is how to use such nucleotide sequences to identify and rank valid targets for drug discovery. Antisense technology provides one means by which this might be accomplished; however, the many manual, labor-intensive and costly steps involved in traditional methods of developing active antisense compounds has limited their use in target validation (Szoka, Nature Biotechnology, 1997, 15, 509). Nevertheless, the great target specificity that is characteristic of antisense compounds makes them ideal choices for target validation, especially when the functional roles of proteins that are highly related are being investigated (Albert et al., Trends in Pharm. Sci., 1994, I5, 250).
Accordingly, the need exists for systems and methods for developing compounds efficiently and effectively that modulate a gene, wherein such compounds can be directly developed from nucleotide sequence information. Such compounds are needed to confirm that modulation of a gene that is thought to be involved in a disease or disorder will in fact cause an in vitro or in vivo effect indicative of the origin, development, spread or growth of the disease or disorder.
The present disclosure answers this need by providing systems and methods for automatically generating active oligonucleotide and other compounds, especially antisense compounds, to a target nucleotide sequence via robotic or other automated means. Such active compounds are contacted with a cell culture, cell-free extract, tissue or animal capable of expressing the gene of interest, and subsequent biochemical or biological parameters indicative of the potential gene product function are measured.
These results are compared to those obtained with a control cell system, cell-free extract, tissue or animal which has not been contacted with an active antisense compound in order to determine whether or not modulation of the gene of interest affects a specific cellular function. The resulting active antisense compounds may be used as positive controls when
6 other, non antisense-based agents directed to the same target nucleic acid, or to its gene product, are screened.
It should be noted that embodiments of the invention drawn to gene function analysis and target validation have parameters that are shared with other embodiments of the invention, but also have unique parameters. For example, antisense drug discovery naturally requires that the toxicity of the antisense compounds be manageable, whereas, for gene function analysis or target validation, overt toxicity resulting from the antisense compounds is acceptable unless it interferes with the assay being used to evaluate the effects of treatment with such compounds.
U.S. Patent 5,563,036 to Peterson et al. describes systems and methods of screening for compounds that inhibit the binding of a transcription factor to a nucleic acid.
In a preferred embodiment, an assay portion of the process is stated to be performed by a computer controlled robot.
U.S. Patent 5,708,158 to Hoey describes systems and methods for identifying I S pharmacological agents stated to be useful for diagnosing or treating a disease associated with a gene the expression of which is modulated by a human nuclear factor of activated T
cells. The methods are stated to be particularly suited to high-thoughput screening wherein one or more steps of the process are performed by a computer controlled robot.
U.S. Patents 5,693,463 and 5,716,780 to Edwards et al. describe systems and methods for identifying non-oligonucleotide molecules that specifically bind to a DNA
molecule based on their ability to compete with a DNA-binding protein that recognizes the DNA molecule.
U.S. Patents 5,463,564 and 5,684,711 to Agrafiotis et al. describe computer based iterative processes for generating chemical entities with defined physical, chemical and/or bioactive properties.
SUMMARY OF THE INVENTION
The present invention is directed to automated systems and methods for defining sets of compounds that modulate the expression of target nucleic acid sequences, and generating sets of oligonucleotides that modulate the expression of target nucleic acid sequences. The present invention is also directed to identifying nucleic acid sequences
7 amenable to antisense binding of oligonucleotides to those nucleic acid sequences by the systems and methods of the invention. For purposes of illustration, the present invention is described herein with respect to the production and identification of active antisense oligonucleotides; however, the present invention is not limited to this embodiment.
S The present invention is directed to iterative processes for defining chemical compounds with prescribed sets of physical, chemical and/or biological properties, and to systems for implementing these processes. During each iteration of a process as contemplated herein, a target nucleic acid sequence is provided or selected, and a library of (candidate) virtual compounds is generated in silico (that is in a computer manipulatible and reliable form) according to defined criteria. A library of virtual compounds is generated. These virtual compounds are reviewed and compounds predicted to have particular desired properties are selected. The selected compounds are synthesized, preferably in a robotic, batchwise manner; and then they are robotically assayed for a desired physical, chemical or biological activity in order to identify compounds with the desired properties. Active compounds are, thus, generated and, at the same time, preferred sequences and regions of the target nucleic acid that are amenable to modulation are identified. The preferred compounds of the invention are oiigonucleotides that bind to a target nucleic acid sequence.
In subsequent iterations of the process, second libraries of candidate compounds are generated and/or selected to give rise to a second virtual compound library. Through multiple iterations of the process, a library of target nucleic acid sequences that are tractable to modulation via binding of these compounds to the nucleic acid sequence are identified. Such modulation includes, but is not limited to, antisense technology, gene function analysis and target validation.
The present invention is also directed to processes for validating the function of a gene or the product of the gene comprising generating in silico a library of nucleobase sequences targeted to the gene and robotically assaying a plurality of synthetic compounds having at least some of the nucleobase sequences for effects on biological function.
Further features and advantages of the present invention, as well as the structure and operation of various embodiments of the present invention, are described in detail below with reference to the accompanying drawings. In the drawings, like reference WO 99/53101 PC'T/US99/08268
8 numbers indicate identical or functionally similar elements.
BRIEF DESCRIPTION OF THE DRAWINGS
The present invention will be described with reference to the accompanying drawings, wherein:
Figures l and 2 are a flow diagram of one method according to the present invention depicting the overall flow of data and materials among various elements of the invention.
Figure 3 is a flow diagram depicting the flow of data and materials among elements of step 200 of Figure 1.
Figures 4 and 5 are a flow diagram depicting the flow of data and materials among elements of step 300 of Figure 1.
Figure 6 is a flow diagram depicting the flow of data and materials among elements of step 306 of Figure 4.
Figure 7 is another flow diagram depicting the flow of data and materials among elements of step 306 of Figure 4.
Figure 8 is a another flow diagram depicting the flow of data and materials among elements of step 306 of Figure 4.
Figure 9 is a flow diagram depicting the flow of data and materials among elements of step 350 of Figure 5.
Figures 10 and 11 are flow diagrams depicting a logical analysis of data and materials among elements of step 400 of Figure 1.
Figure 12 is a flow diagram depicting the flow of data and materials among the elements of step 400 of Figure 1.
Figures 13 and 14 are flow diagrams depicting the flow of data and materials among elements of step 500 of Figure l .
Figure 15 is a flow diagram depicting the flow of data and materials among elements of step 600 of Figure 1.
Figure 16 is a flow diagram depicting the flow of data and materials among elements of step 700 of Figure 1.
Figure 17 is a flow diagram depicting the flow of data and materials among the
9 elements of step 1100 of Figure 2.
Figure 18 is a block diagram showing the interconnecting of certain devices utilized in conjunction with a preferred method of the invention;
Figure 19 is a flow diagram showing a representation of data storage in a relational database utilized in conjunction with one method of the invention;
Figure 20 is a flow diagram depicting the flow of data and materials in effecting a preferred embodiment of the invention as set forth in Example 14;
Figure 21 is a flow diagram depicting the flow of data and materials in effecting a preferred embodiment of the invention as set forth in Example 15;
Figure 22 is a flow diagram depicting the flow of data and materials in effecting a preferred embodiment of the invention as set forth in Example 2;
Figure 23 is a pictorial elevation view of a preferred apparatus used to robotically synthesize oligonucleotides; and Figure 24 is a pictorial plan view of an apparatus used to robotically synthesize oligonucleotides.
DETAILED DESCRIPTION OF THE INVENTION
Certain preferred methods of this invention are now described with reference to the flow diagram of Figures l and 2.
1. Target Nucleic Acid Selection. The target selection process, process step 100, provides a target nucleotide sequence that is used to help guide subsequent steps of the process. It is generally desired to modulate the expression of the target nucleic acid for any of a variety of purposes, such as, e.g., drug discovery, target validation and/or gene function analysis.
One of the primary objectives of the target selection process, step 100, is to identify molecular targets that represent significant therapeutic opportunities, to provide new and efficacious means of drug discovery and to determine the function of genes that are uncharacterized except for nucleotide sequence. To meet these objectives, genes are classified based upon specific sets of selection criteria.
One such set of selection criteria concerns the quantity and quality of target nucleotide sequence. There must be sufficient target nucleic acid sequence information available for oligonucleotide design. Moreover, such information must be of sufficient quality to give rise to an acceptable level of confidence in the data to perform the methods described herein. Thus, the data must not contain too many missing or incorrect base entries. In the case of a target sequence that encodes a polypeptide, such errors can often be detected by virtually translating all three reading frames of the sense strand of the target sequence and confirming the presence of a continuous polypeptide sequence having predictable attributes, e.g., encoding a polypeptide of known size, or encoding a polypeptide that is about the same length as a homologous protein. In any event, only a very high frequency of sequence errors will frustrate the methods of the invention; most
10 oligonucleotides to the target sequence will avoid such errors unless such errors occur frequently throughout the entire target sequence.
Another preferred criterion is that appropriate culturable cell lines or other source of reproducible genetic expression should be available. Such cell lines express, or can be induced to express, the gene comprising the target nucleic acid sequence. The oligonucleotide compounds generated by the process of the invention are assayed using such cell lines and, if such assaying is performed robotically, the cell line is preferably tractable to robotic manipulation such as by growth in 96 well plates. Those skilled in the art will recognize that if an appropriate cell line does not exist, it will nevertheless be possible to construct an appropriate cell line. For example, a cell line can be transfected with an expression vector comprising the target gene in order to generate an appropriate cell line for assay purposes.
For gene function analysis, it is possible to operate upon a genetic system having a lack of information regarding, or incomplete characterization of, the biological functions) of the target nucleic acid or its gene product(s). This is a powerful agent of the invention.
A target nucleic acid for gene function analysis might be absolutely uncharacterized, or might be thought to have a function based on minimal data or homology to another gene.
By application of the process of the invention to such a target, active compounds that modulate the expression of the gene can be developed and applied to cells. The resulting cellular, biochemical or molecular biological responses are observed, and this information is used by those skilled in the art to elucidate the function of the target gene.
For target validation and drug discovery, another selection criterion is disease WO 99/53101 PCTlUS99108268
11 association. Candidate target genes are placed into one of several broad categories of known or deduced disease association. Level 1 Targets are target nucleic acids for which there is a strong correlation with disease. This correlation can come from multiple scientific disciplines including, but not limited to, epidemiology, wherein frequencies of gene abnormalities are associated with disease incidence; molecular biology, wherein gene expression and function are associated with cellular events correlated with a disease; and biochemistry, wherein the in vitro activities of a gene product are associated with disease parameters. Because there is a strong therapeutic rationale for focusing on Level 1 Targets, these targets are most preferred for drug discovery and/or target validation.
Level 2 Targets are nucleic acid targets for which the combined epidemiological, molecular biological, and/or biochemical correlation with disease is not so clear as for Level 1. Level 3 Targets are targets for which there is little or no data to directly link the target with a disease process, but there is indirect evidence for such a link, i.e., homology with a Level 1 or Level 2 target nucleic acid sequence or with the gene product thereof. In order not to prejudice the target selection process, and to ensure that the maximum number of nucleic acids actually involved in the causation, potentiation, aggravation, spread, continuance or after-effects of disease states are investigated, it is preferred to examine a balanced mix of Level 1, 2 and 3 target nucleic acids.
In order to carry out drug discovery, experimental systems and reagents shall be available in order for one to evaluate the therapeutic potential of active compounds generated by the process of the invention. Such systems may be operable in vitro (e.g., in vitro models of cell:cell association) or in vivo (e.g., animal models of disease states). It is also desirable, but not obligatory, to have available animal model systems which can be used to evaluate drug pharmacology.
Candidate targets nucleic acids can also classified by biological processes.
For example, programmed cell death ("apoptosis") has recently emerged as an important biological process that is perturbed in a wide variety of diseases.
Accordingly, nucleic acids that encode factors that play a role in the apoptotic process are identified as candidate targets. Similarly, potential target nucleic acids can be classified as being involved in inflammation, autoimmune disorders, cancer, or other pathological or dysfunctional processes.
12 Moreover, genes can often be grouped into families based on sequence homology and biological function. Individual family members can act redundantly, or can provide specificity through diversity of interactions with downstream effectors, or through expression being restricted to specific cell types. When one member of a gene family is associated with a disease process then the rationale for targeting other members of the same family is reasonably strong. Therefore, members of such gene families are preferred target nucleic acids to which the methods and systems of the invention may be applied.
Indeed, the potent specificity of antisense compounds for different gene family members makes the invention particularly suited for such targets (Albert et al., Trends Pharm. Sci., 1994, 15, 250). Those skilled in the art will recognize that a partial or complete nucleotide sequence of such family members can be obtained using the polymerise chain reaction (PCR) and "universal" primers, i.e., primers designed to be common to all members of a given gene family.
PCR products generated from universal primers can be cloned and sequenced or directly sequenced using techniques known in the art. Thus, although nucleotide sequences from cloned DNAs, or from complementary DNAs (cDNAs) derived from mRNAs, may be used in the process of the invention, there is no requirement that the target nucleotide sequence be isolated from a cloned nucleic acid. Any nucleotide sequence, no matter how determined, of any nucleic acid, isolated or prepared in any fashion, may be used as a target nucleic acid in the process of the invention.
Furthermore, although polypeptide-encoding nucleic acids provide the target nucleotide sequences in one embodiment of the invention, other nucleic acids may be targeted as well. Thus, for example, the nucleotide sequences of structural or enzymatic RNAs may be utilized for drug discovery and/or target validation when such RNAs are associated with a disease state, or for gene function analysis when their biological role is not known.
2. Assembly of Target Nucleotide Sequence. Figure 3 is a block diagram detailing the steps of the target nucleotide sequence assembly process, process step 200 in acccordance with one embodiment of the invention. The oligonucleotide design process, process step 300, is facilitated by the availability of accurate target sequence information.
Because of limitations of automated genome sequencing technology, gene sequences are
13 often accumulated in fragments. Further, because individual genes are often being sequenced by independent laboratories using different sequencing strategies, sequence information corresponding to different fragments is often deposited in different databases.
The target nucleic acid assembly process take advantage of computerized homology search algorithms and sequence fragment assembly algorithms to search available databases for related sequence information and incorporate available sequence information into the best possible representation of the target nucleic acid molecule, for example a RNA
transcript.
This representation is then used to design oligonucleotides, process step 300, which can be tested for biological activity in process step 700.
In the case of genes directing the synthesis of multiple transcripts, i.e. by alternative splicing, each distinct transcript is a unique target nucleic acid for purposes of step~300. In one embodiment of the invention, if active compounds specific for a given transcript isoform are desired, the target nucleotide sequence is limited to those sequences that are unique to that transcript isoform. In another embodiment of the invention, if it is desired to modulate two or more transcript isoforms in concert, the target nucleotide sequence is limited to sequences that are shared between the two or more transcripts.
In the case of a polypeptide-encoding nucleic acid, it is generally preferred that full-length cDNA be used in the oligonucleotide design process step 300 (with full-length cDNA being defined as reading from the 5' cap to the poly A tail). Although full-length cDNA is preferred, it is possible to design oligonucleotides using partial sequence information. Therefore it is not necessary for the assembly process to generate a complete cDNA sequence. Further in some cases it may be desirable to design oligonucleotides targeting introns. In this case the process can be used to identify individual introns at process step 220.
The process can be initiated by entering initial sequence information on a selected molecular target at process step 205. In the case of a polypeptide-encoding nucleic acid, the full-length cDNA sequence is generally preferred for use in oligonucleotide design strategies at process step 300. The first step is to determine if the initial sequence information represents the full-length cDNA, decision step 210. In the case where the full-length cDNA sequence is available the process advances directly to the oligonucleotide design step 300. When the full-length cDNA sequence is not available, databases are
14 searched at process step 212 for additional sequence information.
The algorithm preferably used in process steps 212 and 230 is BLAST (Altschul, et al., J. Mol. Biol., 1990, 215, 403), or "Gapped BLAST" (Altschul et al., Nucl.
Acids Res., 1997, 25, 3389). These are database search tools based on sequence homology used to S identify related sequences in a sequence database. The BLAST search parameters are set to only identify closely related sequences. Some preferred databases searched by BLAST
are a combination of public domain and proprietary databases. The databases, their contents, and sources are listed in Table I .
Table l: Database Sources of Target Sequences Database Contents Source NR All non-redundant GenBank, National Center for Biotechnology EMBL, DDBJ and PDB Information at the National Institutes sequences of Health Month All new or revised GenBank, National Center for Biotechnology EMBL, DDBJ and PDB Information at the National Institutes sequences released in the last of Health 30 days Dbest Non-redundant database of National Center for Biotechnology GenBank, EMBL, DDBJ and Information at the National Institutes EST divisions of Health Dbsts Non-redundant database of National Center for Biotechnology GenBank, EMBL, DDBJ and Information at the National Institutes STS divisions of Health Htgs High throughput genomic National Center for Biotechnology sequences Information at the National Institutes of Health When genomic sequence information is available at decision step 215, introns are removed and exons are assembled into continuous sequence representing the cDNA
sequence in process step 220. Exon assembly occurs using the Phragment Assembly Program "Phrap" (Copyright University of Washington Genome Center, Seattle, WA).
The Phrap algorithm analyzes sets of overlapping sequences and assembles them into one continuous sequence referred to as a "contig." The resulting contig is preferably used to search databases for additional sequence information at process step 230. When genomic information is not available, the results of process step 212 are analyzed for individual exons at decision step 225. Exons are frequently recorded individually in databases. If multiple complete exons are identified, they are prferably assembled into a contig using Phrap at process step 250. If multiple complete exons are not identified at decision step 225, then sequences can be analyzed for partial sequence information in decision step 228.
ESTs identified in the database dbEST are examples of such partial sequence information.
If additional partial information is not found, then the process is advanced to process step 10 230 at decision step 228. If partial sequence information is found in process 212 then that information is advanced to process step 230 via decision step 228.
Process step 230, decision step 240, decision step 260 and process step 250 define a loop designed to extend iteratively the amount of sequence information available for targeting. At the end of each iteration of this loop, the results are analyzed in decision
15 steps 240 and 260. If no new information is found then the process advances at decision step 240 to process step 300. If there is an unexpectedly large amount of sequence information identified, suggesting that the process moved outside the boundary of the gene into repetitive genomic sequence, then the process is preferably cycled back one iteration and that sequence is advanced at decision step 240 to process step 300. If a small amount of new sequence information is identified, then the loop is iterated such as by taking the 100 most S-prime (5') and 100 most 3-prime (3') bases and interating them through the BLAST homology search at process step 230. New sequence information is added to the existing contig at process step 250.
3. In Silico Generation of a Set of Nucleobase Sequences and Virtual Oligonucleotides.
For the following steps 300 and 400, they may be performed in the order described below, i.e., step 300 before step 400, or, in an alternative embodiment of the invention, step 400 before step 300. In this alternate embodiment, each oligonucleotide chemistry is first assigned to each oligonucleotide sequence. Then, each combination of oligonucleotide chemistry and sequence is evaluated according to the parameters of step 300. This embodiment has the desirable feature of taking into account the effect of
16 alternative oligonucleotide chemistries on such parameters. For example, substitution of S-methyl cytosine (SMeC or m5c) for cytosine in an antisense compound may enhance the stability of a duplex formed between that compound and its target nucleic acid. Other oligonucleotide chemistries that enhance oligonucleotide:[target nucleic acid]
duplexes are known in the art (see for example, Freier et al., Nucleic Acids Research, 1997, 25, 4429).
As will be appreciated by those skilled in the art, different oligonucleotide chemistries may be preferred for different target nucleic acids. That is, the optimal oligonucleotide chemistry for binding to a target DNA might be suboptimal for binding to a target RNA
having the same nucleotide sequence.
In effecting the process of the invention in the order step 300 before step 400 as seen in Figure 1, from a target nucleic acid sequence assembled at step 200, a list of oligonucleotide sequences is generated as represented in the flowchart shown in Figures 4 and 5. In step 302, the desired oligonucleotide length is chosen. In a preferred embodiment, oligonucleotide length is between from about 8 to about 30, more preferably from about 12 to about 25, nucleotides. In step 304, all possible oligonucleotide sequences of the desired length capable of hybridizing to the target sequence obtained in step 200 are generated. In this step, a series of oligonucleotide sequences are generated, simply by determining the most 5' oligonucleotide possible and "walking" the target sequence in increments of one base until the 3' most oligonucleotide possible is reached.
In step 305, a virtual oligonucleotide chemistry is applied to the nucleobase sequences of step 304 in order to yield a set of virtual oligonucleotides that can be evaluated in silico. Default virtual oligonucleotide chemistries include those that are well-characterized in terms of their physical and chemical properties, e.g., 2'-deoxyribonucleic acid having naturally occurnng bases (A, T, C and G), unmodified sugar residues and a phosphodiester backbone.
4. In Silico Evaluation of Thermodynamic Properties of Virtual Oligonucleotides.
In step 306, a series of thermodynamic, sequence, and homology scores are preferably calculated for each virtual oligonucleotide obtained from step 305.
Thermodynamic properties are calculated as represented in Figure 6. In step 308, the desired thermodynamic properties are selected. As many or as few as desired can be
17 selected; optionally, none will be selected. The desired properties will typically include step 309, calculation of the free energy of the target structure. If the oligonucleotide is a DNA molecule, then steps 310, 312, and 314 are performed. If the oligonucleotide is an RNA molecule, then steps 311, 313 and 315 are performed. In both cases, these steps correspond to calculation of the free energy of intramolecular oligonucleotide interactions, intermolecular interactions and duplex formation. In addition, a free energy of oligonucleotide-target binding is preferably calculated at step 316.
Other thermodynamic and kinetic properties may be calculated for oligonucleotides as represented at step 317. Such other thermodynamic and kinetic properties may include melting temperatures, association rates, dissociation rates, or any other physical property that may be predictive of oligonucleotide activity.
The free energy of the target structure is defined as the free energy needed to disrupt any secondary structure in the target binding site of the targeted nucleic acid. This region includes any intra-target nucleotide base pairs that need to be disrupted in order for an oligonucleotide to bind to its complementary sequence. The effect of this localized disruption of secondary structure is to provide accessibility by the oligonucleotide. Such structures will include double helices, terminal unpaired and mismatched nucleotides, loops, including hairpin loops, bulge loops, internal loops and multibranch loops (Serra et al., Methods in Enzymology, 1995, 259, 242).
The intermolecular free energies refer to inherent energy due to the most stable structure formed by two oligonucleotides; such structures include dimer formation.
Intermolecular free energies should also be taken into account when, for example, two or more oligonucleotides, of different sequence are to be administered to the same cell in an assay.
The intramolecular free energies refer to the energy needed to disrupt the most stable secondary structure within a single oligonucleotide. Such structures include, for example, hairpin loops, bulges and internal loops. The degree of intramolecular base pairing is indicative of the energy needed to disrupt such base pairing.
The free energy of duplex formation is the free energy of denatured oligonucleotide binding to its denatured target sequence. The oligonucleotide-target binding is the total binding involved, and includes the energies involved in opening up
18 intra- and inter- molecular oligonucleotide structures, opening up target structure, and duplex formation.
The most stable RNA structure is predicted based on nearest neighbor analysis (Xia, T., et al., Biochemistry, 1998, 37, 14719-14735; Serra et al., Methods in Enzymology, 1995, 259, 242). This analysis is based on the assumption that stability of a given base pair is determined by the adjacent base pairs. For each possible nearest neighbor combination, thermodynamic properties have been determined and are provided.
For double helical regions, two additional factors need to be considered, an entropy change required to initiate a helix and a entropy change associated with self complementary strands only. Thus, the free energy of a duplex can be calculated using the equation:
4G°T = 0H° - TOS°
where:
dG is the free energy of duplex formation, dH is the enthalpy change for each nearest neighbor, dS is the entropy change for each nearest neighbor, and T is temperature.
The dH and dS for each possible nearest neighbor combination have been experimentally determined. These letter values are often available in published tables.
For terminal unpaired and mismatched nucleotides, enthalpy and entropy measurements for each possible nucleotide combination are also available in published tables. Such results are added directly to values determined for duplex formation. For loops, while the available data is not as complete or accurate as for base pairing, one known model determines the free energy of loop formation as the sum of free energy based on loop size, the closing base pair, the interactions between the first mismatch of the loop with the closing base pair, and additional factors including being closed by AU or UA
or a first mismatch of GA or UU. Such equations may also be used for oligoribonucleotide-target RNA interactions.
The stability of DNA duplexes is used in the case of intra- or intermolecular oligodeoxyribonucleotide interactions. DNA duplex stability is calculated using similar equations as RNA stability, except experimentally determined values differ between nearest neighbors in DNA and RNA and helix initiation tends to be more favorable in
19 DNA than in RNA (SantaLucia et al., Biochemistry, 1996, 35, 3555).
Additional thermodynamic parameters are used in the case of RNA/DNA hybrid duplexes. This would be the case for an RNA target and oligodeoxynucleotide.
Such parameters were determined by Sugimoto et al. (Biochemistry, 1995, 34, 11211 ). In addition to values for nearest neighbors, differences were seen for values for enthalpy of helix initiation.
5. In Silico Evaluation of Target Accessibility Target accessibility is believed to be an important consideration in selecting oligonucleotides. Such a target site will possess minimal secondary structure and thus, will require minimal energy to disrupt such structure. In addition, secondary structure in oligonucleotides, whether inter- or intra-molecular, is undesirable due to the energy required to disrupt such structures. Oligonucleotide-target binding is dependent on both these factors. It is desirable to minimize the contributions of secondary structure based on these factors. The other contribution to oligonucleotide-target binding is binding affinity.
Favorable binding affinities based on tighter base pairing at the target site is desirable.
Following the calculation of thermodynamic properties ending at step 317, the desired sequence properties to be scored are selected at step 324. As many or as few as desired can be selected; optionally, none will be selected. These properties include the number of strings of four guanosine residues in a row at step 325 or three guanosine in a row at step 326, the length of the longest string of adenosines at step 327, cytidines at step 328 or uridines or thymidines at step 329, the length of the longest string of purines at step 330 or pyrimidine at step 331, the percent composition of adenosine at step 332, cytidine at step 333, guanosine at step 334 or uridines or thymidines at step 335, the percent composition of purines at step 336 or pyrimidines at step 337, the number of CG
dinucleotide repeats at step 338, CA dinucleotide repeats at step 339 or UA or TA
dinucleotide repeats at step 340. In addition, other sequence properties may be used as found to be relevant and predictive of antisense efficacy, as represented at step 341.
These sequence properties may be important in predicting oligonucleotide activity, or lack thereof. For example, U.S. Patent 5,523,389 discloses oligonucleotides containing stretches of three or four guanosine residues in a row. Oligonucleotides having such sequences may act in a sequence-independent manner. For an antisense approach, such a mechanism is not usually desired. In addition, high numbers of dinucleotide repeats may be indicative of low complexity regions which may be present in large numbers of unrelated genes. Unequal base composition, for example, 90% adenosine, can also give non-specific effects. From a practical standpoint, it may be desirable to remove S oligonucleotides that possess long stretches of other nucleotides due to synthesis considerations. Other sequences properties, either listed above or later found to be of predictive value may be used to select oligonucleotide sequences.
Following step 341, the homology scores to be calculated are selected in step 342.
Homology to nucleic acids encoding protein isoforms of the target, as represented at step 10 343, may be desired. For example, oligonucleotides specific for an isoform of protein kinase C can be selected. Also, oligonucleotides can be selected to target multiple isoforms of such genes. Homology to analogous target sequences, as represented at step 344, may also be desired. For example, an oligonucleotide can be selected to a region common to both humans and mice to facilitate testing of the oligonucleotide in both 15 species. Homology to splice variants of the target nucleic acid, as represented at step 345, may be desired. In addition, it may be desirable to determine homology to other sequence variants as necessary, as represented in step 346.
Following step 346, from which scores were obtained in each selected parameter, a desired range is selected to select the most promising oligonucleotides, as represented at
20 step 347. Typically, only several parameters will be used to select oligonucleotide sequences. As structure prediction improves, additional parameters may be used. Once the desired score ranges are chosen, a list of all oligonucleotides having parameters falling within those ranges will be generated, as represented at step 348.
6. Targeting Oligonucleotides to Functional Regions of a Nucleic Acid.
It may be desirable to target oligonucleotide sequences to specific functional regions of the target nucleic acid. A decision is made whether to target such regions, as represented in decision step 349. If it is desired to target functional regions then process step 350 occurs as seen in greater detail in Figure 9. If it is not desired then the process proceeds to step 375.
In step 350, as seen in Figure 9, the desired functional regions are selected.
Such regions include the transcription start site or 5' cap at step 353, the 5' untranslated region WO 99/53101 PCTlUS99/08268
21 at step 354, the start codon at step 355, the coding region at step 356, the stop codon at step 357, the 3' untranslated region at step 358, 5' splice sites at step 359 or 3' splice sites at step 360, specific exons at step 361 or specific introns at step 362, mRNA
stabilization signal at step 363, mRNA destabilization signal at step 364, poly-adenylation signal at step 365, poly-A addition site at step 366, poly-A tail at step 367, or the gene sequence 5' of known pre-mRNA at step 368. In addition, additional functional sites may be selected, as represented at step 369.
Many functional regions are important to the proper processing of the gene and are attractive targets for antisense approaches. For example, the AUG start codon is commonly targeted because it is necessary to initiate translation. In addition, splice sites are thought to be attractive targets because these regions are important for processing of the mRNA. Other known sites may be more accessible because of interactions with protein factors or other regulatory molecules.
After the desired functional regions are selected and determined, then a subset of all previously selected oligonucleotides are selected based on hybridization to only those desired functional regions, as represented by step 370.
7. Uniform Distribution of Oligonucleotides.
Whether or not targeting functional sites is desired, a large number of oligonucleotide sequences may result from the process thus far. In order to reduce the number of oligonucleotide sequences to a manageable number, a decision is made whether to uniformly distribute selected oligonucleotides along the target, as represented in step 375.
A uniform distribution of oligonucleotide sequences will aim to provide complete coverage throughout the complete target nucleic acid or the selected functional regions. A
computer-based program is used to automate the distribution of sequences, as represented in step 380. Such a program factors in parameters such as length of the target nucleic acid, total number of oligonucleotide sequences desired, oligonucleotide sequences per unit length, number of oligonucleotide sequences per functional region. Manual selection of oligonucleotide sequences is also provided for by step 385. In some cases, it may be desirable to manually select oligonucleotide sequences. For example, it may be useful to determine the effect of small base shifts on activity. Once the desired number of oligonucleotide sequences is obtained either from step 380 or step 385, then these
22 oligonucleotide sequences are passed onto step 400 of the process, where oligonucleotide chemistries are assigned.
8. Assignment of Actual Oligonucleotide Chemistry.
Once a set of select nucleobase sequences has been generated according to the preceding process and decision steps, actual oligonucleotide chemistry is assigned to the sequences. An "actual oligonucleotide chemistry" or simply "chemistry" is a chemical motif that is common to a particular set of robotically synthesized oligonucleotide compounds. Preferred chemistries include, but are not limited to, oligonucleotides in which every linkage is a phosphorothioate linkage, and chimeric oligonucleotides in which a defined number of 5' and/or 3' terminal residues have a 2'-methoxyethoxy modification.
Chemistries can be assigned to the nucleobase sequences during general procedure step 400 (Figure 1). The logical basis for chemistry assignment is illustrated in Figures 10 and 11 and an iterative routine for stepping through an oligonucleotide nucleoside by nucleoside is illustrated in Figure 12. Chemistry assignment can be effected by 1 S assignment directly into a word processing program, via an interactive word processing program or via automated programs and devices. In each of these instances, the output file is selected to be in a format that can serve as an input file to automated synthesis devices.
9. Oligonucleotide Compounds.
In the context of this invention, in reference to oligonucleotides, the term "oligonucleotide" is used to refer to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetics thereof. Thus this term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally-occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms, i.e., phosphodiester linked A, C, G, T and U nucleosides, because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target and increased stability in the presence of nucleases.
The oligonucleotide compounds in accordance with this invention can be of various lengths depending on various parameters, including but not limited to those discussed above in reference to the selection criteria of general procedure 300. For use as
23 antisense oligonucleotides compounds of the invention preferably are from about 8 to about 30 nucleobases in length (i.e. frorn about 8 to about 30 linked nucleosides).
Particularly preferred are antisense oligonucleotides comprising from about 12 to about 25 nucleobases. A discussion of antisense oligonucleotides and some desirable modifications can be found in De Mesmaeker et al., Acc. Chem. Res., 1995, 28, 366. Other lengths of oligonucleotides might be selected for non-antisense targeting strategies, for instance using the oliganucleotides as ribozymes. Such ribozymes normally require oligonucleotides of longer length as is known in the art.
A nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocyclic base. The two most common classes of such heterocyclic bases are the purines and the pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a normal (where normal is defined as being found in RNA and DNA) pentofuranosyl sugar, the phosphate group can be linked to either the 2', 3' or 5' hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
In turn the respective ends of this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally preferred.
Within the oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide. The normal linkage or backbone of RNA
and DNA is a 3' to 5' phosphodiester linkage.
Specific examples of preferred oligonucleotides useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages.
As defined in this specification, oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
10. Selection of Oligonucleotide Chemistries.
In a general logic scheme as illustrated in Figures 10 and 11, for each nucleoside position, the user or automated device is interrogated first for a base assignment, followed
24 by a sugar assignment, a linker assignment and finally a conjugate assignment.
Thus for each nucleoside, at process step 410 a base is selected. In selecting the base, base chemistry 1 can be selected at process step 412 or one or more alternative bases are selected at process steps 414, 416 and 418. After base selection is effected, the sugar portion of the nucleoside is selected. Thus for each nucleoside, at process step 420 a sugar is selected that together with the select base will complete the nucleoside.
In selecting the sugar, sugar chemistry 1 can be selected at process 422 or one or more alternative sugars are selected at process steps 424, 426 and 428. For each two adjacent nucleoside units, at process step 430, the internucleoside linker is selected. The linker chemistry for the internucleoside linker can be linker chemistry 1 selected at process step 432 or one or more alternative internucleoside linker chemistries are selected at process steps 434, 436 and 438.
In addition to the base, sugar and internucleoside linkage, at each nucleoside position, one or more conjugate groups can be attached to the oligonucleotide via 1 S attachment to the nucleoside or attachment to the internucleoside linkage.
The addition of a conjugate group is integrated at process step 440 and the assignment of the conjugate group is effected at process step 450.
For illustrative purposes in Figures 10 and 11, for each of the bases, the sugars, the internucleoside linkers, or the conjugates, chemistries 1 though n are illustrated. As described in this specification, it is understood that the number of alternate chemistries between chemistry 1 and alternative chemistry n, for each of the bases, the sugars, the internucleoside linkages and the conjugates, is variable and includes, but is not limited to, each of the specific alternative bases, sugar, internucleoside linkers and conjugates identified in this specification as well as equivalents known in the art.
Utilizing the logic as described in conjunction with Figures 10 and 11, chemistry is assigned, as is shown in Figure 12, to the list of oligonucleotides from general procedure 300. In assigning chemistries to the oligonucleotides in this list, a pointer can be set at process step 452 to the first oligonucleotide in the list and at step 453 to the first nucleotide of that first oligonucleotide. The base chemistry is selected at step 410, as described above, the sugar chemistry is selected at step 420, also as described above, followed by selection of the internucleoside linkage at step 430, also as described above.

