US20030113739A1 - System of components for preparing oligonucleotides - Google Patents

System of components for preparing oligonucleotides Download PDF

Info

Publication number
US20030113739A1
US20030113739A1 US10/116,325 US11632502A US2003113739A1 US 20030113739 A1 US20030113739 A1 US 20030113739A1 US 11632502 A US11632502 A US 11632502A US 2003113739 A1 US2003113739 A1 US 2003113739A1
Authority
US
United States
Prior art keywords
compounds
oligonucleotide
nucleic acid
robotically
generating
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
US10/116,325
Inventor
Lex Cowsert
Brenda Baker
John McNeil
Susan Freier
Henri Sasmor
Douglas Brooks
Cara Ohashi
Jacqueline Wyatt
Alexander Borchers
Timothy 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.)
Individual
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
Application filed by Individual filed Critical Individual
Priority to US10/116,325 priority Critical patent/US20030113739A1/en
Publication of US20030113739A1 publication Critical patent/US20030113739A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6811Selection methods for production or design of target specific oligonucleotides or binding molecules
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1048SELEX
    • 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
    • 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

Definitions

  • the present invention relates generally to the generation 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.
  • Synthetic oligonucleotides of complementarity to targets are known to hybridize with particular, target nucleic acids.
  • 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 target nucleic acid in vivo; such synthetic compounds are described herein as “active oligonucleotide compounds.”
  • Oligonucleotide compounds are also 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 is the antisense-mediated use of an endogenous nuclease, such as RNase H in eukaryotes or RNase P in prokaryotes, to the target nucleic acid (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 RNAs, lanthanide ion complexes, and other reactions species. (Haseloff et al., Nature, 1988, 334, 585; Baker et al., J. Am. Chem. Soc., 1997, 119, 8749).
  • 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).
  • antisense technology does allow for the direct utilization of nucleotide sequence information for gene function analysis.
  • antisense sequences hybridizable to the sequence can be generated using techniques known in the art.
  • ASOs 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 function(s) of the target gene.
  • active antisense oligonucleotides Although the practicality and value of this empirical approach to developing active antisense compounds 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).
  • 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.
  • 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, 15, 250).
  • 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 origin, development, spread or growth of the disease or disorder 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 will have a therapeutic benefit or not.
  • the resulting active antisense compounds may be used as positive controls when other, non antisense-based agents directed to the same target nucleic acid, or to its gene product, are screened.
  • 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.
  • 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. Pat. No. 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.
  • an assay portion of the process is stated to be performed by a computer controlled robot.
  • U.S. Pat. No. 5,708,158 to Hoey describes systems and methods for identifying 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. Pat. Nos. 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.
  • the present invention is directed to automated systems and methods for generating active oligonucleotide compounds, i.e., those having desired physical, chemical and/or biological properties.
  • the present invention is also directed to oligonucleotide-sensitive target sequences identified, by the systems and methods.
  • the present invention is described herein with respect to the production of antisense oligonucleotides; however, the present invention is not limited to this embodiment.
  • the present invention is directed to iterative processes for generating new chemical compounds with prescribed sets of physical, chemical and/or biological properties, and to systems for implementing these processes.
  • a target nucleic acid sequence is provided or selected, and a library of (candidate) nucleobase sequences is generated in silico (that is in a computer manipulatible and reliable form) according to defined criteria a virtual oligonucleotide chemistry is chosen.
  • a library of virtual oligonucleotide compounds having the desired nucleobase sequences 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.
  • second libraries of candidate nucleobase sequences are generated and/or selected to give rise to a second virtual oligonucleotide library.
  • a library of target nucleic acid sequences that are tractable to oligonucleotide technologies are identified. Such modulation includes, but is not limited to, antisense technology, gene function analysis and target validation.
  • FIGS. 1 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.
  • FIG. 3 is a flow diagram depicting the flow of data and materials among elements of step 200 of FIG. 1.
  • FIGS. 4 and 5 are a flow diagram depicting the flow of data and materials among elements of step 300 of FIG. 1.
  • FIG. 6 is a flow diagram depicting the flow of data and materials among elements of step 306 of FIG. 4.
  • FIG. 7 is another flow diagram depicting the flow of data and materials among elements of step 306 of FIG. 4.
  • FIG. 8 is a another flow diagram depicting the flow of data and materials among elements of step 306 of FIG. 4.
  • FIG. 9 is a flow diagram depicting the flow of data and materials among elements of step 350 of FIG. 5.
  • FIGS. 10 and 11 are flow diagrams depicting a logical analysis of data and materials among elements of step 400 of FIG. 1.
  • FIG. 12 is a flow diagram depicting the flow of data and materials among the elements of step 400 of FIG. 1.
  • FIGS. 13 and 14 are flow diagrams depicting the flow of data and materials among elements of step 500 of FIG. 1.
  • FIG. 15 is a flow diagram depicting the flow of data and materials among elements of step 600 of FIG. 1.
  • FIG. 16 is a flow diagram depicting the flow of data and materials among elements of step 700 of FIG. 1.
  • FIG. 17 is a flow diagram depicting the flow of data and materials among the elements of step 1100 of FIG. 2.
  • FIG. 18 is a block diagram showing the interconnecting of certain devices utilized in conjunction with a preferred method of the invention.
  • FIG. 19 is a flow diagram showing a representation of data storage in a relational database utilized in conjunction with one method of the invention.
  • FIG. 20 is a flow diagram depicting the flow of date and materials in effecting a preferred embodiment of the invention as set forth in Example 14;
  • FIG. 21 is a flow diagram depicting the depicting the flow of date and materials in effecting a preferred embodiment of the invention as set forth in Example 15;
  • FIG. 22 is a flow diagram depicting the depicting the flow of date and materials in effecting a preferred embodiment of the invention as set forth in Example 2;
  • FIG. 23 is a pictorial elevation view of a preferred apparatus used to robotically synthesize oligonucleotides.
  • FIG. 24 is a pictorial plan view of an apparatus used to robotically synthesize oligonucleotides.
  • the target selection process 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, provide new medicines to the medical community to fill therapeutic voids or improve upon existing therapies, 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 containing too many missing or incorrect base entries.
  • a target sequence that encodes a polypeptide such errors can 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 oligonucleotides to the target sequence will avoid such errors unless such errors occur frequently throughout the entire target sequence.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • apoptosis programmed cell death
  • 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).
  • PCR polymerase chain reaction
  • ‘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.
  • 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.
  • nucleotide sequences may be targeted as well.
  • 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.
  • FIG. 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 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 .
  • each distinct transcript is a unique target nucleic acid for purposes of step 300 .
  • the target nucleotide sequence is limited to those sequences that are unique to that transcript isoform.
  • the target nucleotide sequence is limited to sequences that are shared between the two or more transcripts.
  • full-length cDNA be used in the oligonucleotide design process step 300 (with full-length cDNA being defined a reading from the 5′ cap to the poly A tail).
  • 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 .
  • 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 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).
  • BLAST Altschul, et al., J. Mol. Biol., 1990, 215, 403
  • Gaped BLAST Altschul et al., Nucl. Acids Res., 1997, 25, 3389.
  • 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, Wash.).
  • 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 .
  • 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.
  • 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 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 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, then the process is preferably cycled back one iteration and that sequence is advanced at decision step 240 to process step 300 .
  • the loop is iterated such as by taking the 100 most 5-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 .
  • This loop is iterated until either no new sequence information is identified at decision step 240 , or an unexpectedly large amount of new information is found at decision step 260 , suggesting that the process moved outside the boundary of the gene into repetitive genomic sequence. In either of these cases, iteration of this loop is preferably stopped and the process advanced to the oligonucleotide design at process step 300 .
  • 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 alternative oligonucleotide chemistries on such parameters.
  • substitution of 5-methyl cytosine (5 MeC or m5 c) 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.
  • a list of oligonucleotide sequences is generated as represented in the flowchart shown in FIGS. 4 and 5.
  • 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.
  • step 304 all possible oligonucleotide sequences of the desired length capable of hybridizing to the target sequence obtained in step 200 are generated.
  • 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.
  • 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 occurring bases (A, T, C and G), unmodified sugar residues and a phosphodiester backbone.
  • 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 FIG. 6.
  • step 308 the desired thermodynamic properties are selected.
  • 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 .
  • thermodynamic and kinetic properties may be calculated for oligonucleotides as represented at step 317 .
  • 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).
  • 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 intra- and inter-molecular oligonucleotide structures, opening up target structure, and duplex formation.
  • RNA structure is predicted based on nearest neighbor analysis (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:
  • ⁇ G is the free energy of duplex formation
  • ⁇ H is the enthalpy change for each nearest neighbor
  • ⁇ S is the entropy change for each nearest neighbor
  • T is temperature
  • DNA duplex stability 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 DNA than in RNA (SantaLucia et al., Biochemistry, 1996, 35, 3555).
  • 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.
  • 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.
  • the desired sequence properties to be scored are selected at step 324 .
  • These properties include the number of strings of four guanosine residues in a row at step 325 ) or three guanosines 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 pyrimidines 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 dinucleot
  • sequence properties may be important in predicting oligonucleotide activity, or lack thereof.
  • U.S. Pat. No. 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.
  • 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.
  • the homology scores to be calculated are selected in step 342 .
  • Homology to nucleic acids encoding protein isoforms of the target may be desired.
  • oligonucleotides specific for an isoform of protein kinase C can be selected.
  • oligonucleotides can be selected to target multiple isoforms of such genes.
  • Homology to analogous target sequences may also be desired.
  • an oligonucleotide can be selected to a region common to both humans and mice to facilitate testing of the oligonucleotide in both species.
  • Homology to splice variants of the target nucleic acid as represented at step 345 , may be desired.
  • a desired range is selected to select the most promising oligonucleotides, as represented at step 347 .
  • 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 .
  • step 349 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 FIG. 9. If it is not desired then the process proceeds to step 375 .
  • step 350 the desired functional regions are selected.
  • Such regions include the transcription start site or 5′ cap at step 353 ), the 5′ untranslated region 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 ).
  • additional functional sites may be selected, as represented at step 369.
  • step 370 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 .
  • oligonucleotide sequences may result from the process thus far.
  • 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 utility is used to automate the distribution of sequences, as represented in step 380 .
  • oligonucleotide sequences are 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.
  • 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 (FIG. 1).
  • the logical basis for chemistry assignment is illustrated in FIGS. 10 and 11 and an iterative routine for stepping through an oligonucleotide nucleoside by nucleoside is illustrated in FIG. 12.
  • Chemistry assignment can be effected by 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.
  • oligonucleotide in reference to oligonucleotides, is used to refer to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetics thereof.
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • 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.
  • 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 .
  • compounds of the invention preferably are from about 8 to about 30 nucleobases in length.
  • antisense oligonucleotides comprising from about 12 to about 25 nucleobases (i.e. from about 8 to about 30 linked nucleosides).
  • oligonucleotides might be selected for non-antisense targeting strategies, for instance using the oligonucleotides 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.
  • the phosphate group can be linked to either the 2′, 3′ or 5′ hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • the respective ends of this linear polymeric structure can be further joined to form a circular structure, however, open linear structures are generally preferred.
  • 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.
  • oligonucleotides useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages.
  • 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.
  • modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
  • a base is selected.
  • 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 .
  • the sugar portion of the nucleoside is selected.
  • a sugar is selected that together with the select base will complete the nucleoside.
  • sugar chemistry 1 can be selected at process 422 or one or more alternative sugars are selected at process steps 424 , 426 and 428 .
  • 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 .
  • one or more conjugate groups can be attached to the oligonucleotide via 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 .
  • chemistries 1 though n are illustrated.
  • the number of alternate chemistries between chemistry 1 and alternative chemistry n, for each of the base, the sugar, the internucleoside linkage and the conjugate 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.
  • chemistry is assigned, as is shown in FIG. 12, to the list of oligonucleotides from general procedure 300 .
  • 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.
  • 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.
  • an inquiry is made at 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.’
  • 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 460 is followed and the pointer is moved to 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 process steps 453 is reiterated.
  • the result at decision step 460 is ‘Yes’, chemistry has been assigned to all of the nucleotides in the list of oligonucleotides.
  • chemistry selection includes selection of the base forming the nucleoside from a large palette of 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).
  • modified nucleobases including other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-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, 5-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 5-substituted a
  • nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in the Concise Encyclopedia Of Polymer Science And Engineering , pages 858-859, Kroschwitz, J. I., 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.
  • 5-substituted pyrimidines include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-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 1 -methoxyethyl sugar modifications, described below.
  • 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.
  • adenine might be selected however other alternative bases that can effect hybridization in a manner mimicking an ‘A’ base such as 2,6-diaminopurine might be selected should other consideration, e.g., stronger hybridization (relative to hybridization achieved with adenine), be desired.
  • 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 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 1 to C 10 alkyl or C 2 to C 10 alkenyl and alkynyl.
  • substituent groups comprise one of the following at the 2′ position: C 1 to C 10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, poly-alkylamino, 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—CH 2 CH 20 CH 3 , also known as 2′—O—(2-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(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2′-DMAOE, as described in co-owned U.S.
  • modifications include 2′-methoxy (2′—O—CH 3 ), 2′-aminopropoxy (2′-OCH 2 CH 2 CH 2 NH 2 ) 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.
  • chemistry selection includes selection of the internucleoside linkage.
  • These internucleoside linkages are also referred to as linkers, backbones or oligonucleotide backbones.
  • linkers backbones or oligonucleotide backbones.
  • a palette of different internucleoside linkages or backbones is available.
  • modified oligonucleotide backbones for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotri-esters, 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.
  • 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.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • alkene containing backbones sulfamate backbones
  • sulfonate and sulfonamide backbones amide backbones; and others having mixed N, 0, S and CH 2 component parts.
  • Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 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, and each of which is herein incorporated by reference.
  • 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.
  • a peptide nucleic acid (PNA) is referred to as a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar-phosphate backbone of an oligonucleotide is replaced with an amide-containing backbone, in particular an aminoethylglycine backbone.
  • 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 herein incorporated by reference. Further teaching of PNA compounds can be found in Nielsen et al., Science, 1991, 254, 1497.
  • the most preferred embodiments of the invention are oligonucleotides with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH 2 —NH—O—CH 2 —, —CH 2 —N(CH3)—O—CH 2 — [known as a methylene (methylimino) or MMI backbone], —CH 2 —O—N(CH 3 )—CH 2 —, —CH 2 —N(CH 3 )—N(CH 3 )—CH 2 — and —O—N(CH 3 )—CH 2 —CH 2 —[wherein the native phosphodiester backbone is represented as —O—P—O—CH 2 —] of the above referenced U.S.
  • oligonucleotide In attaching a conjugate group to one or more nucleosides or internucleoside linkages of an oligo-nucleotide, 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. Sci.
  • cholic acid Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053
  • a thioether e.g., hexyl-S-tritylthiol
  • a thiocholesterol (Oberhauser et al., Nucl.
  • 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.
  • 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.
  • 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. Pat. Nos.
  • oligonucleotides are synthesized on an automated synthesizer.
  • the synthesizer is preferably a variation of the synthesizer described in U.S. Pat. Nos. 5,472,672 and 5,529,756, the entire contents of which are herein incorporated by reference.
  • 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.
  • 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.
  • a synthesis file i.e., a cmd file
  • 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.
  • control software and data files are within the routine skill of persons skilled in anotated nucleotide synthesis. The same will depend upon the hardware employed, the chemistries adopeted and the design paradigm selected by the operator.
  • 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.
  • a tab file is built at process step 512 and stored at process step 514 .
  • 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_l.cmd is linked to synthesis l.tab by use of the same preamble in their names.
  • the automated, multi-well parallel array synthesizer employs a reagent array delivery format, in which each reagent utilized has a dedicated plumbing system. As seen in FIGS. 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.
  • the target reaction vessels 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 .
  • 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 facilitated 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.
  • 2′—O—(methoxyethyl) modified nucleoside 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.
  • oligonucleotides may be left empty (depending on the number of oligonucleotides to be made during an individual synthesis) or some of the well may have oligonucleotides that will serve as standards for comparison or analytical purposes.
  • the modified 96 well polypropylene plate 556 is utilized as the reaction vessel.
  • the working volume in each well is approximately 700 ⁇ l.
  • the bottom of each well is provided with a pressed-fit 20 ⁇ m polypropylene frit and a long capillary exit into a lower collection chamber as is illustrated in FIG. 5 of the above referenced U.S. Pat. No. 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 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 nmole and 10 ⁇ mol scales.
  • a CPG support is preferred, however other medium loading polystyrene-PEG supports such as TentaGelTM or ArgoGelTM can also be used.
  • 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 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 environment.
  • 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.
  • 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 ⁇ l.
  • the minimum delivery volume is about 2 ⁇ l, and flow rate accuracy is ⁇ 5%.
  • 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 ⁇ l 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 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.
  • a non-disposable synthesis plate machined from stainless steel or monel 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 UNC5891A 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 19200 Hz serial channel, and uses an elementary instruction set to communicate valve_number, time_open, motor_number and position_data.
  • TPU timer processing unit
  • 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.
  • 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.
  • [0166] means: Add 300 ⁇ l of the named reagent acetonitrile; ACN to each well of active synthesis
  • [0168] means: If the sequence pointer in the seq file is to a reagent in the list of reagents, independent of scale, add 300 ⁇ l of that particular reagent specified for that well.
  • sequence pointer in the seq file is to a reagent in the list of acids in the Class ACIDS — 1, and PYR is the name of pyridine, and ethyl chloroformate is defined in the tab file to activate the class, ACIDS — 1, then this instruction means:
  • 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.
  • [0175] means: Operate on those wells for which reagents contained in the Acid — 1 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.
  • WASH_1 the procedure name ⁇ BEGIN REPEAT 3 ADD ACN 300 DRAIN 15 END_REPEAT END
  • [0177] 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 ⁇ l 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 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.
  • 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:
  • 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 ( ⁇ mole) 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 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.
  • a MOVE command Not included in the command set is a MOVE command.
  • the controller software and hardware determines the correct nozzle(s) and well(s) 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.
  • the list of oligonucleotides for synthesis can be rearranged or grouped for optimization of synthesis.
  • 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.
  • a nucleotide bearing a 3′ phosphoramite is added to the 5′ 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.
  • an aliquot of a solid support bearing the proper nucleoside thereon is added to the well for synthesis.
  • the oligonucleotides can be grouped by the above described parameter or other parameters that facilitate the synthesis of the oligonucleotides.
  • 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′.
  • 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 multi well synthesizer at process step 588 for oligonucleotide synthesis.
  • the list of oligonucleotides again enters the general procedure flow as indicated in FIG. 1.
  • the plates can be lyophilized at this point if desired. Upon lyophilization, each well contains the oligonucleotides located therein as a dry compound.
  • quality control is performed on the oligonucleotides at process step 600 after a decision is made (decision step 550 ) to perform quality control.
  • quality control may be desired when there is some reason to doubt that some aspect of the synthetic process step 500 has been compromised.
  • 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.
  • 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.
  • step 610 sterile, double-distilled water is transferred by an automated liquid handler ( 2040 of FIG. 18) to each well of a multi-well plate containing a set of lyophilized antisense oligonucleotides.
  • the automated liquid handler ( 2040 of FIG. 18) reads the barcode sticker on the multi-well plate to obtain the plate's identification number.
  • Automated liquid handler 2040 queries Sample Database 520 (which resides in Database Server 2002 of FIG. 18) for the quality control assay instruction set for that plate and executes the appropriate steps.
  • Sample Database 520 which resides in Database Server 2002 of FIG. 18
  • the first illustrative quality control process quantitates the concentration of oligonucleotide in each well. If this quality control step is performed, an automated liquid handler ( 2040 of FIG. 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 FIG. 18). The UV spectrophotometer ( 2016 of FIG. 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 FIG. 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 FIG. 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 FIG. 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.