At decision 440, the process branches depending on whether a conjugate will be added at the current nucleotide position. If a conjugate is desired, the conjugate is selected at step 450, also as described above.
Whether or not a conjugate was added at decision step 440, an inquiry is made at 5 decision step 454. This inquiry asks if the pointer resides at the last nucleotide in the current oligonucleotide. If the result at decision step 454 is "No," the pointer is moved to the next nucleotide in the current oligonucleotide and the loop including steps 410, 420, 430, 440 and 454 is repeated. This loop is reiterated until the result at decision step 454 is "Yes."
10 When the result at decision step 454 is "Yes," a query is made at decision step 460 concerning the location of the pointer in the list of oligonucleotides. If the pointer is not at the last oligonucleotide of the list, the "No" path of the decision step 4b0 is followed and the pointer is moved to the first nucleotide of the next oligonucleotide in the list at process step 458. With the pointer set to the next oligonucleotide in the list, the loop that starts at 15 process steps 453 is reiterated. When the result at decision step 460 is "Yes," chemistry has been assigned to all of the nucleotides in the list of oligonucleotides.
11. Description of Oligonucleotide Chemistries.
As is illustrated in Figure 10, for each nucleoside of an oligonucleotide, chemistry selection includes selection of the base forming the nucleoside from a large palette of 20 different base units available. These may be "modified" or "natural" bases (also reference herein as nucleobases) including the natural purine bases adenine (A) and guanine (G), and the natural pyrimidine bases thymine (T), cytosine (C) and uracil (U). They further can include modified nucleobases including other synthetic and natural nucleobases such as S-methylcytosine (5-me-C), S-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-
25 aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, S-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo uracils and cytosines particularly 5-bromo, 5-trifluoromethyl and other S-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and z6 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in United States Patent No. 3,687,808, those disclosed in the Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J.L, ed. John Wiley &
Sons, 1990, those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y.S., Chapter 15, Antisense Research and Applications, pages 289-302, Crooke, S.T. and Lebleu, B., ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyl-adenine, S-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C
(Sanghvi, Y.S., Crooke, S.T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred for selection as the base.
These are particularly useful when combined with a 2'-O-methoxyethyl sugar modifications, described below.
Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Patent 3,687,808, as well as U.S. Patents 4,845,205;
5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255;
5,484,908;
5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617;
and 5,681,941, each of which is incorporated herein by reference in its entirety.
Reference is also made to allowed United States patent application 08/762,488, filed on December 10, 1996, commonly owned with the present application and which is incorporated herein by reference in its entirety.
In selecting the base for any particular nucleoside of an oligonucleotide, consideration is first given to the need of a base for a particular specificity for hybridization to an opposing strand of a particular target. Thus if an "A"
base is required, adenine might be selected however other alternative bases that can effect hybridization in a manner mimicking an "A" base such as 2-aminoadenine might be selected should other consideration, e.g., stronger hybridization (relative to hybridization achieved with adenine), be desired.

As is illustrated in Figure 10, for each nucleoside of an oligonucleotide, chemistry selection includes selection of the sugar forming the nucleoside from a large palette of different sugar or sugar surrogate units available. These may be modified sugar groups, for instance sugars containing one or more substituent groups. Preferred substituent S groups comprise the following at the 2' position: OH; F; O-, S-, or N-alkyl;
O-, S-, or N-alkenyl; or O, S- or N-alkynyl; wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C, to C,o alkyl or Cz to C,a alkenyl and alkynyl.
Particularly preferred are O~(CHz)nOlmCH3~ O(CHz)nOCH3, O(CHz)n~z. O(CHz)~~~'Hs. O(CHz)"O~z. ~d O(CHz)"ON[(CHz)"CH3)]z, where n and m are from 1 to about 10. Other preferred substituent groups comprise one of the following at the 2' position: C, to C,o lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SOZCH3, ONOz, NOz, N3, NHz, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA
cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. A
preferred modification includes 2'-methoxyethoxy (2'-O-CHZCHZOCH3, also known as 2'-O-(2-methoxyethyl), 2'-O-methoxyethyl, or 2'-MOE) (Martin et al., Helv. Chim.
Acta, 1995, 78, 486) i.e., an alkoxyalkoxy group. A further preferred modification includes 2'-dimethylamino oxyethoxy, i.e., a O(CHz)zON(CH3)z group, also known as 2'-DMAOE, as described in co-owned United States patent application Serial Number 09/016,520, filed on January 30, 1998, which is incorporated herein by reference in its entirety.
Other preferred modifications include 2'-methoxy (2'-O-CH3), 2'-aminopropoxy (2'-OCHZCH2CHzNHz) and 2'-fluoro (2'-F). Similar modifications may also be made at other positions on the sugar group, particularly the 3' position of the sugar on the 3' terminal nucleotide or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide. The nucleosides of the oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
Representative United States patents that teach the preparation of such modified sugars structures include, but are not limited to, U.S. Patents 4,981,957;
5,118,800;
5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134;
5,567,811;

5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053 5,639,873; 5,646,265;
5,658,873;
5,670,633; and 5,700,920, certain of which are commonly owned with the present application, each of which is incorporated herein by reference in its entirety, together with allowed United States patent application 08/468,037, filed on June 5, 1995, which is commonly owned with the present application and which is incorporated herein by reference in its entirety.
As is illustrated in Figure 10, for each adjacent pair of nucleosides of an oligonucleotide, chemistry selection includes selection of the internucleoside linkage.
These internucleoside linkages are also referred to as linkers, backbones or oligonucleotide backbones. For forming these nucleoside linkages, a palette of different internucleoside linkages or backbones is available. These include modified oligonucleotide backbones, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalklyphosphotriesters, and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'. Various salts, mixed salts and free acid forms are also included.
Representative United States patents that teach the preparation of the above phosphorus containing linkages include, but are not limited to, U.S. Patents 3,687,808;
4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019;
5,278,302;
5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677;
5,476,925;
5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361;
5,625,050;
and 5,697,248, certain of which are commonly owned with this application, each of which is incorporated herein by reference in its entirety.
Preferred internucleoside linkages for oligonucleotides that do not include a phosphorus atom therein, i.e., for oligonucleosides, have backbones that are formed by short chain alkyl or cycloalkyl intersugar linkages, mixed heteroatom and alkyl or cycloalkyl intersugar linkages, or one or more short chain heteroatomic or heterocyclic intersugar linkages. These include those having morpholino linkages (formed in part from WO 99!53101 PCT/US99/08268 the sugar portion of a nucleoside); siloxane backbones; sulfide, suifoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones;
methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CHZ component parts.
Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Patents 5,034,506;
5,166,315;
5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938;
5,434,257;
5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240;
5,610,289;
5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360;
5,677,437;
and 5,677,439, certain of which are commonly owned with this application, each of which is incorporated herein by reference in its entirety.
In other preferred oligonucleotides, i.e., oligonucleotide mimetics, both the sugar and the intersugar linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups. The base units are maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar-phosphate backbone of an oligonucleotide is replaced with an amide-containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
Representative United States patents that teach the preparation of PNA compounds include, but are not limited to, U.S.: 5,539,082; 5,714,331; and 5,719,262, each of which is incorporated herein by reference in its entirety. Further teaching of PNA compounds can be found in Nielsen et al., Science, 1991, 254, 1497.
For the internucleoside linkages, the most preferred embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular -CHz-NH-O-CHZ-, -CHZ-N(CH3)-O-CHZ-[known as a methylene (methylimino) or MMI backbone], -CHZ-O-N(CH3)-CHZ-,- CHz-N(CH3)-N(CH3)-CHz- and -O-N(CH3)-CHZ-CHZ- (wherein the native phosphodiester backbone is represented as -O-P-O-CHZ-) of the above referenced U.S. patent 5,489,677, and the amide backbones of the above referenced U.S. patent 5,602,240. Also preferred are oligonucleotides having morpholino backbone structures of the above-referenced U.S.
Patent 5,034,506.
In attaching a conjugate group to one or more nucleosides or internucleoside 5 linkages of an oligonucleotide, various properties of the oligonucleotide are modified.
Thus modification of the oligonucleotides of the invention to chemically link one or more moieties or conjugates to the oligonucleotide are intended to enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad.
10 Sci. USA, 1989, 86, 6553), cholic acid (Manoharan et al., Bioorg. Med.
Chem. Let., 1994, 4, 1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N. Y.
Acad. Sci., 1992, 660, 306; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10, 111;
15 Kabanov et al., FEBS Lett., 1990, 259, 327; Svinarchuk et al., Biochimie, 1993, 75, 49), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651; Shea et al., Nucl. Acids Res., 1990, 18, 3777), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969), or adamantane acetic acid 20 (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923).
Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Patents 4,828,979;
25 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538;
5,578,717, 5,580,731;
5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603;
5,512,439;
5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737;
4,824,941;
4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136;
5,082,830;
5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250;
5,292,873;
30 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475;
5,512,667; 5,514,785;
5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696;
5,599,923;

5,599,928 and 5,688,941, certain of which are commonly owned with the present application, and each of which is herein incorporated by reference in its entirety.
12. Chimeric Compounds.
It is not necessary for all positions in a given compound to be uniformly modified.
In fact, more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an oligonucleotide. The present invention also includes compounds which are chimeric compounds. "Chimeric" compounds or "chimeras," in the context of this invention, are compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an oligonucleotide compound.
These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
By way of example, RNase H is a cellular endonuclease which cleaves the RNA
strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
Chimeric antisense compounds of the invention may be formed as composite structures representing the union of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above.
Such compounds have also been referred to in the art as "hybrids" or "gapmers".
Representative United States patents that teach the preparation of such hybrid structures include, but are not limited to, U.S. Patents 5,013,830; 5,149,797; 5,220,007;
5,256,775;
5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356;
and 5,700,922, certain of which are commonly owned with the present application and each of which is incorporated herein by reference in its entirety, together with commonly owned and allowed United States patent application serial number 08/465,880, filed on June 6, 1995, which is incorporated herein by reference in its entirety.
13. Description of Automated Oligonucleotide Synthesis.
In the next step of the overall process (illustrated in Figures 1 and 2), oligonucleotides are synthesized on an automated synthesizer. Although many devices may be employed, the synthesizer is preferably a variation of the synthesizer described in United States patents 5,472,672 and 5,529,756, each of which is incorporated herein by reference in its entirety. The synthesizer described in those patents is modified to include movement in along the Y axis in addition to movement along the X axis. As so modified, a 96-well array of compounds can be synthesized by the synthesizer. The synthesizer further includes temperature control and the ability to maintain an inert atmosphere during all phases of synthesis. The reagent array delivery format employs orthogonal X-axis motion of a matrix of reaction vessels and Y-axis motion of an array of reagents. Each reagent has its own dedicated plumbing system to eliminate the possibility of cross-contamination of reagents and line flushing and/or pipette washing. This in combined with a high delivery speed obtained with a reagent mapping system allows for the extremely rapid delivery of reagents. This further allows long and complex reaction sequences to be performed in an efficient and facile manner.
The software that operates the synthesizer allows the straightforward programming of the parallel synthesis of a large number of compounds. The software utilizes a general synthetic procedure in the form of a command (.cmd) file, which calls upon certain reagents to be added to certain wells via lookup in a sequence (.seq) file.
The bottle position, flow rate, and concentration of each reagent is stored in a lookup table (.tab) file.
Thus, once any synthetic method has been outlined, a plate of compounds is made by permutating a set of reagents, and writing the resulting output to a text file. The text file is input directly into the synthesizer and used for the synthesis of the plate of compounds.
The synthesizer is interfaced with a relational database allowing data output related to the synthesized compounds to be registered in a highly efficient manner.
Building of the .seq, .cmd and .tab files is illustrated in Figure 13. Thus as a part of the general oligonucleotide synthesis procedure 500, for each linker chemistry at process step 502, a synthesis file, i.e., a .cmd file, is built at process step 504. This file can be built fresh to reflect a completely new set of machine commands reflecting a set of chemical synthesis steps or it can modify an existing file stored at process step 504 by editing that stored file in process step 508. The .cmd files are built using a word processor and a command set of instructions as outlined below.
It will be appreciated that the preparation of control software and data files is within the routine skill of persons skilled in annotated nucleotide synthesis.
The same will depend upon the hardware employed, the chemistries adopted and the design paradigm selected by the operator.
In a like manner to the building the .cmd files, .tab files are built to reflect the necessary reagents used in the automatic synthesizer for the particular chemistries that have been selected for the linkages, bases, sugars and conjugate chemistries.
Thus for each of a set of these chemistries at process step 510, a .tab file is built at process step 512 and stored at process step 514. As with the .cmd files, an existing .tab file can be edited at process step 516.
Both the .cmd files and the .tab files are linked together at process step 518 and stored for later retrieval in an appropriate sample database 520. Linking can be as simple as using like file names to associate a .cmd file to its appropriate .tab file, e.g., synthesis_1.cmd is linked to synthesis_1.tab by use of the same preamble in their names.
The automated, mufti-well parallel array synthesizer employs a reagent array delivery format, in which each reagent utilized has a dedicated plumbing system. As seen in Figures 23 and 24, an inert atmosphere 522 is maintained during all phases of a synthesis. Temperature is controlled via a thermal transfer plate 524, which holds an injection molded reaction block 526. The reaction plate assembly slides in the X-axis direction, while for example eight nozzle blocks (528, 530, 532, 534, 536, 538, 540 and 542) holding the reagent lines slide in the Y-axis direction, allowing for the extremely rapid delivery of any of 64 reagents to 96 wells. In addition, there are for example, six banks of fixed nozzle blocks (544, 546, 548, 550, 552 and 554) which deliver the same reagent or solvent to eight wells at once, for a total of 72 possible reagents.
In synthesizing oligonucleotides for screening, the target reaction vessels, a 96 well plate 556 (a 2-dimensional array), moves in one direction along the X axis, while the series of independently controlled reagent delivery nozzles (528, 530, 532, 534, 536, 538, 540 and 542) move along the Y-axis relative to the reaction vessel 558. As the reaction plate 556 and reagent nozzles (528, 530, 532, 534, 536, 538, 540 and 542) can be moved independently at the same time, this arrangement facilitates the extremely rapid delivery of up to 72 reagents independently to each of the 96 reaction vessel wells.
The system software allows the straightforward programming of the synthesis of a large number of compounds by supplying the general synthetic procedure in the form of the command file to call upon certain reagents to be added to specific wells via lookup in the sequence file with the bottle position, flow rate, and concentration of each reagent being stored in the separate reagent table file. Compounds can be synthesized on various scales. For oligonucleotides, a 200 nmole scale is typically selected while for other compounds larger scales, as for example a 10 mole scale (3-5 mg), might be utilized.
The resulting crude compounds are generally >80% pure, and are utilized directly for high throughput screening assays. Alternatively, prior to use the plates can be subjected to quality control (see general procedure 600 and Example 9) to ascertain their exact purity.
Use of the synthesizer results in a very efficient means for the parallel synthesis of compounds for screening.
The software inputs accept tab delimited text files {as discussed above for file 504 and 512) from any text editor. A typical command file, a .cmd file, is shown in Example 3 at Table 2. Typical sequence files, .seq files, are shown in Example 3 at Tables 3 and 4 (.SEQ file), and a typical reagent file, a .tab file, is shown in Example 3 at Table 5. Table 3 illustrates the sequence file for an oligonucleotide having 2'-deoxy nucleotides at each position with a phosphorothioate backbone throughout. Table 4 illustrates the sequence file for an oligonucleotide, again having a phosphorothioate backbone throughout, however, certain modified nucleoside are utilized in portions of the oligonucleotide. As shown in this table, 2'-O-(2-methoxyethyl) modified nucleosides are utilized in a first region (a wing) of the oligonucleotide, followed by a second region (a gap) of 2'-deoxy nucleotides and finally a third region (a further wing) that has the same chemistry as the first region. Typically some of the wells of the 96 well plate 556 may be left empty (depending on the number of oligonucleotides to be made during an individual synthesis) or some of the wells may have oligonucleotides that will serve as standards for comparison or analytical purposes.
Prior to loading reagents, moisture sensitive reagent lines are purged with argon at 522 for 20 minutes. Reagents are dissolved to appropriate concentrations and installed on the synthesizer. Large bottles, collectively identified as 558 in Figure 23 (containing 8 delivery lines) are used for wash solvents and the delivery of general activators, trityl group cleaving reagents and other reagents that may be used in multiple wells during any particular synthesis. Small septa bottles, collectively identified as 560 in Figure 23, are utilized to contain individual nucleotide amidite precursor compounds. This allows for anhydrous preparation and efficient installation of multiple reagents by using needles to 10 pressurize the bottle, and as a delivery path. After all reagents are installed, the lines are primed with reagent, flow rates measured, then entered into the reagent table (.tab file). A
dry resin loaded plate is removed from vacuum and installed in the machine for the synthesis.
The modified 96 well polypropylene plate 556 is utilized as the reaction vessel.
15 The working volume in each well is approximately 700 ~sl. The bottom of each well is provided with a pressed-fit 20 ~cm polypropylene frit and a long capillary exit into a lower collection chamber as is illustrated in Figure S of the above referenced United States Patent 5,372,672. The solid support for use in holding the growing oligonucleotide during synthesis is loaded into the wells of the synthesis plate 556 by pipetting the desired 20 volume of a balanced density slurry of the support suspended in an appropriate solvent, typically an acetonitrile-methylene chloride mixture. Reactions can be run on various scales as for instance the above noted 200 nmoie and 10 ~cmol scales. For oligonucleotide synthesis a CPG support is preferred, however other medium loading polystyrene-PEG
supports such as TENTAGELT"' or ARGOGELT"" can also be used.
25 As seen in Figure 24, the synthesis plate is transported back and forth in the X-direction under an array of 8 moveable banks (530, 532, 534, 536, 538, 540, 542 and 544) of 8 nozzles (64 total) in the Y-direction, and 6 banks (544, 546, 548, 550, 552 and 554) of 48 fixed nozzles, so that each well can receive the appropriate amounts of reagents and/or solvents from any reservoir (large bottle or smaller septa bottle). A sliding balloon-type 30 seal 562 surrounds this nozzle array and joins it to the reaction plate headspace 564. A
slow sweep of nitrogen or argon 522 at ambient pressure across the plate headspace is used to preserve an anhydrous envirorunent.
The liquid contents in each well do not drip out until the headspace pressure exceeds the capillary forces on the liquid in the exit nozzle. A slight positive pressure in the lower collection chamber can be added to eliminate residual slow leakage from filled wells, or to effect agitation by bubbling inert gas through the suspension. In order to empty the wells, the headspace gas outlet valve is closed and the internal pressure raised to about 2 psi. Normally, liquid contents are blown directly to waste 566.
However, a 96 well microtiter plate can be inserted into the lower chamber beneath the synthesis plate in order to collect the individual well eluents for spectrophotometric monitoring (trityl, etc.) of reaction progress and yield.
The basic plumbing scheme for the machine is the gas-pressurized delivery of reagents. Each reagent is delivered to the synthesis plate through a dedicated supply line, collectively identified at 568, solenoid valve collectively identified at 570 and nozzle, collectively identified at 572. Reagents never cross paths until they reach the reaction well. Thus, no line needs to be washed or flushed prior to its next use and there is no possibility of cross-contamination of reagents. The liquid delivery velocity is sufficiently energetic to thoroughly mix the contents within a well to form a homogeneous solution, even when employing solutions having drastically different densities. With this mixing, once reactants are in homogeneous solution, diffusion carries the individual components into and out of the solid support matrix where the desired reaction takes place. Each reagent reservoir can be plumbed to either a single nozzle or any combination of up to 8 nozzles. Each nozzle is also provided with a concentric nozzle washer to wash the outside of the delivery nozzles in order to eliminate problems of crystallized reactant buildup due to slow evaporation of solvent at the tips of the nozzles. The nozzles and supply lines can be primed into a set of dummy wells directly to waste at any time.
The entire plumbing system is fabricated with teflon tubing, and reagent reservoirs are accessed via syringe needle/septa or direct connection into the higher capacity bottles.
The septum vials 560 are held in removable 8-bottle racks to facilitate easy setup and cleaning. The priming volume for each line is about 350 ~cl. The minimum delivery volume is about 2 ~cl, and flow rate accuracy is t5%. The actual amount of material delivered depends on a timed flow of liquid. The flow rate for a particular solvent will depend on its viscosity and wetting characteristics of the teflon tubing. The flow rate (typically 200-350 ~cl per sec) is experimentally determined, and this information is contained in the reagent table setup file.
Heating and cooling of the reaction block 526 is effected utilizing a recirculating S heat exchanger plate 524, similar to that found in PCR thermocyclers, that nests with the polypropylene synthesis plate 556 to provide good thermal contact. The liquid contents in a well can be heated or cooled at about 10°C per minute over a range of +5 to +80°C, as polypropylene begins to soften and deform at about 80°C. For temperatures greater than this, a non-disposable synthesis plate machined from stainless steel or morel with replaceable frits can be utilized.
The hardware controller can be any of a wide variety, but conveniently can be designed around a set of three 1 MHz 86332 chips. This controller is used to drive the single X-axis and 8 Y-axis stepper motors as well as provide the timing functions for a total of 154 solenoid valves. Each chip has 16 bidirectional timer I/O and 8 interrupt channels in its timer processing unit (TPU). These are used to provide the step and direction signals, and to read 3 encoder inputs and 2 limit switches for controlling up to three motors per chip. Each 86332 chip also drives a serial chain of 8 darlington array chips to provide power to 64 valves with msec resolution. The controller communicates with the Windows software interface program running on a PC via a Hz serial channel, and uses an elementary instruction set to communicate valve number, time open, motor number and position data.
The three components of the software program that run the array synthesizer are the generalized procedure or command (.cmd) file which specifies the synthesis instructions to be performed, the sequence (.seq) file which specifies the scale of the reaction and the order in which variable groups will be added to the core synthon, and the reagent table (.tab) file which specifies the name of a chemical, its location (bottle number), flow rate, and concentration are utilized in conjunction with a basic set of command instructions.
One basic set of command instructions can be:
ADD
IF {block of instructions} END IF

REPEAT {block of instructions} END REPEAT
PRIME, NOZZLE WASH
WAIT, DRAIN
LOAD, REMOVE
NEXT SEQUENCE
LOOP BEGIN, LOOP END
The ADD instruction has two forms, and is intended to have the look and feel of a standard chemical equation. Reagents are specified to be added by a molar amount if the number proceeds the name identifier, or by an absolute volume in microliters if the number follows the identifier. The number of reagents to be added is a parsed list, separated by the "+" sign. For variable reagent identifiers, the key word, <seq>, means look in the sequence table for the identity of the reagent to be added, while the key word, <act>, means add the reagent which is associated with that particular <seq>.
Reagents are delivered in the order specified in the list.
Thus:

means: Add 300 ,ul of the named reagent acetonitrile; ACN to each well of active synthesis ADD <seq> 300 means: If the sequence pointer in the .seq file is to a reagent in the list of reagents, independent of scale, add 300 ,ul of that particular reagent specified for that well.
ADD 1.1 PYR + 1.0 <seq> + I .1 <act 1 >
means: If the sequence pointer in the .seq file is to a reagent in the list of acids in the Class ACIDS_l, and PYR is the name of pyridine, and ethyl chloroformate is defined in the .tab file to activate the class, ACIDS l, then this instruction means:
Add 1.1 equiv. pyridine 1.0 equiv. of the acid specified for that well and 1.1 equiv. of the activator, ethyl chloroformate The IF command allows one to test what type of reagent is specified in the <seq> variable and process the succeeding block of commands accordingly.
Thus:
ACYLATION {the procedure name}
BEGIN
IF CLASS = ACIDS_1 ADD 1.0 <seq> + 1.1 <actl > + 1.1 PYR

ENDIF
IF CLASS = ACIDS 2 ADD 1.0 <seq> + 1.2 <actl> + 1.2 TEA
ENDIF

END
1 S means: Operate on those wells for which reagents contained in the Acid_l class are specified, WAIT 60 sec, then operate on those wells for which reagents contained in the Acid 2 class are specified, then WAIT 60 sec longer, then DRAIN the whole plate. Note that the Acid 1 group has reacted for a total of 120 sec, while the Acid 2 group has reacted for only 60 sec.
The REPEAT command is a simple way to execute the same block of commands multiple times.
Thus:
WASH_1 {the procedure name}
BEGIN

END REPEAT
END
means: repeats the add acetonitrile and drain sequence for each well three times.
The PRIME command will operate either on specific named reagents or on nozzles which will be used in the next associated <seq> operation. The ,ul amount dispensed into a prime port is a constant that can be specified in a config.dat file.
The NOZZLE WASH command for washing the outside of reaction nozzles free from residue due to evaporation of reagent solvent will operate either on specific named 5 reagents or on nozzles which have been used in the preceding associated <seq> operation.
The machine is plumbed such that if any nozzle in a block has been used, all the nozzles in that block will be washed into the prime port.
The WAIT and DRAIN commands are by seconds, with the drain command applying a gas pressure over the top surface of the plate in order to drain the wells.
10 The LOAD and REMOVE commands are instructions for the machine to pause for operator action.
The NEXT SEQUENCE command increments the sequence pointer to the next group of substituents to be added in the sequence file. The general form of a .seq file entry is the definition:
Well No Weli ID Scate Sequence The sequence information is conveyed by a series of columns, each of which represents a variable reagent to be added at a particular position. The scale (~cmole) variable is included so that reactions of different scale can be run at the same time if desired. The reagents are defined in a lookup table (the .tab file), which specifies the name of the reagent as referred to in the sequence and command files, its location (bottle number), flow rate, and concentration. This information is then used by the controller software and hardware to determine both the appropriate slider motion to position the plate and slider arms for delivery of a specific reagent, as well as the specific valve and time required to deliver the appropriate reagents. The adept classification of reagents allows the use of conditional IF loops from within a command file to perform addition of different reagents differently during a "single step" performed across 96 wells simultaneously. The special class ACTIVATORS defines certain reagents that always get added with a particular class of reagents (for example tetrazole during a phosphitylation reaction in adding the next nucleotide to a growing oligonucleotide).

The general form of the .tab file is the definition:
Class Bottle Reagent Name Flow rate Conc.
The LOOP BEGIN and LOOP END commands define the block of commands which will continue to operate until a NEXT SEQUENCE command points past the end of the longest list of reactants in any well.
Not included in the command set is a MOVE command. For all of the above commands, if any plate or nozzle movement is required, this is automatically executed in order to perform the desired solvent or reagent delivery operation. This is accomplished by the controller software and hardware, which determines the correct nozzles) and wells) required for a particular reagent addition, then synchronizes the position of the requisite nozzle and well prior to adding the reagent.
A MANUAL mode can also be utilized in which the synthesis plate and nozzle blocks can be "homed" or moved to any position by the operator, the nozzles primed or washed, the various reagent bottles depressurized or washed with solvent, the chamber pressurized, etc. The automatic COMMAND mode can be interrupted at any point, MANUAL commands executed, and then operation resumed at the appropriate location.
The sequence pointer can be incremented to restart a synthesis anywhere within a command file.
In reference to Figure 14, the list of oligonucleotides for synthesis can be rearranged or grouped for optimization of synthesis. Thus at process step 574, the oligonucleotides are grouped according to a factor on which to base the optimization of synthesis. As illustrated in the Examples below, one such factor is the 3' most nucleoside of the oligonucleotide. Using the amidite approach for oligonucleotide synthesis, a nucleotide bearing a 3' phosphoramite is added to the S' hydroxyl group of a growing nucleotide chain. The first nucleotide (at the 3' terminus of the oligonucleotide - the 3' most nucleoside) is first connected to a solid support. This is normally done batchwise on a large scale as is standard practice during oligonucleotide synthesis.
Such solid supports pre-loaded with a nucleoside are commercially available.
In utilizing the multi well format for oligonucleotide synthesis, for each oligonucleotide to be synthesized, an aliquot of a solid support bearing the proper nucleoside thereon is added to the well for synthesis. Prior to loading the sequence of oligonucleotides to be synthesized in the .seq file, they are sorted by the 3' terminal nucleotide. Based on that sorting, all of the oligonucleotide sequences having an "A" nucleoside at their 3' end are grouped together, those with a "C" nucleoside are grouped together as are those with "G" or "T"
nucleosides. Thus in loading the nucleoside-bearing solid support into the synthesis wells, machine movements are conserved.
The oligonucleotides can be grouped by the above described parameter or other parameters that facilitate the synthesis of the oligonucleotides. Thus in Figure 14, sorting is noted as being effected by some parameter of type 1, as for instance the above described 3' most nucleoside, or other types of parameters from type 2 to type n at process steps 576, 578 and 580. Since synthesis will be from the 3' end of the oligonucleotides to the 5' end, the oligonucleotide sequences are reverse sorted to read 3' to 5'. The oligonucleotides are entered in the .seq file in this form, i.e., reading 3' to 5'.
Once sorted into types, the position of the oligonucleotides on the synthesis plates is specified at process step 582 by the creation of a .seq file as described above. The .seq file is associated with the respective .cmd and .tab files needed for synthesis of the particular chemistries specified for the oligonucleotides at process step 584 by retrieval of the .cmd and .tab files at process step 586 from the sample database 520.
These files are then input into the mufti well synthesizer at process step 588 for oligonucleotide synthesis.
Once physically synthesized, the list of oligonucleotides again enters the general procedure flow as indicated in Figure 1. For shipping, storage or other handling purposes, the plates can be lyophilized at this point if desired. Upon lyophilization, each well contains the oligonucleotides located therein as a dry compound.
14. Quality Control.
In an optional step, quality control is performed on the oligonucleotides at process step 600 after a decision is made (decision step 550) to perform quality control. Although optional, quality control may be desired when there is some reason to think that some aspect of the synthetic process step 500 has been compromised. Alternatively, samples of the oligonucleotides may be taken and stored in the event that the results of assays conducted using the oligonucleotides (process step 700) yield confusing results or suboptimal data. In the latter event, for example, quality control might be performed after decision step 800 if no oligonucleotides with sufficient activity are identified. In either event, decision step 650 follows quality control step process 600. If one or more of the oligonucleotides do not pass quality control, process step 500 can be repeated, i.e., the oligonucleotides are synthesized for a second time.
The operation of the quality control system general procedure 600 is detailed in steps 610-660 of Figure 1 S. Also referenced in the following discussion are the robotics and associated analytical instrumentation as shown in Figure 18.
During step 610 (Figure 15), sterile, double-distilled water is transferred by an automated liquid handler (2040 of Figure 18) to each well of a multi-well plate containing a set of lyophilized antisense oligonucleotides. The automated liquid handler (2040 of Figure 18) reads the barcode sticker on the mufti-well plate to obtain the plate's identification number. Automated liquid handler 2040 then queries Sample Database 520 (which resides in Database Server 2002 of Figure 18) for the quality control assay instruction set for that plate and executes the appropriate steps. Three quality control processes are illustrated, however, it is understood that other quality control processes or steps maybe practiced in addition to or in place of the processes illustrated.
The first illustrative quality control process (steps 622 to 626) quantitates the concentration of oligonucleotide in each well. If this quality control step is performed, an automated liquid handler (2040 of Figure 18) is instructed to remove an aliquot from each well of the master plate and generate a replicate daughter plate for transfer to the UV
spectrophotometer (2016 of Figure 18). The UV spectrophotometer (2016 of Figure 18) then measures the optical density of each well at a wavelength of 260 nanometers. Using standardized conversion factors, a microprocessor within UV spectrophotometer (2016 of Figure 18) then calculates a concentration value from the measured absorbance value for each well and output the results to Sample Database 520.
The second illustrative quality control process steps 632 to 636) quantitates the percent of total oligonucleotide in each well that is full length. If this quality control step is performed, an automated liquid handler (2040 of Figure 18) is instructed to remove an aliquot from each well of the master plate and generate a replicate daughter plate for transfer to the multichannel capillary gel electrophoresis apparatus (2022 of Figure 18).