  • 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 636 quantitates the mass of total oligonucleotide in each well that is full length. If this quality control step is performed, an automated liquid handler ( 2040 of FIG. 2018) 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 FIG. 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 arrive at the mass of oligonucleotide product in each well. The results are then output to Sample Database 520 .
  • 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 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 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 FIG. 15).
  • an automated liquid handler ( 2040 of FIG. 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.
  • 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 [0204] 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 5A 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). Cells are routinely passaged by trysinization 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.
  • 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 trysinization and dilution when they reach 90% confluence.
  • NHDF Human neonatal dermal fibroblast
  • HEK Human embryonic keratinocytes
  • Oligonucleotide-mediated modulation of expression of a target nucleic acid can be assayed in a variety of ways known in the art.
  • 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.
  • poly(A)+ mRNA is preferred.
  • RT-PCR Reverse transcriptase polymerase chain reaction
  • 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, Mich. or via the internet at http://www.ANTIBODIES-PROBES.com/), or can be prepared via conventional antibody generation methods.
  • antipeptide monospecific 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).
  • the assaying step is described in detail in FIG. 16.
  • 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.
  • alternative assay methods 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.
  • such assays are tractable to semi-automated or robotic means.
  • RT-PCR 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 .
  • process step 785 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 .
  • data from all the assays on each oligonucleotide are complied and statistical parameters are automatically determined at process step 790 .
  • 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.
  • 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.
  • 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 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.
  • 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.
  • 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 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.
  • oligonucleotide compounds of the invention are optimized is by reiterating portions of the process of the invention using marginal compounds from the first iteration and selecting additional chemistries to the nucleobase sequences thereof.
  • 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 .
  • a second oligonucleotide chemistry is assigned at procedure 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 procedure steps 800 and 1000 .
  • FIG. 18 An embodiment of computer, network and instrument resources for effecting the methods of the invention is shown in FIG. 18.
  • four computer servers are provided.
  • a large database server 2002 stores all chemical structure, sample tracking and genomic, assay, quality control, and program status data. Further, this database server serves as the platform for a document management system.
  • a compute engine 2004 runs computational programs including RNA folding, oligonucleotide walking, and genomic searching.
  • a file server 2006 allows raw instrument output storage and sharing of robot instructions.
  • a groupware server 2008 enhances staff communication and process scheduling.
  • 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 is 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 .
  • FIG. 19 illustrates the data structure of a sample relational database. Various elements of data are segregated among linked storage elements of the database.
  • Cell-cell interactions are a feature of a variety of biological processes.
  • 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).
  • 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 as SEQ ID NO:85).
  • CD40 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 (Ibid.). 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 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.
  • CD40 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.
  • 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.
  • 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 FIG. 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 5.0TM, as represented in step 2504 . Desired thermodynamic properties were selected in step 2508 . 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 5.0TM.
  • 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.
  • 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.
  • a spreadsheet macro removed all oligonucleotides containing the text string ‘GGGG’.
  • 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.
  • Table 2 is a command file for synthesis of oligonucleotide having regions of 2′-O-(methoxyethyl) nucleosides and region of 2′-deoxy nucleosides each linked by phosphorothioate internucleotide linkages.
  • 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.
  • Table 4 is a seq file for oligonucleotides having regions of 2′-O-(methoxyethyl)nucleosides and 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-(methoxyethy) substituted nucleoside). The columns wrap around to next line when longer than one line.
  • Table 5 is a tab for reagents necessary for synthesizing an oligonucleotides having both 2′-O-(methoxy-ethy)nucleosides and 2′-deoxy nucleosides located therein. TABLE 5 Identity of columns: GroupName, Bottle ID, ReagentName, FlowRate, Concentration. Wherein reagent name is identified using base identifier, ‘moe’ indicated a 2′-O- (methoxyethy) substituted nucleoside and ‘cpg’ indicates a control pore glass solid support medium. The columns wrap around to next line when longer than one line.
  • Oligonucleotides were synthesized via solid phase P(III) phosphoramidite chemistry using a multi 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-cyanoethyldiisopropyl 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.
  • Oligonucleotides were cleaved from support and deprotected with concentrated NH 4 OH 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.
  • Unsubstituted and substituted phosphodiester oligo-nucleotides 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.
  • the oligonucleotides were purified by precipitating twice with 2.5 volumes of ethanol from a 0.5 M NaCl solution.
  • Phosphinate oligonucleotides are prepared as described in U.S. Pat. No. 5,508,270, herein incorporated by reference.
  • Alkyl phosphonate oligonucleotides are prepared as described in U.S. Pat. No. 4,469,863, herein incorporated by reference.
  • 3′-Deoxy-3′-methylene phosphonate oligonucleotides are prepared as described in U.S. Pat. Nos. 5,610,289 or 5,625,050, herein incorporated by reference.
  • Phosphoramidite oligonucleotides are prepared as described in U.S. Pat. No. 5,256,775 or U.S. Pat. No. 5,366,878, hereby incorporated by reference.
  • Alkylphosphonothioate oligonucleotides are prepared as described in published PCT applications PCT/US94/00902 and PCT/US93/06976 (published as WO 94/17093 and WO 94/02499, respectively).
  • 3′,-Deoxy-3′-amino phosphoramidate oligonucleotides are prepared as described in U.S. Pat. No. 5,476,925, herein incorporated by reference.
  • Phosphotriester oligonucleotides are prepared as described in U.S. Pat. No. 5,023,243, herein incorporated by reference.
  • Boranophosphate oligonucleotides are prepared as described in U.S. Pat. Nos. 5,130,302 and 5,177,198, both herein incorporated by reference.
  • Methylenemethylimino linked oligonucleosides also identified as MMI linked oligonucleosides, methylenedi-methylhydrazo 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 oligonucleo-sides, as well as mixed backbone compounds having, for instance, alternating MMI and PO or PS linkages are prepared as described in U.S. Pat. Nos. 5,378,825; 5,386,023; 5,489,677; 5,602,240 and 5,610,289, all of which are herein incorporated by reference.
  • Formacetal and thioformacetal linked oligonucleosides are prepared as described in U.S. Pat. Nos. 5,264,562 and 5,264,564, herein incorporated by reference.
  • Ethylene oxide linked oligonucleosides are prepared as described in U.S. Pat. No. 5,223,618, herein incorporated by reference.
  • PNAs Peptide nucleic acids
  • PNA Peptide nucleic acids
  • 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.’
  • Chimeric oligonucleotides having 2′-0-alkyl phosphorothioate and 2′-deoxy phosphorothioate oligo-nucleotide segments are synthesized using 2′-deoxy-5′-dimethoxytrityl-3′-O-phosphoramidites for the DNA portion and 5′-dimethoxytrityl-2′-O-methyl-3′-0-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.
  • [2′-O-(2-methoxyethyl)]—[2′-deoxy]—[-2′-O-(methoxy-ethyl)] chimeric phosphorothioate oligonucleotides are prepared as per the procedure above for the 2′-O-methyl chimeric oligonucleotide, with the substitution of 2′-O-(methoxyethyl) amidites for the 2′-O-methyl amidites.
  • [0279] [2′-0-(2-methoxyethyl phosphodiester]—[2′-deoxy phosphorothioate]—[2′-O-(methoxyethyl) phosphodiester] chimeric oligonucleotides are prepared as per the above procedure for the 2′-O-methyl chimeric oligonucleotide with the substitution of 2′-O-(methoxyethyl) amidites for the 2′-O-methyl amidites in the wing portions.
  • Sulfurization 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.
  • oligonucleotides were prepared as per the protocol of the 96 well format of Example 4.
  • the 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-(methoxyethyl)nucleotides on either side of a center gap region of 2′-deoxy nucleotides.
  • 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.
  • Tables 6-a and 6-b show the sequences of an intial first library, i.e., a library of phosphoroethioate oligonucleotides targeted to a CD40 target.
  • the compounds of Table 6-a shows the members of this listed in compliance with the established rule for listing SEQ ID NO:, i.e., in numerical SEQ ID NO: order.
  • 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.
  • Table 6-b the oligonucleotide 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.
  • oligonucleotide concentration 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 multi-channel robotic pipettors.
  • oligonucleotides or oligonucleosides are purified by precipitation twice out of 0.5 M NaCl with 2.5 volumes ethanol. Synthesized oligonucleotides are analyzed by polyacrylamide gel electrophoresis on denaturing gels. Oligonucleotide purity is checked by 31 P nuclear magnetic resonance spectroscopy, and/or by HPLC, as described by Chiang et al., J. Biol. Chem. 1991, 266, 18162.
  • 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 ⁇ l cold PBS. 60 ⁇ l lysis buffer (10 mM Tris-HCl, pH 7.6, 1 mM EDTA, 0.5 M NaCl, 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 ⁇ l of lysate was transferred to Oligo d(T) coated 96 well plates (AGCT Inc., Irvine, Calif.).
  • RT-PCR reverse transcriptase polymerase chain reaction
  • ABI PRISMTM 7700 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions. This is a closed-tube, non-gel-based, fluorescence detection system which allows high-throughput quantitation of polymerase chain reaction (PCR) products in real-time.
  • RNA 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, Calif.
  • a quencher dye e.g., TAMRA, PE-Applied Biosystems, Foster City, Calif.
  • annealing of the probe to the target sequence creates a substrate that can be cleaved by the 5′-exonuclease activity of Taq polymerase.
  • cleavage of the probe by Taq polymerase releases the reporter dye from the remainder of the probe (and hence from the quencher moiety) and a sequence-specific fluorescent signal is generated.
  • RT-PCR reagents were obtained from PE-Applied Biosystems, Foster City, Calif. RT-PCR reactions were carried out by adding 25 ⁇ l PCR cocktail (1 ⁇ TaqmanTM buffer A, 5.5 mM MgCl 2 , 300 uM each of DATP, dCTP and dGTP, 600 uM 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 ⁇ l poly(A) mRNA solution. The RT reaction was carried 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 15 seconds (denaturation) followed by 60° C. for 1.5 minutes (annealing/extension).
  • PCR primers were:
  • forward primer forward primer: CAGAGTTCACTGAAACGGAATGC (SEQ ID NO:86)
  • reverse primer GGTGGCAGTGTGTCTCTCTGTTC (SEQ ID NO:87)
  • reverse primer GAAGGTGAAGGTCGGAGTC (SEQ ID NO:89) GAAGATGGTGATGGGATTTC, (SEQ ID NO:90)
  • PCR probe 5′ JOE-CAAGCTTCCCGTTCTCAGCC-TAMRA 3′ (SEQ ID No. 91) where JOE (PE-Applied Biosystems, Foster City, Calif.) is the fluorescent reporter dye and TAMRA (PE-Applied Biosystems, Foster City, Calif.) is the quencher dye.
  • oligonucleotides complementary to mRNA were designed to target different regions of the human CD40 mRNA, using published sequences (GenBank accession number X60592, incorporated herein 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.
  • oligonucleotides complementary to mRNA were designed to target different regions of the human CD40 mRNA, 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′-methoxyethyl (2′-MOE) nucleotides.
  • the intersugar (backbone) linkages are phosphorothioate (P ⁇ S) throughout the oligonucleotide. Cytidine residues in the 2′-MOE wings are 5-methylcytidines.
  • 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.
  • 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.
  • 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.
  • the assembled reverse complement of SEQ ID NOS: 1 and 2 is represented below as SEQ ID NO:92: 5′- AGTGGTCCTGCCGCCTGGTC -3′ SEQ ID NO:92
  • an application is deployed which facilitates the selection process for determining the target positions of 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 interface allows users to skip from process to process at will, in any order.
  • the target Before running the site picking program, the target must have all relevant properties computed as described previously and indicated in process step 2204 .
  • the site picking program 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. 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 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 .
  • 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 .
  • 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 finding option, the user must pick a site at process step 2238 by clicking on its position on the linear representation of target.
  • a dynamic property is calculated for all possible sites (not yet eliminated) at process step 2230 . This property indicates the nearness of the site two a picked site allowing the user to pick sites in subsequent iterations based on target coverage.
  • 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 he 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.
  • FIG. 21 In a preferred embodiment of the invention, illustrated in FIG. 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.
  • the user launches it at process step 2302 .
  • the user selects from the previously selected sets of oligonucleotides at process step 2304 , registered to the database in site picker's process step 2238 .
  • 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 .
  • 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.
  • 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.
  • 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.
  • the software applies the template at process step 2312 to each of the complements of the sites in the list.
  • the templates it is possible that chemistries will be defined which are impossible to make with the chemical precursors presently used on the automatic synthesizer.
  • a database is maintained of all precursors previously designed, and their availability for automated synthesis.
  • the templates are applied, the resulting molecules are tested at process step 2316 against this database to see if they are readily synthesized.
  • a molecule is added to a list that the user inspects.
  • the user decides whether to modify the chemistry to make it compatible with the currently recognized list of available chemistries or to ignore it.
  • 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.
  • the results of the registration are displayed at process step 2328 with the new IDs 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 than 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.
  • a gene walk is executed using a CD40 antisense oligonucleotide having SEQ ID NO:15 (5′-CTGGCACAAAGAACAGCA.
  • SEQ ID NO:15 5′-CTGGCACAAAGAACAGCA.
  • Oligonucleotide Sequence ID 15 Name of Gene Target: CD40 Scope of Gene Walk: 20 Sequence Shift Increment: 1
  • 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 at which the 5 ′ terminus of each oligonucleotide hybridizes. This, the first ten oligonucleotides are single-base frame shift sequences directed against the CD40 sequence upstream of compound SEQ ID NO: 15 and the latter ten are single-base frame shift sequences directed against the CD40 sequence downstream of compound SEQ ID NO: 15.

Abstract

Interative, 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

    FIELD OF THE INVENTION
  • The present invention relates generally to the generation 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. [0001]
  • BACKGROUND OF THE INVENTION
  • 1. Oligonucleotide Technology [0002]
  • Synthetic oligonucleotides of complementarity to targets are known to hybridize with particular, target nucleic acids. In one example, 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 target nucleic acid in vivo; such synthetic compounds are described herein as “active oligonucleotide compounds.”[0003]
  • Oligonucleotide compounds are also 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 is the antisense-mediated use of an endogenous nuclease, such as RNase H in eukaryotes or RNase P in prokaryotes, to the target nucleic acid (Chiang et al., [0004] 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 RNAs, lanthanide ion complexes, and other reactions 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 sequences amenable to antisense technologies through an empirical approach (Szoka, [0005] Nature Biotechnology, 1997, 15, 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 [0006]
  • 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 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., [0007] Biochemistry, 1992, 31, 12055).
  • Heretofore, the search for lead antisense compounds has been limited to 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. [0008]
  • Accordingly, the need exists for systems and methods for efficiently and effectively generating new active antisense and other olgonucleotide compounds targeted to specific nucleic acid sequences. The present disclosure answers this need by providing systems and methods for automatically generating active antisense compounds via robotic and other automated means. [0009]
  • 3. Gene Function Analysis [0010]
  • 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, [0011] 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 (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 function(s) of the target gene. Although the practicality and value of this empirical approach to developing active antisense compounds 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, [0012] 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. [0013]
  • 4. Target Validation [0014]
  • 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, [0015] 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.
  • 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, [0016] 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, 15, 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. [0017]
  • 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 origin, development, spread or growth of the disease or disorder 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 will have a therapeutic benefit or not. The resulting active antisense compounds may be used as positive controls when other, non antisense-based agents directed to the same target nucleic acid, or to its gene product, are screened. [0018]
  • 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. [0019]
  • U.S. Pat. No. 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. [0020]
  • U.S. Pat. No. 5,708,158 to Hoey describes systems and methods for identifying 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. [0021]
  • U.S. Pat. Nos. 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. [0022]
  • SUMMARY OF THE INVENTION
  • The present invention is directed to automated systems and methods for generating active oligonucleotide compounds, i.e., those having desired physical, chemical and/or biological properties. The present invention is also directed to oligonucleotide-sensitive target sequences identified, by the systems and methods. For purposes of illustration, the present invention is described herein with respect to the production of antisense oligonucleotides; however, the present invention is not limited to this embodiment. [0023]
  • The present invention is directed to iterative processes for generating new 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) nucleobase sequences is generated in silico (that is in a computer manipulatible and reliable form) according to defined criteria a virtual oligonucleotide chemistry is chosen. A library of virtual oligonucleotide compounds having the desired nucleobase sequences 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. [0024]
  • In subsequent iterations of the process, second libraries of candidate nucleobase sequences are generated and/or selected to give rise to a second virtual oligonucleotide library. Through multiple iterations of the process, a library of target nucleic acid sequences that are tractable to oligonucleotide technologies are identified. Such modulation includes, but is not limited to, antisense technology, gene function analysis and target validation. [0025]
  • 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 numbers indicate identical or functionally similar elements.[0026]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The present invention will be described with reference to the accompanying drawings, wherein: [0027]
  • FIGS. 1 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. [0028]
  • FIG. 3 is a flow diagram depicting the flow of data and materials among elements of [0029] step 200 of FIG. 1.
  • FIGS. 4 and 5 are a flow diagram depicting the flow of data and materials among elements of [0030] step 300 of FIG. 1.
  • FIG. 6 is a flow diagram depicting the flow of data and materials among elements of [0031] step 306 of FIG. 4.
  • FIG. 7 is another flow diagram depicting the flow of data and materials among elements of [0032] step 306 of FIG. 4.
  • FIG. 8 is a another flow diagram depicting the flow of data and materials among elements of [0033] step 306 of FIG. 4.
  • FIG. 9 is a flow diagram depicting the flow of data and materials among elements of [0034] step 350 of FIG. 5.
  • FIGS. 10 and 11 are flow diagrams depicting a logical analysis of data and materials among elements of [0035] step 400 of FIG. 1.
  • FIG. 12 is a flow diagram depicting the flow of data and materials among the elements of [0036] step 400 of FIG. 1.
  • FIGS. 13 and 14 are flow diagrams depicting the flow of data and materials among elements of [0037] step 500 of FIG. 1.
  • FIG. 15 is a flow diagram depicting the flow of data and materials among elements of [0038] step 600 of FIG. 1.
  • FIG. 16 is a flow diagram depicting the flow of data and materials among elements of [0039] step 700 of FIG. 1.
  • FIG. 17 is a flow diagram depicting the flow of data and materials among the elements of [0040] step 1100 of FIG. 2.
  • FIG. 18 is a block diagram showing the interconnecting of certain devices utilized in conjunction with a preferred method of the invention; [0041]
  • FIG. 19 is a flow diagram showing a representation of data storage in a relational database utilized in conjunction with one method of the invention; [0042]
  • FIG. 20 is a flow diagram depicting the flow of date and materials in effecting a preferred embodiment of the invention as set forth in Example 14; [0043]
  • FIG. 21 is a flow diagram depicting the depicting the flow of date and materials in effecting a preferred embodiment of the invention as set forth in Example 15; [0044]
  • FIG. 22 is a flow diagram depicting the depicting the flow of date and materials in effecting a preferred embodiment of the invention as set forth in Example 2; [0045]
  • FIG. 23 is a pictorial elevation view of a preferred apparatus used to robotically synthesize oligonucleotides; and [0046]
  • FIG. 24 is a pictorial plan view of an apparatus used to robotically synthesize oligonucleotides.[0047]
  • Certain preferred methods of this invention are now described with reference to the flow diagram of FIGS. 1 and 2. [0048]
  • 1. Target Nucleic Acid Selection. [0049]
  • The target selection process, [0050] 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 [0051] 100, is to identify molecular targets that represent significant therapeutic opportunities, provide new medicines to the medical community to fill therapeutic voids or improve upon existing therapies, 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 containing too many missing or incorrect base entries. In the case of a target sequence that encodes a polypeptide, such errors can 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 oligonucleotides to the target sequence will avoid such errors unless such errors occur frequently throughout the entire target sequence. [0052]
  • 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 [0053] 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 I-ossible to operate upon a genetic system having a lack of information regarding, or incomplete characterization of, the biological function(s) 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. [0054]
  • For target validation and drug discovery, another selection criterion is disease association. Candidate target genes are placed into one of several broad categories of known or deduced disease association. [0055] 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.