The apparatus electrophoretically resolves in capillary tube gels the oligonucleotide product in each well. As the product reaches the distal end of the tube gel during electrophoresis, a detection window dynamically measures the optical density of the product that passes by it. Following electrophoresis, the value of percent product that passed by the detection window with respect to time is utilized by a built in microprocessor to calculate the relative size distribution of oligonucleotide product in each well. These results are then output to the Sample Database (520.
The third illustrative quality control process steps 632 to 63b) quantitates the mass of the oligonucleotide in each well that is full length. If this quality control step is performed, an automated liquid handler (2040 of Figure 18) is instructed to remove an aliquot from each well of the master plate and generate a replicate daughter plate for transfer to the multichannel liquid electrospray mass spectrometer (2018 of Figure 18).
The apparatus then uses electrospray technology to inject the oligonucleotide product into the mass spectrometer. A built in microprocessor calculates the mass-to-charge ratio to arnve at the mass of oligonucleotide product in each well. The results are then output to Sample Database 520.
Following completion of the selected quality control processes, the output data is manually examined or is examined using an appropriate algorithm and a decision is made as to whether or not the plate receives "Pass" or "Fail" status. The current criteria for acceptance, for 18 mer oligonucleotides, is that at least 85% of the oligonucleotides in a multi-well plate must be 85% or greater full length product as measured by both capillary gel electrophoresis and mass spectrometry. An input (manual or automated) is then made into Sample Database 520 as to the pass/fail status of the plate. If a plate fails, the process cycles back to step 500, and a new plate of the same oligonucleotides is automatically placed in the plate synthesis request queue (process 554 of Figure 15). If a plate receives "Pass" status, an automated liquid handler (2040 of Figure 18) is instructed to remove appropriate aliquots from each well of the master plate and generate two replicate daughter plates in which the oligonucleotide in each well is at a concentration of 30 micromolar.
The plate then moves on to process 700 for oligonucleotide activity evaluation.
15. Cell Lines for Assaying Oligonucleotide Activity. The effect of antisense compounds on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid, or its gene product, is present at measurable levels.
This can be routinely determined using, for example, PCR or Northern blot analysis. The following four cell types are provided for illustrative purposes, but other cell types can be routinely used.
T-24 cells: The transitional cell bladder carcinoma cell line T-24 is obtained from the American Type Culture Collection {ATCC) (Manassas, VA). T-24 cells were routinely cultured in complete McCoy's SA basal media (Life Technologies, Gaithersburg, MD) supplemented with 10% fetal calf serum, penicillin 100 units per milliliter, and streptomycin 100 micrograms per milliliter (all from Life Technologies).
10 Cells are routinely passaged by trypsinization and dilution when they reach 90%
confluence. Cells are routinely seeded into 96-well plates (Falcon-Primaria #3872) at a density of 7000 cells/well for use in RT-PCR analysis. For Northern blotting or other analysis, cells are seeded onto 100 mm or other standard tissue culture plates and treated similarly, using appropriate volumes of medium and oligonucleotide.
15 A549 cells: The human lung carcinoma cell line A549 is obtained from the ATCC (Manassas, VA). A549 cells were routinely cultured in DMEM basal media (Life Technologies) supplemented with 10% fetal calf serum, penicillin 100 units per milliliter, and streptomycin 100 micrograms per milliliter (all from Life Technologies).
Cells are routinely passaged by trypsinization and dilution when they reach 90%
confluence.
20 NHDF cells: Human neonatal dermal fibroblast {NHDF) were obtained from the Clonetics Corporation (Walkersville, MD). NHDFs were routinely maintained in Fibroblast Growth Medium (Clonetics Corp.) as provided by the supplier. Cells are maintained for up to 10 passages as recommended by the supplier.
HEK cells: Human embryonic keratinocytes (HEK) were obtained from 25 the Clonetics Corp. HEKs were routinely maintained in Keratinocyte Growth Medium (Clonetics Corp.) as provided by the suppliex. Cell are routinely maintained for up to 10 passages as recommended by the supplier.
16. Treatment of Cells with Candidate Compounds:
When cells reach about 80% confluency, they are treated with oligonucleotide.
For 30 cells grown in 96-well plates, wells are washed once with 200 ~cl OPTI-MEM-1T"' reduced-serum medium (Life Technologies) and then treated with 130 ~cl of OPTI-MEM-1T'" containing 3.75 ~g/ml LIPOFECTINT"' (Life Technologies) and the desired oligonucleotide at a final concentration of 150 nM. After 4 hours of treatment, the medium was replaced with fresh medium. Cells were harvested 16 hours after oligonucleotide treatment.
Alternatively, for cells resistant to cationic mediated transfection, oligonucleotides can be introduced by electroporation. Electroporation conditions must be optimized for every cell type. In general, oligonucleotide is added directly to complete growth media to a final concentration between 1 and 20 micromolar. An electronic pulse is delivered to the cells using a BTX T820 ELECTRO SQUARE PORATORTM using a Multi-coaxial 96-well electrode (BT840) (BTX Corporation, San Diego, California). Following electroporation, the cells are returned to the incubator for 16 hours.
17. Assaying Oligonucleotide Activity:
Oligonucleotide-mediated modulation of expression of a target nucleic acid can be assayed in a variety of ways known in the art. For example, target RNA levels can be quantitated by, e.g., Northern blot analysis, competitive PCR, or reverse transcriptase polymerase chain reaction (RT-PCR). RNA analysis can be performed on total cellular RNA or, preferably in the case of polypeptide-encoding nucleic acids, poly(A)+
mRNA.
For RT-PCR, poly(A)+ mRNA is preferred. Methods of RNA isolation are taught in, for example, Ausubel et al. (Short Protocols in Molecular Biology, 2nd Ed., pp. 4-1 to 4-13, Greene Publishing Associates and John Wiley & Sons, New York, 1992). Northern blot analysis is routine in the art (Id., pp. 4-14 to 4-29).
Alternatively, total RNA can be prepared from cultured cells or tissue using the QIAGEN RNeasy~-96 kit for the high throughput preparation of RNA (QIAGEN, Inc., Valencia, CA). Essentially, protocols are carried out according to the manufacturer's directions. Optionally, a DNase step is included to remove residual DNA prior to RT-PCR.
To improve efficiency and accuracy the repetitive pipeting steps and elution step have been automated using a QIAGEN Bio-Robot 9604. Essentially after lysing of the oligonucleotide treated cell cultures in situ, the plate is transferred to the robot deck where the pipeting, DNase treatment, and elution steps are carried out.
Reverse transcriptase polymerase chain reaction (RT-PCR) can be conveniently accomplished using the commercially available ABI PRISM~ 7700 Sequence Detection System (PE-Applied Biosystems, Foster City, CA) according to the manufacturer's instructions. Other methods of PCR are also known in the art.
Target protein levels can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), Enzyme-linked immunosorbent assay (ELISA) or fluorescence-activated cell sorting (FACS).
Antibodies directed to a protein encoded by a target nucleic acid can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies, (Aerie Corporation, Birmingham, MI or via the Internet at http://www.ANTIBODIES-PROBES.com/), or can be prepared via conventional antibody generation methods. Methods fox preparation of polyclonal, monospecific ("antipeptide") and monoclonal antisera are taught by, for example, Ausubel et al. (Short Protocols in Molecular Biology, 2nd Ed., pp. 11-3 to 11-54, Greene Publishing Associates and John Wiley & Sons, New York, 1992).
Immunoprecipitation methods are standard in the art and are described by, for example, Ausubel et al. {Id., pp. 10-57 to 10-63). Western blot (immunoblot) analysis is standard in the art (Id., pp. 10-32 to 10-10-35). Enzyme-linked immunosorbent assays (ELISA) are standard in the art (Id., pp. 11-5 to 11-17).
Because it is preferred to assay the compounds of the invention in a batchwise fashion, i.e., in parallel to the automated synthesis process described above, preferred means of assaying are suitable for use in 96-well plates and with robotic means.
Accordingly, automated RT-PCR is preferred for assaying target nucleic acid levels, and automated ELISA is preferred for assaying target protein levels.
The assaying step, general procedure step 700, is described in detail in Figure 16.
After an appropriate cell line is selected at process step 710, a decision is made at decision step 714 as to whether RT-PCR will be the only method by which the activity of the compounds is evaluated. In some instances, it is desirable to run alternative assay methods at process step 718; for example, when it is desired to assess target polypeptide levels as well as target RNA levels, an immunoassay such as an ELISA is run in parallel with the RT-PCR assays. Preferably, such assays are tractable to semi-automated or robotic means.
When RT-PCR is used to evaluate the activities of the compounds, cells are plated into multi-well plates (typically, 96-well plates) in process step 720 and treated with test or control oligonucleotides in process step 730. Then, the cells are harvested and lysed in process step 740 and the lysates are introduced into an apparatus where RT-PCR
is carried out in process step 750. A raw data file is generated, and the data is downloaded and compiled at step 760. Spreadsheet files with data charts are generated at process step 770, and the experimental data is analyzed at process step 780. Based on the results, a decision is made at process step 785 as to whether it is necessary to repeat the assays and, if so, the process begins again with step 720. In any event, data from all the assays on each oligonucleotide are compiled and statistical parameters are automatically determined at process step 790.
18. Classification of Compounds Based on Their Activity:
Following assaying, general procedure step 700, oligonucleotide compounds are classified according to one or more desired properties. Typically, three classes of compounds are used: active compounds, marginally active (or "marginal") compounds and inactive compounds. To some degree, the selection criteria for these classes vary from target to target, and members of one or more classes may not be present for a given set of oligonucleotides.
However, some criteria are constant. For example, inactive compounds will typically comprise those compounds having 5% or less inhibition of target expression (relative to basal levels). Active compounds will typically cause at least 30%
inhibition of target expression, although lower levels of inhibition are acceptable in some instances.
Marginal compounds will have activities intermediate between active and inactive compounds, with preferred marginal compounds having activities more like those of active compounds.
19. Optimization of Lead Compounds by Sequence.
One means by which oligonucleotide compounds are optimized for activity is by varying their nucleobase sequences so that different regions of the target nucleic acid are targeted. Some such regions will be more accessible to oligonucleotide compounds than others, and "sliding" a nucleobase sequence along a target nucleic acid only a few bases can have significant effects on activity. Accordingly, varying or adjusting the nucleobase sequences of the compounds of the invention is one means by which suboptimal compounds can be made optimal, or by which new active compounds can be generated.

The operation of the gene walk process 1100 detailed in steps 1104-1112 of Figure 17 is detailed as follows. As used herein, the term "gene walk" is defined as the process by which a specified oligonucleotide sequence x that binds to a specified nucleic acid target y is used as a frame of reference around which a series of new oligonucleotides sequences capable of hybridizing to nucleic acid target y are generated that are sequence shifted increments of oligonucleotide sequence x. Gene walking can be done "downstream", "upstream" or in both directions from a specified oligonucleotide.
During step 1104 the user manually enters the identification number of the oligonucleotide sequence around which it is desired to execute gene walk process 1100 and the name of the corresponding target nucleic acid. The user then enters the scope of the gene walk at step 1104, by which is meant the number of oligonucleotide sequences that it is desired to generate. The user then enters in step 1108 a positive integer value for the sequence shift increment. Once this data is generated, the gene walk is effected. This causes a subroutine to be executed that automatically generates the desired list of sequences by walking along the target sequence. At that point, the user proceeds to process 400 to assign chemistries to the selected oligonucleotides.
Example 16 below, details a gene walk. In subsequent steps, this new set of nucleobase sequences generated by the gene walk is used to direct the automated synthesis at general procedure step 500 of a second set of candidate oligonucleotides.
These compounds are then taken through subsequent process steps to yield active compounds or reiterated as necessary to optimize activity of the compounds.
20. Optimization of Lead Compounds by Chemistry.
Another means by which oligonucleotide compounds of the invention are optimized is by reiterating portions of the process of the invention using marginal or active compounds from the first iteration and selecting additional chemistries to the nucleobase sequences thereof.
Thus, for example, an oligonucleotide chemistry different from that of the first set of oligonucleotides is assigned at general procedure step 400. The nucleobase sequences of marginal compounds are used to direct the synthesis at general procedure step 500 of a second set of oligonucleotides having the second assigned chemistry. The resulting second set of oligonucleotide compounds is assayed in the same manner as the first set at procedure process step 700 and the results are examined to determine if compounds having sufficient activity have been generated at decision step 800.
21. Identification of Sites Amenable to Antisense Technologies.
In a related process, a second oligonucleotide chemistry is assigned at procedure 5 step 400 to the nucleobase sequences of all of the oligonucleotides (or, at least, all of the active and marginal compounds) and a second set of oligonucleotides is synthesized at procedure step 500 having the same nucleobase sequences as the first set of compounds.
The resulting second set of oligonucleotide compounds is assayed in the same manner as the first set at procedure step 700 and active and marginal compounds are identified at 10 procedure steps 800 and 1000.
In order to identify sites on the target nucleic acid that are amenable to a variety of antisense technologies, the following mathematically simple steps are taken.
The sequences of active and marginal compounds from two or more such automated syntheses/assays are compared and a set of nucleobase sequences that are active, or 15 marginally so, in both sets of compounds is identified. The reverse complements of these nucleobase sequences corresponds to sequences of the target nucleic acid that are tractable to a variety of antisense and other sequence-based technologies. These antisense-sensitive sites are assembled into contiguous sequences (contigs) using the procedures described for assembling target nucleotide sequences (at procedure step 200).
20 22. Systems for Executing Preferred Methods of the Invention.
An embodiment of computer, network and instrument resources for effecting the methods of the invention is shown in Figure 18. In this embodiment, four computer servers are provided. First, a large database server 2002 stores all chemical structure, sample tracking and genomic, assay, quality control, and program status data.
Further, this 25 database server serves as the platform for a document management system.
Second, a compute engine 2004 runs computational programs including RNA folding, oligonucleotide walking, and genomic searching. Third, a file server 2006 allows raw instrument output storage and sharing of robot instructions. Fourth, a groupware server 2008 enhances staff communication and process scheduling.
30 A redundant high-speed network system is provided between the main servers and the bridges 2026, 2028 and 2030. These bridges provide reliable network access to the many workstations and instruments deployed for this process. The instruments selected to support this embodiment are all designed to sample directly from standard 96 well microtiter plates, and include an optical density reader 2016, a combined liquid chromatography and mass spectroscopy instrument 2018, a gel fluorescence and scintillation imaging system 2032 and 2042, a capillary gel electrophoreses system 2022 and a real-time PCR system 2034.
Most liquid handling is accomplished automatically using robots with individually controllable robotic pipetters 2038 and 2020 as well as a 96-well pipette system 2040 for duplicating plates. Windows NT or Macintosh workstations 2044, 2024, and 2036 are deployed for instrument control, analysis and productivity support.
23. Relational Database.
Data is stored in an appropriate database. For use with the methods of the invention, a relational database is preferred. Figure 19 illustrates the data structure of a sample relational database. Various elements of data are segregated among linked storage elements of the database.
EXAMPLES
The following examples illustrate the invention and are not intended to limit the same. Those skilled in the art will recognize, or be able to ascertain through routine experimentation, numerous equivalents to the specific procedures, materials and devices described herein. Such equivalents are considered to be within the scope of the present invention.
EXAMPLE 1: Selection of CD40 as a Target Cell-cell interactions are a feature of a variety of biological processes. In the activation of the immune response, for example, one of the earliest detectable events in a normal inflammatory response is adhesion of leukocytes to the vascular endothelium, followed by migration of leukocytes out of the vasculature to the site of infection or injury.
The adhesion of leukocytes to vascular endothelium is an obligate step in their migration out of the vasculature (for a review, see Albelda et al., FASEB J., 1994, 8, 504). As is well known in the art, cell-cell interactions are also critical for propagation of both B-lymphocytes and T-lymphocytes resulting in enhanced humoral and cellular immune responses, respectively (for a reviews, see Makgoba et al., Immunol. Today, 1989, 10, 417;
Janeway, Sci. Amer., 1993, 269, 72).
CD40 was first characterized as a receptor expressed on B-lymphocytes. It was later found that engagement of B-cell CD40 with CD40L expressed on activated T-cells is essential for T-cell dependent B-cell activation (i.e. proliferation, immunoglobulin secretion, and class switching) (for a review, see Gruss et al. Leuk.
Lymphoma, 1997, 24, 393). A full cDNA sequence for CD40 is available (GenBank accession number X60592, incorporated herein by reference as SEQ ID N0:85).
As interest in CD40 mounted, it was subsequently revealed that functional CD40 is expressed on a variety of cell types other than B-cells, including macrophages, dendritic cells, thymic epithelial cells, Langerhans cells, and endothelial cells (/bid.). These studies have led to the current belief that CD40 plays a much broader role in immune regulation by mediating interactions of T-cells with cell types other than B-cells. In support of this I S notion, it has been shown that stimulation of CD40 in macrophages and dendritic results is required for T-cell activation during antigen presentation (Id.). Recent evidence points to a role for CD40 in tissue inflammation as well. Production of the inflammatory mediators IL-12 and nitric oxide by macrophages has been shown to be CD40 dependent (Buhlmann et al., J. Clin. Ir~munol., 1996, 16, 83). In endothelial cells, stimulation of CD40 by CD40L has been found to induce surface expression of E-selectin, /CAM-1, and VCAM-1, promoting adhesion of leukocytes to sites of inflammation (Buhlmann et al., J.
Clin.
Immunol, 1996, 16, 83; Gruss et al., Leuk Lymphoma, 1997, 24, 393). Finally, a number of reports have documented overexpression of CD40 in epithelial and hematopoietic tumors as well as tumor infiltrating endothelial cells, indicating that CD40 may play a role in tumor growth and/or angiogenesis as well (Gruss et al., Leuk Lymphoma, 1997, 24, 393-422; Kluth et al. Cancer Res, 1997, 57, 891).
Due to the pivotal role that CD40 plays in humoral immunity, the potential exists that therapeutic strategies aimed at downregulating CD40 may provide a novel class of agents useful in treating a number of immune associated disorders, including but not limited to graft versus host disease, graft rejection, and autoimmune diseases such as multiple sclerosis, systemic lupus erythematosus, and certain forms of arthritis. Inhibitors of CD40 may also prove useful as an anti-inflammatory compound, and could therefore be useful as treatment for a variety of diseases with an inflammatory component such as asthma, rheumatoid arthritis, allograft rejections, inflammatory bowel disease, and various dermatological conditions, including psoriasis. Finally, as more is learned about the association between CD40 overexpression and tumor growth, inhibitors of CD40 may prove useful as anti-tumor agents as well.
Currently, there are no known therapeutic agents which effectively inhibit the synthesis of CD40. To date, strategies aimed at inhibiting CD40 function have involved the use of a variety of agents that disrupt CD40/CD40L binding. These include monoclonal antibodies directed against either CD40 or CD40L, soluble forms of CD40, and synthetic peptides derived from a second CD40 binding protein, A20. The use of neutralizing antibodies against CD40 and/or CD40L in animal models has provided evidence that inhibition of CD40 stimulation would have therapeutic benefit for GVHD, allograft rejection, rheumatoid arthritis, SLE, MS, and B-cell lymphoma (Buhlmann et al., J. Clin. Immunol, 1996, 16, 83). However, due to the expense, short half life, and bioavailability problems associated with the use of large proteins as therapeutic agents, there is a long felt need for additional agents capable of effectively inhibiting CD40 function. Oligonucleotides compounds avoid many of the pitfalls of current agents used to block CD40/CD40L interactions and may therefore prove to be uniquely useful in a number of therapeutic applications.
EXAMPLE 2: Generation of Virtual Oligonucleotides Targeted to CD40 The process of the invention was used to select oligonucleotides targeted to CD40, generating the list of oligonucleotide sequences with desired properties as shown in Figure 22. From the assembled CD40 sequence, the process began with determining the desired oligonucleotide length to be eighteen nucleotides, as represented in step 2500. All possible oligonucleotides of this length were generated by Oligo S.OTM, as represented in step 2504. Desired thermodynamic properties were selected in step 250$. The single parameter used was oligonucleotides of melting temperature less than or equal to 40°C
were discarded. In step 2512, oligonucleotide melting temperatures were calculated by Oligo 5.0~'. Oligonucleotide sequences possessing an undesirable score were discarded. It is believed that oligonucleotides with melting temperatures near or below physiological and cell culture temperatures will bind poorly to target sequences. All oligonucleotide sequences remaining were exported into a spreadsheet. In step 2516, desired sequence properties are selected. These include discarding oligonucleotides with at least one stretch of four guanosines in a row and stretches of six of any other nucleotide in a row. In step 2520, a spreadsheet macro removed all oligonucleotides containing the text string "GGGG." In step 2524, another spreadsheet macro removed all oligonucleotides containing the text strings "AAAAAA" or "CCCCCC" or "TTTTTT." From the remaining oligonucleotide sequences, 84 sequences were selected manually with the criteria of having an uniform distribution of oligonucleotide sequences throughout the target sequence, as represented in step 2528. These oligonucleotide sequences were then passed to the next step in the process, assigning actual oligonucleotide chemistries to the sequences.
EXAMPLE 3: Input Files For Automated Oligonucleotide Synthesis Command File (.cmd File) Table 2 is a command file for synthesis of an oligonucleotide having regions of 2'-O-(2-methoxyethyl) nucleosides and a central region of 2'-deoxy nucleosides each linked by phosphorothioate internucleotide linkages.
Table 2 SOLID SUPPORT SKIP
BEGIN
Next Sequence END
ITTITIAL-WASH
BEGIN
Add ACN 300 Drain 10 END

LOOP-BEGIN
DEBLOCK
BEGIN
Prime TCA
5 Load Tray Repeat 2 Add TCA 150 Wait 10 Drain 8 10 End Repeat Remove Tray Add TCA 125 Wait 10 Drain 8 WASH AFTER DEBLOCK
BEGIN
Repeat 3 Add ACN 250 To All 20 Drain 10 End Repeat END
COUPLING
BEGIN
25 if class = DEOXY THIOATE
Nozzle wash <actl>
prime <actl>
prime <seq>
Add <actl> 70 + <seq> 70 Wait 40 Drain 5 end-if if class = MOE THIOATE
Nozzle wash <actl>
Prime <act 1 >
prime <seq>
Add <actl> 120 + <seq> 120 Wait 230 Drain 5 End if END
WASH AFTER COUPLING
BEGIN
Add ACN 200 To All Drain 10 END
OXIDIZE
BEGIN
if class = DEOXY THIOATE
Add BEAU 180 Wait 40 Drain 7 end if if class = MOE THIOATE
Add BEAU 200 Wait 120 Drain 7 end if END
CAP
BEGIN
Add CAP_B 80 + CAP_A 80 Wait 20 Drain 7 END
WASH AFTER CAP
BEGIN
Add ACN 150 To All Drain 5 Add ACN 250 To All 1 S Drain 11 END
BASE COUNTER
BEGIN
Next Sequence END
LOOP END
DEBLOCK FINAL
BEGIN
Prime TCA
Load Tray Repeat 2 Add TCA 150 To All Wait 10 Drain 8 End Repeat Remove Tray Add TCA 125 To All Wait 10 Drain 10 END
FINAL WASH
BEGIN
Repeat 4 Add ACN 300 to All Drain 12 End Repeat END
ENDALL
BEGIN
Wait 3 END
Sequence files (.seq Files) Table 3 is a .seq file for oligonucleotides having 2'-deoxy nucleosides linked by phosphorothioate internucleotide linkages.
Table 3 Identity of columns: Syn #, Well, Scale, Nucleotide at particular position (identified using base identifier followed by backbone identifier where "s" is phosphorothioate).
Note the columns wrap around to next line when longer than one line.
1 A01 200 As Cs Cs As Gs Gs As Cs Gs Gs Cs Gs Gs As Cs Cs As G

2 A02 200 As Cs Gs Gs Cs Gs Gs As Cs Cs As Gs As Gs Ts Gs Gs A

3 A03 200 As Cs Cs As As Gs Cs As Gs As Cs Gs Gs As Gs As Cs G

4 A04 200 As Gs Gs As Gs As Cs Cs Cs Cs Gs As Cs Gs As As Cs G

5 A05 200 As Cs Cs Cs Cs Gs As Cs Gs As As Cs Gs As Cs Ts Gs G

6 A06 200 As Cs Gs As As Cs Gs As Cs Ts Gs Gs Cs Gs As Cs As G

7 A07 200 As Cs Gs As Cs Ts Gs Gs Cs Gs As Cs As Gs Gs Ts As G

8 A08 200 As Cs As Gs Gs Ts As Gs Gs Ts Cs Ts Ts Gs Gs Ts Gs G

9 A09 200 As Gs Gs Ts Cs Ts Ts Gs Gs Ts Gs Gs Gs Ts Gs As Cs G

10 A10 200 As Gs Ts Cs As Cs Gs As Cs As As Gs As As As Cs As C

11 All 200 As Cs Gs As Cs As As Gs As As As Cs As Cs Gs Gs Ts C

12 A12 200 As Gs As As As Cs As Cs Gs Gs Ts Cs Gs Gs Ts Cs Cs T

13 BO1 200 As As Cs As Cs Gs Gs Ts Cs Gs Gs Ts Cs Cs Ts Gs Ts C

14 B02 200 As Cs Ts Cs As Cs Ts Gs As Cs Gs Ts Gs Ts Cs Ts Cs A

15 B03 200 As Cs Gs Gs As As Gs Gs As As Cs Gs Cs Cs As Cs Ts T

16 B04 200 As Ts Cs Ts Gs Ts Gs Gs As Cs Cs Ts Ts Gs Ts Cs Ts C

17 BOS 200 As Cs As Cs Ts Ts Cs Ts Ts Cs Cs Gs As Cs Cs Gs Ts G

18 B06 200 As Cs Ts Cs Ts Cs Gs As Cs As Cs As Gs Gs As Cs Gs T

19 B07 200 As As As Cs Cs Cs Cs As Gs 5 Ts Ts Cs Gs Ts Cs Ts As A

20 B08 200 As Ts Gs Ts Cs Cs Cs Cs As As As Gs As Cs Ts As Ts G

21 B09 200 As Cs Gs Cs Ts Cs Gs Gs Gs As Cs Gs Gs Gs Ts Cs As G

10 22 B10 200 As Gs Cs Cs Gs As As Gs As As Gs As Gs Gs Ts Ts As C

23 B11 200 As Cs As Cs As Gs Ts As Gs As Cs Gs As As As Gs Cs T

24 B12 200 As Cs As Cs Ts Cs Ts Gs Gs 15 Ts Ts Ts Cs Ts Gs Gs As C

25 C01 200 As Cs Gs As Cs Cs As Gs As As As Ts As Gs Ts Ts Ts T
26 C02 200 As Gs Ts Ts As As As As Gs Gs Gs Cs Ts Gs Cs Ts As G

20 27 C03 200 As Gs Gs Ts Ts Gs Ts Gs As Cs Gs As Cs Gs As Gs Gs T

28 C04 200 As As Ts Gs Ts As Cs Cs Ts As Cs Gs Gs Ts Ts Gs Gs C

29 COS 200 As Gs Ts Cs As Cs Gs Ts Cs 25 Cs Ts Cs Ts Cs Ts Gs Ts C

30 C06 200 Cs Ts Gs Gs Cs Gs As Cs As Gs Gs Ts As Gs Gs Ts Cs T

31 C07 200 Cs Ts Cs Ts Gs Ts Gs Ts Gs As Cs Gs Gs Ts Gs Gs Ts C

30 32 C08 200 Cs As Gs Gs Ts Cs Gs Ts Cs Ts Ts Cs Cs Cs Gs Ts Gs G

WO 99/53101 PCT/(IS99/08268 33 C09 200 Cs Ts Gs Ts Gs Gs Ts As Gs As Cs Gs Ts Gs Gs As Cs A

34 C10 200 Cs Ts As As Cs Gs As Ts Gs Ts Cs Cs Cs Cs As As As G

35 C11 200 Cs Ts Gs Ts Ts Cs Gs As Cs As Cs Ts Cs Ts Gs Gs Ts T

36 C12 200 Cs Ts Gs Gs As Cs Cs As As Cs As Cs Gs Ts Ts Gs Ts C

37 DOl 200 Cs Cs Gs Ts Cs Cs Gs Ts Gs Ts Ts Ts Gs Ts Ts Cs Ts G

38 D02 200 Cs Ts Gs As Cs Ts As Cs As As Cs As Gs As Cs As Cs C

39 D03 200 Cs As As Cs As Gs As Cs As Cs Cs As Gs Gs Gs Gs Ts C

40 D04 200 Cs As Gs Gs Gs Gs Ts Cs Cs Ts As Gs Cs Cs Gs As Cs T

41 DOS 200 Cs Ts Cs Ts As Gs Ts Ts As As As As Gs Gs Gs Cs Ts G

42 D06 200 Cs Ts Gs Cs Ts As Gs As As Gs Gs As Cs Cs Gs As Gs G

43 D07 200 Cs Ts Gs As As As Ts Gs Ts As Cs Cs Ts As Cs Gs Gs T

44 D08 200 Cs As Cs Cs Cs Gs Ts Ts Ts Gs Ts Cs Cs Gs Ts Cs As A

45 D09 200 Cs Ts Cs Gs As Ts As Cs Gs Gs Gs Ts Cs As Gs Ts Cs A

46 D10 200 Gs Gs Ts As Gs Gs Ts Cs Ts Ts Gs Gs Ts Gs Gs Gs Ts G

47 D11 200 Gs As Cs Ts Ts Ts Gs Cs Cs Ts Ts As Cs Gs Gs As As G

48 D12 200 Gs Ts Gs Gs As Gs Ts Cs Ts Ts Ts Gs Ts Cs Ts Gs Ts G

49 E01 200 Gs Gs As Gs Ts Cs Ts Ts Ts Gs Ts Cs Ts Gs Ts Gs Gs T

50 E02 200 Gs Gs As Cs As Cs Ts Cs Ts Cs Gs As Cs As Cs As Gs G

51 E03 200 Gs As Cs As Cs As Gs Gs As Cs Gs Ts Gs Gs Cs Gs As G

52 E04 200 Gs As Gs Ts As Cs Gs As Gs Cs Gs Gs Gs Cs Cs Gs As A

53 EOS 200 Gs As Cs Ts As Ts Gs Gs Ts As Gs As Cs Gs Cs Ts Cs G

54 E06 200 Gs As As Gs As Gs Gs Ts Ts As Cs As Cs As Gs Ts As G

55 E07 200 Gs As Gs Gs Ts Ts As Cs As Cs As Gs Ts As Gs As Cs G

56 E08 200 Gs Ts Ts Gs Ts Cs Cs Gs Ts Cs Cs Gs Ts Gs Ts Ts Ts G

57 E09 200 Gs As Cs Ts Cs Ts Cs Gs Gs Gs As Cs Cs As Cs Cs As C

58 E10 200 Gs Ts As Gs Gs As Gs As As Cs Cs As Cs Gs As Cs Cs A

59 E11 200 Gs Gs Ts Ts Cs Ts Ts Cs Gs Gs Ts Ts Gs Gs Ts Ts As T

60 E12 200 Gs Ts Gs Gs Gs Gs Ts Ts Cs Gs Ts Cs Cs Ts Ts Gs Gs G

61 FOl 200 Gs Ts Cs As Cs Gs Ts Cs Cs Ts Cs Ts Gs As As As Ts G

62 F02 200 Gs Ts Cs Cs Ts Cs Cs Ts As Cs Cs Gs Ts Ts Ts Cs Ts C

63 F03 200 Gs Ts Cs Cs Cs Cs As Cs Gs Ts Cs Cs Gs Ts Cs Ts Ts C

64 F04 200 Ts Cs As Cs Cs As Gs Gs As Cs Gs Gs Cs Gs Gs As Cs C

65 FOS 200 Ts As Cs Cs As As Gs Cs As Gs As Cs Gs Gs As Gs As C

66 F06 200 Ts Cs Cs Ts Gs Ts Cs Ts Ts Ts Gs As Cs Cs As Cs Ts C

67 F07 200 Ts Gs Ts Cs Ts Ts Ts Gs As Cs Cs As Cs Ts Cs As Cs T

68 F08 200 Ts Gs As Cs Cs As Cs Ts Cs As Cs Ts Gs As Cs Gs Ts G

69 F09 200 Ts Gs As Cs Gs Ts Gs Ts Cs Ts Cs As As Gs Ts Gs As C

70 F10 200 Ts Cs As As Gs Ts Gs As Cs Ts Ts Ts Gs Cs Cs Ts Ts A

71 FI1 200 Ts Gs Ts Ts Ts As Ts Gs As Cs Gs Cs Ts Gs Gs Gs Gs T

72 F12 200 Ts Ts As Ts Gs As Cs Gs Cs Ts Gs Gs Gs Gs Ts Ts Gs G

73 GO1 200 Ts Gs As Cs Gs Cs Ts Gs Gs Gs Gs Ts Ts Gs Gs As Ts C

74 G02 200 Ts Cs Gs Ts Cs Ts Ts Cs Cs Cs Gs Ts Gs Gs As Gs Ts C

75 G03 200 Ts Gs Gs Ts As Gs As Cs Gs Ts Gs Gs As Cs As Cs Ts T

76 G04 200 Ts Ts Cs Ts Ts Cs Cs Gs As Cs Cs Gs Ts Gs As Cs As T

77 GOS 200 Ts Gs Gs Ts As Gs As Cs Gs Cs Ts Cs Gs Gs Gs As Cs G

78 G06 200 Ts As Gs As Cs Gs Cs Ts Cs Gs Gs Gs As Cs Gs Gs Gs T

79 G07 200 Ts Ts Ts Ts As Cs As Gs Ts Gs Gs Gs As As Cs Cs Ts G

80 G08 200 Ts Gs Gs Gs As As Cs Cs Ts Gs Ts Ts Cs Gs As Cs As C

81 G09 200 Ts Cs Gs Gs Gs As Cs Cs As Cs Cs As Cs Ts As Gs Gs G

82 G10 200 Ts As Gs Gs As Cs As As As Cs Gs Gs Ts As Gs Gs As G

83 G11 200 Ts Gs Cs Ts As Gs As As Gs Gs As Cs Cs Gs As Gs Gs T

84 G12 200 Ts Cs Ts Gs Ts Cs As Cs Ts Cs Cs Gs As Cs Gs Ts Gs G

Table 4 is a .seq file for oligonucleotides having regions of 2'-O-(2-methoxyethyl)-nucleosides and a central region of 2'-deoxy nucleosides each linked by phosphorothioate internucleotide linkages.
Table 4 Identity of columns: Syn #, Well, Scale, Nucleotide at particular position (identified using base identifier followed by backbone identifier where "s" is phosphorothioate and "moe" indicated a 2'-O-(2-methoxyethyl) substituted nucleoside). The columns wrap around to next line when longer than one line.
1 A01 200 moeAs moeCs moeCs moeAs Gs Gs As Cs Gs Gs Cs Gs Gs As moeCs moeCs moeAs moeG

2 A02 200 moeAs moeCs moeGsmoeGsCs GsGs AsCs Cs AsGs As Gs moeTs moeGs moeGs moeA

3 A03 200 moeAs moeCs moeCsmoeAsAs GsCs AsGs As CsGs Gs As moeGs moeAs moeCs moeG

4 A04 200 moeAs moeGs moeGsmoeAsGs AsCs CsCs Cs GsAs Cs Gs moeAs moeAs moeCs moeG

5 A05 200 moeAs moeCs moeCsmoeCsCs GsAs CsGs As AsCs Gs As moeCs moeTs moeGs moeG
6 A06 200 moeAs moeCs moeGs moeAs As Cs Gs As Cs Ts Gs Gs Cs Gs moeAs moeCs moeAs moeG
7 A07 200 moeAs moeCs moeGs moeAs Cs Ts Gs Gs Cs Gs As Cs As Gs 5 moeGs moeTs moeAs moeG
8 A08 200 moeAs moeCs moeAs moeGs Gs Ts As Gs Gs Ts Cs Ts Ts Gs moeGs moeTs moeGs moeG
9 A09 200 moeAs moeGs moeGs moeTs Cs Ts Ts Gs Gs Ts Gs Gs Gs Ts moeGs moeAs moeCs moeG
10 10 A10 200 moeAs moeGs moeTs moeCs As Cs Gs As Cs As As Gs As As moeAs moeCs moeAs moeC
11 All 200 moeAs moeCs moeGs moeAs Cs As As Gs As As As Cs As Cs moeGs moeGs moeTs moeC
12 A12 200 moeAs moeGs moeAs moeAs As Cs As Cs Gs Gs Ts Cs Gs Gs 15 moeTs moeCs moeCs moeT
13 BO1 200 moeAs moeAs moeCs moeAs Cs Gs Gs Ts Cs Gs Gs Ts Cs Cs moeTs moeGs moeTs moeC
14 B02 200 moeAs moeCs moeTs moeCs As Cs Ts Gs As Cs Gs Ts Gs Ts moeCs moeTs moeCs moeA
20 15 B03 200 moeAs moeCs moeGs moeGs As As Gs Gs As As Cs Gs Cs Cs moeAs moeCs moeTs moeT
16 B04 200 moeAs moeTs moeCs moeTs Gs Ts Gs Gs As Cs Cs Ts Ts Gs moeTs moeCs moeTs moeC
17 B05 200 moeAs moeCs moeAs moeCs Ts Ts Cs Ts Ts Cs Cs Gs As Cs 25 moeCs moeGs moeTs moeG
18 B06 200 moeAs moeCs moeTs moeCs Ts Cs Gs As Cs As Cs As Gs Gs moeAs moeCs moeGs moeT
19 B07 200 moeAs moeAs moeAs moeCs Cs Cs Cs As Gs Ts Ts Cs Gs Ts moeCs moeTs moeAs moeA
30 20 B08 200 moeAs moeTs moeGs moeTs Cs Cs Cs Cs As As As Gs As Cs moeTs moeAs moeTs moeG