  • [0056] 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. [0057]
  • 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. [0058]
  • 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., [0059] 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 polymerase 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. [0060]
  • 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. [0061]
  • 2. Assembly of Target Nucleotide Sequence. [0062]
  • FIG. 3 is a block diagram detailing the steps of the target nucleotide sequence assembly process, [0063] 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 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 [0064] 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 [0065] 300 (with full-length cDNA being defined a 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 [0066] 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 searched at process step 212 for additional sequence information.
  • The algorithm preferably used in process steps [0067] 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 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 1.
    TABLE 1
    Database Sources of Target Sequences
    Database Contents Source
    NR All non-redundant National Center for
    GenBank, EMBL, DDBJ Biotechnology Information
    and PDB sequences at the National
    Institutes of Health
    Month All new or revised National Center for
    GenBank, EMBL, DDBJ Biotechnology Information
    and PDB sequences at the National
    released in the last Institutes of Health
    30 days
    Dbest Non-redundant National Center for
    database of GenBank, Biotechnology Information
    EMBL, DDBJ and EST at the National
    divisions Institutes of Health
    Dbsts Non-redundant National Center for
    database of GenBank, Biotechnology Information
    EMBL, DDBJ and STS at the National
    divisions Institutes of Health
    Htgs High throughput National Center for
    genomic sequences Biotechnology Information
    at the National
    Institutes of Health
  • When genomic sequence information is available at [0068] 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, Wash.). 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 preferably 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 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.
  • [0069] 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 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, 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 5-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.
  • This loop is iterated until either no new sequence information is identified at [0070] decision step 240, or an unexpectedly large amount of new information is found at decision step 260, suggesting that the process moved outside the boundary of the gene into repetitive genomic sequence. In either of these cases, iteration of this loop is preferably stopped and the process advanced to the oligonucleotide design at process step 300.
  • 3. In Silico Generation of a Set of Nucleobase Sequences and Virtual Oligonucleotides. [0071]
  • For the following [0072] 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 alternative oligonucleotide chemistries on such parameters. For example, substitution of 5-methyl cytosine (5 MeC or m5 c) 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 [0073] order step 300 before step 400 as seen in FIG. 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 FIGS. 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 [0074] 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 occurring bases (A, T, C and G), unmodified sugar residues and a phosphodiester backbone.
  • 4. In Silico Evaluation of Thermodynamic Properties of Virtual Oligonucleotides. [0075]
  • In [0076] 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 FIG. 6. In step 308, the desired thermodynamic properties are selected.
  • This will typically include [0077] 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 [0078] 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. [0079]
  • 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., [0080] 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. [0081]
  • 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. [0082]
  • 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 intra- and inter-molecular oligonucleotide structures, opening up target structure, and duplex formation. [0083]
  • The most stable RNA structure is predicted based on nearest neighbor analysis (Serra et al., [0084] 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:
  • Δ T =ΔH°−TΔS°
  • where: [0085]
  • ΔG is the free energy of duplex formation, [0086]
  • ΔH is the enthalpy change for each nearest neighbor, [0087]
  • ΔS is the entropy change for each nearest neighbor, and [0088]
  • T is temperature. [0089]
  • The ΔH and ΔS 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. [0090]
  • 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 DNA than in RNA (SantaLucia et al., [0091] 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. ([0092] 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 [0093]
  • 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. [0094]
  • Following the calculation of thermodynamic properties ending at [0095] step 317, the desired sequence properties to be scored are selected at step 324. These properties include the number of strings of four guanosine residues in a row at step 325) or three guanosines 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 pyrimidines 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. Pat. No. 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 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. [0096]
  • Following [0097] 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 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 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 [0098] 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 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. [0099]
  • 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 [0100] decision step 349. If it is desired to target functional regions then process step 350 occurs as seen in greater detail in FIG. 9. If it is not desired then the process proceeds to step 375.
  • In [0101] step 350, as seen in FIG. 9, the desired functional regions are selected. Such regions include the transcription start site or 5′ cap at step 353), the 5′ untranslated region 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. [0102]
  • 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 [0103] step 370.
  • 7. Uniform Distribution of Oligonucleotides. [0104]
  • 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 [0105] 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 utility is used to automate the distribution of sequences, as represented in step 380. Such a utility 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 oligonucleotide sequences are passed onto step 400 of the process, where oligonucleotide chemistries are assigned.
  • 8. Assignment of Actual Oligonucleotide Chemistry. [0106]
  • 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. [0107]
  • Chemistries can be assigned to the nucleobase sequences during general procedure step [0108] 400 (FIG. 1). The logical basis for chemistry assignment is illustrated in FIGS. 10 and 11 and an iterative routine for stepping through an oligonucleotide nucleoside by nucleoside is illustrated in FIG. 12. Chemistry assignment can be effected by 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. [0109]
  • 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. [0110]
  • 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 [0111] general procedure 300. For use as antisense oligonucleotides compounds of the invention preferably are from about 8 to about 30 nucleobases in length. Particularly preferred are antisense oligonucleotides comprising from about 12 to about 25 nucleobases (i.e. from about 8 to about 30 linked nucleosides). 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 oligonucleotides 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. [0112]
  • 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. [0113]
  • 10. Selection of Oligonucleotide Chemistries. [0114]
  • In a general logic scheme as illustrated in FIGS. 10 and 11, for each nucleoside position, the user or automated device is interrogated first for a base assignment, followed by a sugar assignment, a linker assignment and finally a conjugate assignment. Thus for each nucleoside, at process step [0115] 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 attachment to the nucleoside or attachment to the internucleoside linkage. The addition of a conjugate group is integrated at [0116] process step 440 and the assignment of the conjugate group is effected at process step 450.
  • For illustrative purposes in FIGS. 10 and 11, for each of the base, the sugar, the internucleoside linkers, or the conjugate, [0117] 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 base, the sugar, the internucleoside linkage and the conjugate, 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 FIGS. 10 and 11, chemistry is assigned, as is shown in FIG. 12, to the list of oligonucleotides from [0118] 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 [0119] decision step 440, an inquiry is made at 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.’
  • When the result at [0120] 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 460 is followed and the pointer is moved to 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 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. [0121]
  • As is illustrated in FIG. 10, for each nucleoside of an oligonucleotide, chemistry selection includes selection of the base forming the nucleoside from a large palette of 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 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-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, 5-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 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in the [0122] Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. I., 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-aminopropyladenine, 5-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 21-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. Pat. No. 3,687,808, as well as U.S. Pat. Nos. 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, Reference is also made to allowed U.S. patent application Ser. No. 08/762,488, filed on Dec. 10, 1996, commonly owned with the present application and herein incorporated by reference. [0123]
  • 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 2,6-diaminopurine might be selected should other consideration, e.g., stronger hybridization (relative to hybridization achieved with adenine), be desired. [0124]
  • As is illustrated in FIG. 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 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[0125] 1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Particularly preferred are O[(CH2)nO]mCH3, O(CH2)nOCH3, O(CH2)nNH2, O(CH2)n, CH3, O(CH2)nONH2, and O (CH2)nON [(CH2)nCH3)]2, where n and m are from 1 to about 10. Other preferred substituent groups comprise one of the following at the 2′ position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, poly-alkylamino, 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—CH2CH20CH3, also known as 2′—O—(2-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(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, as described in co-owned U.S. patent application Ser. No. 09/016,520, filed on Jan. 30, 1998, the contents of which are herein incorporated by reference.
  • Other preferred modifications include 2′-methoxy (2′—O—CH[0126] 3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2) 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. Pat. Nos. 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 herein incorporated by reference, together with allowed U.S. patent application Ser. No. 08/468,037, filed on Jun. 5, 1995, which is commonly owned with the present application and is herein incorporated by reference. [0127]
  • As is illustrated in FIG. 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, phosphotri-esters, 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. [0128]
  • Representative United States patents that teach the preparation of the above phosphorus containing linkages include, but are not limited to, U.S. Pat. Nos. 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, and each of which is herein incorporated by reference. [0129]
  • 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 the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide 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, 0, S and CH[0130] 2 component parts.
  • Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 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, and each of which is herein incorporated by reference. [0131]
  • In other preferred oligonucleotides, i.e., oligo-nucleotide 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 herein incorporated by reference. Further teaching of PNA compounds can be found in Nielsen et al., [0132] 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 —CH[0133] 2—NH—O—CH2—, —CH2—N(CH3)—O—CH2— [known as a methylene (methylimino) or MMI backbone], —CH2—O—N(CH3)—CH2—, —CH2—N(CH3)—N(CH3)—CH2— and —O—N(CH3)—CH2—CH2—[wherein the native phosphodiester backbone is represented as —O—P—O—CH2—] of the above referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above referenced U.S. Pat. No. 5,602,240. Also preferred are oligonucleotides having morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.
  • In attaching a conjugate group to one or more nucleosides or internucleoside linkages of an oligo-nucleotide, 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., [0134] Proc. Natl. Acad. 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; 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 (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. Pat. Nos. 4,828,979; 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; 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. [0135]
  • 12. Chimeric Compounds. [0136]
  • 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. [0137]
  • 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. [0138]
  • 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. Pat. Nos. 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 herein incorporated by reference, together with commonly owned and allowed U.S. patent application Ser. No. 08/465,880, filed on Jun. 6, 1995, also herein incorporated by reference. [0139]
  • 13. Description of Automated Oligonucleotide Synthesis. [0140]
  • In the next step of the overall process (illustrated in FIGS. 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 U.S. Pat. Nos. 5,472,672 and 5,529,756, the entire contents of which are herein incorporated by reference. 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. [0141]
  • 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. [0142]
  • Building of the seq, .cmd and tab files is illustrated in FIG. 13. Thus as a part of the general [0143] 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 anotated nucleotide synthesis. The same will depend upon the hardware employed, the chemistries adopeted and the design paradigm selected by the operator. [0144]
  • 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 [0145] 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 [0146] 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_l.cmd is linked to synthesis l.tab by use of the same preamble in their names.