21 B09 200 moeAs moeCs moeGs moeCs Ts Cs Gs Gs Gs As Cs Gs Gs Gs moeTs moeCs moeAs moeG
22 B 10 200 moeAs moeGs moeCs moeCs Gs As As Gs As As Gs As Gs Gs moeTs moeTs moeAs moeC
23 B11 200 moeAs moeCs moeAs moeCs As Gs Ts As Gs As Cs Gs As As moeAs moeGs moeCs moeT
24 B12 200 moeAs moeCs moeAs moeCs Ts Cs Ts Gs Gs Ts Ts Ts Cs Ts moeGs moeGs moeAs moeC
25 COl 200 moeAs moeCs moeGs moeAs Cs Cs As Gs As As As Ts As Gs moeTs moeTs moeTs moeT
26 C02 200 moeAs moeGs moeTs moeTs As As As As Gs Gs Gs Cs Ts Gs moeCs moeTs moeAs moeG
27 C03 200 moeAs moeGs moeGs moeTs Ts Gs Ts Gs As Cs Gs As Cs Gs moeAs moeGs moeGs moeT
28 C04 200 moeAs moeAs moeTs moeGs Ts As Cs Cs Ts As Cs Gs Gs Ts moeTs rnoeGs moeGs moeC
29 COS 200 moeAs moeGs moeTs moeCs As Cs Gs Ts Cs Cs Ts Cs Ts Cs moeTs moeGs moeTs moeC
30 C06 200 moeCs moeTs moeGs moeGs Cs Gs As Cs As Gs Gs Ts As Gs moeGs moeTs moeCs moeT
31 C07 200 moeCs moeTs moeCs moeTs Gs Ts Gs Ts Gs As Cs Gs Gs Ts moeGs moeGs moeTs moeC
32 C08 200 moeCs moeAs moeGs moeGs Ts Cs Gs Ts Cs Ts Ts Cs Cs Cs moeGs moeTs moeGs moeG
33 C09 200 moeCs moeTs moeGs moeTs Gs Gs Ts As Gs As Cs Gs Ts Gs moeGs moeAs moeCs moeA
34 C10 200 moeCs moeTs moeAs moeAs Cs Gs As Ts Gs Ts Cs Cs Cs Cs moeAs moeAs moeAs moeG
C11 200 moeCs rnoeTs moeGs moeTs Ts Cs Gs As Cs As Cs Ts Cs Ts 30 moeGs moeGs moeTs moeT
36 C12 200 moeCs moeTs rnoeGs moeGs As Cs Cs As As Cs As Cs Gs Ts moeTs moeGs moeTs moeC
37 DOl 200 moeCs moeCs moeGs moeTs Cs Cs Gs Ts Gs Ts Ts Ts Gs Ts moeTs moeCs moeTs moeG
38 D02 200 moeCs moeTs moeGs moeAs Cs Ts As Cs As As Cs As Gs As moeCs moeAs moeCs moeC
39 D03 200 moeCs moeAs moeAs moeCs As Gs As Cs As Cs Cs As Gs Gs moeGs moeGs moeTs moeC
40 D04 200 moeCs moeAs moeGs moeGs Gs Gs Ts Cs Cs Ts As Gs Cs Cs moeGs moeAs moeCs moeT
41 DOS 200 moeCs moeTs moeCs moeTs As Gs Ts Ts As As As As Gs Gs moeGs moeCs moeTs moeG
42 D06 200 moeCs moeTs moeGs moeCs Ts As Gs As As Gs Gs As Cs Cs moeGs moeAs moeGs moeG
43 D07 200 moeCs moeTs moeGs moeAs As As Ts Gs Ts As Cs Cs Ts As moeCs moeGs moeGs moeT
44 D08 200 moeCs moeAs moeCs moeCs Cs Gs Ts Ts Ts Gs Ts Cs Cs Gs moeTs moeCs moeAs moeA
45 D09 200 moeCs moeTs moeCs moeGs As Ts As Cs Gs Gs Gs Ts Cs As moeGs moeTs moeCs moeA
46 D 10 200 moeGs moeGs moeTs moeAs Gs Gs Ts Cs Ts Ts Gs Gs Ts Gs moeGs moeGs moeTs moeG
47 D11 200 moeGs moeAs moeCs moeTs Ts Ts Gs Cs Cs Ts Ts As Cs Gs moeGs moeAs moeAs moeG
48 D12 200 moeGs moeTs moeGs moeGs As Gs Ts Cs Ts Ts Ts Gs Ts Cs moeTs moeGs moeTs moeG
49 E01 200 moeGs moeGs moeAs moeGs Ts Cs Ts Ts Ts Gs Ts Cs Ts Gs moeTs moeGs moeGs moeT
50 E02 200 moeGs moeGs moeAs moeCs As Cs Ts Cs Ts Cs Gs As Cs As moeCs moeAs moeGs moeG
51 E03 200 moeGs moeAs moeCs moeAs Cs As Gs Gs As Cs Gs Ts Gs Gs moeCs moeGs moeAs moeG

52 E04 200 moeGs moeAs moeGs moeTs As Cs Gs As Gs Cs Gs Gs Gs Cs moeCs moeGs moeAs moeA
53 EOS 200 moeGs moeAs moeCs moeTs As Ts Gs Gs Ts As Gs As Cs Gs moeCs moeTs moeCs moeG
54 E06 200 moeGs moeAs moeAs moeGs As Gs Gs Ts Ts As Cs As Cs As moeGs moeTs moeAs moeG
55 E07 200 moeGs moeAs moeGs moeGs Ts Ts As Cs As Cs As Gs Ts As moeGs moeAs moeCs moeG
56 E08 200 moeGs moeTs moeTs moeGs Ts Cs Cs Gs Ts Cs Cs Gs Ts Gs moeTs moeTs moeTs moeG
57 E09 200 moeGs moeAs moeCs moeTs Cs Ts Cs Gs Gs Gs As Cs Cs As moeCs moeCs moeAs rnoeC
58 E10 200 moeGs moeTs moeAs moeGs Gs As Gs As As Cs Cs As Cs Gs moeAs moeCs moeCs moeA
59 E11 200 moeGs moeGs moeTs moeTs Cs Ts Ts Cs Gs Gs Ts Ts Gs Gs moeTs moeTs moeAs moeT
60 E12 200 moeGs moeTs moeGs moeGs Gs Gs Ts Ts Cs Gs Ts Cs Cs Ts moeTs moeGs moeGs moeG
61 FO1 200 moeGs moeTs moeCs moeAs Cs Gs Ts Cs Cs Ts Cs Ts Gs As moeAs moeAs moeTs moeG
62 F02 200 moeGs moeTs moeCs moeCs Ts Cs Cs Ts As Cs Cs Gs Ts Ts moeTs moeCs moeTs moeC
63 F03 200 moeGs moeTs moeCs moeCs Cs Cs As Cs Gs Ts Cs Cs Gs Ts moeCs moeTs moeTs moeC
64 F04 200 moeTs moeCs moeAs moeCs Cs As Gs Gs As Cs Gs Gs Cs Gs moeGs moeAs moeCs moeC
65 FOS 200 moeTs moeAs moeCs moeCs As As Gs Cs As Gs As Cs Gs Gs moeAs moeGs moeAs moeC
66 F06 200 moeTs moeCs moeCs moeTs Gs Ts Cs Ts Ts Ts Gs As Cs Cs moeAs moeCs moeTs moeC
67 F07 200 moeTs moeGs moeTs moeCs Ts Ts Ts Gs As Cs Cs As Cs Ts moeCs moeAs moeCs moeT
68 F08 200 moeTs moeGs moeAs moeCs Cs As Cs Ts Cs As Cs Ts Gs As moeCs moeGs moeTs moeG
69 F09 200 moeTs moeGs moeAs moeCs Gs Ts Gs Ts Cs Ts Cs As As Gs moeTs moeGs moeAs moeC
70 F10 200 moeTs moeCs moeAs moeAs Gs Ts Gs As Cs Ts Ts Ts Gs Cs moeCs moeTs moeTs moeA
71 F11 200 moeTs moeGs moeTs moeTs Ts As Ts Gs As Cs Gs Cs Ts Gs moeGs moeGs moeGs moeT
72 F12 200 moeTs moeTs moeAs moeTs Gs As Cs Gs Cs Ts Gs Gs Gs Gs moeTs moeTs moeGs moeG
73 GO1 200 moeTs moeGs moeAs moeCs Gs Cs Ts Gs Gs Gs Gs Ts Ts Gs moeGs moeAs moeTs moeC
74 G02 200 moeTs moeCs moeGs moeTs Cs Ts Ts Cs Cs Cs Gs Ts Gs Gs 1 S moeAs moeGs moeTs moeC
75 G03 200 moeTs moeGs moeGs moeTs As Gs As Cs Gs Ts Gs Gs As Cs moeAs moeCs moeTs moeT
76 G04 200 moeTs moeTs moeCs moeTs Ts Cs Cs Gs As Cs Cs Gs Ts Gs moeAs moeCs moeAs moeT
77 GOS 200 moeTs moeGs moeGs moeTs As Gs As Cs Gs Cs Ts Cs Gs Gs moeGs moeAs moeCs moeG
78 G06 200 moeTs moeAs moeGs moeAs Cs Gs Cs Ts Cs Gs Gs Gs As Cs moeGs moeGs moeGs moeT
79 G07 200 moeTs moeTs rnoeTs moeTs As Cs As Gs Ts Gs Gs Gs As As moeCs moeCs moeTs moeG
80 G08 200 moeTs moeGs moeGs moeGs As As Cs Cs Ts Gs Ts Ts Cs Gs moeAs moeCs moeAs moeC
81 G09 200 moeTs moeCs moeGs moeGs Gs As Cs Cs As Cs Cs As Cs Ts moeAs moeGs moeGs moeG
82 G10 200 moeTs moeAs moeGs moeGs As Cs As As As Cs Gs Gs Ts As moeGs moeGs moeAs moeG

WO 99/53101 PC'TNS99/0$268 83 G11 200 moeTs rnoeGs moeCs moeTs As Gs As As Gs Gs As Cs Cs Gs moeAs moeGs moeGs moeT
84 G12 200 moeTs moeCs moeTs moeGs Ts Cs As Cs Ts Cs Cs Gs As Cs moeGs moeTs moeGs moeG
5 Reagent file (.tab File) Table 5 is a .tab file for reagents necessary for synthesizing an oligonucleotides having both 2'-O-(2-methoxyethyl)nucleosides and 2'-deoxy nucleosides located therein.
Table 5 Identity of columns: GroupName, Bottle ID, ReagentName, FlowRate, Concentration.
10 Wherein reagent name is identified using base identifier, "moe" indicated a 2'-O-(2-methoxyethyl) substituted nucleoside and "cpg" indicates a control pore glass solid support medium. The columns wrap around to next line when longer than one line..
SUPPORT
BEGIN

15 0 moeG moeG cpg 100 0 moeSmeC moeSmeC cpg 100 0 moeA moeA cpg 100 0 moeT moeT cpg 100 END

BEGIN

END
WASH

END
OXIDIZERS

BEGIN

END

CAPPING

BEGIN

67 CAP_A CAP_A 230 END

DEOXY THIOATE

BEGIN

31,32 Gs deoxyG 270 1 39,40 SmeCs SmethyldeoxyC 270 1 37,38 As deoxyA 270 1 29,30 Ts deoxyT 270 1 END

MOE-THIOATE

BEGIN

15,16 moeGs methoxyethoxyG 240 1 23,24 moeSmeCs methoxyethoxyC 240 1 21,22 moeAs methoxyethoxyA 240 1 13,14 moeTs methoxyethoxyT 240 1 END

ACTIVATORS

BEGIN

5,6,7,8 SET s-ethyl-tet 280 Activates DEOXY THIOATE
MOE THIOATE
END
EXAMPLE 4: Oligonucleotide Synthesis - 96 Well Plate Format Oligonucleotides were synthesized via solid phase P(III) phosphoramidite chemistry using a mufti well automated synthesizer utilizing input files as described in EXAMPLE 3 above. The oligonucleotides were synthesized by assembling 96 sequences simultaneously in a standard 96 well format. Phosphodiester internucleotide linkages were afforded by oxidation with aqueous iodine. Phosphorothioate internucleotide linkages were generated by sulfurization utilizing 3,H-1,2 benzodithiole-3-one 1,1 dioxide (Beaucage Reagent) in anhydrous acetonitrile. Standard base-protected beta-cyanoethyl-diisopropyl phosphoramidites were purchased from commercial vendors (e.g.
PE/ABI, Pharmacia). Non-standard nucleosides are synthesized as per known literature or patented methods. They are utilized as base protected beta-cyanoethyldiisopropyl phosphoramidites.
Following synthesis, oligonucleotides were cleaved from support and deprotected with concentrated NH40H at elevated temperature (55-60°C) for 12-16 hours and the released product then dried in vacuo. The dried product was then re-suspended in sterile water to afford a master plate from which all analytical and test plate samples are then diluted utilizing robotic pipettors.
EXAMPLE 5: Alternative Oligonucleotide Synthesis Unsubstituted and substituted phosphodiester oligonucleotides are alternately synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine.
Phosphorothioates are synthesized as per the phosphodiester oligonucleotides except the standard oxidation bottle was replaced by 0.2 M solution of 3H-1,2-benzodithiole-3-one 1,1-dioxide in acetonitrile for the stepwise thiation of the phosphite linkages. The thiation wait step was increased to 68 sec and was followed by the capping step. After cleavage from the CPG column and deblocking in concentrated ammonium hydroxide at 55°C (18 hr), the oligonucleotides were purified by precipitating twice with 2.5 volumes of ethanol from a 0.5 M NaCI solution.
Phosphinate oligonucleotides are prepared as described in U.S. Patent 5,508,270, incorporated herein by reference in its entirety.
Alkyl phosphonate oligonucleotides are prepared as described in U.S. Patent 4,469,863, incorporated herein by reference in its entirety.
3'-Deoxy-3'-methylene phosphonate oligonucleotides are prepared as described in U.S. Patents 5,610,289 or 5,625,050, each of which is incorporated herein by reference in its entirety.
Phosphoramidite oligonucleotides are prepared as described in U.S. Patent, 5,256,775 or U.S. Patent 5,366,878, incorporated herein by reference in its entirety.
Alkylphosphonothioate oligonucleotides are prepared as described in published PCT applications PCT/US94/00902 and PCT/LTS93/06976 (published as WO 94/17093 and WO 94/02499, respectively), each of which is incorporated herein by reference in its entirety.
3'-Deoxy-3'-amino phosphoramidate oligonucieotides are prepared as described in U.S. Patent 5,476,925, incorporated herein by reference in its entirety.
Phosphotriester oligonucleotides are prepared as described in U.S. Patent 5,023,243, incorporated herein by reference in its entirety.
Boranophosphate oligonucleotides are prepared as described in U.S. Patents 5,130,302 and 5,177,198, each of which is incorporated herein by reference in its entirety.
Methylenemethylimino linked oligonucleosides, also identified as MMI linked oligonucleosides, methylenedimethylhydrazo linked oligonucleosides, also identified as MDH linked oligonucleosides, and methylenecarbonylamino linked oligonucleosides, also identified as amide-3 linked oligonucleosides, and methyleneaminocarbonyl linked oligo-nucleosides, also identified as amide-4 linked oligonucleosides, as well as mixed backbone compounds having, for instance, alternating MMI and PO or PS linkages are prepared as described in U.S. Patents 5,378,825; 5,386,023; 5,489,677; 5,602,240 and 5,610,289, each of which is incorporated herein by reference in its entirety.
Formacetal and thioformacetal linked oligonucleosides are prepared as described in U.S. Patents 5,264,562 and 5,264,564, each of which is incorporated herein by reference in its entirety.
Ethylene oxide linked oligonucleosides are prepared as described in U.S.
Patent 5,223,618, incorporated herein by reference in its entirety.
EXAMPLE 6: PNA Synthesis Peptide nucleic acids (PNAs) are prepared in accordance with any of the various procedures referred to in Peptide Nucleic Acids (PNA): Synthesis, Properties and Potential Applications, Bioorganic & Medicinal Chemistry, 1996, 4, 5. They may also be prepared in accordance with U.S. Patents 5,539,082; 5,700,922, and 5,719,262, each of which is incorporated herein by reference in its entirety.
EXAMPLE 7: Chimeric Oligonucleotide Synthesis Chimeric oligonucleotides, oligonucleosides or mixed oligonucleotides/oligonucleosides of the invention can be of several different types. These include a first type wherein the "gap" segment of linked nucleosides is positioned between 5' and 3' "wing" segments of linked nucleosides and a second "open end" type wherein the "gap" segment is located at either the 3' or the 5' terminus of the oligomeric compound.
Oligonucleotides of the first type are also known in the art as "gapmers" or gapped oligonucleotides. Oligonucleotides of the second type are also known in the art as "hemimers" or "wingmers."
A. [2'-O-Me]--[2'-deoxyJ~[2'-O-Me] Chimeric Phosphorothioate Oligonucleotides Chimeric oligonucleotides having 2'-O-alkyl phosphorothioate and 2'-deoxy phosphorothioate oligonucleotide segments are synthesized using 2'-deoxy-5'-dimethoxytrityl-3'-O-phosphoramidites for the DNA portion and 5'-dimethoxytrityl-2'-O-methyl-3'-O-phosphoramidites for 5' and 3' wings. The standard synthesis cycle is modified by increasing the wait step after the delivery of tetrazole and base to 600 s repeated four times for DNA and twice for 2'-0-methyl. The fully protected oligonucleotide was cleaved from the support and the phosphate group is deprotected in 3:1 Ammonia/Ethanol at room temperature overnight then lyophilized to dryness.

Treatment in methanolic ammonia for 24 hrs at room temperature is done to deprotect all bases and the samples are again lyophilized to dryness.
B. [2'-O-(2-Methoxyethyl)]--[2'-deoxy]--[2'-O-(2-Methoxyethyl)]
Chimeric Phosphorothioate Oligonucleotides [2'-O-(2-methoxyethyl)]--[2'-deoxy]--[-2'-O-(2-methoxyethyl)] chimeric phosphorothioate oligonucleotides are prepared as per the procedure above for the 2'-O-methyl chimeric oligonucleotide, with the substitution of 2'-O-(2-methoxyethyl) amidites for the 2'-O-methyl amidites.
C. [2'-O-(2-Methoxyethyl)Phosphodiester]--[2'-deoxy Phosphorothioate]--10 [2'-O-(2-Methoxyethyl) Phosphodiester] Chimeric Oligonucleotide [2'-O-(2-methoxyethyl phosphodiester]--[2'-deoxy phosphorothioate]--[2'-O-(2-methoxyethyl) phosphodiester] chimeric oligonucleotides are prepared as per the above procedure for the 2'-O-methyl chimeric oligonucleotide with the substitution of 2'-O-(2-methoxyethyl) amidites for the 2'-O-methyl amidites in the wing portions.
Sulfurization 15 utilizing 3,H-1,2 benzodithiole-3-one 1,1 dioxide (Beaucage Reagent) is used to generate the phosphorothioate internucleotide linkages within the wing portions of the chimeric structures. Oxidization with iodine is used to generate the phosphodiester internucleotide linkages for the center gap.
Other chimeric oligonucleotides, chimeric oligonucleosides and mixed chimeric 20 oligonucleotides/oligonucleosides are synthesized according to United States Patent 5,623,065, which is incorporated herein by reference in its entirety.
EXAMPLE 8: Output Oligonucleotides From Automated Oligonucleotide Synthesis Using the .seq files, the .cmd files and .tab file of Example 3, oligonucleotides were prepared as per the protocol of the 96 well format of Example 4. The 25 oligonucleotides were prepared utilizing phosphorothioate chemistry to give in one instance a first library of phosphorothioate oligodeoxynucleotides. The oligonucleotides were prepared in a second instance as a second library of hybrid oligonucleotides having phosphorothioate backbones with a first and third "wing" region of 2'-O-(2-methoxyethyl)nucleotides on either side of a center gap region of 2'-deoxy nucleotides.
30 The two libraries contained the same set of oligonucleotide sequences. Thus the two libraries are redundant with respect to sequence but are unique with respect to the combination of sequence and chemistry. Because the sequences of the second library of compounds is the same as the first (however the chemistry is different), for brevity sake, the second library is not shown.
For illustrative purposes Tables 6-a and 6-b show the sequences of an initial first library, i.e., a library of phosphorothioate oligonucleotides targeted to a CD40 target. The compounds of Table 6-a shows the members of this library listed in compliance with the established rule for listing SEQ ID NO:, i.e., in numerical SEQ ID NO: order.
Table 6-a Sequences of Oligonucleotides Targeted to CD40 by SEQ ID NO.:
NUCLEOBASE SEQUENCE SEQ ID NO.

GCAGAGGCAGACGAACCA

GACAGCGGTCAGCAAGCA

GATGGACAGCGGTCAGCA

CTCACCAGTTTCTGTCCT 1g GTGCCAGCCTTCTTCACA 3~

GCAGGGCTCGCAGATGGT 4~

The sequences shown in Table 6-a, above, and Table 6-b, below, are in a 5' to 3' direction. This is reversed with respect to 3' to 5' direction shown in the .seq files of Example 3. For synthesis purposes, the .seq files are generated reading from 3' to 5'. This allows for aligning all of the 3' most "A" nucleosides together, all of the 3' most "G"
nucleosides together, all of the 3' most "C" nucleosides together and all of the 3' most "T"
nucleosides together. Thus when the first nucleoside of each particular oligonucleotide (attached to the solid support) is added to the wells on the plates, machine movement is reduced since an automatic pipette can move in a linear manner down one row and up another on the 96 well plate.
The location of the well holding each particular oligonucleotides is indicated by row and column. There are eight rows designated A to G and twelve columns designated 1 to 12 in a typical 96 well format plate. Any particular well location is indicated by its "Well No." which is indicated by the combination of the row and the column, e.g. A08 is the well at row A, column 8.
In Table 6-b below, the oligonucleotides of Table 6-a are shown reordered according to the Well No. on their synthesis plate. The order shown in Table 6-b is the actually order as synthesized on an automated synthesizer taking advantage of the preferred placement of the first nucleoside according to the above alignment criteria.
Table 6-b:
Sequences of Oligonucleotides Targeted to CD40 Order by Synthesis Well No.
Well No. ~ SE ID NO:

C02 GATCGTCGGGAA.AATTGA 72 D05 GTCGGGAA.AATTGATCTC 71
35 D07 TGGCATCCATGTAAAGTC 77 EXAMPLE 9: Oligonucleotide Analysis A. Oligonucleotide Analysis - 96 Well Plate Format The concentration of oligonucleotide in each well was assessed by dilution of samples and UV absorption spectroscopy. The full-length integrity of the individual products was evaluated by capillary electrophoresis (CE) in either the 96 well format (Beckman MDQ) or, for individually prepared samples, on a commercial CE
apparatus (e.g., Beckman 5000, ABI 270). Base and backbone composition was confirmed by mass analysis of the compounds utilizing electrospray-mass spectroscopy. All assay test plates were diluted from the master plate using single and mufti-channel robotic pipettors.
B. Alternative Oligonucleotide Analysis After cleavage from the controlled pore glass support (Applied Biosystems) and deblocking in concentrated ammonium hydroxide at 55°C for 18 hours, the oligonucleotides or oligonucleosides are purified by precipitation twice out of 0.5 M NaCI
with 2.5 volumes ethanol. Synthesized oligonucleotides are analyzed by polyacrylamide gel electrophoresis on denaturing gels. Oligonucleotide purity is checked by 3'P nuclear s2 magnetic resonance spectroscopy, and/or by HPLC, as described by Chiang et al., J. Biol.
Chem. 1991, 266, 18162.
EXAMPLE 10: Automated Assay of CD40 Oligonucleotide Activity S A. Poly(A)+ mRNA isolation.
Poly(A)+ mRNA was isolated according to Miura et al. (Clin. Chem., 1996, 42, 1758). Briefly, for cells grown on 96-well plates, growth medium was removed from the cells and each well was washed with 200 ~1 cold PBS. 60 ~.1 lysis buffer ( 10 mM Tris-HCI, pH 7.6, 1 mM EDTA, 0.5 M NaCI, 0.5% NP-40, 20 mM vanadyl-ribonucleoside complex) was added to each well, the plate was gently agitated and then incubated at room temperature for five minutes. 55 pl of lysate was transferred to Oligo d(T) coated 96 well plates (ACCT Inc., Irvine, CA). Plates were incubated for 60 minutes at room temperature, washed 3 times with 200 ml of wash buffer ( 10 mM Tris-HCl pH
7.6, 1 mM
EDTA, 0.3 M NaCI). After the final wash, the plate was blotted on paper towels to remove excess wash buffer and then air-dried for 5 minutes. 60 ml of elution buffer (5 mM Tris-HCl pH 7.6), preheated to 70 ° C was added to each well, the plate was incubated on a 90 ° C plate for 5 minutes, and the eluate then transferred to a fresh 96-well plate.
Cells grown on 100 mm or other standard plates may be treated similarly, using appropriate volumes of all solutions.
B. Total RNA isolation Total mRNA was isolated using an RNEASY 96TM kit and buffers purchased from Qiagen Inc. (Valencia CA) following the manufacturer's recommended procedures.
Briefly, for cells grown on 96-well plates, growth medium was removed from the cells and each well was washed with 200 mL cold PBS. 100 mL Buffer RLT was added to each well and the plate vigorously agitated for 20 seconds. 100 mL of 70% ethanol was then added to each well and the contents mixed by pipetting three times up and down. The samples were then transferred to the RNEASY 96TM well plate attached to a QIAVACTM
manifold fitted with a waste collection tray and attached to a vacuum source. Vacuum was applied for 15 seconds. 1 mL of Buffer RW 1 was added to each well of the RNEASY 96TM
plate and the vacuum again applied for 15 seconds. 1 mL of Buffer RPE was then added to each well of the RNEASY 96TM plate and the vacuum applied for a period of 15 seconds. The AMENDED SHEET

. ~''~rrr~~rCA 0 2 3 2 5 013 2 0 0 0 - 10 - O S ~~~ ~ 7 N ov ~9sg Buffer RPE wash was then repeated and the vacuum was applied for an additional minutes. The plate was then removed from the QIAVACTM manifold and blotted dry on paper towels. The plate was then re-attached to the QIAVACTM manifold fitted with a collection tube rack containing 1.2 mL collection tubes. RNA was then eluted by pipetting 60 mL water into each well, incubating 1 minute, and then applying the vacuum for 30 seconds. The elution step was repeated with an additional 60 mL water.
C. RT-PCR Analysis of CD40 mRNA Levels Quantitation of CD40 mRNA levels was determined by reverse transcriptase polymerise chain reaction (RT-PCR) using the ABI PRISMTM 7700 Sequence Detection System (PE-Applied Biosystems, Foster City, CA) according to manufacturer's r , instructions. This is a closed-tube, non-gel-based, fluorescence detection system which allows high-throughput quantitation of polymerise chain reaction (PCR) products in real-time.
As opposed to standard PCR, in which amplification products are quantitated after the PCR is completed, products in RT-PCR are quantitated as they accumulate.
This is accomplished by including in the PCR reaction an oligonucleotide probe that anneals specifically between the forward and reverse PCR primers, and contains two fluorescent dyes. A reporter dye (e.g., JOE or FAM, PE-Applied Biosystems, Foster City, CA) is attached to the 5' end of the probe and a quencher dye (e.g., TAMRA, PE-Applied Biosystems, Foster City, CA) is attached to the 3' end of the probe. When the probe and __ dyes are intact, reporter dye emission is quenched by the proximity of the 3' quencher dye.
During amplification, annealing of the probe to the target sequence creates a substrate that can be cleaved by the 5'-exonuclease activity of Taq polymerise. During the extension phase of the PCR amplification cycle, cleavage of the probe by Taq polymerise releases the reporter dye from the remainder of the probe (and hence from the quencher moiety) and a sequence-specific fluorescent signal is generated.
With each cycle, additional reporter dye molecules are cleaved from their respective probes, and the fluorescence intensity is monitored at regular (six-second) intervals by laser optics built into the ABI PRISMTM 7700 Sequence Detection System. In each assay, a series of parallel reactions containing serial dilutions of mRNA
from AMENDED S,yEET

untreated control samples generates a standard curve that is used to quantitate the percent inhibition after antisense oligonucleotide treatment of test samples.
RT-PCR reagents were obtained from PE-Applied Biosystems, Foster City, CA.
RT-PCR reactions were carried out by adding 25 ml PCR cocktail ( 1 x TAQMANTM
buffer A, 5.5 mM MgCIZ, 300 mM each of dATP, dCTP and dGTP, 600 mM of dUTP, 100 nM
each of forward primer, reverse primer, and probe, 20 U RNAse inhibitor, 1.25 units AMPLITAQ GOLDTM, and 12.5 U MuLV reverse transcriptase) to 96 well plates containing 25 ml poly(A) mRNA solution. The RT reaction was carned out by incubation for 30 minutes at 48°C. following a 10 minute incubation at 95°C
to activate the AMPLITAQ GOLDTM, 40 cycles of a two-step PCR protocol were carried out: 95 °C for seconds (denaturation) followed by 60°C for 1.5 minutes (annealing/extension).
For CD40, the PCR primers were:
forward: 5' CAGAGTTCACTGAAACGGAATGC 3' (SEQ ID N0:86) 15 reverse: S' GGTGGCAGTGTGTCTCTCTGTTC 3' (SEQ ID N0:87), and PCR probe: S' FAM TTCCTTGCGGTGAAAGCGAATTCCT-TAMRA 3' (SEQ ID
N0:88) where FAM (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.
For GAPDH, the PCR primers were:
forward: 5' GAAGGTGAAGGTCGGAGTC 3' (SEQ ID N0:89) reverse: 5' GAAGATGGTGATGGGATTTC 3' (SEQ ID N0:90), and PCR probe: 5' JOE-CAAGCTTCCCGTTCTCAGCC-TAMRA 3' (SEQ ID NO. 91) where JOE (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.
EXAMPLE 11: Inhibition of CD40 Expression by Phosphorothioate Oligodeoxynucleotides In accordance with the present invention, a series of oligonucleotides complementary to mRNA were designed to target different regions of the human mRNA, using published sequences (GenBank accession number X60592, incorporated herein by reference as SEQ ID NO: 85). The oligonucleotides are shown in Table 7.

Target sites are indicated by the beginning nucleotide numbers, as given in the sequence source reference (X60592), to which the oligonucleotide binds. All compounds in Table 7 are oligodeoxynucleotides with phosphorothioate backbones (internucleoside linkages) throughout. Data are averages from three experiments.
Table 7:
Inhibition of CD40 mRNA Levels by Phosphorothioate Oligodeoxynucleotides TARGET SEQ ID
ISIS# SITE SEQUENCE % INHIB. NO.
18623 18 CCAGGCGGCAGGACCA 30.71 1 10 18624 20 GACCAGGCGGCAGGAC 28.09 2 18625 26 AGGTGAGACCAGGCGG 21.89 3 18626 48 CAGAGGCAGACGAACC 0.00 4 18627 49 GCAGAGGCAGACGAAC 0.00 5 18628 73 GCAAGCAGCCCCAGAG 0.00 6 15 18629 78 GGTCAGCAAGCAGCCC 29.96 7 18630 84 GACAGCGGTCAGCAAG 0.00 8 18631 88 GATGGACAGCGGTCAG 0.00 9 18632 92 TCTGGATGGACAGCGG 0.00 10 18633 98 GGTGGTTCTGGATGGA 0.00 11 20 18634 101 GTGGGTGGTTCTGGAT 0.00 12 18635 104 GCAGTGGGTGGTTCTG 0.00 13 18636 152 CACAAAGAACAGCACT 0.00 14 18637 156 CTGGCACAAAGAACAG 0.00 1 S

18638 162 TCCTGGCTGGCACAAA 0.00 16 25 18639 165 CTGTCCTGGCTGGCAC 4.99 17 18640 176 CTCACCAGTTTCTGTCC 0.00 18 18641 179 TCACTCACCAGTTTCTG 0.00 19 18642 185 GTGCAGTCACTCACCA 0.00 20 18643 190 ACTCTGTGCAGTCACTC 0.00 21 30 18644 196 CAGTGAACTCTGTGCA 5.30 22 18645 205 ATTCCGTTTCAGTGAAC 0.00 23 18646 211 GAAGGCATTCCGTTTC 9.00 24 18647 222 TTCACCGCAAGGAAGG 0.00 25 18648 250 CTCTGTTCCAGGTGTCT 0.00 26 35 18649 267 CTGGTGGCAGTGTGTC 0.00 27 18650 286 TGGGGTCGCAGTATTT 0.00 28 18651 289 GGTTGGGGTCGCAGTA 0.00 29 18652 292 CTAGGTTGGGGTCGCA 0.00 30 18653 318 GGTGCCCTTCTGCTGG 19.67 31 40 18654 322 CTGAGGTGCCCTTCTGC 15.63 32 18655 332 GTGTCTGTTTCTGAGGT 0.00 33 18656 334 TGGTGTCTGTTTCTGAG 0.00 34 18657 345 ACAGGTGCAGATGGTG 0.00 35 18658 348 TTCACAGGTGCAGATG 0.00 36 18659 360 GTGCCAGCCTTCTTCAC 5.67 37 18660 364 TACAGTGCCAGCCTTCT 7.80 38 18661 391 GGACACAGCTCTCACA 0.00 39 18662 395 TGCAGGACACAGCTCT 0.00 40 18663 401 GAGCGGTGCAGGACAC 0.00 41 18664 416 AAGCCGGGCGAGCATG 0.00 42 18665 432 AATCTGCTTGACCCCA 5.59 43 18666 446 GAAACCCCTGTAGCAA 0.10 44 18667 452 GTATCAGAAACCCCTG 0.00 45 18668 463 GCTCGCAGATGGTATC 0.00 46 18669 468 GCAGGGCTCGCAGATG 34.05 47 18670 471 TGGGCAGGGCTCGCAG 0.00 48 18671 474 GACTGGGCAGGGCTCG 2.71 49 18672 490 CATTGGAGAAGAAGCC 0.00 50 18673 497 GATGACACATTGGAGA 0.00 S

18674 500 GCAGATGACACATTGG 0.00 52 18675 506 TCGAAAGCAGATGACA 0.00 53 18676 524 GTCCAAGGGTGACATT 8.01 54 18677 532 CACAGCTTGTCCAAGG 0.00 55 18678 539 TTGGTCTCACAGCTTGT 0.00 56 18679 546 CAGGTCTTTGGTCTCAC 6.98 57 18680 558 CTGTTGCACAACCAGG 18.76 SS