  • The automated, multi-well parallel array synthesizer employs a reagent array delivery format, in which each reagent utilized has a dedicated plumbing system. As seen in FIGS. 23 and 24, an [0147] 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 [0148] 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 facilitated 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 μmole 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 [0149] 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 [0150] 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—(methoxyethyl) modified nucleoside 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 well 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 [0151] 522 for 20 minutes. Reagents are dissolved to appropriate concentrations and installed on the synthesizer. Large bottles, collectively identified as 0.558 in FIG. 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 FIG. 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 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 [0152] well polypropylene plate 556 is utilized as the reaction vessel. The working volume in each well is approximately 700 μl. The bottom of each well is provided with a pressed-fit 20 μm polypropylene frit and a long capillary exit into a lower collection chamber as is illustrated in FIG. 5 of the above referenced U.S. Pat. No. 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 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 nmole and 10 μmol scales. For oligonucleotide synthesis a CPG support is preferred, however other medium loading polystyrene-PEG supports such as TentaGel™ or ArgoGel™ can also be used.
  • As seen in FIG. 24, the synthesis plate is transported back and forth in the X-direction under an array of 8 moveable banks ([0153] 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 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 environment.
  • 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. [0154]
  • Normally, liquid contents are blown directly to [0155] 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. [0156]
  • 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 [0157] 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 [0158] 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 μl. The minimum delivery volume is about 2 μl, and flow rate accuracy is ±5%. 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 μl per sec) is experimentally determined, and this information is contained in the reagent table setup file.
  • Heating and cooling of the [0159] reaction block 526 is effected utilizing a recirculating 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 monel 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 [0160] 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 UNC5891A 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 19200 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, 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. [0161]
  • One basic set of command instructions can be: [0162]
    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. [0163]
  • Thus: [0164]
  • [0165] ADD ACN 300
  • means: Add 300 μl of the named reagent acetonitrile; ACN to each well of active synthesis [0166]
  • ADD <seq>300 [0167]
  • means: If the sequence pointer in the seq file is to a reagent in the list of reagents, independent of scale, add 300 μl of that particular reagent specified for that well. [0168]
  • ADD 1.1 PYR +1.0 <seq>+1.1 <actl>[0169]
  • means: If the sequence pointer in the seq file is to a reagent in the list of acids in the [0170] Class ACIDS 1, and PYR is the name of pyridine, and ethyl chloroformate is defined in the tab file to activate the class, ACIDS 1, then this instruction means:
  • Add 1.1 equiv. pyridine [0171]
  • 1.0 equiv. of the acid specified for that well and [0172]
  • 1.1 equiv. of the activator, ethyl chloroformate [0173]
  • 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. [0174]
    Thus:
    ACYLATION   {the procedure name}
    BEGIN
    IF CLASS = ACIDS_1
    ADD 1.0 <seq> + 1.1 <act1> + 1.1 PYR
    WAIT 60
    ENDIF
    IF CLASS = ACIDS_2
    ADD 1.0 <seq> + 1.2 <act1> + 1.2 TEA
    ENDIF
    WAIT 60
    DRAIN 10
    END
  • means: Operate on those wells for which reagents contained in the [0175] Acid 1 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. [0176]
    Thus:
    WASH_1   {the procedure name}
    BEGIN
    REPEAT
    3
    ADD ACN 300
    DRAIN 15
    END_REPEAT
    END
  • means: repeats the add acetonitrile and drain sequence for each well three times. [0177]
  • The PRIME command will operate either on specific named reagents or on nozzles which will be used in the next associated <seq>operation. The μl amount dispensed into a prime port is a constant that can be specified in a config.dat file. [0178]
  • 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 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. [0179]
  • 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. [0180]
  • The LOAD and REMOVE commands are instructions for the machine to pause for operator action. [0181]
  • 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: [0182]
  • Well_No Well_ID Scale Sequence [0183]
  • 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 (μmole) 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). [0184]
  • The general form of the .tab file is the definition: [0185]
  • Class Bottle Reagent Name Flow_rate Conc. [0186]
  • 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. [0187]
  • 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 nozzle(s) and well(s) required for a particular reagent addition, then synchronizes the position of the requisite nozzle and well prior to adding the reagent. [0188]
  • 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. [0189]
  • In reference to FIG. 14, the list of oligonucleotides for synthesis can be rearranged or grouped for optimization of synthesis. Thus at [0190] 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 5′ 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. [0191]
  • The oligonucleotides can be grouped by the above described parameter or other parameters that facilitate the synthesis of the oligonucleotides. Thus in FIG. 14, sorting is noted as being effected by some parameter of [0192] 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 [0193] 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 multi 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 FIG. 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. [0194]
  • In an optional step, quality control is performed on the oligonucleotides at [0195] 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 doubt 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 [0196] general procedure 600 is detailed in steps 610-660 of FIG. 15. Also referenced in the following discussion are the robotics and associated analytical instrumentation as shown in FIG. 18.
  • During step [0197] 610 (FIG. 15), sterile, double-distilled water is transferred by an automated liquid handler (2040 of FIG. 18) to each well of a multi-well plate containing a set of lyophilized antisense oligonucleotides. The automated liquid handler (2040 of FIG. 18) reads the barcode sticker on the multi-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 FIG. 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 ([0198] 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 FIG. 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 FIG. 18). The UV spectrophotometer (2016 of FIG. 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 FIG. 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 [0199] 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 FIG. 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 FIG. 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 [0200] 632 to 636) quantitates the mass of total oligonucleotide in each well that is full length. If this quality control step is performed, an automated liquid handler (2040 of FIG. 2018) 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 FIG. 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 arrive 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 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 [0201] 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 FIG. 15). If a plate receives ‘Pass’ status, an automated liquid handler (2040 of FIG. 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. [0202]
  • 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. [0203]
  • T-24 Cells: [0204]
  • 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 5A basal media (Life Technologies, Gaithersburg, Md.) supplemented with 10% fetal calf serum, [0205] penicillin 100 units per milliliter, and streptomycin 100 micrograms per milliliter (all from Life Technologies). Cells are routinely passaged by trysinization 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.
  • A549 Cells: [0206]
  • 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, [0207] penicillin 100 units per milliliter, and streptomycin 100 micrograms per milliliter (all from Life Technologies). Cells are routinely passaged by trysinization and dilution when they reach 90% confluence.
  • NHDF Cells: [0208]
  • 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. [0209]
  • HEK Cells: [0210]
  • Human embryonic keratinocytes (HEK) were obtained from the Clonetics Corp. HEKs were routinely maintained in Keratinocyte Growth Medium (Clonetics Corp.) as provided by the supplier. Cell are routinely maintained for up to 10 passages as recommended by the supplier. [0211]
  • 16. Treatment of Cells with Candidate Compounds: [0212]
  • When cells reach about 80% confluency, they are treated with oligonucleotide. For cells grown in 96-well plates, wells are washed once with 200 μl Opti-MEM-1™ reduced-serum medium (Life Technologies) and then treated with 130 μl of Opti-MEM-1™ containing 3.75 μg/ml LIPOFECTIN (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. [0213]
  • 17. Assaying Oligonucleotide Activity: [0214]
  • 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. ([0215] 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). 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, Calif.) according to 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, Mich. or via the internet at http://www.ANTIBODIES-PROBES.com/), or can be prepared via conventional antibody generation methods. Methods for preparation of polyclonal, monospecific (‘antipeptide’) and monoclonal antisera are taught by, for example, Ausubel et al. ([0216] 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). [0217]
  • 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. [0218]
  • The assaying step, [0219] general procedure step 700, is described in detail in FIG. 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 [0220] 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 complied and statistical parameters are automatically determined at process step 790.
  • 18. Classification of Compounds Based on Their Activity: [0221]
  • Following assaying, [0222] 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. [0223]
  • 19. Optimization of Lead Compounds by Sequence. [0224]
  • 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. [0225]
  • The operation of the [0226] gene walk process 1100 detailed in steps 1104-1112 of FIG. 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 [0227] 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 [0228] 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. [0229]
  • Another means by which oligonucleotide compounds of the invention are optimized is by reiterating portions of the process of the invention using marginal compounds from the first iteration and selecting additional chemistries to the nucleobase sequences thereof. [0230]
  • Thus, for example, an oligonucleotide chemistry different from that of the first set of oligonucleotides is assigned at [0231] 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. [0232]
  • In a related process, a second oligonucleotide chemistry is assigned at [0233] procedure 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 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 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. [0234]
  • These antisense-sensitive sites are assembled into contiguous sequences (contigs) using the procedures described for assembling target nucleotide sequences (at procedure step [0235] 200).
  • 22. Systems for Executing Preferred Methods of the Invention. [0236]
  • An embodiment of computer, network and instrument resources for effecting the methods of the invention is shown in FIG. 18. In this embodiment, four computer servers are provided. First, a [0237] large database server 2002 stores all chemical structure, sample tracking and genomic, assay, quality control, and program status data. Further, this 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.