18681 570 GTTTGTGCCTGCCTGTT 2.43 59 18682 575 GTCTTGTTTGTGCCTGC 0.00 60 18683 590 CCACAGACAACATCAG 0.00 61 18684 597 CTGGGGACCACAGACA 0.00 62 18685 607 TCAGCCGATCCTGGGG 0.00 63 18686 621 CACCACCAGGGCTCTC 23.31 64 18687 626 GGGATCACCACCAGGG 0.00 65 18688 657 GAGGATGGCAAACAGG 0.00 66 18689 668 ACCAGCACCAAGAGGA 0.00 67 18690 679 TTTTGATAAAGACCAG 0.00 68 18691 703 TATTGGTTGGCTTCTTG 0.00 69 18692 729 GGGTTCCTGCTTGGGG 0.00 70 18693 750 GTCGGGAAAATTGATC 0.00 71 18694 754 GATCGTCGGGAAA.ATT 0.00 72 18695 765 GGAGCCAGGAAGATCG 0.00 73 18696 766 TGGAGCCAGGAAGATC 0.00 74 18697 780 TGGAGCAGCAGTGTTG 0.00 75 18698 796 GTAAAGTCTCCTGCAC 0.00 76 18699 806 TGGCATCCATGTAAAG 0.00 77 18700 810 CGGTTGGCATCCATGT 0.00 78 18701 834 CTCTTTGCCATCCTCCT 4.38 79 18702 861 CTGTCTCTCCTGCACTG 0.00 80 18703 873 GGTGCAGCCTCACTGT 0.00 81 18704 910 AACTGCCTGTTTGCCCA 33.89 82 18705 954 CTTCTGCCTGCACCCCT 0.00 83 18706 976 ACTGACTGGGCATAGC 0.00 84 As shown in Table 7, SEQ ID NOS: 1, 2, 7, 47 and 82 demonstrated at least 25%
inhibition of CD40 expression and are therefore preferred compounds of the invention.
EXAMPLE 12: Inhibition of CD40 Expression by Phosphorothioate 2'-MOE
Gapmer Oligonucleotides In accordance with the present invention, a second series of oligonucleotides complementary to mRNA were designed to target different regions of the human mltNA, using published sequence X60592. The oligonucleotides are shown in Table 8.
Target sites are indicated by the beginning or initial nucleotide numbers, as given in the sequence source reference (X60592), to which the oligonucleotide binds.
All compounds in Table 8 are chimeric oligonucleotides ("gapmers") 18 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by four-nucleotide "wings." The wings are composed of 2'-O-(2-methoxyethyl) (2'-MOE) nucleotides.
The intersugar (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide.
1 S Cytidine residues in the 2'-MOE wings are 5-methylcytidines.
Data are averaged from three experiments.
Table 8:
Inhibition of CD40 mRNA Levels by Chimeric Phosphorothioate Oligonucleotides ISIS# TARGET SEQUENCE % Inhibition SEQ ID
19211 18 CCAGGCGGCAGGACCA 75.71 1 19212 20 GACCAGGCGGCAGGA 77.23 2 19213 26 AGGTGAGACCAGGCG 80.82 3 19214 48 CAGAGGCAGACGAAC 23.68 4 19215 49 GCAGAGGCAGACGAA 45.97 5 19216 73 GCAAGCAGCCCCAGAG 65.80 6 19217 78 GGTCAGCAAGCAGCCC 74.73 7 19218 84 GACAGCGGTCAGCAAG 67.21 8 19219 88 GATGGACAGCGGTCAG 65.14 9 19220 92 TCTGGATGGACAGCGG 78.71 10 19221 98 GGTGGTTCTGGATGGA 81.33 11 19222 101 GTGGGTGGTTCTGGAT 57.79 12 19223 104 GCAGTGGGTGGTTCTG 73.70 13 19224 152 CACAAAGAACAGCACT 40.25 14 19225 156 CTGGCACAAAGAACAG 60.11 15 $8 19226 162 TCCTGGCTGGCACAAA 10.18 16 19227 165 CTGTCCTGGCTGGCAC 24.37 17 19228 176 CTCACCAGTTTCTGTC 22.30 18 19229 179 TCACTCACCAGTTTCT 40.64 19 19230 185 GTGCAGTCACTCACCA 82.04 20 19231 190 ACTCTGTGCAGTCACT 37.59 21 19232 196 CAGTGAACTCTGTGCA 40.26 22 19233 205 ATTCCGTTTCAGTGAA 56.03 23 19234 211 GAAGGCATTCCGTTTC 32.21 24 19235 222 TTCACCGCAAGGAAGG 61.03 25 19236 250 CTCTGTTCCAGGTGTCT 62.19 26 19237 267 CTGGTGGCAGTGTGTC 70.32 27 19238 286 TGGGGTCGCAGTATTT 0.00 28 19239 289 GGTTGGGGTCGCAGTA 19.40 29 19240 292 CTAGGTTGGGGTCGCA 36.32 30 19241 318 GGTGCCCTTCTGCTGG 78.91 31 19242 322 CTGAGGTGCCCTTCTG 69.84 32 19243 332 GTGTCTGTTTCTGAGG 63.32 33 19244 334 TGGTGTCTGTTTCTGA 42.83 34 19245 345 ACAGGTGCAGATGGTG 73.31 35 19246 348 TTCACAGGTGCAGATG 47.72 36 19247 360 GTGCCAGCCTTCTTCA 61.32 37 19248 364 TACAGTGCCAGCCTTC 46.82 38 19249 391 GGACACAGCTCTCACA 0.00 39 19250 395 TGCAGGACACAGCTCT 52.05 40 19251 401 GAGCGGTGCAGGACAC 50.15 41 19252 416 AAGCCGGGCGAGCATG 32.36 42 19253 432 AATCTGCTTGACCCCA 0.00 43 19254 446 GAAACCCCTGTAGCAA 0.00 44 19255 452 GTATCAGAAACCCCTG 36.13 45 19256 463 GCTCGCAGATGGTATC 64.65 46 19257 468 GCAGGGCTCGCAGATG 74.95 47 19258 471 TGGGCAGGGCTCGCAG 0.00 48 19259 474 GACTGGGCAGGGCTCG 82.00 49 19260 490 CATTGGAGAAGAAGCC 41.31 50 19261 497 GATGACACATTGGAGA 13.81 51 19262 500 GCAGATGACACATTGG 78.48 52 19263 506 TCGAAAGCAGATGACA 59.28 53 19264 524 GTCCAAGGGTGACATT 70.99 54 19265 532 CACAGCTTGTCCAAGG 0.00 55 19266 539 TTGGTCTCACAGCTTG 45.92 56 19267 546 CAGGTCTTTGGTCTCA 63.95 57 19268 558 CTGTTGCACAACCAGG 82.32 58 19269 570 GTTTGTGCCTGCCTGTT 70.10 59 19270 575 GTCTTGTTTGTGCCTGC 68.95 60 19271 590 CCACAGACAACATCAG 11.22 61 19272 597 CTGGGGACCACAGACA 9.04 62 19273 607 TCAGCCGATCCTGGGG 0.00 63 19274 621 CACCACCAGGGCTCTC 23.08 64 19275 626 GGGATCACCACCAGGG 57.94 65 19276 657 GAGGATGGCAAACAG 49.14 66 19277 668 ACCAGCACCAAGAGG 3.48 67 19278 679 TTTTGATAAAGACCAG 30.58 68 19279 703 TATTGGTTGGCTTCTTG 49.26 69 19280 729 GGGTTCCTGCTTGGGG 13.95 70 19281 750 GTCGGGAAAATTGATC 54.78 71 19282 754 GATCGTCGGGAAAATT 0.00 72 19283 765 GGAGCCAGGAAGATC 69.47 73 19284 766 TGGAGCCAGGAAGATC 54.48 74 19285 780 TGGAGCAGCAGTGTTG 15.17 75 19286 796 GTAAAGTCTCCTGCAC 30.62 76 19287 806 TGGCATCCATGTAAAG 65.03 77 19288 810 CGGTTGGCATCCATGT 34.49 78 19289 834 CTCTTTGCCATCCTCCT 41.84 79 19290 861 CTGTCTCTCCTGCACT 25.68 80 19291 873 GGTGCAGCCTCACTGT 76.27 81 19292 910 AACTGCCTGTTTGCCC 63.34 82 19293 954 CTTCTGCCTGCACCCC 0.00 83 19294 976 ACTGACTGGGCATAGC 11.55 84 As shown in Table 8, SEQ ID NOS: 1, 2, 3, 6, 7, 8, 9, 10, 11, 12, 13, 15, 20, 23, 25, 26, 27, 31, 32, 33, 35, 37, 40, 41, 46, 47, 49, 52, 53, 54, 57, 58, 59, 60, 65, 71, 73, 74, 77, 81 and 82 demonstrated at least 50% inhibition of CD40 expression and are therefore preferred compounds of the invention.
EXAMPLE 13: Oligonucleotide-Sensitive Sites of the CD40 Target Nucleic Acid As the data presented in the preceding two Examples shows, several sequences were present in preferred compounds of two distinct oligonucleotide chemistries.
Specifically, compounds having SEQ ID NOS: 1, 2, 7, 47 and 82 are preferred in both instances. These compounds map to different regions of the CD40 transcript but nevertheless define accessible sites of the target nucleic acid.
For example, SEQ ID NOS: 1 and 2 overlap each other and both map to the 5-untranslated region (5'-UTR) of CD40. Accordingly, this region of CD40 is particularly preferred for modulation via sequence-based technologies. Similarly, SEQ ID
NOS: 7 and 47 map to the open reading frame of CD40, whereas SEQ ID NO: 82 maps to the 3'-untranslated region (3'-UTR). Thus, the ORF and 3'-UTR of CD40 may be targeted by sequence-based technologies as well.
The reverse complements of the active CD40 compounds are easily determined by those skilled in the art and may be assembled to yield nucleotide sequences corresponding to accessible sites on the target nucleic acid. For example, the assembled reverse complement of SEQ ID NOS: 1 and 2 is represented below as SEQ ID N0:92:
S'- AGTGGTCCTGCCGCCTGGTC -3' SEQ ID N0:92 TCACCAGGACGGCGGACC -5' SEQ ID NO:1 ACCAGGACGGCGGACCAG -5' SEQ ID N0:2 Through multiple iterations of the process of the invention, more extensive "footprints" are generated. A library of this information is compiled and may be used by those skilled in the art in a variety of sequence-based technologies to study the molecular and biological functions of CD40 and to investigate or confirm its role in various diseases 10 and disorders.
EXAMPLE 14: Site Selection Program In a preferred embodiment of the invention, illustrated in Figure 20, an application is deployed which facilitates the selection process for determining the target positions of 15 the oligos to be synthesized, or "sites." This program is written using a three-tiered object-oriented approach. All aspects of the software described, therefore, are tightly integrated with the relational database. For this reason, explicit database read and write steps are not shown. It should be assumed that each step described includes database access. The description below illustrates one way the program can be used. The actual 20 interface allows users to skip from process to process at will, in any order.
Before running the site picking program, the target must have all relevant properties computed as described previously and indicated in process step 2204. When the site picking program is launched at process step 2206 the user is presented with a panel showing targets which have previously been selected and had their properties calculated.
25 The user selects one target to work with at process step 2208 and proceeds to decide if any derived properties will be needed at process step 2210. Derived properties are calculated by performing mathematical operations on combinations of pre-calculated properties as defined by the user at process step 2212.
The derived properties are made available as peers with all the pre-calculated 30 properties. The user selects one of the properties to view plotted versus target position at process step 2214. This graph is shown above a linear representation of the target. The horizontal or position axis of both the graph and target are linked and scalable by the user.
The zoom range goes from showing the full target length to showing individual target bases as letters and individual property points. The user next selects a threshold value below or above which all sites will be eliminated from future consideration at process step 2216. The user decides whether to eliminate more sites based on any other properties at process step 2218. If they choose to eliminate more, they return to pick another property to display at process step 2214 and threshold at process step 2216.
After eliminating sites, the user selects from the remaining list by choosing any property at process step 2220 and then choosing a manual or automatic selection technique at process step 2222. In the automatic technique, the user decides whether they want to pick from maxima or minima and the number of maxima or minima to be selected as sites at process step 2224. The software automatically finds and picks the points.
When picking manually the user must decide if they wish to use automatic peak finding at process step 2226. If the user selects automatic peak finding, then user must click on the graphed property with the mouse at process step 2236. The nearest maxima or minima, depending on the modifier key held down, to the selected point will be picked as the site.
Without the peak fording option, the user must pick a site at process step 2238 by clicking on its position on the linear representation of target.
Each time a site, or group of sites, is picked, a dynamic property is calculated for all possible sites (not yet eliminated) at process step 2230. This property indicates the nearness of the site to a picked site allowing the user to pick sites in subsequent iterations based on target coverage. After new sites are picked, the user determines if the desired number of sites has been picked. If too few sites have been picked the user returns to pick more 2220. If too many sites have been picked, the user may eliminate them by selecting and deleting them on the target display at process step 2234. If the correct number of sites is picked, and the user is satisfied with the set of picked sites, the user registers these sites to the database along with their name, notebook number, and page number at process step 2238. The database time stamps this registration event.

EXAMPLE 15: Site Selection Program In a preferred embodiment of the invention, illustrated in Figure 21, an application is deployed which facilitates the assignment of specific chemical structure to the complement of the sequence of the sites previously picked and facilitates the registration and ordering of these now fully defined antisense compounds. This program is written using a three-tiered object-oriented approach. All aspects of the software described, therefore, are tightly integrated with the relational database. For this reason, explicit database read and write steps are not shown, it being understood that each step described also includes appropriate database read/write access.
To begin using the oligonucleotide chemistry assignment program, the user launches it at process step 2302. The user then selects from the previously selected sets of oligonucleotides at process step 2304, registered to the database in site picker's process step 2238. Next, the user must decide whether to manually assign the chemistry a base at a time, or run the sites through a template at process step 2306. If the user chooses to use a template, they must determine if a desired template is available at process step 2308. If a template is not available with the desired chemistry modifications and the correct length, the user can define one at process step 2314.
To define a template, the user must select the length of the oligonucleotide the template is to define. This oligonucleotide is then represented as a bar with selectable regions. The user sets the number of regions on the oligonucleotide, and the positions and lengths of these regions by dragging them back and forth on the bar. Each region is represented by a different color.
For each region, the user defines the chemistry modifications for the sugars, the linkers, and the heterocycles at each base position in the region. At least four heterocycle chemistries must be given, one for each of the four possible base types (A, G, C or T or U) in the site sequence the template will be applied to. A user interface is provided to select these chemistries which show the molecular structure of each component selected and its modification name. By pushing on a pop-up list next to each of the pictures, the user may choose from a list of structures and names, those possible to put in this place. For example, the heterocycle that represents the base type G is shown as a two dimensional structure diagram. If the user clicks on the pop-up list, a row of other possible structures and names is shown. The user drags the mouse to the desired chemistry and releases the mouse. Now the newly selected molecule is displayed as the choice for G type heterocycle modifications.
Once the user has created a template, or selected an existing one, the software applies the template at process step 2312 to each of the complements of the sites in the list.
When the templates are applied, it is possible that chemistries will be defined which are impossible to make with the chemical precursors presently used on the automatic synthesizer. To check this, a database is maintained of all precursors previously designed, and their availability for automated synthesis. When the templates are applied, the resulting molecules are tested at process step 2316 against this database to see if they are readily synthesized.
If a molecule is not readily synthesized, it is added to a list that the user inspects.
At process step 2318, the user decides whether to modify the chemistry to make it compatible with the currently recognized list of available chemistries or to ignore it. To modify a chemistry, the user must use the base at a time interface at process step 2322.
The user can also choose to go directly to this step, bypassing templates all together at process step 2306.
The base at a time interface at process step 2322 is very similar to the template editor at process step 2314 except that instead of specifying chemistries for regions, they are defined one base at a time. This interface also differs in that it dynamically checks to see if the design is readily synthesized as the user makes selections. In other words, each choice made limits the choices the software makes available on the pop-up selection lists.
To accommodate this function, an additional choice is made available on each pop-up of "not defined." For example, this allows the user to inhibit linker choice from restricting the sugar choices by first setting the linker to "not defined." The user would then pick the sugar, and then pick from the remaining linker choices available.
Once all of the sites on the list are assigned chemistries or dropped, they are registered at process step 2324 to a commercial chemical structure database.
Registering to this database makes sure the structure is unique, assigns it a new identifier if it is unique, and allows future structure and substructure searching by creating various hash-tables. The compound definition is also stored at process step 2326 to various hash tables referred to as chemistry/position tables. These allow antisense compound searching and categorization based on oligonucleotide chemistry modification sequences and equivalent base sequences.
The results of the registration are displayed at process step 2328 with the new IDs S if they are new compounds and with the old IDs if they have been previously registered.
The user next selects which of the compounds processed they wish to order for synthesis at process step 2330 and registers an order list at process step 2332 by including scientist name, notebook number and page number. The database time-stamps this entry.
The user may then choose at process step 2334, to quit the program at process step 2338, go back to the beginning and choose a new site list to work with process step 2304, or start the oligonucleotide ordering interface at process step 2336.
EXAMPLE 16: Gene Walk to Optimize Oligonucleotide Sequence A gene walk is executed using a CD40 antisense oligonucleotide having SEQ ID NO: l S (5'-CTGGCACAAAGAACAGCA-3'). In effecting this gene walk, the following parameters are used:
Gene Walk Parameter Entered value Oligonucleotide Sequence ID: 1 S
Name of Gene Target: CD40 Scope of Gene Walk: 20 Sequence Shift Increment: 1 Entering these values and effecting the gene walk centered on SEQ ID NO: 15 automatically generates the following new oligonucleotides:
Table 9:
Oligonucleotide Generated By Gene Walk SEQ Se uence ID

- GAACAGCACTGACTG

98 ACAAAGAAC_AGCACT

The list shown above contains 20 oligonucleotide sequences directed against the CD40 nucleic acid sequence. They are ordered by the position along the CD40 sequence 20 at which the 5' terminus of each oligonucleotide hybridizes. Thus, the first ten oligonucleotides are single-base frame shift sequences directed against the CD40 sequence upstream of compound SEQ ID NO: 1 S and the latter ten are single-base frame shift sequences directed against the CD40 sequence downstream of compound SEQ ID NO:
15.
25 EXAMPLE 17: Automated Assay of RhoC Oligonucleotide Activity RhoC, a member of the Rho subfamily of small GTPases, is a protein that has been shown to be involved in a diverse set of signaling pathways including the ultimate regulation of the dynamic organization of the cytoskeleton.
Oligonucleotides were designed as described in Example 2, synthesized as 30 described in Examples 3 through 8, analyzed as described in Examples 9 and assayed as described in Example 10 except for target specific primer and probes.
RhoC probes and primers were designed to hybridize to the human RhoC sequence, using published sequence information (GenBank accession number L25081, incorporated herein by reference as SEQ ID N0:113).
35 For RhoC the PCR primers were:
forward primer: TGATGTCATCCTCATGTGCTTCT (SEQ ID NO: 114) reverse primer: CCAGGATGATGGGCACGTT (SEQ ID NO: 115) and the PCR probe was: FAM-CGACAGCCCTGACAGCCTGGAAA-TAMRA (SEQ ID NO: 116) where FAM (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye) and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.
EXAMPLE 18: Antisense inhibition of RhoC expression- phosphorothioate oligodeoxynucleotides In accordance with the present invention, a series of oligonucleotides were designed to target different regions of the human lthoC RNA, using published sequences (GenBank accession number L25081, incorporated herein by reference as SEQ ID
NO:
113). The oligonucleotides are shown in Table 10. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no.
L25081), to which the oligonucleotide binds. All compounds in Table 10 are oligodeoxynucleotides with phosphorothioate backbones (internucleoside linkages) throughout. The compounds were analyzed for effect on RhoC mIRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments. If present, "N.D." indicates "no data".
Table 10 Inhibition of RhoC mRNA levels by phosphorothioate oligodeoxynucleotides ISIS# REGION SEQIJENCE % SEQ ID
TARGET

SITE Inhibition NO.

25304 5' UTR 4 gagctgagatgaagtcaa29 117 25305 5' UTR 44 gctgaagttcccaggctg25 118 25306 5' UTR 47 ccggctgaagttcccagg42 119 25307 Coding 104 ggcaccatccccaacgat81 120 25308 Coding 105 aggcaccatccccaacga81 121 25309 Coding 111 tcccacaggcaccatccc70 122 25310 Coding 117 aggtcttcccacaggcac40 123 25311 Coding 127 atgaggaggcaggtcttc41 124 25312 Coding 139 ttgctgaagacgatgagg23 125 25313 Coding 178 tcaaagacagtagggacg0 126 25314 Coding 181 ttctcaaagacagtaggg2 127 25315 Coding 183 agttctcaaagacagtag38 128 25316 Coding 342 tgttttccaggctgtcag59 129 25317 Coding 433 tcgtcttgcctcaggtcc 79 130 25318 Coding 439 gtgtgctcgtcttgcctc 67 131 25319 Coding 445 ctcctggtgtgctcgtct 67 132 25320 Coding 483 cagaccgaacgggctcct 65 133 25321 Coding 488 ttcctcagaccgaacggg 57 134 25322 Coding 534 actcaaggtagccaaagg 33 135 25323 Coding 566 ctcccgcactccctcctt 91 136 25324 Coding 575 ctcaaacacctcccgcac 34 137 25325 Coding 58I ggccatctcaaacacctc 64 138 25326 Coding 614 cttgttcttgcggacctg 72 139 25327 Coding 625 cccctccgacgcttgttc 66 140 25328 3' UTR 737 gtatggagccctcaggag 60 141 25329 3' UTR 746 gagccttcagtatggagc 63 142 25330 3' UTR 753 gaaaatggagccttcagt 24 143 I5 25331 3' UTR 759 ggaactgaaaatggagcc 2 144 25332 3' UTR 763 ggagggaactgaaaatgg 13 145 25333 3' UTR 766 gcaggagggaactgaaaa 27 146 25334 3' UTR 851 agggcagggcataggcgt 31 147 25335 3' UTR 854 ggaagggcagggcatagg 21 148 25336 3' UTR 859 catgaggaagggcagggc 0 149 25337 3' UTR 920 taaagtgctggtgtgtga 39 150 25338 3' UTR 939 cctgtgagccagaagtgt 69 151 25339 3' UTR 941 ttcctgtgagccagaagt 69 I52 25340 3' UTR 945 cactttcctgtgagccag 82 153 25341 3' UTR 948 agacactttcctgtgagc 69 154 25342 3' UTR 966 actctgggtccctactgc 20 155 25343 3' UTR 992 tgcagaaacaactccagg 0 156 Example 19: Antisense inhibition of RhoC expression- phosphorothioate 2'-MOE
gapmer oligonucleotides In accordance with the present invention, a second series of oligonucleotides targeted to human RhoC were synthesized. The oligonucleotide sequences are shown in Table 11. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession No. L25081 ), to which the oligonucleotide binds.
All compounds in Table 11 are chimeric oligonucleotides ("gapmers") 18 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by four-nucleotide "wings". The wings are composed of 2'-methoxyethyl (2'-MOE) nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. Cytidine residues in the 2'-MOE wings are 5-methylcytidines.
Data were obtained by real-time quantitative PCR as described in other examples herein and are averaged from three experiments. If present, "N.D." indicates "no data".
Table 11 Inhibition of RhoC mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS# REGION SEQUENCE % SEQ ID
TARGET

SITE Inhibition NO.

25344 5' UTR 4 gagctgagatgaagtcaa0 117 25345 5' UTR 44 gctgaagttcccaggctg35 118 25346 5' UTR 47 ccggctgaagttcccagg53 119 25347 Coding 104 ggcaccatccccaacgat50 120 25348 Coding 105 aggcaccatccccaacga56 121 25349 Coding ~lll tcccacaggcaccatccc4 122 25350 Coding 117 aggtcttcccacaggcac11 123 25351 Coding 127 atgaggaggcaggtcttc6 124 25352 Coding 139 ttgctgaagacgatgagg15 125 25353 Coding 178 tcaaagacagtagggacg32 126 25354 Coding 181 ttctcaaagacagtaggg7 127 25355 Coding 183 agttctcaaagacagtag39 128 25356 Coding 342 tgttttccaggctgtcag59 129 25357 Coding 433 tcgtcttgcctcaggtcc48 130 25358 Coding 439 gtgtgctcgtcttgcctc36 131 25359 Coding 445 ctcctggtgtgctcgtct61 132 25360 Coding 483 cagaccgaacgggctcct50 133 25361 Coding 488 ttcctcagaccgaacggg14 134 25362 Coding 534 actcaaggtagccaaagg32 135 25363 Coding 566 ctcccgcactccctcctt21 136 25364 Coding 575 ctcaaacacctcccgcac9 137 25365 Coding 581 ggccatctcaaacacctc66 138 25366 Coding 614 cttgttcttgcggacctg61 139 25367 Coding 625 cccctccgacgcttgttc0 140 25368 3' UTR 737 gtatggagccctcaggag28 141 25369 3' UTR 746 gagccttcagtatggagc32 142 25370 3' UTR 753 gaaaatggagccttcagt0 143 25371 3' UTR 759 ggaactgaaaatggagcc40 144 25372 3' UTR 763 ggagggaactgaaaatgg45 145 25373 3' UTR 766 gcaggagggaactgaaaa35 146 25374 3' UTR 851 agggcagggcataggcgt5 147 25375 3' UTR 854 ggaagggcagggcatagg0 148 WO 99/53101 PCT/US99/082b8 25376 3' UTR 859 catgaggaagggcagggc 0 149 25377 3' UTR 920 taaagtgctggtgtgtga 20 150 25378 3' UTR 939 cctgtgagccagaagtgt 67 151 25379 3' UTR 941 ttcctgtgagccagaagt 61 152 25380 3' UTR 945 cactttcctgtgagccag 80 153 25381 3' UTR 948 agacactttcctgtgagc 0 154 25382 3' UTR 966 actctgggtccctactgc 0 155 25383 3' UTR 992 tgcagaaacaactccagg 0 156 EXAMPLE 20: Automated Assay of Cellular Inhibitor of Apoptosis-2 Expression Oligonucleotide Activity Cellular Inhibitor of Apoptosis-2 (also known as c-IAP-2, apoptosis inhibitor 2, API-2, hIAP-1, and MIHC) is a member of the inhibitor of apoptosis (IAP) family of anti-apoptotic proteins which interfere with the transmission of intracellular death signals.
Oligonucleotides were designed as described in Example 2, synthesized as described in Examples 3 through 8, analyzed as described in Examples 9 and assayed as described in Example 10 except for target specific primer and probes. Cellular Inhibitor of Apoptosis-2 probes and primers were designed to hybridize to the human Cellular Inhibitor of Apoptosis-2 sequence, using published sequence information (GenBank accession number U37546, incorporated herein by reference as SEQ ID N0:157).
For Cellular Inhibitor of Apoptosis-2 the PCR primers were:
forward primer: GGACTCAGGTGTTGGGAATCTG (SEQ ID NO: 158) reverse primer: CAAGTACTCACACCTTGGAAACCA (SEQ ID NO: 159) and the PCR
probe was: FAM-AGATGATCCATGGGTTCAACATGCCAA-TAMRA (SEQ ID NO:
160) where FAM (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye) and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.
EXAMPLE 21: Antisense inhibition of Cellular Inhibitor of Apoptosis-2 expression-phosphorothioate oligodeoxynucleotides In accordance with the present invention, a series of oligonucleotides were designed to target different regions of the human Cellular Inhibitor of Apoptosis-2 RNA, using published sequences (GenBank accession number U37546, incorporated herein by reference as SEQ ID NO: 157). The oligonucleotides are shown in Table 12.
Target sites are indicated by nucleotide numbers, as given in the sequence source reference {Genbank accession no. LT37546), to which the oligonucleotide binds. All compounds in Table 12 are oligodeoxynucleotides with phosphorothioate backbones (internucleoside linkages) throughout. The compounds were analyzed for effect on Cellular Inhibitor of Apoptosis-2 mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments. If present, "N.D." indicates "no data".
Table 12 Inhibition of Cellular Inhibitor of Apoptosis-2 mRNA levels by phosphorothioate oligodeoxynucleotides ISIS# REGION SEQUENCE % SEQ ID
TARGET

SITE Inhibition NO.

23412 5' UTR 3 actgaagacattttgaat62 161 23413 5' UTR 37 cttagaggtacgtaaaat29 162 23414 5' UTR 49 gcacttttatttcttaga70 163 2341 5' UTR 62 attttaattagaagcact0 164 S

23416 S' UTR 139 accatatttcactgattc70 165 23417 5' UTR 167 ctaactcaaaggaggaaa0 166 23418 5' UTR 175 cacaagacctaactcaaa27 167 23419 5' UTR 268 gctctgctgtcaagtgtt57 168 23420 5' UTR 303 tgtgtgactcatgaagct23 169 23421 5' UTR 335 ttcagtggcattcaatca23 170 23422 5' UTR 357 cttctccaggctactaga50 171 23423 5' UTR 363 ggtcaacttctccaggct65 172 23424 5' UTR 437 taaaacccttcacagaag0 173 23425 5' UTR 525 ttaaggaagaaatacaca0 174 23426 5' UTR 651 gcatggctttgcttttat0 175 23427 Coding 768 caaacgtgttggcgcttt35 176 23428 Coding 830 agcaggaaaagtggaata0 177 23429 Coding 1015 ttaacggaatttagactc0 178 23430 Coding 1064 atttgttactgaagaagg0 179 23431 Coding 1118 agagccacggaaatatcc9 180 23432 Coding 1168 aaatcttgatttgctctg7 181 23433 Coding 1231 gtaagtaatctggcattt0 182 23434 Coding 1323 agcaagccactctgtctc50 183 23435 Coding 1436 tgaagtgtcttgaagctg0 184 23436 Coding 1580 tttgacatcatcactgtt0 185 23437 Coding 1716 tggcttgaacttgacgga0 186 23438 Coding 1771 tcatctcctgggctgtct40 187 23439 Coding 1861 gcagcattaatcacagga 0 188 23440 Coding 2007 tttctctctcctcttccc 10 189 23441 Coding 21 SO aacatcatgttcttgttc 9 190 23442 Coding 2273 atataacacagcttcagc 0 191 23443 Coding 2350 aattgttcttccactggt 0 192 23444 Coding 2460 aagaaggagcacaatctt 70 193 23445 3' UTR 2604 gaaaccaaattaggataa 12 194 23446 3' UTR 2753 tgtagtgctacctctttt 69 195 23447 3' UTR 2779 ctgaaattttgattgaat 14 196 23448 3' UTR 2795 tacaatttcaataatgct 38 197 23449 3' UTR 2920 gggtctcagtatgctgcc 21 198 23450 3' UTR 3005 ccttcgatgtataggaca 0 199 23451 3' UTR 3040 catgtccctaaaatgtca 0 200 EXAMPLE 22: Antisense inhibition of Cellular Inhibitor of Apoptosis-2 expression-phosphorothioate 2'-MOE gapmer oligonucleotides In accordance with the present invention, a second series of oligonucleotides targeted to human Cellular Inhibitor of Apoptosis-2 were synthesized. The oligonucleotide sequences are shown in Table 13. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no.
U37546), to which the oligonucleotide binds.
All compounds in Table 13 are chimeric oligonucleotides ("gapmers") 18 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by four-nucleotide "wings". The wings are composed of 2'-methoxyethyl (2'-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. Cytidine residues in the 2'-MOE wings are 5-methylcytidines.
Data were obtained by real-time quantitative PCR as described in other examples herein and are averaged from three experiments. If present, "N.D." indicates "no data".

Table 13 Inhibition of Cellular Inhibitor of Apoptosis-2 mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS# REGION SEQUENCE % SEQ ID
TARGET

SITE Inhibition NO.

23452 5' UTR 3 actgaagacattttgaat35 161 23453 5' UTR 37 cttagaggtacgtaaaat26 162 23454 S' UTR 49 gcacttttatttcttaga76 163 23455 5' UTR 62 attttaattagaagcact0 164 23456 S' UTR 139 accatatttcactgattc0 165 23457 5' UTR 167 ctaactcaaaggaggaaa5 166 23458 S' UTR 175 cacaagacctaactcaaa0 167 23459 S' UTR 268 gctctgctgtcaagtgtt57 168 23460 5' UTR 303 tgtgtgactcatgaagct67 169 23461 5' UTR 335 ttcagtggcattcaatca59 170 ~

23462 5' UTR 357 cttctccaggctactaga0 171 23463 S' UTR 363 ggtcaacttctccaggct75 172 23464 S' UTR 437 taaaacccttcacagaag11 173 23465 5' UTR 525 ttaaggaagaaatacaca0 174 23466 S' UTR 651 gcatggctttgcttttat46 175 23467 Coding 768 caaacgtgttggcgcttt47 176 23468 Coding 830 agcaggaaaagtggaata39 177 23469 Coding 1015 ttaacggaatttaga.ctc12 178 23470 Coding 1064 atttgttactgaagaagg34 179 23471 Coding 1118 agagccacggaaatatcc54 180 23472 Coding 1168 aaatcttgatttgctctg34 181 23473 Coding 1231 gtaagtaatctggcattt0 182 23474 Coding 1323 agcaagccactctgtctc42 183 23475 Coding 1436 tgaagtgtcttgaagctg0 184 23476 Coding 1580 tttgacatcatcactgtt57 185 23477 Coding 1716 tggcttgaacttgacgga23 186 23478 Coding 1771 tcatctcctgggctgtct66 187 23479 Coding 1861 gcagcattaatcacagga65 188 23480 Coding 2007 tttctctctcctcttccc0 189 23481 Coding 2150 aacatcatgttcttgttc13 190 23482 Coding 2273 atataacacagcttcagc0 191 23483 Coding 2350 aattgttcttccactggt60 192 23484 Coding 2460 aagaaggagcacaatctt65 193 23485 3' UTR 2604 gaaaccaaattaggataa0 194 23486 3' UTR 2753 tgtagtgctacctctttt73 195 23487 3' UTR 2779 ctgaaattttgattgaat4 196 23488 3' UTR 2795 tacaatttcaataatgct 0 197 23489 3' UTR 2920 gggtctcagtatgctgcc 42 198 23490 3' UTR 3005 ccttcgatgtataggaca 71 199 23491 3' UTR 3040 catgtccctaaaatgtca 45 200 EXAMPLE 23: Automated Assay of ELK-1 Oligonucleotide Activity ELK-1 (also known as p62TCF) is a member of the ternary complex factor {TCF) subfamily of Ets domain proteins and utilizes a bipartite recognition mechanism mediated by both protein-DNA and protein-protein interactions. This results in gene regulation not only by direct DNA binding but also by indirect DNA binding through recruitment by other factors (Rao et al., Science, 1989, 244, 66-70). The formation of ternary complexes with an array of proteins allows the differential regulation of many genes.
The mechanism by which ELK-1 controls various signal transduction pathways involves regulating the activity of the Egr-1, pip92, nur77 and c-fos promoters by binding to the serum response element (SRE) in these promoters in response to extracellular stimuli such as growth factors, mitogens and oncogene products (Sharrocks et al., Int. J. Biochem.
Cell Biol., 1997, 29, 1371-1387). ELK-1 has also been shown to mediate other functions within the cell including apoptosis.
Oligonucleotides were designed as described in Example 2, synthesized as described in Examples 3 through 8, analyzed as described in Examples 9 and assayed as described in Example 10 except for target specific primer and probes.
ELK-1 probes and primers were designed to hybridize to the human ELK-1 sequence, using published sequence information (GenBank accession number M25269, incorporated herein by reference as SEQ ID N0:201).
For ELK-1 the PCR primers were:
forward primer: GCAAGGCAATGGCCACAT (SEQ ID NO: 202) reverse primer: CTCCTCTGCATCCACCAGCTT (SEQ ID NO: 203) and the PCR probe was: FAM-TCTCCTGGACTTCACGGGATGGTGGT-TAMRA (SEQ ID NO: 204) where FAM (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye) and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.

EXAMPLE 24: Antisense inhibition of ELK-1 expression-phosphorothioate oligodeoxynucleotides In accordance with the present invention, a series of oligonucleotides were designed to target different regions of the human ELK-I RNA, using published sequences (GenBank accession number M25269, incorporated herein by reference as SEQ ID
NO:
201 ). The oligonucleotides are shown in Table 14. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no.
M25269), to which the oligonucleotide binds. All compounds in Table 14 are oligodeoxynucleotides with phosphorothioate backbones (internucleoside linkages) throughout. The compounds were analyzed for effect on ELK-1 mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments. If present, "N.D." indicates "no data".
Table 14 Inhibition of ELK-1 mRNA levels by phosphorothioate oligodeoxynucleotides ISIS# REGION SEQUENCE % SEQ ID
TARGET

SITE Inhibition NO.