  • A redundant high-speed network system is provided between the main servers and the [0238] bridges 2026, 2028 and 2030. These bridges provide reliable network access to the is 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 [0239] 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 [0240] 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. [0241]
  • Data is stored in an appropriate database. For use with the methods of the invention, a relational database is preferred. FIG. 19 illustrates the data structure of a sample relational database. Various elements of data are segregated among linked storage elements of the database. [0242]
  • 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. [0243]
  • 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., [0244] 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. [0245] Leuk. Lymphoma, 1997, 24, 393). A full cDNA sequence for CD40 is available (GenBank accession number X60592, incorporated herein as SEQ ID NO: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 (Ibid.). 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 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., [0246] J. Clin. Immunol., 1996, 16, 83). In endothelial cells, stimulation of CD40 by CD40L has been found to induce surface expression of E-selectin, ICAM-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. [0247]
  • 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., [0248] 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 FIG. 22. From the assembled CD40 sequence, the process began with determining the desired oligonucleotide length to be eighteen nucleotides, as represented in [0249] step 2500. All possible oligonucleotides of this length were generated by Oligo 5.0™, as represented in step 2504. Desired thermodynamic properties were selected in step 2508. 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) [0250]
  • Table 2 is a command file for synthesis of oligonucleotide having regions of 2′-O-(methoxyethyl) nucleosides and region of 2′-deoxy nucleosides each linked by phosphorothioate internucleotide linkages. [0251]
    TABLE 2
    SOLID_SUPPORT_SKIP
    BEGIN
    Next_Sequence
    END
     INITIAL-WASH
    BEGIN
    Add ACN
    300
    Drain 10
    END
     LOOP-BEGIN
     DEBLOCK
    BEGIN
    Prime TCA
    Load Tray
    Repeat
    2
    Add TCA 150
    Wait 10
    Drain 8
    End_Repeat
    Remove Tray
    Add TCA 125
    Wait 10
    Drain 8
    END
    WASH_AFTER_DEBLOCK
    BEGIN
    Repeat
    3
    Add ACN 250 To_All
    Drain 10
    End_Repeat
    END
    COUPLING
    BEGIN
    if class = DEOXY_THIOATE
    Nozzle wash <act1>
    prime <act1>
    prime <seq>
    Add <act1> 70 + <seq> 70
    Wait 40
    Drain 5
    end-if
    if class = MOE_THIOATE
    Nozzle wash <act1>
    Prime <act1>
    prime <seq>
    Add <act1> 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
    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) [0252]
  • Table 3 is a seq file for oligonucleotides having 2′-deoxy nucleosides linked by phosphorothioate internucleotide linkages. [0253]
    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 Gs
    2 A02 200 As Cs Gs Gs Cs Gs Gs As Cs
    Cs  As  Gs As Gs Ts Gs Gs As
    3 A03 200 As Cs Cs As As Gs Cs As Gs
    As  Cs  Gs Gs As Gs As Cs Gs
    4 A04 200 As Gs Gs As Gs As Cs Cs Cs
    Cs  Gs  As Cs Gs As As Cs Gs
    5 A05 200 As Cs Cs Cs Cs Gs As Cs Gs
    As  As  Cs Gs As Cs Ts Gs Gs
    6 A06 200 As Cs Gs As As Cs Gs As Cs
    Ts  Gs  Gs Cs Gs As Cs As Gs
    7 A07 200 As Cs Gs As Cs Ts Gs Gs Cs
    Gs  As  Cs As Gs Gs Ts As Gs
    8 A08 200 As Cs As Gs Gs Ts As Gs Gs
    Ts  Cs  Ts Ts Gs Gs Ts Gs Gs
    9 A09 200 As Gs Gs Ts Cs Ts Ts Gs Gs
    Ts  Gs  Gs Gs Ts Gs As Cs Gs
    10 A10 200 As Gs Ts Cs As Cs Gs As Cs
    As  As  Gs As As As Cs As Cs
    11 A11 200 As Cs Gs As Cs As As Gs As
    As  As  Cs As Cs Gs Gs Ts Cs
    12 A12 200 As Gs As As As Cs As Cs Gs
    Gs  Ts  Cs Gs Gs Ts Cs Cs Ts
    13 B01 200 As As Cs As Cs Gs Gs Ts Cs
    Gs  Gs  Ts Cs Cs Ts Gs Ts Cs
    14 B02 200 As Cs Ts Cs As Cs Ts Gs As
    Cs  Gs  Ts Gs Ts Cs Ts Cs As
    15 B03 200 As Cs Gs Gs As As Gs Gs As
    As  Cs  Gs Cs Cs As Cs Ts Ts
    16 B04 200 As Ts Cs Ts Gs Ts Gs Gs As
    Cs  Cs  Ts Ts Gs Ts Cs Ts Cs
    17 B05 200 As Cs As Cs Ts Ts Cs Ts Ts
    Cs  Cs  Gs As Cs Cs Gs Ts Gs
    18 B06 200 As Cs Ts Cs Ts Cs Gs As Cs
    As  Cs  As Gs Gs As Cs Gs Ts
    19 B07 200 As As As Cs Cs Cs Cs As Gs
    Ts  Ts  Cs Gs Ts Cs Ts As As
    20 B08 200 As Ts Gs Ts Cs Cs Cs Cs As
    As  As  Gs As Cs Ts As Ts Gs
    21 B09 200 As Cs Gs Cs Ts Cs Gs Gs Gs
    As  Cs  Gs Gs Gs Ts Cs As Gs
    22 B10 200 As Gs Cs Cs Gs As As Gs As
    As  Gs  As Gs Gs Ts Ts As Cs
    23 B11 200 As Cs As Cs As Gs Ts As Gs
    As  Cs  Gs As As As Gs Cs Ts
    24 B12 200 As Cs As Cs Ts Cs Ts Gs Gs
    Ts  Ts  Ts Cs Ts Gs Gs As Cs
    25 C01 200 As Cs Gs As Cs Cs As Gs As
    As  As  Ts As Gs Ts Ts Ts Ts
    26 C02 200 As Gs Ts Ts As As As As Gs
    Gs  Gs  Cs Ts Gs Cs Ts As Gs
    27 C03 200 As Gs Gs Ts Ts Gs Ts Gs As
    Cs  Gs  As Cs Gs As Gs Gs Ts
    28 C04 200 As As Ts Gs Ts As Cs Cs Ts
    As  Cs  Gs Gs Ts Ts Gs Gs Cs
    29 C05 200 As Gs Ts Cs As Cs Gs Ts Cs
    Cs  Ts  Cs Ts Cs Ts Gs Ts Cs
    30 C06 200 Cs Ts Gs Gs Cs Gs As Cs As
    Gs  Gs  Ts As Gs Gs Ts Cs Ts
    31 C07 200 Cs Ts Cs Ts Gs Ts Gs Ts Gs
    As  Cs  Gs Gs Ts Gs Gs Ts Cs
    32 C08 200 Cs As Gs Gs Ts Cs Gs Ts Cs
    Ts  Ts  Cs Cs Cs Gs Ts Gs Gs
    33 C09 200 Cs Ts Gs Ts Gs Gs Ts As Gs
    As  Cs  Gs Ts Gs Gs As Cs As
    34 C10 200 Cs Ts As As Cs Gs As Ts Gs
    Ts  Cs  Cs Cs Cs As As As Gs
    35 C11 200 Cs Ts Gs Ts Ts Cs Gs As Cs
    As  Cs  Ts Cs Ts Gs Gs Ts Ts
    36 C12 200 Cs Ts Gs Gs As Cs Cs As As
    Cs  As  Cs Gs Ts Ts Gs Ts Cs
    37 D01 200 Cs Cs Gs Ts Cs Cs Gs Ts Gs
    Ts  Ts  Ts Gs Ts Ts Cs Ts Gs
    38 D02 200 Cs Ts Gs As Cs Ts As Cs As
    As  Cs  As Gs As Cs As Cs Cs
    39 D03 200 Cs As As Cs As Gs As Cs As
    Cs  Cs  As Gs Gs Gs Gs Ts Cs
    40 D04 200 Cs As Gs Gs Gs Gs Ts Cs Cs
    Ts  As  Gs Cs Cs Gs As Cs Ts
    41 D05 200 Cs Ts Cs Ts As Gs Ts Ts As
    As  As  As Gs Gs Gs Cs Ts Gs
    42 D06 200 Cs Ts Gs Cs Ts As Gs As As
    Gs  Gs  As Cs Cs Gs As Gs Gs
    43 D07 200 Cs Ts Gs As As As Ts Gs Ts
    As  Cs  Cs Ts As Cs Gs Gs Ts
    44 D08 200 Cs As Cs Cs Cs Gs Ts Ts Ts
    Gs  Ts  Cs Cs Gs Ts Cs As As
    45 D09 200 Cs Ts Cs Gs As Ts As Cs Gs
    Gs  Gs  Ts Cs As Gs Ts Cs As
    46 D10 200 Gs Gs Ts As Gs Gs Ts Cs Ts
    Ts  Gs  Gs Ts Gs Gs Gs Ts Gs
    47 D11 200 Gs As Cs Ts Ts Ts Gs Cs Cs
    Ts  Ts  As Cs Gs Gs As As Gs
    48 D12 200 Gs Ts Gs Gs As Gs Ts Cs Ts
    Ts  Ts  Gs Ts Cs Ts Gs Ts Gs
    49 E01 200 Gs Gs As Gs Ts Cs Ts Ts Ts
    Gs  Ts  Cs Ts Gs Ts Gs Gs Ts
    50 E02 200 Gs Gs As Cs As Cs Ts Cs Ts