24752 5' UTR 11 cccctgcgtttccctaca15 205 24753 5' UTR 50 ggtggtggtggcggtggc29 206 24754 5' UTR 139 ggcgttggcaatgttggc82 207 24755 5' UTR 167 aagttgaggctgtgtgta0 208 24756 5' UTR 189 aggccacggacgggtctc92 209 24757 5' UTR 229 gattgattcgctacgatg71 210 24758 5' UTR 255 gggatgcggaggagtgcg74 211 24759 5' UTR 289 agtgctcacgccatccca22 212 24760 Coding 328 aaactgccacagcgtcac64 213 24761 Coding 381 gaagtccaggagatgatg62 214 24762 Coding 395 caccaccatcccgtgaag88 215 24763 Coding 455 tcttgttcttgcgtagtc62 216 24764 Coding 512 tgttcttgtcatagtagt52 217 24765 Coding 527 tcaccttgcggatgatgt57 218 24766 Coding 582 gagcaccctgcgacctca72 219 24767 Coding 600 ggcgggcagtcctcagtg82 220 24768 Coding 787 ggtgaaggtggaatagag58 221 24769 Coding 993 tccgatttcaggtttggg55 222 24770 Coding 1110 ttggtggtttctggcaca67 223 24771 Coding 1132 tggagggacttctggctc 69 224 24772 Coding 1376 gcgtaggaagcagggatg 34 225 24773 Coding 1440 gtgctccagaagtgaatg 64 226 24774 Coding 1498 actggatggaaactggaa 34 227 24775 Coding 1541 ggccatccacgctgatag 74 228 24776 3' UTR 1701 ccaccacaatcagagcat 74 229 24777 3' UTR 1711 gatccccaccccaccaca 16 230 24778 3' UTR 1765 tgttttctgtggaggaga 48 231 24779 3' UTR 1790 aaacagagaagttgtgga 11 232 24780 3' UTR 1802 gggactgacagaaaacag 0 233 24781 3' UTR 1860 ataaataaataaaccgcc 18 234 24782 3' UTR 1894 gttaggtcaggctcatcc 56 235 24783 3' UTR 1974 gttctcaagccagacctc 52 236 24784 3' UTR 1992 aataaagaaagaaaggtc 41 237 24785 3' UTR 2006 agggcaggctgagaaata 29 238 24786 3' UTR 2053 cttctactcacatccaaa 54 239 24787 3' UTR 2068 caaaacaaactaactctt 24 240 24788 3' UTR 2080 ggaataataaaacaaaac 40 241 24789 3' UTR 2107 ttcttcctggacccctga 93 242 24790 3' UTR 2161 ccaagggtgtgattcttc 81 243 24791 3' UTR 2200 tgtctgagagaaaggttg 55 244 EXAMPLE 25: Antisense inhibition of ELK-1 expression- phosphorothioate 2'-MOE gapmer oligonucleotides In accordance with the present invention, a second series of oligonucleotides targeted to human ELK-1 were synthesized. The oligonucleotide sequences are shown in Table 15. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no. M25269), to which the oligonucleotide binds.
All compounds in Table 15 are chimeric oligonucleotides ("gapmers") 18 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by four-nucleotide "wings". The wings are composed of 2'-methoxyethyl (2'-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. Cytidine residues in the 2'-MOE wings are 5-methylcytidines.
Data were obtained by real-time quantitative PCR as described in other examples herein and are averaged from three experiments. If present, "N.D." indicates "no data".

Table 15 Inhibition of ELK-1 mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS# REGION SEQUENCE % SEQ
TARGET ID

SITE Inhibition NO.

24792 5' UTR 11 cccctgcgtttccctaca23 205 24793 5' UTR 50 ggtggtggtggcggtggc80 206 24794 5' UTR 139 ggcgttggcaatgttggc91 207 24795 5' UTR 167 aagttgaggctgtgtgta27 208 24796 5' UTR 189 aggccacggacgggtctc79 209 24797 5' UTR 229 gattgattcgctacgatg69 210 24798 5' UTR 255 gggatgcggaggagtgcg42 211 24799 5' UTR 289 agtgctcacgccatccca45 212 24800 Coding 328 aaactgccacagcgtcac57 213 24801 Coding 381 gaagtccaggagatgatg55 214 24802 Coding 395 caccaccatcccgtgaag41 215 24803 Coding 455 tcttgttcttgcgtagtc80 216 24804 Coding 512 tgttcttgtcatagtagt65 217 24805 Coding 527 tcaccttgcggatgatgt70 218 24806 Coding 582 gagcaccctgcgacctca64 219 24807 Coding 600 ggcgggcagtcctcagtg67 220 24808 Coding 787 ggtgaaggtggaatagag45 221 24809 Coding 993 tccgatttcaggtttggg75 222 24810 Coding 1110 ttggtggtttctggcaca82 223 24811 Coding 1132 tggagggacttctggctc60 224 24812 Coding 1376 gcgtaggaagcagggatg49 225 24813 Coding 1440 gtgctccagaagtgaatg71 226 24814 Coding 1498 actggatggaaactggaa62 227 24815 Coding 1541 ggccatccacgctgatag78 228 24816 3' UTR 1701 ccaccacaatcagagcat54 229 24817 3' UTR 1711 gatccccaccccaccaca44 230 24818 3' UTR 1765 tgltttctgtggaggaga74 231 24819 3' UTR 1790 aaacagagaagttgtgga64 232 24820 3' UTR 1802 gggactgacagaaaacag16 233 24821 3' UTR 1860 ataaataaataaaccgcc38 234 24822 3' UTR 1894 gttaggtcaggctcatcc59 235 24823 3' UTR 1974 gttctcaagccagacctc62 236 24824 3' UTR 1992 aataaagaaagaaaggtc35 237 24825 3' UTR 2006 agggcaggctgagaaata0 238 24826 3' UTR 2053 cttctactcacatccaaa46 239 24827 3' UTR 2068 caaaacaaactaactctt38 240 24828 3' UTR 2080 ggaataataaaacaaaac37 241 24829 3' UTR 2107 ttcttcctggacccctga 71 242 24830 3' UTR 2161 ccaagggtgtgattcttc 88 243 24831 3' UTR 2200 tgtctgagagaaaggttg 65 244 EXAMPLE 26: Automated Assay of Gi alpha proteins Oligonucleotide Activity G-alpha-11 is a member of the Gq subfamily of G proteins whose primary function is to activate PLC-b isoforms producing second messengers and affecting intracellular calcium stores.
Oligonucleotides were designed as described in Example 2, synthesized as described in Examples 3 through 8, analyzed as described in Examples 9 and assayed as described in Example 10 except for target specific primer and probes. G-alpha-11 probes and primers were designed to hybridize to the human G-alpha-11 sequence, using published sequence information (GenBank accession number AF011497, incorporated herein by reference as SEQ ID N0:245). For G-alpha-11 the PCR primers were:
forward primer: TGACCACCTTCGAGCATCAG (SEQ ID NO: 246) reverse primer: CGGTCGTAGCATTCCTGGAT (SEQ ID NO: 247) and the PCR probe was: FAM-TCAGTGCCATCAAGACCCTGTGGGAG-TAMRA (SEQ ID NO: 248) where FAM (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye) and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.
EXAMPLE 27: Antisense inhibition of G-alpha-11 expression- phosphorothioate oligodeoxynucleotides In accordance with the present invention, a series of oligonucleotides were designed to target different regions of the human G-alpha-11 RNA, using published sequences (GenBank accession number AF011497, incorporated herein by reference as SEQ ID NO: 245). The oligonucleotides are shown in Table 16. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no.
AFOl 1497), to which the oligonucleotide binds. All compounds in Table 16 are oligodeoxynucleotides with phosphorothioate backbones (internucleoside linkages) throughout. The compounds were analyzed for effect on G-alpha-11 mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments. If present, "N.D." indicates "no data".
Table 16 Inhibition of G-alpha-11 mRNA levels by phosphorothioate oligodeoxynucleotides ISIS# REGION TARGET SEQUENCE % SEQ ID

SITE Inhibition NO.

20576 Coding 1 gatggactccagagtcat0 249 20577 Coding 6 gccatgatggactccaga75 250 20578 Coding 9 cacgccatgatggactcc0 251 20579 Coding 25 ctcatcgctcaggcaaca61 252 20580 Coding 31 cttcacctcatcgctcag20 253 20581 Coding 36 gactccttcacctcatcgi 5 254 20582 Coding 45 atccgcttggactccttc17 255 20583 Coding 50 cgttgatccgcttggact0 256 20584 Coding 61 ctcgatctcggcgttgat0 257 20585 Coding 77 cccgccgcagctgcttct58 258 20586 Coding 106 cttgagctcgcgccgggc31 259 20587 Coding 116 gcagcagcagcttgagct0 260 20588 Coding 127 gcccgtgccgagcagcag0 261 20589 Coding 146 acgtgctcttcccgctct28 262 20590 Coding 159 atctgcttgatgaacgtg0 263 20591 Coding 162 cgcatctgcttgatgaac0 264 20592 Coding 184 gtagccggcgccgtggat1 265 20593 Coding 197 tgtcctcctccgagtagc0 266 20594 Coding 199 cttgtcctcctccgagta79 267 20595 Coding 207 aagccgcgcttgtcctcc56 268 20596 Coding 222 tagacgagcttggtgaag0 269 20597 Coding 230 tgttctggtagacgagct0 270 20598 Coding 242 tggcggtgaagatgttct0 271 20599 Coding 258 cggatcatggcctgcatg1 272 20600 Coding 271 cgtctccatggcccggat49 273 20601 Coding 285 tagaggatcttgagcgtc0 274 20602 Coding 287 tgtagaggatcttgagcg0 275 20603 Coding 297 tgctcgtacttgtagagg7 276 20604 Coding 306 gccttgttctgctcgtac25 277 20605 Coding 309 ttggccttgttctgctcg0 278 20606 Coding 319 caggagcgcattggcctt0 279 20607 Coding 340 ctccacgtccacctcccg69 280 20608 Coding 349 ggtcaccttctccacgtc27 281 20609 Coding 362 gatgctcgaaggtggtca33 282 20610 Coding 373 actgacgtactgatgctc36 283 20611 Coding 382 cttgatggcactgacgta78 284 20612 Coding 388 cagggtcttgatggcact0 285 20613 Coding 409 ctggatgcccgggtcctc 0 286 20614 Coding 411 tcctggatgcccgggtcc 30 287 20615 Coding 429 cgcctgcggtcgtagcat 0 288 20616 Coding 440 gctggtactcgcgcctgc 41 289 20617 Coding 459 tacttggcagagtcggag 34 290 20618 Coding 468 gtcaggtagtacttggca 76 291 20619 Coding 479 ggtcaacgtcggtcaggt 18 292 20620 Coding 489 gtggcgatgcggtcaacg 1 293 20621 Coding 503 gcaggtagcccaaggtgg 20 294 20622 Coding 518 cgtcctgctgggtgggca 40 295 20623 Coding 544 ggtggtgggcacgcggac 0 296 20624 Coding 555 tcgatgatgccggtggtg 0 297 20625 Coding 572 ccaggtcgaaagggtact 0 298 20626 Coding 578 tgttctccaggtcgaaag 33 299 20627 Coding 584 agatgatgttctccaggt 0 300 20628 Coding 591 atccggaagatgatgttc 0 301 20629 Coding 624 ctccgctccgaccgctgg 56 302 20630 Coding 634 gatccacttcctccgctc 59 303 20631 Coding 655 tgtcacgttctcaaagca 0 304 20632 Coding 663 atgatggatgtcacgttc 0 305 20633 Coding 671 cgagaaacatgatggatg 0 306 20634 Coding 682 gctgagggcgacgagaaa 75 307 20635 Coding 709 cgactccaccaggacttg 40 308 20636 Coding 726 atccggttctcgttgtcc 22 309 20637 Coding 728 ccatccggttctcgttgt 19 310 20638 Coding 744 agggctttgctctcctcc 77 311 20639 Coding 754 ggtccggaacagggcttt 26 312 20640 Coding 766 gtaggtgatgatggtccg 0 313 20641 Coding 787 ggaggagttctggaacca 64 314 20642 Coding 803 tgaggaagaggatgacgg 0 315 20643 Coding 818 gcaggtccttcttgttga 6 316 20644 Coding 831 atcttgtcctccagcagg 4 317 20645 Coding 842 gcgagtacaggatcttgt 17 318 20646 Coding 858 aagtagtccaccaggtgc 0 319 20647 Coding 910 gatgaactcccgcgccgc 52 320 20648 Coding 935 ggttcaggtccacgaaca 71 321 20649 Coding 958 gtagatgatcttgtcgct 0 322 20650 Coding 972 cacgtgaagtgtgagtag 0 323 20651 Coding 993 atgttctccgtgtcggtg 0 324 20652 Coding 1014 acggccgcgaacacgaag 6 325 20653 Coding 1027 gatggtgtccttcacggc 0 326 20654 Coding 1043 tcaggttcagctgcagga 3 327 20655 Coding 1059 accagattgtactccttc 0 328 EXAMPLE 28: Antisense inhibition of G-alpha-11 expression- phosphorothioate 2'-MOE gapmer oligonucleotides In accordance with the present invention, a second series of oligonucleotides targeted to human G-alpha-11 were synthesized. The oligonucleotide sequences are shown in Table 17. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no. AF011497), to which the oligonucleotide binds.
All compounds in Table 17 are chimeric oligonucleotides ("gapmers") 18 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by four-nucleotide "wings". The wings are composed of 2'-methoxyethyl (2'-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. Cytidine residues in the 2'-MOE wings are 5-methylcytidines.
Data were obtained by real-time quantitative PCR as described in other examples herein and are averaged from three experiments. If present, "N.D." indicates "no data".
Table 17 Inhibition of G-alpha-11 mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS# REGION TARGET SEQUENCE % SEQ ID

SITE Inhibition NO.

20981 Coding 1 gatggactccagagtcat0 249 20982 Coding 6 gccatgatggactccaga0 250 20983 Coding 9 cacgccatgatggactcc0 251 20984 Coding 25 ctcatcgctcaggcaaca0 252 20985 Coding 31 cttcacctcatcgctcag2 253 20986 Coding 36 gactccttcacctcatcg0 254 20987 Coding 45 atccgcttggactccttc19 255 20988 Coding 50 cgttgatccgcttggact15 256 20989 Coding 61 ctcgatctcggcgttgat0 257 20990 Coding 77 cccgccgcagctgcttct41 258 20991 Coding 106 cttgagctcgcgccgggc19 259 20992 Coding 116 gcagcagcagcttgagct23 260 20993 Coding 127 gcccgtgccgagcagcag38 261 20994 Coding 146 acgtgctcttcccgctct34 262 20995 Coding 159 atctgcttgatgaacgtg56 263 20996 Coding 162 cgcatctgcttgatgaac 31 264 20997 Coding 184 gtagccggcgccgtggat 0 265 20998 Coding 197 tgtcctcctccgagtagc 42 266 20999 Coding 199 cttgtcctcctccgagta 0 267 21000 Coding 207 aagccgcgcttgtcctcc 73 268 21001 Coding 222 tagacgagcttggtgaag 0 269 21002 Coding 230 tgttctggtagacgagct 61 270 21003 Coding 242 tggcggtgaagatgttct 14 271 21004 Coding 258 cggatcatggcctgcatg 84 272 21005 Coding 271 cgtctccatggcccggat 70 273 21006 Coding 285 tagaggatcttgagcgtc 39 274 21007 Coding 287 tgtagaggatcttgagcg 28 275 21008 Coding 297 tgctcgtacttgtagagg 70 276 21009 Coding 306 gccttgttctgctcgtac 76 277 21010 Coding 309 ttggccttgttctgctcg 0 278 21011 Coding 319 caggagcgcattggcctt 87 279 21012 Coding 340 ctccacgtccacctcccg 0 280 21013 Coding 349 ggtcaccttctccacgtc 69 281 21014 Coding 362 gatgctcgaaggtggtca 0 282 21015 Coding 373 actgacgtactgatgctc 69 283 21016 Coding 382 cttgatggcactgacgta 32 284 21017 Coding 388 cagggtcttgatggcact 19 285 21018 Coding 409 ctggatgcccgggtcctc 63 286 21019 Coding 411 tcctggatgcccgggtcc 56 287 21020 Coding 429 cgcctgcggtcgtagcat 73 288 21021 Coding 440 gctggtactcgcgcctgc 68 289 21022 Coding 459 tacttggcagagtcggag 50 290 21023 Coding 468 gtcaggtagtacttggca 13 291 21024 Coding 479 ggtcaacgtcggtcaggt 64 292 21025 Coding 489 gtggcgatgcggtcaacg 52 293 21026 Coding 503 gcaggtagcccaaggtgg 52 294 21027 Coding 518 cgtcctgctgggtgggca 0 295 21028 Coding 544 ggtggtgggcacgcggac 81 296 21029 Coding 555 tcgatgatgccggtggtg 48 297 21030 Coding 572 ccaggtcgaaagggtact 61 298 21031 Coding 578 tgttctccaggtcgaaag 0 299 21032 Coding 584 agatgatgttctccaggt 0 300 21033 Coding 591 atccggaagatgatgttc 0 301 21034 Coding 624 ctccgctccgaccgctgg 59 302 21035 Coding 634 gatccacttcctccgctc 17 303 21036 Coding 655 tgtcacgttctcaaagca 9 304 21037 Coding 663 atgatggatgtcacgttc 41 305 21038 Coding 671 cgagaaacatgatggatg 0 306 WO 99/53101 PCT/US99/0$268 21039 Coding 682 gctgagggcgacgagaaa 11 307 21040 Coding 709 cgactccaccaggacttg 0 308 21041 Coding 726 atccggttctcgttgtcc 67 309 21042 Coding 728 ccatccggttctcgttgt 30 310 21043 Coding 744 agggctttgctctcctcc 61 311 21044 Coding 754 ggtccggaacagggcttt 72 312 21045 Coding 766 gtaggtgatgatggtccg 68 313 21046 Coding 787 ggaggagttctggaacca 54 314 21047 Coding 803 tgaggaagaggatgacgg 23 31 S

21048 Coding 818 gcaggtccttcttgttga 0 316 21049 Coding 831 atcttgtcctccagcagg 39 317 21050 Coding 842 gcgagtacaggatcttgt 74 318 21051 Coding 858 aagtagtccaccaggtgc 36 319 21052 Coding 910 gatgaactcccgcgccgc 67 320 21053 Coding 935 ggttcaggtccacgaaca 37 321 21054 Coding 958 gtagatgatcttgtcgct 64 322 21055 Coding 972 cacgtgaagtgtgagtag 37 323 21056 Coding 993 atgttctccgtgtcggtg 0 324 21057 Coding 1014 acggccgcgaacacgaag 0 325 21058 Coding 1027 gatggtgtccttcacggc 69 326 21059 Coding 1043 tcaggttcagctgcagga 0 327 21060 Coding 1059 accagattgtactccttc 0 328 EXAMPLE 29: Automated Assay of AKT-1 Oligonucleotide Activity Akt-1 {also known as PKB alpha and RAC-PK alpha) is a member of the AKT/PKB family of serine/threonine kinases and has been shown to be involved in a diverse set of signaling pathways.
Oligonucleotides were designed as described in Example 2, synthesized as described in Examples 3 through 8, analyzed as described in Examples 9 and assayed as described in Example 10 except for target specific primer and probes. AKT-1 probes and primers were designed to hybridize to the human AKT-1 sequence, using published sequence information (GenBank accession number M63167, incorporated herein by reference as SEQ ID N0:329). For Akt-1 the PCR primers were:
forward primer: CGTGACCATGAACGAGTTTGA (SEQ ID NO: 330) reverse primer: CAGGATCACCTTGCCGAAA (SEQ ID NO: 331) and the PCR probe was: FAM-CTGAAGCTGCTGGGCAAGGGCA-TAMRA (SEQ ID NO: 332) where FAM (PE-Applied Biosystems, Foster City, CA) is the fluorescent reporter dye) and TAMRA (PE-Applied Biosystems, Foster City, CA) is the quencher dye.

EXMAMPLE 30: Antisense inhibition of Akt-1 expression- phosphorothioate oligodeoxynucleotides In accordance with the present invention, a series of oligonucleotides were designed to target different regions of the human Akt-1 RNA, using published sequences S (GenBank accession number M63167, incorporated herein by reference as SEQ ID
NO:
329). The oligonucleotides are shown in Table 18. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no.
M63167), to which the oligonucleotide binds. Ail compounds in Table 18 are oligodeoxynucleotides with phosphorothioate backbones (internucleoside linkages) throughout. The compounds were analyzed for effect on Akt-1 mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments. If present, "N.D." indicates "no data".
Table 18 1 S Inhibition of Akt-1 mRNA levels by phosphorothioate oligodeoxynucleotides ISIS# REGION TARGET SEQUENCE % SEQ
ID

SITE Inhibition NO.

28880 S' UTR 4 ccctgtgccctgtcccagSS 333 28881 S' UTR 27 cctaagcccctggtgaca1S 334 28882 S' UTR 62 ctttgacttctttgaccc68 33S

28883 S' UTR 70 ggcagcccctttgacttcS3 336 28884 Coding 213 caaccctccttcacaata24 337 28885 Coding 234 tactcccctcgtttgtgc0 338 28886 Coding 281 tgccatcattcttgagga6S 339 2S 28887 Coding 293 agccaatgaaggtgccat67 340 28888 Coding 3S2 cacagagaagttgttgag22 341 28889 Coding 496 agtctggatggcggttgt49 342 28890 Coding S31 tcctcctcctcctgcttc9 343 28891 Coding S70 cctgagttgtcactgggt49 344 28892 Coding 666 ccgaaagtgcccttgcccS6 34S

28893 Coding 744 gccacgatgacttccttc60 346 28894 Coding 927 cggtcctcggagaacaca0 347 28895 Coding 990 acgttcttctccgagtgc30 348 28896 Coding 1116 gtgccgcaaaaggtcttc66 349 3S 28897 Coding 1125 tactcaggtgtgccgcaa66 3S0 28898 Coding 1461 ggcttgaagggtgggctg41 3 S

28899 Coding 1497 tcaaaatacctggtgtcaS 1 3S2 28900 Coding 1512 gccgtgaactcctcatea 56 353 28901 Coding 1541 ggtcaggtggtgtgatgg 0 354 28902 Coding 1573 ctcgetgtccacaeactc 61 355 28903 3' UTR 1671 gcctctccatccctccaa 76 356 28904 3' UTR 1739 acagcgtggcttctctca 12 357 28905 3' UTR 1814 ttttcttccctaccccgc 64 358 28906 3' UTR 1819 gatagttttettccetac 0 359 28907 3' UTR 1831 taaaacccgcaggatagt 74 360 28908 3' UTR 1856 ggagaacaaactggatga 0 361 28909 3' UTR 1987 ctggctgacagagtgagg 59 362 28910 3' UTR 1991 gcggctggctgacagagt 61 363 28911 3' UTR 2031 cccagagagatgacagat 46 364 28912 3' UTR 2127 gctgctgtgtgcctgcca 38 365 28913 3' UTR 2264 cataataeacaataacaa 39 366 28914 3' UTR 2274 atttgaacaacataatac 11 367 28915 3' UTR 2397 aagtgctaccgtggagag 57 368 28916 3' UTR 2407 cgaaaaggtcaagtgeta 41 369 28917 3' UTR 2453 cagggagtcagggagggc 13 370 28918 3' UTR 2545 aaagttgaatgttgtaaa 10 371 28919 3' UTR 2553 aaaatactaaagttgaat 25 372 EXAMPLE 31: Antisense inhibition of Akt-1 expression- phosphorothioate 2'-MOE
gapmer oligonucleotides In accordance with the present invention, a second series of oligonucleotides targeted to human Akt-1 were synthesized. The oligonucleotide sequences are shown in Table 19. Target sites are indicated by nucleotide numbers, as given in the sequence source reference (Genbank accession no. M63167), to which the oligonucleotide binds.
All compounds in Table 19 are chimeric oligonucleotides ("gapmers") 18 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by four-nucleotide "wings". The wings are composed of 2'-methoxyethyl (2'-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. Cytidine residues in the 2'-MOE wings are 5-methylcytidines.
Data were obtained by real-time quantitative PCR as described in other examples herein and are averaged from three experiments. If present, "N.D." indicates "no data".

Table 19 Inhibition of Akt-1 mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS# REGION TARGET SEQUENCE % SEQ ID

SITE Inhibition NO.

28920 5' UTR 4 ccctgtgccctgtcccag88 333 28921 S' UTR 27 cctaagcccctggtgaca44 334 28922 S' UTR 62 ctttgacttctttgaccc61 335 28923 5' UTR 70 ggcagcccctttgacttc79 336 28924 Coding 213 caaccctccttcacaata72 337 28925 Coding 234 tactcccctcgtttgtgc39 338 28926 Coding 281 tgccatcattcttgagga73 339 28927 Coding 293 agccaatgaaggtgccat62 340 28928 Coding 352 cacagagaagttgttgag48 341 28929 Coding 496 agtctggatggcggttgt43 342 28930 Coding 531 tcctcctcctcctgcttc49 343 28931 Coding 570 cctgagttgtcactgggt71 344 28932 Coding 666 ccgaaagtgcccttgccc64 345 28933 Coding 744 gccacgatgacttccttc66 346 28934 Coding 927 cggtcctcggagaacaca77 347 28935 Coding 990 acgttcttctccgagtgc89 348 28936 Coding 1116 gtgccgcaaaaggtcttc61 349 28937 Coding 1125 tactcaggtgtgccgcaa74 350 28938 Coding 1461 ggcttgaagggtgggctg54 351 28939 Coding 1497 tcaaaatacctggtgtca78 352 28940 Coding 1512 gccgtgaactcctcatca88 353 28941 Coding 1541 ggtcaggtggtgtgatgg71 354 28942 Coding 1573 ctcgctgtccacacactc83 355 28943 3' UTR 1671 gcctctccatccctccaa86 356 28944 3' UTR 1739 acagcgtggcttctctca73 357 28945 3' UTR 1814 ttttcttccctaccccgc77 358 28946 3' UTR 1819 gatagttttcttccctac43 359 28947 3' UTR 1831 taaaacccgcaggatagt64 360 28948 3' UTR 1856 ggagaacaaactggatga70 361 28949 3' UTR 1987 ctggctgacagagtgagg90 362 28950 3' UTR 1991 gcggctggctgacagagt82 363 28951 3' UTR 2031 cccagagagatgacagat53 364 28952 3' UTR 2127 gctgctgtgtgcctgcca80 365 28953 3' UTR 2264 cataatacacaataacaa48 366 28954 3' UTR 2274 atttgaacaacataatac39 367 28955 3' UTR 2397 aagtgctaccgtggagag38 368 28956 3' UTR 2407 cgaaaaggtcaagtgcta83 369 28957 3' UTR 2453 cagggagtcagggagggc 59 370 28958 3' UTR 2545 aaagttgaatgttgtaaa 25 371 28959 3' UTR 2553 aaaatactaaagttgaat 45 372 SEQUENCE LISTING
<110> ISIS Pharmaceuticals, Inc.
Cowsert, Lex M.
Baker, Brenda F.
McNeil, John Freier, Susan M.
Sasmor, Henri M.
Brooks, Douglas G.
Ohashi, Cara Wyatt, Jacqueline R.
Borchers, Alexander Vickers, Timothy A.
<120> Identification of Genetic Targets for Modulation by Oligonucleotides and Generation of Oligonucleotides for Gene Modulation <130> ISIS-3456 <140>
<141>
<150> US 09/067,638 <151> 1998-04-28 <150> US 60/081,483 <151 > 1998-04-13 <160> 372 <210> 1 <21l> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 1 ccaggcggca ggaccact 18 <210> 2 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 2 gaccaggcgg caggacca 18 <210> 3 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 3 aggtgagacc aggcggca 18 <210> 4 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 4 cagaggcaga cgaaccat 18 <210> 5 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 5 gcagaggcag acgaacca 18 <210> 6 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 6 gcaagcagcc ccagagga 18 <210> 7 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 7 ggtcagcaag cagcccca 18 <210> 8 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 8 gacagcggtc agcaagca 18 <210> 9 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 9 gatggacagc ggtcagca 18 <210>10 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 10 tctggatgga cagcggtc 18 <210> 11 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 11 ggtggttctg gatggaca 18 <210>12 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 12 gtgggtggtt ctggatgg 18 <210>13 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 13 gcagtgggtg gttctgga 18 <210>14 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 14 cacaaagaac agcactga 18 <210> 15 <211>18 <212>DNA

<213>Artificial Sequence <223>Antisense Oligonucleotide <400> 15 ctggcacaaa gaacagca 18 <210>16 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 16 tcctggctgg cacaaaga 18 <210>17 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 17 ctgtcctggc tggcacaa 18 <210> 18 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 18 ctcaccagtt tctgtcct 18 <210> 19 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 19 tcactcacca gtttctgt 18 <210> 20 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 20 gtgcagtcac tcaccagt 18 <210> 21 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 21 actctgtgca gtcactca 18 <210> 22 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 22 cagtgaactc tgtgcagt 18 <210> 23 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 23 attccgtttc agtgaact 18 <210> 24 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 24 gaaggcattc cgtttcag 18 <210> 25 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 25 ttcaccgcaa ggaaggca 18 <210> 26 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 26 ctctgttcca ggtgtcta 18 <210> 27 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 27 ctggtggcag tgtgtctc 18 <210> 28 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 28 tggggtcgca gtatttgt 18 <210> 29 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 29 ggttggggtc gcagtatt 18 <210>30 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 30 ctaggttggg gtcgcagt 18 <210> 31 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 31 ggtgcccttc tgctggac 18 <210>32 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 32 ctgaggtgcc cttctgct 18 <210> 33 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 33 gtgtctgttt ctgaggtg 18 <210> 34 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 34 tggtgtctgt ttctgagg 18 <210> 35 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 35 acaggtgcag atggtgtc 18 <210> 36 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 36 ttcacaggtg cagatggt 18 <210> 37 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 37 gtgccagcct tcttcaca 18 <210> 38 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 38 tacagtgcca gccttctt 18 <210> 39 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 39 ggacacagct ctcacagg 18 <210> 40 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 40 tgcaggacac agctctca 18 <210> 41 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 41 gagcggtgca ggacacag 18 <210>42 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 42 aagccgggcg agcatgag 18 <210> 43 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 43 aatctgcttg accccaaa 18 <210> 44 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 44 gaaacccctg tagcaatc 18 <210> 45 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 45 gtatcagaaa cccctgta 18 <210> 46 <211> 18 --<212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 46 gctcgcagat ggtatcag 18 <210> 47 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 47 gcagggctcg cagatggt 18 <210> 48 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 48 tgggcagggc tcgcagat 18 <210>49 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 49 gactgggcag ggctcgca 18 <210>50 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 50 cattggagaa gaagccga 18 <210> 51 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 51 gatgacacat tggagaag 18 <210> 52 <211> 18 <2I2> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 52 gcagatgaca cattggag 18 <210>53 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 53 tcgaaagcag atgacaca 18 <210> 54 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 54 gtccaagggt gacatttt 18 <210>55 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 55 cacagcttgt ccaagggt 18 <210>56 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 56 ttggtctcac agcttgtc 18 <210> 57 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 57 caggtctttg gtctcaca 18 <210>58 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 58 ctgttgcaca accaggtc 18 <210>59 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucieotide <400> 59 gtttgtgcct gcctgttg 18 <210>60 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 60 gtcttgtttg tgcctgcc 18 <210> 61 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 61 ccacagacaa catcagtc 18 <210>62 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 62 ctggggacca cagacaac 18 <210> 63 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 63 tcagccgatc ctggggac 18 <210>64 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 64 caccaccagg gctctcag 18 <210>65 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 65 gggatcacca ccagggct 18 <210>66 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 66 gaggatggca aacaggat 18 <210> 67 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 67 accagcacca agaggatg 18 <210>68 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 68 ttttgataaa gaccagca 18 <210>69 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 69 tattggttgg cttcttgg 18 <210>70 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 70 gggttcctgc ttggggtg 18 <210> 71 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 71 gtegggaaaa ttgatetc 18 <210> 72 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 72 gatcgtcggg aaaattga 18 <210>73 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 73 ggagccagga agatcgtc 18 <210> 74 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 74 tggagccagg aagatcgt 18 <210> 75 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 75 tggagcagca gtgttgga 18 <210> 76 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 76 gtaaagtctc ctgcactg 18 <210>77 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 77 tggcatccat gtaaagtc 18 <210> 78 <211>18 <212>DNA

<213>Artificial Sequence <223>Antisense Oligonucleotide <400> 78 cggttggcat ccatgtaa 18 <210>79 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 79 ctctttgcca tcctcctg 18 <210> 80 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 80 ctgtctctcc tgcactga 18 <210> 81 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 81 ggtgcagcct cactgtct 18 <210> 82 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 82 aactgcctgt ttgcccac 18 <210>83 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 83 cttctgcctg cacccctg 18 <210>84 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 84 actgactggg catagctc 18 <210>85 <211>1004 <212>DNA

<213>Homo sapiens <220>
<221> CDS
<222> (48)..(881) <400> 85 gcctcgctcg ggcgcccagt ggtcctgccg cctggtctca cctcgcc atg gtt cgt 56 Met Val Arg ctg cct ctg cag tgc gtc ctc tgg ggc tgc ttg ctg acc get gtc cat 104 Leu Pro Leu Gln Cys Val Leu Trp Gly Cys Leu Leu Thr Ala Val His cca gaa cca ccc act gca tgc aga gaa aaa cag tac cta ata aac agt 152 Pro Glu Pro Pro Thr AIa Cys Arg Glu Lys Gln Tyr Leu Ile Asn Ser cag tgc tgt tct ttg tgc cag cca gga cag aaa ctg gtg agt gac tgc 200 Gln Cys Cys Ser Leu Cys Gln Pro Gly Gln Lys Leu Val Ser Asp Cys aca gag ttc act gaa acg gaa tgc ctt cct tgc ggt gaa agc gaa ttc 248 Thr Glu Phe Thr Glu Thr Glu Cys Leu Pro Cys Gly Glu Ser Glu Phe cta gac acc tgg aac aga gag aca cac tgc cac cag cac aaa tac tgc 296 Leu Asp Thr Trp Asn Arg Glu Thr His Cys His Gln His Lys Tyr Cys gac ccc aac cta ggg ctt cgg gtc cag cag aag ggc acc tca gaa aca 344 Asp Pro Asn Leu Gly Leu Arg Val Gln Gln Lys Gly Thr Ser Glu Thr gac acc atc tgc acc tgt gaa gaa ggc tgg cac tgt acg agt gag gcc 392 Asp Thr Ile Cys Thr Cys Glu Glu Gly Trp His Cys Thr Ser Glu Ala tgt gag agc tgt gtc ctg cac cgc tca tgc tcg ccc ggc ttt ggg gtc 440 Cys GIu Ser Cys Val Leu His Arg Ser Cys Ser Pro Gly Phe Gly Val aag cag att get aca ggg gtt tct gat acc atc tgc gag ccc tgc cca 488 Lys Gln Ile Ala Thr Gly Val Ser Asp Thr Ile Cys Glu Pro Cys Pro gtc ggc ttc ttc tcc aat gtg tca tct get ttc gaa aaa tgt cac cct 536 Val Gly Phe Phe Ser Asn Val Ser Ser Ala Phe Glu Lys Cys His Pro tgg aca agc tgt gag acc aaa gac ctg gtt gtg caa cag gca ggc aca 584 Trp Thr Ser Cys Glu Thr Lys Asp Leu Val Val Gln Gln Ala Gly Thr aac aag act gat gtt gtc tgt ggt ccc cag gat cgg ctg aga gcc ctg 632 Asn Lys Thr Asp Val Val Cys Gly Pro Gln Asp Arg Leu Arg Ala Leu gtg gtg atc ccc atc atc ttc ggg atc ctg ttt gcc atc ctc ttg gtg 680 Val Val Ile Pro Ile Ile Phe Gly Ile Leu Phe Ala Ile Leu Leu Val ctg gtc ttt atc aaa aag gtg gcc aag aag cca acc aat aag gcc ccc 728 Leu Val Phe Ile Lys Lys Val Ala Lys Lys Pro Thr Asn Lys Ala Pro cac ccc aag cag gaa ccc cag gag atc aat ttt ccc gac gat ctt cct 776 His Pro Lys Gln Glu Pro Gln Glu Ile Asn Phe Pro Asp Asp Leu Pro ggc tcc aac act get get cca gtg cag gag act tta cat gga tgc caa 824 Gly Ser Asn Thr Ala Ala Pro Val Gln Glu Thr Leu His Gly Cys Gln ccg gtc acc cag gag gat ggc aaa gag agt cgc atc tca gtg cag gag 872 Pro Val Thr Gln Glu Asp Gly Lys Glu Ser Arg Ile Ser Val Gln Glu aga cag tga ggctgcaccc acccaggagt gtggccacgt gggcaaacag 921 Arg Gln gcagttggcc agagagcctg gtgctgctgctgcaggggtg caggcagaag cggggagcta 981 tgcccagtca gtgccagccc ctc 1004 <210>86 <211>23 <212>DNA

<213>Artificial Sequence <223> PCR Primer <400> 86 cagagttcactgaaacggaatgc 23 <210> 87 <211> 23 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 87 ggtggcagtg tgtctctctg ttc 23 <210>88 <211>25 <212>DNA

<213>Artificial Sequence <223> PCR Probe <400> 88 ttccttgcgg tgaaagcgaa ttcct 25 <210> 89 <211> 19 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 89 gaaggtgaag gtcggagtc 19 <210> 90 <211> 20 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 90 gaagatggtg atgggatttc 20 <210> 91 <211> 20 <212> DNA
<213> Artificial Sequence <223> PCR Probe <400> 91 caagcttccc gttctcagcc 20 <210> 92 <211> 20 <212> DNA
<213> Artificial Sequence <223> Assembled Target Region <400> 92 agtggtcctg ccgcctggtc 20 <210> 93 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 93 gaacagcact gactgttt 18 <210> 94 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 94 agaacagcac tgactgtt 18 <210>95 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 95 aagaacagca ctgactgt 18 <210> 96 <211>18 <212>DNA

<213>Artificial Sequence <223>Antisense Oligonucleotide <400> 96 aaagaacagc actgactg 18 <210> 97 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 97 caaagaacag cactgact 18 <210> 98 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 98 acaaagaaca gcactgac 18 <210> 99 <400> 99 <210>100 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 100 gcacaaagaa cagcactg 18 <210>101 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 101 ggcacaaaga acagcact 18 <210> 102 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 102 tggcacaaag aacagcac 18 <210> 103 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 103 gctggcacaa agaacagc 18 <210>104 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 104 ggctggcaca aagaacag 18 <210>105 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 105 tggctggcac aaagaaca 18 <210>106 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 106 ctggctggca caaagaac 18 <210>107 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 107 cctggctggc acaaagaa 18 <210> 108 <400> 108 <210>109 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 109 gtcctggctg gcacaaag 18 <210>110 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 110 tgtcctggct ggcacaaa 18 <210> 111 <400> 111 <210>112 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 112 tctgtcctgg ctggcaca 18 <210>113 <211>1058 <212>DNA

<213>Homo Sapiens <220>
<221 > CDS
<222> (77)..(658) <400> 113 gccttgactt catctcagct ccagagcccg ccctctcttc ctgcagcctg ggaacttcag 60 ccggctggag cccacc atg get gca atc cga aag aag ctg gtg atc gtt 109 Met Ala Ala Ile Arg Lys Lys Leu Val Ile Val ggg gat ggt gcc tgt ggg aag acc tgc ctc ctc atc gtc ttc agc aag 157 Gly Asp Gly Ala Cys Gly Lys Thr Cys Leu Leu Ile Val Phe Ser Lys gat cag ttt ccg gag gtc tac gtc cct act gtc ttt gag aac tat att 205 Asp Gln Phe Pro Glu Val Tyr Val Pro Thr Val Phe Glu Asn Tyr Ile gcg gac att gag gtg gac ggc aag cag gtg gag ctg get ctg tgg gac 253 Ala Asp Ile Glu Val Asp Gly Lys Gln Val Glu Leu Ala Leu Trp Asp aca gca ggg cag gaa gac tat gat cga ctg cgg cct ctc tcc tac ccg 301 Thr Ala Gly Gln Glu Asp Tyr Asp Arg Leu Arg Pro Leu Ser Tyr Pro gac act gat gtc atc ctc atg tgc ttc tcc atc gac agc cct gac agc 349 Asp Thr Asp Val Ile Leu Met Cys Phe Ser Ile Asp Ser Pro Asp Ser ctg gaa aac att cct gag aag tgg acc cca gag gtg aag cac ttc tgc 397 Leu Glu Asn Ile Pro Glu Lys Trp Thr Pro Glu Val Lys His Phe Cys ccc aac gtg ccc atc atc ctg gtg ggg aat aag aag gac ctg agg caa 445 Pro Asn Val Pro Ile Ile Leu Val Gly Asn Lys Lys Asp Leu Arg Gln gac gag cac acc agg aga gag ctg gcc aag atg aag cag gag ccc gtt 493 Asp Glu His Thr Arg Arg Glu Leu Ala Lys Met Lys Gln Glu Pro Val cgg tct gag gaa ggc cgg gac atg gcg aac cgg atc agt gcc ttt ggc 541 Arg Ser Glu Glu Gly Arg Asp Met Ala Asn Arg Ile Ser Ala Phe Gly tac ctt gag tgc tca gcc aag acc aag gag gga gtg cgg gag gtg ttt 589 Tyr Leu Glu Cys Ser Ala Lys Thr Lys Glu Gly Val Arg Glu Val Phe gag atg gcc act cgg get ggc ctc cag gtc cgc aag aac aag cgt cgg 637 Glu Met Ala Thr Arg Ala Gly Leu Gln Val Arg Lys Asn Lys Arg Arg agg ggc tgt ccc att ctc tga gatcccccca aagggccctt ttcctacatg 688 Arg Gly Cys Pro Ile Leu ccccctccct tcacaggggt acagaaatta tccccctaca accccagcct cctgagggct 748 ccatactgaa ggctccattt tcagttccct cctgcccagg actgcattgt tttctagccc 808 cgaggtgtgg cacgggccct ccctcccagc gctctgggag ccacgcctat gccctgccct 868 tcctcatggg cccctgggga tcttgcccct ttgaccttcc ccaaaggatg gtcacacacc 928 agcactttat acacttctgg ctcacaggaa agtgtctgca gtagggaccc agagtcccag 988 gcccctggag ttgtttctgc aggggccttg tctctcactg catttggtca ggggggcatg 1048 aataaaggct 1 OS 8 <210> 114 <211> 23 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 114 tgatgtcatc ctcatgtgct tct 23 <210> 115 <211> 19 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 115 ccaggatgat gggcacgtt 19 <210>116 <211>23 <212>DNA

<213>Artificial Sequence <223> PCR Probe <400> 116 cgacagccctgacagcctgg aaa 23 <210> 117 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 117 gagctgagat gaagtcaa 18 <210>118 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 118 gctgaagttc ccaggctg 18 <210>119 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 119 ccggctgaag ttcccagg 18 <210>120 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 120 ggcaccatcc ccaacgat 18 <210>121 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 121 aggcaccatc cccaacga 18 <210>122 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 122 tcccacaggc accatccc 18 <210>123 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 123 aggtcttccc acaggcac 18 <210>124 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 124 atgaggaggc aggtcttc 18 <210>125 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 125 ttgctgaaga cgatgagg 18 <210>126 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 126 tcaaagacag tagggacg 18 <210>127 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 127 ttctcaaaga cagtaggg 18 <210>128 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 128 agttctcaaa gacagtag 18 <210> 129 <211>18 <212>DNA

<213>Artificial Sequence <223>Antisense Oligonucleotide <400> 129 tgttttccag gctgtcag 18 <210>130 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 130 tcgtcttgcc tcaggtcc 18 <210>131 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 131 gtgtgctcgt cttgcctc 18 <210>132 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 132 ctcctggtgt gctcgtct 18 <210> 133 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 133 cagaccgaac gggctcct 18 <210>134 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 134 ttcctcagac cgaacggg 18 <210> 135 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 135 actcaaggta gccaaagg 18 <210>136 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oiigonucleotide <400> 136 ctcccgcact ccctcctt 18 <210>137 <211>18 <212>DNA

<213>Artificial Sequence
36 <223> Antisense Oligonucleotide <400> 137 ctcaaacacc tcccgcac 18 <210> 138 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 138 ggccatctca aacacctc 18 <210> 139 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 139 cttgttcttg cggacctg 18 <210>140 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 140 cccctccgac gcttgttc 18 <210>141 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide
37 <400> 141 gtatggagcc ctcaggag 18 <2I0>142 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 142 gagccttcag tatggagc 18 <210>143 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 143 gaaaatggag ccttcagt 18 <210>144 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 144 ggaactgaaa atggagcc 18 <210> 145 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 145 ggagggaact gaaaatgg 18
38 <210>146 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 146 gcaggaggga actgaaaa 18 <210>147 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 147 agggcagggc ataggcgt 18 <210>148 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 148 ggaagggcag ggcatagg 18 <210>149 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 149 catgaggaag ggcagggc 18 <210> 150
39 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 150 taaagtgctg gtgtgtga 18 <210> 151 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 151 cctgtgagcc agaagtgt 18 <210> 152 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 152 ttcctgtgag ccagaagt 18 <210> 153 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 153 cactttcctg tgagccag 18 <210> 154 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 154 agacactttc ctgtgagc 18 <210>155 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 155 actctgggtc cctactgc 18 <210>156 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> i56 tgcagaaaca actccagg 18 <210> 157 <211> 3076 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (725)..(2539) <400> 157 gaattcaaaa tgtcttcagt tgtaaatctt accattattt tacgtacctc taagaaataa 60 aagtgcttct aattaaaata tgatgtcatt aattatgaaa tacttcttga taacagaagt 120 tttaaaatag ccatcttaga atcagtgaaa tatggtaatg tattattttc ctcctttgag 180 ttaggtcttg tgcttttttt tcctggccac taaatttcac aatttccaaa aagcaaaata 240 aacatattct gaatattttt gctgtgaaac acttgacagc agagctttcc accatgaaaa 300 gaagcttcat gagtcacaca ttacatcttt gggttgattg aatgccactg aaacattcta 360 gtagcctgga gaagttgacc tacctgtgga gatgcctgcc attaaatggc atcctgatgg 420 cttaatacac atcactcttc tgtgaagggt tttaattttc aacacagctt actctgtagc 480 atcatgttta cattgtatgt ataaagatta tacaaaggtg caattgtgta tttcttcctt 540 aaaatgtatc agtataggat ttagaatctc catgttgaaa ctctaaatgc atagaaataa 600 aaataataaa aaatttttca ttttggcttt tcagcctagt attaaaactg ataaaagcaa 660 agccatgcac aaaactacct ccctagagaa aggctagtcc cttttcttcc ccattcattt 720 catt atg aac ata gta gaa aac agc ata ttc tta tca aat ttg atg 766 Met Asn Ile Val Glu Asn Ser Ile Phe Leu Ser Asn Leu Met aaa agc gcc aac acg ttt gaa ctg aaa tac gac ttg tca tgt gaa ctg 814 Lys Ser Ala Asn Thr Phe Glu Leu Lys Tyr Asp Leu Ser Cys Glu Leu tac cga atg tct acg tat tcc act ttt cct get ggg gtt cct gtc tca 862 Tyr Arg Met Ser Thr Tyr Ser Thr Phe Pro Ala Gly Val Pro Val Ser gaa agg agt ctt get cgt get ggt ttc tat tac act ggt gtg aat gac 91 U
Glu Arg Ser Leu Ala Arg Ala Gly Phe Tyr Tyr Thr Gly Val Asn Asp aag gtc aaa tgc ttc tgt tgt ggc ctg atg ctg gat aac tgg aaa aga 958 Lys Val Lys Cys Phe Cys Cys Gly Leu Met Leu Asp Asn Trp Lys Arg gga gac agt cct act gaa aag cat aaa aag ttg tat cct agc tgc aga 1006 Gly Asp Ser Pro Thr Glu Lys His Lys Lys Leu Tyr Pro Ser Cys Arg ttc gtt cag agt cta aat tcc gtt aac aac ttg gaa get acc tct cag 1054 Phe Val Gln Ser Leu Asn Ser Val Asn Asn Leu Glu Ala Thr Ser Gln cct act ttt cct tct tca gta aca aat tcc aca cac tca tta ctt ccg 1102 Pro Thr Phe Pro Ser Ser Val Thr Asn Ser Thr His Ser Leu Leu Pro ggt aca gaa aac agt gga tat ttc cgt ggc tct tat tca aac tct cca 1150 Gly Thr Glu Asn Ser Gly Tyr Phe Arg Gly Ser Tyr Ser Asn Ser Pro tca aat cct gta aac tcc aga gca aat caa gat ttt tct gcc ttg atg 1198 Ser Asn Pro Val Asn Ser Arg Ala Asn Gln Asp Phe Ser Ala Leu Met aga agt tcc tac cac tgt gca atg aat aac gaa aat gcc aga tta ctt 1246 Arg Ser Ser Tyr His Cys Ala Met Asn Asn Glu Asn Ala Arg Leu Leu act ttt cag aca tgg cca ttg act ttt ctg tcg cca aca gat ctg gca 1294 Thr Phe Gln Thr Trp Pro Leu Thr Phe Leu Ser Pro Thr Asp Leu Ala aaa gca ggc ttt tac tac ata gga cct gga gac aga gtg get tgc ttt 1342 Lys Ala Gly Phe Tyr Tyr Ile Gly Pro Gly Asp Arg Val Ala Cys Phe gcc tgt ggt gga aaa ttg agc aat tgg gaa ccg aag gat aat get atg 1390 Ala Cys Gly Gly Lys Leu Ser Asn Trp Glu Pro Lys Asp Asn Ala Met tca gaa cac ctg aga cat ttt ccc aaa tgc cca ttt ata gaa aat cag 1438 Ser Glu His Leu Arg His Phe Pro Lys Cys Pro Phe Ile Glu Asn Gln ctt caa gac act tca aga tac aca gtt tct aat ctg agc atg cag aca 1486 Leu Gln Asp Thr Ser Arg Tyr Thr Val Ser Asn Leu Ser Met Gln Thr cat gca gcc cgc ttt aaa aca ttc ttt aac tgg ccc tct agt gtt cta 1534 His Ala Ala Arg Phe Lys Thr Phe Phe Asn Trp Pro Ser Ser Val Leu gtt aat cct gag cag ctt gca agt gcg ggt ttt tat tat gtg ggt aac 1582 Val Asn Pro Glu Gln Leu Ala Ser Ala Gly Phe Tyr Tyr Val Gly Asn agt gat gat gtc aaa tgc ttt tgc tgt gat ggt gga ctc agg tgt tgg 1630 Ser Asp Asp Val Lys Cys Phe Cys Cys Asp Gly Gly Leu Arg Cys Trp gaa tct gga gat gat cca tgg gtt caa cat gcc aag tgg ttt cca agg 1678 Glu Ser Gly Asp Asp Pro Trp Val Gln His Ala Lys Trp Phe Pro Arg tgt gag tac ttg ata aga att aaa gga cag gag ttc atc cgt caa gtt 1726 Cys Glu Tyr Leu Ile Arg Ile Lys Gly Gln Glu Phe Ile Arg Gln Val caa gcc agt tac cct cat cta ctt gaa cag ctg cta tcc aca tca gac 1774 Gln Ala Ser Tyr Pro His Leu Leu Glu Gln Leu Leu Ser Thr Ser Asp agc cca gga gat gaa aat gca gag tca tca att atc cat ttt gaa cct 1822 Ser Pro Gly Asp Glu Asn Ala Glu Ser Ser Ile Ile His Phe Glu Pro gga gaa gac cat tca gaa gat gca atc atg atg aat act cct gtg att 1870 Gly Glu Asp His Ser Glu Asp Ala Ile Met Met Asn Thr Pro Val Ile aat get gcc gtg gaa atg ggc ttt agt aga agc ctg gta aaa cag aca 1918 Asn Ala Ala Val Glu Met Gly Phe Ser Arg Ser Leu Val Lys Gln Thr gtt caa aga aaa atc cta gca act gga gag aat tat aga cta gtc aat 1966 Val Gln Arg Lys Ile Leu Ala Thr Gly Glu Asn Tyr Arg Leu Val Asn gat ctt gtg tta gac tta ctc aat gca gaa gat gaa ata agg gaa gag 2014 Asp Leu Val Leu Asp Leu Leu Asn Ala Glu Asp Glu Ile Arg Glu Glu gag aga gaa aga gca act gag gaa aaa gaa tca aat gat tta tta tta 2062 Glu Arg Glu Arg Ala Thr Glu Glu Lys Glu Ser Asn Asp Leu Leu Leu atc cgg aag aat aga atg gca ctt ttt caa cat ttg act tgt gta att 2110 Ile Arg Lys Asn Arg Met Ala Leu Phe Gln His Leu Thr Cys Val Ile cca atc ctg gat agt cta cta act gcc gga att att aat gaa caa gaa 2158 Pro Ile Leu Asp Ser Leu Leu Thr Ala Gly Ile Ile Asn Glu Gln Glu WO 99!53101 PCT/US99/08268 cat gat gtt att aaa cag aag aca cag acg tct tta caa gca aga gaa 2206 His Asp Val Ile Lys Gln Lys Thr Gln Thr Ser Leu Gln Ala Arg Glu ctg att gat acg att tta gta aaa gga aat att gca gcc act gta ttc 2254 Leu Ile Asp Thr Ile Leu Val Lys Gly Asn Ile Ala Ala Thr Val Phe aga aac tct ctg caa gaa get gaa get gtg tta tat gag cat tta ttt 2302 Arg Asn Ser Leu Gln Glu Ala Glu Ala Val Leu Tyr Glu His Leu Phe gtg caa cag gac ata aaa tat att ccc aca gaa gat gtt tca gat cta 2350 Val Gln Gln Asp Ile Lys Tyr Ile Pro Thr Glu Asp Val Ser Asp Leu cca gtg gaa gaa caa ttg cgg aga cta caa gaa gaa aga aca tgt aaa 2398 Pro Val Glu Glu Gln Leu Arg Arg Leu Gln Glu Glu Arg Thr Cys Lys gtg tgt atg gac aaa gaa gtg tcc ata gtg ttt att cct tgt ggt cat 2446 Val Cys Met Asp Lys Glu Val Ser Ile Val Phe Ile Pro Cys Gly His cta gta gta tgc aaa gat tgt get cct tct tta aga aag tgt cct att 2494 Leu Val Val Cys Lys Asp Cys Ala Pro Ser Leu Arg Lys Cys Pro Ile tgt agg agt aca atc aag ggt aca gtt cgt aca ttt ctt tca tga 2539 Cys Arg Ser Thr Ile Lys Gly Thr Val Arg Thr Phe Leu Ser agaagaacca aaacatcatc taaactttag aattaattta ttaaatgtat tataacttta 2599 acttttatcc taatttggtt tccttaaaat ttttatttat ttacaactca aaaaacattg 2659 ttttgtgtaa catatttata tatgtatcta aaccatatga acatatattt tttagaaact 2719 aagagaatga taggcttttg ttcttatgaa cgaaaaagag gtagcactac aaacacaata 2779 ttcaatcaaa atttcagcat tattgaaatt gtaagtgaag taaa,acttaa gatatttgag 2839 ttaaccttta agaattttaa atattttggc attgtactaa tacctggttt tttttttgtt 2899 ttgttttttt gtacagacag ggcagcatac tgagaccctg cctttaaaaa caaacagaac 2959 aaaaacaaaa caccagggac acatttctct gtcttttttg atcagtgtcc tatacatcga 3019 aggtgtgcat atatgttgaa tgacatttta gggacatggt gtttttataa agaattc 3076 <210> 158 <211> 22 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 158 ggactcaggt gttgggaatc tg 22 <210> 159 <211> 24 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 159 caagtactca caccttggaa acca 24 <210> 160 <211> 27 <212> DNA
<213> Artificial Sequence <223> PCR Probe <400> 160 agatgatcca tgggttcaac atgccaa 27 <210>161 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 161 actgaagaca ttttgaat 18 <210> 162 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 162 cttagaggta cgtaaaat 18 <210> 163 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 163 gcacttttat ttcttaga 18 <210> 164 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 164 attttaatta gaagcact 18 <210> 165 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 165 accatatttc actgattc 18 <210>166 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 166 ctaactcaaa ggaggaaa 18 <210>167 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 167 cacaagacct aactcaaa 18 <210>168 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 168 gctctgctgt caagtgtt 18 <210>169 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 169 tgtgtgactc atgaagct 18 <210> 170 <211>18 <212>DNA

<213>Artificial Sequence <223>Antisense Oligonucleotide <400> 170 ttcagtggca ttcaatca 18 <210>171 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 171 cttctccagg ctactaga 18 <210>172 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 172 ggtcaacttc tccaggct 18 <210>173 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 173 taaaaccctt cacagaag 18 <210> 174 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 174 ttaaggaaga aatacaca 18 <210>175 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 175 gcatggcttt gcttttat 18 <210> 176 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 176 caaacgtgtt ggcgcttt 18 <210>177 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 177 agcaggaaaa gtggaata 18 <210>178 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 178 ttaacggaat ttagactc 18 <210> 179 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 179 atttgttact gaagaagg 18 <210>180 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 180 agagccacgg aaatatcc 18 <210>181 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 181 aaatcttgat ttgctctg 18 <210> 182 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 182 gtaagtaatc tggcattt 18 <210> 183 <211> 18 <212> DNA
<2I3> Artificial Sequence <223> Antisense Oligonucleotide <400> 183 agcaagccac tctgtctc 18 <210> 184 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 184 tgaagtgtct tgaagctg 18 <210> 185 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 185 tttgacatca tcactgtt 18 <210> 186 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 186 tggcttgaac ttgacgga 18 <210> 187 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 187 tcatctcctg ggctgtct 18 <210>188 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 188 gcagcattaa tcacagga 18 <210>189 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 189 tttctctctc ctcttccc 18 <210>190 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 190 aacatcatgt tcttgttc 18 <210> 191 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 191 atataacaca gcttcagc 18 <210>192 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 192 aattgttctt ccactggt 18 <210>193 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 193 aagaaggagc acaatctt 18 <210> 194 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 194 gaaaccaaat taggataa 18 <210> 195 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 195 tgtagtgcta cctctttt 18 <210>196 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 196 ctgaaatttt gattgaat 18 <210> 197 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 197 tacaatttca ataatgct 18 <210>198 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 198 gggtctcagt atgctgcc 18 <210>199 <211>18 <212>DNA

<213>Artificial Sequence WO 99/53101 PCT/US99/082b8 <223> Antisense Oligonucleotide <400> 199 ccttcgatgt ataggaca 18 <210> 200 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 200 catgtcccta aaatgtca 18 <210>201 <211>2266 <212>DNA

<213>Homo sapiens <220>

<221 CDS
>

<222>(316)..(1602) <400> 201 aattccgagc tgtagggaaa cgcaggggcg gcttctaggt gctgccgccg ccaccgccac 60 caccacctcc accgccgcct cggaacccag gcctgggggg cggtggggcc gcgtatggag 120 cccccgcccc ccggagctgc caacattgcc aacgccaccg ccacgctaca cacagcctca 180 actttcagga gacccgtccg tggccttatt tattccaccc ttcctgtaca tcgtagcgaa 240 tcaatccgtg gcgccgcact cctccgcatc cctctttaac agtacccctg ggatggcgtg 300 agcactcccc cagcg atg gac cca tct gtg acg ctg tgg cag ttt ctg 348 Met Asp Pro Ser Val Thr Leu Trp Gln Phe Leu ctg cag ctg ctg aga gag caa ggc aat ggc cac atc atc tcc tgg act 396 Leu Gln Leu Leu Arg Glu Gln Gly Asn Gly His Ile Ile Ser Trp Thr is zo 2s tca cgg gat ggt ggt gaa ttc aag ctg gtg gat gca gag gag gtg gcc 444 Ser Arg Asp Gly Gly Glu Phe Lys Leu Val Asp Ala Glu Glu Val Ala cgg ctg tgg gga cta cgc aag aac aag acc aac atg aat tac gac aag 492 Arg Leu Trp Gly Leu Arg Lys Asn Lys Thr Asn Met Asn Tyr Asp Lys 45 s0 ss ctc agc cgg gcc ttg cgg tac tac tat gac aag aac atc atc cgc aag 540 Leu Ser Arg Ala Leu Arg Tyr Tyr Tyr Asp Lys Asn Ile Ile Arg Lys 60 6s 70 7s gtg agc ggc cag aag ttc gtc tac aag ttt gtg tcc tac cct gag gtc s88 Val Ser Gly Gln Lys Phe Val Tyr Lys Phe Val Ser Tyr Pro Glu Val gca ggg tgc tcc act gag gac tgc ccg ccc cag cca gag gtg tct gtt 636 Ala Gly Cys Ser Thr Glu Asp Cys Pro Pro Gln Pro Glu Val Ser Val acc tcc acc atg cca aat gtg gcc cct get get ata cat gcc gcc cca 684 Thr Ser Thr Met Pro Asn Val Ala Pro Ala Ala Ile His Ala Ala Pro 110 lls 120 ggg gac act gtc tct gga aag cca ggc aca ccc aag ggt gca gga atg 732 Gly Asp Thr Val Ser Gly Lys Pro Gly Thr Pro Lys Gly Ala Gly Met gca ggc cca ggc ggt ttg gca cgc agc agc cgg aac gag tac atg cgc 780 Ala Gly Pro Gly Gly Leu Ala Arg Ser Ser Arg Asn Glu Tyr Met Arg 140 14s 150 1s5 tcg ggc ctc tat tcc acc ttc acc atc cag tct ctg cag ccg cag cca 828 Ser Gly Leu Tyr Ser Thr Phe Thr Ile Gln Ser Leu Gln Pro Gln Pro 160 16s 170 ccc cct cat cct cgg cct get gtg gtg ctc ccc aat gca get cct gca 876 Pro Pro His Pro Arg Pro Ala Val Val Leu Pro Asn Ala Ala Pro Ala ggg gca gca gcg ccc ccc tcg ggg agc agg agc acc agt cca agc ccc 924 Gly Ala Ala Ala Pro Pro Ser Gly Ser Arg Ser Thr Ser Pro Ser Pro ttg gag gcc tgt ctg gag get gaa gag gcc ggc ttg cct ctg cag gtc 972 Leu Glu Ala Cys Leu Glu Ala Glu Glu Ala Gly Leu Pro Leu Gln Val atc ctg acc ccg ccc gag gcc cca aac ctg aaa tcg gaa gag ctt aat 1020 Ile Leu Thr Pro Pro Glu Ala Pro Asn Leu Lys Ser Glu Glu Leu Asn gtg gag ccg ggt ttg ggc cgg get ttg ccc cca gaa gtg aaa gta gaa 1068 Val Glu Pro Gly Leu Gly Arg Ala Leu Pro Pro Glu Val Lys Val Glu ggg ccc aag gaa gag ttg gaa gtt gcg ggg gag aga ggg ttt gtg cca 1116 Gly Pro Lys Glu Glu Leu Glu Val Ala Gly Glu Arg Gly Phe Val Pro gaa acc acc aag gcc gag cca gaa gtc cct cca cag gag ggc gtg cca 1164 Glu Thr Thr Lys Ala Glu Pro Glu Val Pro Pro Gln Glu Gly Val Pro gcc cgg ctg ccc gcg gtt gtt atg gac acc gca ggg cag gcg ggc ggc 1212 Ala Arg Leu Pro Ala Val Val Met Asp Thr Ala Gly Gln Ala Gly Gly cat gcg get tcc agc cct gag atc tcc cag ccg cag aag ggc cgg aag 1260 His Ala Ala Ser Ser Pro Glu Ile Ser Gln Pro Gln Lys Gly Arg Lys ccc cgg gac cta gag ctt cca ctc agc ccg agc ctg cta ggt ggg ccg 1308 Pro Arg Asp Leu Glu Leu Pro Leu Ser Pro Ser Leu Leu Gly Gly Pro gga ccc gaa cgg acc cca gga tcg gga agt ggc tcc ggc ctc cag get 1356 Gly Pro Glu Arg Thr Pro Gly Ser Gly Ser Gly Ser Gly Leu Gln Ala ccg ggg ccg gcg ctg acc cca tcc ctg ctt cct acg cat aca ttg acc 1404 Pro Gly Pro Ala Leu Thr Pro Ser Leu Leu Pro Thr His Thr Leu Thr ccg gtg ctg ctg aca ccc agc tcg ctg cct cct agc att cac ttc tgg 1452 Pro Val Leu Leu Thr Pro Ser Ser Leu Pro Pro Ser Ile His Phe Trp agc acc ctg agt ccc att gcg ccc cgt agc ccg gcc aag ctc tcc ttc 1500 Ser Thr Leu Ser Pro Ile Ala Pro Arg Ser Pro Ala Lys Leu Ser Phe cag ttt cca tcc agt ggc agc gcc cag gtg cac atc cct tct atc agc 1548 Gln Phe Pro Ser Ser Gly Ser Ala Gln Val His Ile Pro Ser Ile Ser gtg gat ggc ctc tcg acc ccc gtg gtg ctc tcc cca ggg ccc cag aag 1596 Val Asp Gly Leu Ser Thr Pro Val Val Leu Ser Pro Gly Pro Gln Lys cca tga ctactaccac caccaccacc accccttctg gggtcactcc atccatgctc 1652 Pro tctccagcca gccatctcaa ggagaaacat agttcaactg aaagactcat gctctgattg 1712 tggtggggtg gggatccttg ggaagaatta ctcccaagag taactctcat tatctcctcc 1772 acagaaaaca cacagcttcc acaacttctc tgttttctgt cagtccccca gtggccgccc 1832 ttacacgtct cctacttcaa tggtaggggc ggtttattta tttatttttt gaaggccact 1892 gggatgagcc tgacctaacc ttttagggtg gttaggacat ctcccccacc tccccacttt 1952 tttccccaag acaagacaat cgaggtctgg cttgagaacg acctttcttt ctttatttct 2012 cagcctgccc ttggggagat gagggagccc tgtctgcgtt tttggatgtg agtagaagag 2072 ttagtttgtt ttgttttatt attcctggcc atactcaggg gtccaggaag aatttgtacc 2132 atttaatggg ttgggagtct tggccaagga agaatcacac ccttggaata gaaatttcca 2192 cctcccccaa cctttctctc agacagctta tcctttttca accaactttt tggccaggga 2252 ggaatgtccc tttt 2266 <210> 202 <211> 18 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 202 gcaaggcaat ggccacat 18 <210> 203 <211> 21 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 203 ctcctctgca tccaccagct t 21 <210> 204 <211> 26 <212> DNA
<213> Artificial Sequence <223> PCR Probe <400> 204 tctcctggac ttcacgggat ggtggt 26 <210> 205 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 205 cccctgcgtt tccctaca 18 <210> 206 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 206 g~gt~ggtg gcggtggc 18 <210> 207 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 207 ggcgttggca atgttggc 18 <210> 208 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 208 aagttgaggc tgtgtgta 18 <210>209 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 209 aggccacgga cgggtctc 18 <210> 210 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 210 gattgattcg ctacgatg 18 <210>211 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 211 gggatgcgga ggagtgcg 18 <210> 212 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 212 agtgctcacg ccatccca 18 <210>213 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 213 aaactgccac agcgtcac 18 <210>214 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 214 gaagtccagg agatgatg 18 <210> 215 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 215 caccaccatc ccgtgaag 18 <210> 216 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 216 tcttgttctt gcgtagtc 18 <210>217 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 217 tgttcttgtc atagtagt 18 <210>218 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 218 tcaccttgcg gatgatgt 18 <210>219 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 219 gagcaccctg cgacctca 18 <210>220 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 220 ggcgggcagt cctcagtg 18 <210> 221 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 221 ggtgaaggtg gaatagag 18 <210>222 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 222 tccgatttca ggtttggg 18 <210> 223 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 223 ttggtggttt ctggcaca 18 <210> 224 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 224 tggagggact tctggctc 18 <210> 225 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 225 gcgtaggaag cagggatg 18 <210>226 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 226 gtgctccaga agtgaatg 18 <210>227 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 227 actggatgga aactggaa 18 <210> 228 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 228 ggccatccac gctgatag 18 <210>229 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 229 ccaccacaat cagagcat 18 <210> 230 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 230 gatccccacc ccaccaca 18 <210>231 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 231 tgttttctgt ggaggaga 18 <210>232 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 232 aaacagagaa gttgtgga 18 <210>233 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 233 gggactgaca gaaaacag 18 <210> 234 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 234 ataaataaat aaaccgcc 18 <210>235 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 235 gttaggtcag gctcatcc 18 <210>236 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 236 gttctcaagc cagacctc 18 <210> 237 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 237 aataaagaaa gaaaggtc 18 <210>238 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 238 agggcaggct gagaaata 18 <210> 239 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 239 cttctactca catccaaa 18 <210> 240 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 240 caaaacaaac taactctt 18 <210> 241 <21i> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 241 ggaataataa aacaaaac 18 <210>242 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 242 ttcttcctgg acccctga 18 <210> 243 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 243 ccaagggtgt gattcttc 18 <210> 244 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 244 tgtctgagag aaaggttg 18 <210> 245 WO 99/53101 PCTlUS99/08268 <211> 1080 <212> DNA
<213> Homo sapiens <220>
<221 > CDS
<222> {1)..(1080) <400> 245 atg act ctg gag tcc atc atg gcg tgt tgc ctg agc gat gag gtg aag 48 Met Thr Leu Glu Ser Ile Met Ala Cys Cys Leu Ser Asp Glu Val Lys gag tcc aag cgg atc aac gcc gag atc gag aag cag ctg cgg cgg gac 96 Glu Ser Lys Arg Ile Asn Ala Glu Ile Glu Lys Gln Leu Arg Arg Asp aag cgc gac gcc cgg cgc gag ctc aag ctg ctg ctg ctc ggc acg ggc 144 Lys Arg Asp Ala Arg Arg Glu Leu Lys Leu Leu Leu Leu Gly Thr Gly gag agc ggg aag agc acg ttc atc aag cag atg cgc atc atc cac ggc 192 Glu Ser Gly Lys Ser Thr Phe Ile Lys Gln Met Arg Ile Ile His Gly gcc ggc tac tcg gag gag gac aag cgc ggc ttc acc aag ctc gtc tac 240 Ala Gly Tyr Ser Glu Glu Asp Lys Arg Gly Phe Thr Lys Leu Val Tyr cag aac atc ttc acc gcc atg cag gcc atg atc cgg gcc atg gag acg 288 Gln Asn Ile Phe Thr Ala Met Gln Ala Met Ile Arg Ala Met Glu Thr ctc aag atc ctc tac aag tac gag cag aac aag gcc aat gcg ctc ctg 336 Leu Lys Ile Leu Tyr Lys Tyr Glu Gln Asn Lys Ala Asn Ala Leu Leu atc cgg gag gtg gac gtg gag aag gtg acc acc ttc gag cat cag tac 384 Ile Arg Glu Val Asp Val Glu Lys Val Thr Thr Phe Glu His Gln Tyr gtc agt gcc atc aag acc ctg tgg gag gac ccg ggc atc cag gaa tgc 432 Val Ser Ala Ile Lys Thr Leu Trp Glu Asp Pro Gly Ile Gln Glu Cys tac gac cgc agg cgc gag tac cag ctc tcc gac tct gcc aag tac tac 480 Tyr Asp Arg Arg Arg Glu Tyr Gln Leu Ser Asp Ser Ala Lys Tyr Tyr ctg acc gac gtt gac cgc atc gcc acc ttg ggc tac ctg ccc acc cag 528 Leu Thr Asp Val Asp Arg Ile Ala Thr Leu Gly Tyr Leu Pro Thr Gln cag gac gtg ctg cgg gtc cgc gtg ccc acc acc ggc atc atc gag tac 576 Gln Asp Val Leu Arg Val Arg Val Pro Thr Thr Gly Ile Ile Glu Tyr cct ttc gac ctg gag aac atc atc ttc cgg atg gtg gat gtg ggg ggc 624 Pro Phe Asp Leu Glu Asn Ile Ile Phe Arg Met Val Asp Val Gly Gly cag cgg tcg gag cgg agg aag tgg atc cac tgc ttt gag aac gtg aca 672 Gln Arg Ser Glu Arg Arg Lys Trp Ile His Cys Phe Glu Asn Val Thr tcc atc atg ttt ctc gtc gcc ctc agc gaa tac gac caa gtc ctg gtg 720 Ser Ile Met Phe Leu Val Ala Leu Ser Glu Tyr Asp Gln Val Leu Val gag tcg gac aac gag aac cgg atg gag gag agc aaa gcc ctg ttc cgg 768 Glu Ser Asp Asn Glu Asn Arg Met Glu Glu Ser Lys Ala Leu Phe Arg acc atc atc acc tac ccc tgg ttc cag aac tcc tcc gtc atc ctc ttc 816 Thr Ile Ile Thr Tyr Pro Trp Phe Gln Asn Ser Ser Val Ile Leu Phe ctc aac aag aag gac ctg ctg gag gac aag atc ctg tac tcg cac ctg 864 Leu Asn Lys Lys Asp Leu Leu Glu Asp Lys Ile Leu Tyr Ser His Leu gtg gac tac ttc ccc gag ttc gat ggt ccc cag cgg gac gcc cag gcg 912 Val Asp Tyr Phe Pro Glu Phe Asp Gly Pro Gln Arg Asp Ala Gln Ala gcg cgg gag ttc atc ccg aag atg ttc gtg gac ctg aac ccc gac agc 960 Ala Arg Glu Phe Ile Pro Lys Met Phe Val Asp Leu Asn Pro Asp Ser gac aag atc atc tac tca cac ttc acg tgt gcc acc gac acg gag aac 1008 Asp Lys Ile Ile Tyr Ser His Phe Thr Cys Ala Thr Asp Thr Glu Asn atc cgc ttc gtg ttc gcg gcc gtg aag gac acc atc ctg cag ctg aac 1056 Ile Arg Phe Val Phe Ala Ala Val Lys Asp Thr Ile Leu Gln Leu Asn ctg aag gag tac aat ctg gtc taa 1080 Leu Lys Glu Tyr Asn Leu Val <210> 246 <211> 20 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 246 tgaccacctt cgagcatcag 20 <210> 247 <211> 20 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 247 cggtcgtagc attcctggat 20 <210> 248 <211> 26 <212> DNA
<213> Artificial Sequence <223> PCR Probe <400> 248 tcagtgccat caagaccctg tgggag 26 <210> 249 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 249 gatggactcc agagtcat 18 <210>250 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 250 gccatgatgg actccaga 18 <210>251 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 251 cacgccatga tggactcc 18 <210>252 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 252 ctcatcgctc aggcaaca 18 <210>253 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 253 cttcacctca tcgctcag 18 <210>254 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 254 gactccttca cctcatcg 18 <210>255 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 255 atccgcttgg actccttc 18 <210>256 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 256 cgttgatccg cttggact 18 <210> 257 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 257 ctcgatctcg gcgttgat 18 <210> 258 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 258 cccgccgcag ctgcttct 18 <210> 259 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 259 cttgagctcg cgccgggc 18 <210> 260 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 260 gcagcagcag cttgagct 18 <210> 261 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 261 gcccgtgccg agcagcag 18 <210> 262 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 262 acgtgctctt cccgctct 18 <210> 263 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 263 atctgcttga tgaacgtg 18 <210> 264 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 264 cgcatctgct tgatgaac 18 <210>265 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 265 gtagccggcg ccgtggat 18 <210> 266 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 266 tgtcctcctc cgagtagc 18 <210> 267 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 267 cttgtcctcc tccgagta 18 <210> 268 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 268 aagccgcgct tgtcctcc 18 <210> 269 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 269 tagacgagct tggtgaag 18 <210> 270 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 270 tgttctggta gacgagct 18 <210>271 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 271 tggcggtgaa gatgttct 18 <210> 272 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 272 cggatcatgg cctgcatg 18 <210>273 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 273 cgtctccatg gcccggat 18 <210>274 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oiigonucleotide <400> 274 tagaggatct tgagcgtc 18 <210>275 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 275 tgtagaggat cttgagcg 18 <210>276 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 276 tgctcgtact tgtagagg 18 <210>277 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 277 gccttgttct gctcgtac 18 <210>278 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 278 ttggccttgt tctgctcg 18 <210>279 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 279 caggagcgca ttggcctt 18 <210> 280 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 280 ctccacgtcc acctcccg 18 <210>281 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 281 ggtcaccttc tccacgtc 18 <210>282 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 282 gatgctcgaa ggtggtca 18 <210>283 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 283 actgacgtac tgatgctc 18 <210>284 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 284 cttgatggca ctgacgta 18 <210> 285 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 285 cagggtcttg atggcact 18 <210> 286 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 286 ctggatgccc gggtcctc 18 <210> 287 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 287 tcctggatgc ccgggtcc 18 <210>288 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 288 cgcctgcggt cgtagcat 18 <210>289 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 289 gctggtactc gcgcctgc 18 <210>290 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 290 tacttggcag agtcggag 18 <210> 291 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 291 gtcaggtagt acttggca 18 <210> 292 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 292 ggtcaacgtc ggtcaggt 18 <210>293 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 293 gtggcgatgc ggtcaacg 18 <210>294 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 294 gcaggtagcc caaggtgg 18 <210>295 <211>18 <212>DNA

<213>Artificial Sequence WO 99!53101 PCT/US99/08268 <223> Antisense Oligonucleotide <400> 295 cgtcctgctg ggtgggca 18 <210> 296 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 296 ggtggtgggc acgcggac 18 <210>297 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 297 tcgatgatgc cggtggtg 18 <210>298 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 298 ccaggtcgaa agggtact 18 <210>299 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 299 tgttctccag gtcgaaag 18 <210> 300 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 300 agatgatgtt ctccaggt 18 <210> 301 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 301 atccggaaga tgatgttc 18 <210> 302 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 302 ctccgctccg accgctgg 18 <210> 303 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 303 gatccacttc ctccgctc 18 <210>304 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 304 tgtcacgttc tcaaagca 18 <210>305 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 305 atgatggatg tcacgttc 18 <210>306 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 306 cgagaaacat gatggatg 18 <210>307 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 307 gctgagggcg acgagaaa 18 <210> 308 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 308 cgactccacc aggacttg 18 <210> 309 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 309 atccggttct cgttgtcc 18 <210>310 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 310 ccatccggtt ctcgttgt 18 <210> 311 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 311 agggctttgc tctcctcc 18 <210> 312 <211> 18 <212> DNA

<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 312 ggtccggaac agggcttt 18 <210> 313 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 313 gtaggtgatg atggtccg 18 <210>314 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 314 ggaggagttc tggaacca 18 <210>315 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 315 tgaggaagag gatgacgg 18 <210>316 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 316 gcaggtcctt cttgttga 18 <210>317 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 317 atcttgtcct ccagcagg 18 <210> 318 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 318 gcgagtacag gatcttgt 18 <210> 319 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 319 aagtagtcca ccaggtgc 18 <210>320 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 320 gatgaactcc cgcgccgc 18 <210> 321 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 321 ggttcaggtc cacgaaca 18 <210>322 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 322 gtagatgatc ttgtcgct 18 <210> 323 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 323 cacgtgaagtgtgagtag 18 <210>324 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 324 atgttctccg tgtcggtg 18 <210> 325 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 325 acggccgcga acacgaag 18 <210> 326 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 326 gatggtgtcc ttcacggc 18 <210> 327 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 327 tcaggttcag ctgcagga 18 <210> 328 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 328 accagattgt actccttc 18 <210> 329 <211> 2610 <212> DNA
<213> Homo sapiens <220>
<221 > CDS
<222> (199)..(1641) <400> 329 atcctgggac agggcacagg gccatctgtc accaggggct tagggaaggc cgagccagcc 60 tgggtcaaag aagtcaaagg ggctgcctgg aggaggcagc ctgtcagctg gtgcatcaga 120 ggctgtggcc aggccagctg ggctcgggga gcgccagcct gagaggagcg cgtgagcgtc I80 gcgggagcct cgggcacc atg agc gac gtg get att gtg aag gag ggt tgg 231 Met Ser Asp Val Ala Ile Val Lys Glu Gly Trp ctg cac aaa cga ggg gag tac atc aag acc tgg cgg cca cgc tac ttc 279 Leu His Lys Arg Gly Glu Tyr Ile Lys Thr Trp Arg Pro Arg Tyr Phe ctc ctc aag aat gat ggc acc ttc att ggc tac aag gag cgg ccg cag 327 Leu Leu Lys Asn Asp Gly Thr Phe Ile Gly Tyr Lys Glu Arg Pro Gln gat gtg gac caa cgt gag get ccc ctc aac aac ttc tct gtg gcg cag 375 Asp Val Asp Gln Arg Glu Ala Pro Leu Asn Asn Phe Ser Val Ala Gln tgc cag ctg atg aag acg gag cgg ccc cgg ccc aac acc ttc atc atc 423 Cys Gln Leu Met Lys Thr Glu Arg Pro Arg Pro Asn Thr Phe Ile Ile cgc tgc ctg cag tgg acc act gtc atc gaa cgc acc ttc cat gtg gag 471 Arg Cys Leu Gln Trp Thr Thr Val Ile Glu Arg Thr Phe His Val Glu act cct gag gag cgg gag gag tgg aca acc gcc atc cag act gtg get 519 Thr Pro Glu Glu Arg Glu Glu Trp Thr Thr Ala Ile Gln Thr Val Ala gac ggc ctc aag aag cag gag gag gag gag atg gac ttc cgg tcg ggc 567 Asp Gly Leu Lys Lys Gln Glu Glu Glu Glu Met Asp Phe Arg Ser Gly WO 99!53101 PCT/US99J08268 tca ccc agt gac aac tca ggg get gaa gag atg gag gtg tcc ctg gcc 615 Ser Pro Ser Asp Asn Ser Gly Ala Glu Glu Met Glu Val Ser Leu Ala aag ccc aag cac cgc gtg acc atg aac gag ttt gag tac ctg aag ctg 663 Lys Pro Lys His Arg Val Thr Met Asn Glu Phe Glu Tyr Leu Lys Leu ctg ggc aag ggc act ttc ggc aag gtg atc ctg gtg aag gag aag gcc 711 Leu Gly Lys Gly Thr Phe Gly Lys Val Ile Leu Val Lys Glu Lys Ala aca ggc cgc tac tac gcc atg aag atc ctc aag aag gaa gtc atc gtg 759 Thr Gly Arg Tyr Tyr Ala Met Lys Ile Leu Lys Lys Glu Val Ile Val gcc aag gac gag gtg gcc cac aca ctc acc gag aac cgc gtc ctg cag 807 Ala Lys Asp Glu Val Ala His Thr Leu Thr Glu Asn Arg Val Leu Gln aac tcc agg cac ccc ttc ctc aca gcc ctg aag tac tct ttc cag acc 855 Asn Ser Arg His Pro Phe Leu Thr Ala Leu Lys Tyr Ser Phe Gln Thr cac gac cgc ctc tgc ttt gtc atg gag tac gcc aac ggg ggc gag ctg 903 His Asp Arg Leu Cys Phe Val Met Glu Tyr Ala Asn Gly Gly Glu Leu ttc ttc cac ctg tcc cgg gaa cgt gtg ttc tcc gag gac cgg gcc cgc 951 Phe Phe His Leu Ser Arg Glu Arg Val Phe Ser Glu Asp Arg Ala Arg ttc tat ggc get gag att gtg tca gcc ctg gac tac ctg cac tcg gag 999 Phe Tyr Gly Ala Glu Ile Val Ser Ala Leu Asp Tyr Leu His Ser Glu aag aac gtg gtg tac cgg gac ctc aag ctg gag aac ctc atg ctg gac 1047 Lys Asn Val Val Tyr Arg Asp Leu Lys Leu Glu Asn Leu Met Leu Asp aag gac ggg cac att aag atc aca gac ttc ggg ctg tgc aag gag ggg 1095 Lys Asp Gly His Ile Lys Ile Thr Asp Phe Gly Leu Cys Lys Glu Gly atc aag gac ggt gcc acc atg aag acc ttt tgc ggc aca cct gag tac 1143 Ile Lys Asp Gly Ala Thr Met Lys Thr Phe Cys Gly Thr Pro Glu Tyr ctg gcc ccc gag gtg ctg gag gac aat gac tac ggc cgt gca gtg gac 1191 Leu Ala Pro Glu Val Leu Glu Asp Asn Asp Tyr Gly Arg Ala Val Asp tgg tgg ggg ctg ggc gtg gtc atg tac gag atg atg tgc ggt cgc ctg 1239 Trp Trp Gly Leu Gly Val Val Met Tyr Glu Met Met Cys Gly Arg Leu ccc ttc tac aac cag gac cat gag aag ctt ttt gag ctc atc ctc atg 1287 Pro Phe Tyr Asn Gln Asp His Glu Lys Leu Phe Glu Leu Ile Leu Met gag gag atc cgc ttc ccg cgc acg ctt ggt ccc gag gcc aag tcc ttg 1335 Glu Glu Ile Arg Phe Pro Arg Thr Leu Gly Pro Glu Ala Lys Ser Leu ctt tca ggg ctg ctc aag aag gac ccc aag cag agg ctt ggc ggg ggc 1383 Leu Ser Gly Leu Leu Lys Lys Asp Pro Lys Gln Arg Leu Gly Gly Gly tcc gag gac gcc aag gag atc atg cag cat cgc ttc ttt gcc ggt atc 1431 Ser Glu Asp Ala Lys Glu Ile Met Gln His Arg Phe Phe Ala Gly Ile gtg tgg cag cac gtg tac gag aag aag ctc agc cca ccc ttc aag ccc 1479 Val Trp Gln His Val Tyr Glu Lys Lys Leu Ser Pro Pro Phe Lys Pro cag gtc acg tcg gag act gac acc agg tat ttt gat gag gag ttc acg 1527 Gln Val Thr Ser Glu Thr Asp Thr Arg Tyr Phe Asp Glu Glu Phe Thr gcc cag atg atc acc atc aca cca cct gac caa gat gac agc atg gag 1575 Ala Gln Met Ile Thr Ile Thr Pro Pro Asp Gln Asp Asp Ser Met Glu tgt gtg gac agc gag cgc agg ccc cac ttc ccc cag ttc tcc tac tcg 1623 Cys Val Asp Ser Glu Arg Arg Pro His Phe Pro Gln Phe Ser Tyr Ser gcc agc agc acg gcc tga ggcggcggtg gactgcgctg gacgatagct 1671 Ala Ser Ser Thr Ala tggagggatg gagaggcggc ctcgtgccat gatctgtatt taatggtttt tatttctcgg 1731 gtgcatttga gagaagccac gctgtcctct cgagcccaga tggaaagacg tttttgtgct 1791 gtgggcagca ccctcccccg cagcggggta gggaagaaaa ctatcctgcg ggttttaatt 1851 tatttcatcc agtttgttct ccgggtgtgg cctcagccct cagaacaatc cgattcacgt 1911 agggaaatgt taaggacttc tacagctatg cgcaatgtgg cattgggggg ccgggcaggt 1971 cctgcccatg tgtcccctca ctctgtcagc cagccgccct gggctgtctg tcaccagcta 2031 tctgtcatct ctctggggcc ctgggcctca gttcaacctg gtggcaccag atgcaacctc 2091 actatggtat gctggccagc accctctcct gggggtggca ggcacacagc agccccccag 2151 cactaaggcc gtgtctctga ggacgtcatc ggaggctggg cccctgggat gggaccaggg 2211 atgggggatg ggccagggtt tacccagtgg gacagaggag caaggtttaa atttgttatt 2271 gtgtattatg ttgttcaaat gcattttggg ggtttttaat ctttgtgaca ggaaagccct 2331 cccccttccc cttctgtgtc acagttcttg gtgactgtcc caccggagcc tccccctcag 2391 atgatctctc cacggtagca cttgaccttt tcgacgctta acctttccgc tgtcgcccca 2451 ggccctccct gactccctgt gggggtggcc atccctgggc ccctccacgc ctcctggcca 2511 gacgctgccg ctgccgctgc accacggcgt ttttttacaa cattcaactt tagtattttt 2571 actattataa tataatatgg aaccttccct ccaaattct 2610 <210> 330 <211> 21 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 330 cgtgaccatg aacgagtttg a 21 <210> 331 <211> 19 <212> DNA
<213> Artificial Sequence <223> PCR Primer <400> 331 caggatcacc ttgccgaaa 19 <210> 332 <211> 22 <212> DNA
<213> Artificial Sequence <223> PCR Probe <400> 332 ctgaagctgc tgggcaaggg ca 22 <210> 333 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 333 ccctgtgccc tgtcccag 18 <210> 334 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 334 cctaagcccc tggtgaca 18 <210> 335 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 335 ctttgacttc tttgaccc 18 <210> 336 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 336 ggcagcccct ttgacttc 18 <210>337 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 337 caaccctcct tcacaata 18 <210> 338 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 338 tactcccctc gtttgtgc 18 <210>339 <211>18 <212>DNA

<213>Artificial Sequence WO 99/53101 PC'T/US99108268 <223> Antisense Oligonucleotide <400> 339 tgccatcatt cttgagga 18 <210>340 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 340 agccaatgaa ggtgccat 18 <210>341 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 341 cacagagaag ttgttgag 18 <210>342 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 342 agtctggatg gcggttgt 18 <210> 343 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 343 tcctcctcct cctgcttc 18 <210>344 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 344 cctgagttgt cactgggt 18 <210> 345 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 345 ccgaaagtgc ccttgccc 18 <210>346 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 346 gccacgatga cttccttc 18 <210> 347 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 347 cggtcctcgg agaacaca 18 WO 99/53101 PCT/US99/0$26$
<210> 348 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 348 acgttcttct ccgagtgc 18 <210>349 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 349 gtgccgcaaa aggtcttc 18 <210>350 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 350 tactcaggtg tgccgcaa 18 <210>351 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 351 ctt as gtgggctg 18 gg g gg <210> 352 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 352 tcaaaatacc tggtgtca 18 <210> 353 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 353 gccgtgaact cctcatca 18 <210> 354 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 354 ggtcaggtgg tgtgatgg 18 <210> 355 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 355 ctcgctgtcc acacactc 18 <210> 356 <211> 18 <212> DNA

WO 99/53101 PC'T/US99/08268 <213> Artificial Sequence <223> Antisense Oligonucleotide <440> 356 gcctctccat ccctccaa 18 <210> 357 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 357 acagcgtggc ttctctca 18 <210>358 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 358 ttttcttccc taccccgc 18 <210> 359 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 359 gatagttttc ttccctac 18 <210>360 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 360 taaaacccgc aggatagt 18 <210>361 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 361 ggagaacaaa ctggatga 18 <210>362 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 362 ctggctgaca gagtgagg 18 <210>363 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 363 gcggctggct gacagagt 18 <210> 364 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 364 cccagagagatgacagat 18 <210> 365 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 365 gctgctgtgt gcctgcca 18 <210>366 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 366 cataatacac aataacaa 18 <210>367 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 367 atttgaacaa cataatac 18 <210>368 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 368 aagtgctacc gtggagag 18 <210>369 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 369 cgaaaaggtc aagtgcta 18 <210> 370 <211> 18 <212> DNA
<213> Artificial Sequence <223> Antisense Oligonucleotide <400> 370 cagggagtca gggagggc 18 <210>371 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 371 aaagttgaat gttgtaaa 18 <210>372 <211>18 <212>DNA

<213>Artificial Sequence <223> Antisense Oligonucleotide <400> 372 aaaatactaa agttgaat 18

Claims (54)

What is claimed is:
1. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising generating a library of virtual compounds in silico according to defined criteria, and evaluating in silico the binding of said virtual compounds with said target nucleic acid according to defined criteria.
2. A method of defining a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence comprising generating in silico a plurality of virtual oligonucleotides according to defined criteria, and evaluating in silico the binding of said plurality of virtual oligonucleotides with said target nucleic acid according to defined criteria.
3. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid comprising, generating in silico a library of virtual compounds according to defined criteria wherein said virtual compounds modulate the expression of said target nucleic acid sequence, and robotically synthesizing synthetic compounds corresponding to at least some of said virtual compounds.
4. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid comprising generating in silico virtual compounds according to defined criteria wherein said virtual compounds modulate the expression of said target nucleic acid sequence, synthesizing synthetic compounds corresponding to at least some of said virtual compounds, and robotically assaying said synthetic compounds for one or more desired physical, chemical or biological properties.
A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising generating in silico a library of nucleobase sequences according to defined criteria and evaluating in silico a plurality of virtual oligonucleotides having said nucleobase sequences according to defined criteria.
6. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising evaluating in silico a plurality of virtual compounds according to defined criteria and robotically synthesizing a plurality of synthetic compounds corresponding to said plurality of virtual compounds.
7. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising evaluating in silico a plurality of virtual compounds according to defined criteria and robotically assaying a plurality of synthetic compounds corresponding to at least some of said virtual compounds for one or more desired physical, chemical or biological properties.
8. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically synthesizing a plurality of synthetic compounds compounds having said nucleobase sequences.
9. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising robotically synthesizing a plurality of synthetic compounds and robotically assaying said plurality of synthetic compounds for one or more desired physical, chemical or biological properties.
10. A method of defining a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically assaying a plurality of synthetic compounds having at least some of said nucleobase sequences for one or more desired physical, chemical or biological properties.
11. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria; and (c) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides.
12. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria; and (c) robotically assaying a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides for one or more desired physical, chemical or biological properties.
13. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucletoides with said target nucleic acid sequence, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) robotically synthesizing a plurality of synthetic oligonucleotides having at least some of said nucleobase sequences; and (c) robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
14. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence, comprising the steps of:
(a) evaluating in silico a plurality of virtual oligonucleotides according to defined criteria;
(b) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides; and (c) robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
15. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria;
(c) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides; and (d) robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
16. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;

(b) choosing an oligonucleotide chemistry;
(c) robotically synthesizing a set of synthetic oligonucleotides having said nucleobase sequences of step (a) and said oligonucleotide chemistry of step (b);
(d) robotically assaying said set of synthetic oligonucleotides of step (c) for a physical, chemical or biological activity; and (e) selecting a subset of said set of synthetic oligonucleotides of step (c) having a desired level of physical, chemical or biological activity in order to generate said set of compounds.
17. A method of generating a set of oligonucleotides that modulate the expression of a target nucleic acid sequence via binding of said oligonucleotides with said target nucleic acid sequence, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) choosing an oligonucleotide chemistry;
(c) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) and the oligonucleotide chemistry of (b) according to defined criteria, and selecting those having preferred characteristics, in order to generate a set of preferred nucleobase sequences;
(d) robotically synthesizing a set of synthetic oligonucleotides having said preferred nucleobase sequences of step (c) and said oligonucleotide chemistry of step (b);
(e) robotically assaying said set of synthetic oligonucleotides of step (d) for a physical, chemical or biological activity; and (f) selecting a subset of said set of synthetic oligonucleotides of step (d) having a desired level of physical, chemical or biological activity in order to generate said set of oligonucleotides.
18. The method of claim 12, wherein said step of robotically assaying said plurality of synthetic oligonucleotide compounds is performed by computer-controlled real-time polymerase chain reaction or by computer-controlled enzyme-linked immunosorbent assay.
19. The method of claim 11, wherein said target nucleic acid sequence is that of a genomic DNA, a cDNA, a product of a polymerase chain reaction, an expressed sequence tag, an mRNA or a structural RNA.
20. The method of claim 11, wherein said target nucleic acid sequence is a human nucleic acid.
21. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising generating a library of antisense nucleobase sequences in silico according to defined criteria.
22. A method of identifying a set of compounds that modulate the expression of a target nucleic acid sequence via binding of said compounds with said target nucleic acid sequence comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria.
23. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an compound to said nucleic acid sequences comprising robotically synthesizing a plurality of synthetic antisense compounds.
24. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an compound to said nucleic acid sequences comprising robotically assaying a plurality of synthetic antisense compounds for one or more desired physical, chemical or biological properties.
25. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising generating in silico a library of nucleobase sequences according to defined criteria and evaluating in silico a plurality of virtual oligonucleotides having said nucleobase sequences according to defined criteria.
26. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria and robotically synthesizing a plurality of synthetic oligonucleotides corresponding to least some of said virtual oligonucleotides.
27. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria and robotically assaying a plurality of synthetic oligonucleotides corresponding to least some of said virtual oligonucleotides for one or more desired physical, chemical or biological properties.
28. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically synthesizing a plurality of synthetic oligonucleotides having said nucleobase sequences.
29. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising robotically synthesizing a plurality of synthetic oligonucleotides and robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
30. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically assaying a plurality of synthetic oligonucleotides having said nucleobase sequences for one or more desired physical, chemical or biological properties.
31. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria; and (c) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides.
32. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria; and (c) robotically assaying a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides for one or more desired physical, chemical or biological properties.
33. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) robotically synthesizing a plurality of synthetic oligonucleotides having at least some of said nucleobase sequences; and (c) robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
34. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences, comprising the steps of:
(a) evaluating in silico a plurality of virtual oligonucleotides according to defined criteria;

(b) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides; and (c) robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
35. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria;
(c) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to least some of said plurality of virtual oligonucleotides; and (d) robotically assaying said plurality of synthetic oligonucleotides for one or more desired physical, chemical or biological properties.
36. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) choosing an oligonucleotide chemistry;
(c) robotically synthesizing a set of synthetic oligonucleotides having said nucleobase sequences of step (a) and said oligonucleotide chemistry of step (b);
(d) robotically assaying said set of synthetic oligonucleotides of step (c) for a physical, chemical or biological activity; and (e) selecting a subset of said set of synthetic oligonucleotides of step (c) having a desired level of physical, chemical or biological activity.
37. A method of identifying one or more nucleic acid sequences amenable to antisense binding of an oligonucleotide to said nucleic acid sequences, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) choosing an oligonucleotide chemistry;
(c) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria, and selecting those having preferred characteristics, in order to generate a set of preferred nucleobase sequences;
(d) robotically synthesizing a set of synthetic oligonucleotides having said preferred nucleobase sequences of step (b) and said oligonucleotide chemistry of step (c);
(e) robotically assaying said set of synthetic oligonucleotides of step (d) for a physical, chemical or biological activity; and (f) selecting a subset of said set of oligonucleotides of step (d) having a desired level of physical, chemical or biological activity.
38. The method of claim 32, wherein said step of robotically assaying said plurality of synthetic antisense oligonucleotides is performed by computer-controlled real-time polymerase chain reaction or by computer-controlled enzyme-linked immunosorbent assay.
39. The method of claim 31, wherein said nucleic acid sequence is that of a genomic DNA, a cDNA, a product of a polymerase chain reaction, an expressed sequence tag, an mRNA or a structural RNA.
40. The method of claim 31, wherein said nucleic acid sequence is a human nucleic acid.
41. A computer formatted medium comprising computer readable instructions for identifying compounds that have one or more desired properties according to defined criteria and that bind to a genomic DNA, a cDNA, a product of a polymerase chain reaction, an expressed sequence tag, an mRNA or a structural RNA.
42. A computer formatted medium comprising computer readable instructions for performing the method of any one of claims 1 to 20.
43. A computer formatted medium comprising computer readable instructions for performing a method of identifying one or more nucleic acid sequences amenable to antisense binding of a compound to said nucleic acid sequences.
44. A computer formatted medium comprising computer readable instructions for performing the method of any one of claims 21 to 40.
45. A computer formatted medium comprising one or more nucleic acid sequences amenable to antisense binding of a compound to said nucleic acid sequences in computer readable form.
46. A computer formatted medium comprising one or more nucleic acid sequences amenable to antisense binding of a compound to said nucleic acid sequences in computer readable form, wherein said one or more nucleic acid sequences is identified according to the method of any one of claims 21-40.
47. A process for validating the function of a gene or the product of said gene comprising generating in silico a library of nucleobase sequences targeted to said gene and robotically assaying a plurality of synthetic compounds having at least some of said nucleobase sequences for effects on biological function.
48. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria; and (c) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides.
49. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria; and (c) robotically assaying a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides for effects on biological function.
50. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) robotically synthesizing a plurality of synthetic oligonucleotides having at least some of said nucleobase sequences; and (c) robotically assaying said plurality of synthetic oligonucleotides for effects on biological function.
51. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) evaluating in silico a plurality of virtual oligonucleotides according to defined criteria;
(b) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides; and (c) robotically assaying said plurality of synthetic oligonucleotides for effects on biological function.
52. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) according to defined criteria;
(c) robotically synthesizing a plurality of synthetic oligonucleotides corresponding to at least some of said virtual oligonucleotides; and (d) robotically assaying said plurality of synthetic oligonucleotides for effects on biological function.
53. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) choosing an oligonucleotide chemistry;
(c) robotically synthesizing a set of synthetic oligonucleotides having said nucleobase sequences of step (a) and said oligonucleotide chemistry of step (b);
(d) robotically assaying said set of synthetic oligonucleotides of step (c) for effects on biological function; and (e) selecting a subset of said set of synthetic oligonucleotides of step (c) having a desired level of physical, chemical or biological activity in order to generate said set of compounds.
54. A process for validating the function of a gene or the product of said gene, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) choosing an oligonucleotide chemistry;
(c) evaluating in silico a plurality of virtual oligonucleotides having the nucleobase sequences of (a) and the oligonucleotide chemistry of (b) according to defined criteria, and selecting those having preferred characteristics, in order to generate a set of preferred nucleobase sequences;
(d) robotically synthesizing a set of synthetic oligonucleotides having said preferred nucleobase sequences of step (c) and said oligonucleotide chemistry of step (b);
(e) robotically assaying said set of synthetic oligonucleotides of step (d) for effects on biological function; and (f) selecting a subset of said set of synthetic oligonucleotides of step (d) having a desired level of physical, chemical or biological activity in order to generate said set of oligonucleotides.
CA002325013A 1998-04-13 1999-04-13 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation Abandoned CA2325013A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US8148398P 1998-04-13 1998-04-13
US09/067,638 1998-04-28
US09/067,638 US7321828B2 (en) 1998-04-13 1998-04-28 System of components for preparing oligonucleotides
US60/081,483 1998-04-28
PCT/US1999/008268 WO1999053101A1 (en) 1998-04-13 1999-04-13 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation

Publications (1)

Publication Number Publication Date
CA2325013A1 true CA2325013A1 (en) 1999-10-21

Family

ID=26748106

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002325013A Abandoned CA2325013A1 (en) 1998-04-13 1999-04-13 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation

Country Status (6)

Country Link
EP (1) EP1071826B1 (en)
JP (1) JP2002511276A (en)
AT (1) ATE339517T1 (en)
AU (1) AU762397B2 (en)
CA (1) CA2325013A1 (en)
DE (1) DE69933205T2 (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7332597B2 (en) 2004-06-28 2008-02-19 University Of Kentucky Research Foundation Primers and probe to identify mycobacterium tuberculosis complex
TWI695067B (en) 2013-08-05 2020-06-01 美商扭轉生物科技有限公司 De novo synthesized gene libraries
US10669304B2 (en) 2015-02-04 2020-06-02 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
US9981239B2 (en) 2015-04-21 2018-05-29 Twist Bioscience Corporation Devices and methods for oligonucleic acid library synthesis
MY192997A (en) * 2015-07-10 2022-09-20 Ionis Pharmaceuticals Inc Modulators of diacyglycerol acyltransferase 2 (dgat2)
KR20180050411A (en) 2015-09-18 2018-05-14 트위스트 바이오사이언스 코포레이션 Oligonucleotide mutant library and its synthesis
CN108698012A (en) 2015-09-22 2018-10-23 特韦斯特生物科学公司 Flexible substrates for nucleic acid synthesis
US10417457B2 (en) 2016-09-21 2019-09-17 Twist Bioscience Corporation Nucleic acid based data storage
US11550939B2 (en) 2017-02-22 2023-01-10 Twist Bioscience Corporation Nucleic acid based data storage using enzymatic bioencryption
KR102628876B1 (en) 2017-06-12 2024-01-23 트위스트 바이오사이언스 코포레이션 Methods for seamless nucleic acid assembly
WO2018231864A1 (en) 2017-06-12 2018-12-20 Twist Bioscience Corporation Methods for seamless nucleic acid assembly
CN111566125A (en) 2017-09-11 2020-08-21 特韦斯特生物科学公司 GPCR binding proteins and synthesis thereof
CA3079613A1 (en) 2017-10-20 2019-04-25 Twist Bioscience Corporation Heated nanowells for polynucleotide synthesis
AU2019270243A1 (en) 2018-05-18 2021-01-07 Twist Bioscience Corporation Polynucleotides, reagents, and methods for nucleic acid hybridization
KR20210144698A (en) 2019-02-26 2021-11-30 트위스트 바이오사이언스 코포레이션 Variant Nucleic Acid Libraries for Antibody Optimization
US11332738B2 (en) 2019-06-21 2022-05-17 Twist Bioscience Corporation Barcode-based nucleic acid sequence assembly

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0673435A1 (en) * 1991-08-23 1995-09-27 Isis Pharmaceuticals, Inc. Synthetic unrandomization of oligomer fragments
AU3811897A (en) * 1996-07-24 1998-02-10 Oglios Therapeutics, Inc. Antisense oligonucleotides as antibacterial agents
EP0975795A1 (en) * 1997-02-24 2000-02-02 TM Technologies Inc. Process for selecting anti-sense oligonucleotides

Also Published As

Publication number Publication date
EP1071826A1 (en) 2001-01-31
ATE339517T1 (en) 2006-10-15
DE69933205T2 (en) 2007-09-20
DE69933205D1 (en) 2006-10-26
EP1071826B1 (en) 2006-09-13
EP1071826A4 (en) 2003-05-21
AU762397B2 (en) 2003-06-26
AU3965099A (en) 1999-11-01
JP2002511276A (en) 2002-04-16

Similar Documents

Publication Publication Date Title
US7321828B2 (en) System of components for preparing oligonucleotides
EP1071826B1 (en) Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US6221587B1 (en) Identification of molecular interaction sites in RNA for novel drug discovery
Bucher Regulatory elements and expression profiles
Noller Structure of ribosomal RNA
US20030228597A1 (en) Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
AU2003304278B2 (en) Bead bound combinatorial oligonucleoside phosphorothioate and phosphorodithioate aptamer libraries
Juan et al. RNA secondary structure prediction based on free energy and phylogenetic analysis
US20020045175A1 (en) Gene recombination and hybrid protein development
US20050142581A1 (en) Microrna as ligands and target molecules
JP2004533228A (en) Computer-based assembly of a polynucleotide encoding a target polypeptide
EP1083980B1 (en) Modulation of molecular interaction sites on rna and other biomolecules
US20030032059A1 (en) Gene recombination and hybrid protein development
US20030017483A1 (en) Modulation of molecular interaction sites on RNA and other biomolecules
Wiley Genomics in the real world
WO2005003314A2 (en) A method of selecting an active oligonucleotide predictive model
Marshall et al. Oligonucleotide synthesis as a tool in drug discovery research
Várnai et al. Modeling DNA deformation
Flood Novel Fragmentation Method for Automated Next Generation Sequencing Exome Library Preparation
AU756906B2 (en) Identification of molecular interaction sites in RNA for novel drug discovery
WO1999058722A1 (en) Characterization of interactions between molecular interaction sites of rna and ligands therefor
WO2003016498A2 (en) Molecular interaction sites of rnase p rna and methods of modulating the same
Papana Tools for Comprehensive Statistical Analysis of Microarray Data
Karsten et al. Methods in Expression Analysis of Stem Cells.
Fabris et al. The problem of finding transcription factor binding sites and its impact in defining co-regulated gene-networks

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued
FZDE Discontinued

Effective date: 20110901