    Cs  Gs  As Cs As Cs As Gs Gs
    51 E03 200 Gs As Cs As Cs As Gs Gs As
    Cs  Gs  Ts Gs Gs Cs Gs As Gs
    52 E04 200 Gs As Gs Ts As Cs Gs As Gs
    Cs  Gs  Gs Gs Cs Cs Gs As As
    53 E05 200 Gs As Cs Ts As Ts Gs Gs Ts
    As  Gs  As Cs Gs Cs Ts Cs Gs
    54 E06 200 Gs As As Gs As Gs Gs Ts Ts
    As  Cs  As Cs As Gs Ts As Gs
    55 E07 200 Gs As Gs Gs Ts Ts As Cs As
    Cs  As  Gs Ts As Gs As Cs Gs
    56 E08 200 Gs Ts Ts Gs Ts Cs Cs Gs Ts
    Cs  Cs  Gs Ts Gs Ts Ts Ts Gs
    57 E09 200 Gs As Cs Ts Cs Ts Cs Gs Gs
    Gs  As  Cs Cs As Cs Cs As Cs
    55 E10 200 Gs Ts As Gs Gs As Gs As As
    Cs  Cs  As Cs Gs As Cs Cs As
    59 E11 200 Gs Gs Ts Ts Cs Ts Ts Cs Gs
    Gs  Ts  Ts Gs Gs Ts Ts As Ts
    60 E12 200 Gs Ts Gs Gs Gs Gs Ts Ts Cs
    Gs  Ts  Cs Cs Ts Ts Gs Gs Gs
    61 F01 200 Gs Ts Cs As Cs Gs Ts Cs Cs
    Ts  Cs  Ts Gs As As As Ts Gs
    62 F02 200 Gs Ts Cs Cs Ts Cs Cs Ts As
    Cs  Cs  Gs Ts Ts Ts Cs Ts Cs
    63 F03 200 Gs Ts Cs Cs Cs Cs As Cs Gs
    Ts  Cs  Cs Gs Ts Cs Ts Ts Cs
    64 F04 200 Ts Cs As Cs Cs As Gs Gs As
    Cs  Gs  Gs Cs Gs Gs As Cs Cs
    65 F05 200 Ts As Cs Cs As As Gs Cs As
    Gs  As  Cs Gs Gs As Gs As Cs
    66 F06 200 Ts Cs Cs Ts Gs Ts Cs Ts Ts
    Ts  Gs  As Cs Cs As Cs Ts Cs
    67 F07 200 Ts Gs Ts Cs Ts Ts Ts Gs As
    Cs  Cs  As Cs Ts Cs As Cs Ts
    68 F08 200 Ts Gs As Cs Cs As Cs Ts Cs
    As  Cs  Ts Gs As Cs Gs Ts Gs
    69 F09 200 Ts Gs As Cs Gs Ts Gs Ts Cs
    Ts  Cs  As As Gs Ts Gs As Cs
    70 F10 200 Ts Cs As As Gs Ts Gs As Cs
    Ts  Ts  Ts Gs Cs Cs Ts Ts As
    71 F11 200 Ts Gs Ts Ts Ts As Ts Gs As
    Cs  Gs  Cs Ts Gs Gs Gs Gs Ts
    72 F12 200 Ts Ts As Ts Gs As Cs Gs Cs
    Ts  Gs  Gs Gs Gs Ts Ts Gs Gs
    73 G01 200 Ts Gs As Cs Gs Cs Ts Gs Gs
    Gs  Gs  Ts Ts Gs Gs As Ts Cs
    74 G02 200 Ts Cs Gs Ts Cs Ts Ts Cs Cs
    Cs  Gs  Ts Gs Gs As Gs Ts Cs
    75 G03 200 Ts Gs Gs Ts As Gs As Cs Gs
    Ts  Gs  Gs As Cs As Cs Ts Ts
    76 G04 200 Ts Ts Cs Ts Ts Cs Cs Gs As
    Cs  Cs  Gs Ts Gs As Cs As Ts
    77 G05 200 Ts Gs Gs Ts As Gs As Cs Gs
    Cs  Ts  Cs Gs Gs Gs As Cs Gs
    78 G06 200 Ts As Gs As Cs Gs Cs Ts Cs
    Gs  Gs  Gs As Cs Gs Gs Gs Ts
    79 G07 200 Ts Ts Ts Ts As Cs As Gs Ts
    Gs  Gs  Gs As As Cs Cs Ts Gs
    80 G08 200 Ts Gs Gs Gs As As Cs Cs Ts
    Gs  Ts  Ts Cs Gs As Cs As Cs
    81 G09 200 Ts Cs Gs Gs Gs As Cs Cs As
    Cs  Cs  As Cs Ts As Gs Gs Gs
    82 G10 200 Ts As Gs Gs As Cs As As As
    Cs  Gs  Gs Ts As Gs Gs As Gs
    83 G11 200 Ts Gs Cs Ts As Gs As As Gs
    Gs  As  Cs Cs Gs As Gs Gs Ts
    84 G12 200 Ts Cs Ts Gs Ts Cs As Cs Ts
    Cs  Cs  Gs As Cs Gs Ts Gs Gs
  • Table 4 is a seq file for oligonucleotides having regions of 2′-O-(methoxyethyl)nucleosides and region of 2′-deoxy nucleosides each linked by phosphorothioate internucleotide linkages. [0254]
    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-(methoxyethy) 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    moeGs
    2 A02 200 moeAs   moeCs   moeGs   moeGs   Cs
    Gs  Gs  As   Cs   Cs   As   Gs   As   Gs   moeTs
    moeGs    moeGs    moeAs
    3 A03 200 moeAs   moeCs   moeCs   moeAs   As
    Gs  Cs  As   Gs   As   Cs   Gs   Gs   As   moeGs
    moeAs    moeCs    moeGs
    4 A04 200 moeAs   moeGs   moeGs   moeAs   Gs
    As  Cs  Cs   Cs   Cs   Gs   As   Cs   Gs   moeAs
    moeAs    moeCs    moeGs
    5 A05 200 moeAs   moeCs   moeCs   moeCs   Cs
    Gs  As  Cs   Gs   As   As   Cs   Gs   As   moeCs
    moeTs    moeGs   moeGs
    6 A06 200 moeAs   moeCs   moeGs   moeAs   As
    Cs  Gs  As   Cs   Ts   Gs   Gs   Cs   Gs   moeAs
    moeCs    moeAs    moeGs
    7 A07 200 moeAs   moeCs   moeGs   moeAs   Cs
    Ts  Gs  Gs   Cs   Gs   As   Cs   As   Gs   moeGs
    moeTs    moeAs    moeGs
    8 A08 200 moeAs   moeCs   moeAs   moeGs   Gs
    Ts  As  Gs   Gs   Ts   Cs   Ts   Ts   Gs   moeGs
    moeTs    moeGs    moeGs
    9 A09 200 moeAs   moeGs   moeGs   moeTs   Cs
    Ts  Ts  Gs   Gs   Ts   Gs   Gs   Gs   Ts   moeGs
    moeAs    moeCs    moeGs
    10 A10 200 moeAs   moeGs   moeTs   moeCs   As
    Cs  Gs  As   Cs   As   As   Gs   As   As   moeAs
    moeCs    moeAs    moeCs
    11 A11 200 moeAs   moeCs   moeGs   moeAs   Cs
    As  As  Gs   As   As   As   Cs   As   Cs   moeGs
    moeGs    moeTs    moeCs
    12 A12 200 moeAs   moeGs   moeAs   moeAs   As
    Cs  As  Cs   Gs   Gs   Ts    Cs   Gs   Gs   moeTs
    moeCs    moeCs    moeTs
    13 B01 200 moeAs   moeAs   moeCs   moeAs   Cs
    Gs  Gs  Ts   Cs   Gs   Gs   Ts   Cs   Cs   moeTs
    moeGs    moeTs    moeCs
    14 B02 200 moeAs   moeCs   moeTs   moeCs   As
    Cs  Ts  Gs   As   Cs   Gs   Ts   Gs   Ts   moeCs
    moeTs    moeCs    moeAs
    15 B03 200 moeAs   moeCs   moeGs   moeGs   As
    As  Gs  Gs   As   As   Cs   Gs   Cs   Cs   moeAs
    moeCs    moeTs    moeTs
    16 B04 200 moeAs   moeTs   moeCs   moeTs   Gs
    Ts  Gs  Gs   As   Cs   Cs   Ts   Ts   Gs   moeTs
    moeCs    moeTs    moeCs
    17 B05 200 moeAs   moeCs   moeAs   moeCs   Ts
    Ts  Cs  Ts   Ts   Cs   Cs   Gs   As   Cs   moeCs
    moeGs    moeTs    moeGs
    18 206 200 moeAs   moeCs   moeTs   moeCs   Ts
    Cs  Gs  As   Cs   As   Cs   As   Gs   Gs   moeAs
    moeCs    moeGs    moeTs
    19 B07 200 moeAs   moeAs   moeAs   moeCs   Cs
    Cs  Cs  As   Gs   Ts   Ts   Cs   Gs   Ts   moeCs
    moeTs    moeAs    moeAs
    20 B08 200 moeAs   moeTs   moeGs   moeTs   Cs
    Cs  Cs  Cs   As   As   As   Gs   As   Cs   moeTs
    moeAs    moeTs    moeGs
    21 B09 200 moeAs   moeCs   moeGs   moeCs   Ts
    Cs  Gs  Gs   Gs   As   Cs   Gs   Gs   Gs   moeTs
    moeCs    moeAs    moeGs
    22 B10 200 moeAs   moeGs   moeCs   moeCs   Gs
    As  As  Gs   As   As   Gs   As   Gs   Gs   moeTs
    moeTs    moeAs    moeCs
    23 B11 200 moeAs   moeCs   moeAs   moeCs   As
    Gs  Ts  As   Gs   As   Cs   Gs   As   As   moeAs
    moeGs    moeCs    moeTs
    24 B12 200 moeAs   moeCs   moeAs   moeCs   Ts
    Cs  Ts  Gs   Gs   Ts   Ts   Ts   Cs   Ts   moeGs
    moeGs    moeAs    moeCs
    25 C01 200 moeAs   moeCs   moeGs   moeAs   Cs
    Cs  As  Gs   As   As   As   Ts   As   Gs   moeTs
    moeTs    moeTs    moeTs
    26 C02 200 moeAs   moeGs   moeTs   moeTs   As
    As  As  As   Gs   Gs   Gs   Cs   Ts   Gs   moeCs
    moeTs    moeAs    moeGs
    27 C03 200 moeAs   moeGs   moeGs   moeTs   Ts
    Gs  Ts  Gs   As   Cs   Gs   As   Cs   Gs   moeAs
    moeGs    moeGs    moeTs
    28 C04 200 moeAs   moeAs   moeTs   moeGs   Ts
    As  Cs  Cs   Ts   As   Cs   Gs   Gs   Ts   moeTs
    moeGs    moeGs    moeCs
    29 C05 200 moeAs   moeGs   moeTs   moeCs   As
    Cs  Gs  Ts   Cs   Cs   Ts   Cs   Ts   Cs   moeTs
    moeGs    moeTs    moeCs
    30 C06 200 moeCs   moeTs   moeGs   moeGs   Cs
    Gs  As  Cs   As   Gs   Gs   Ts   As   Gs   moeGs
    moeTs    moeCs    moeTs
    31 C07 200 moeCs   moeTs   moeCs   moeTs   Gs
    Ts  Gs  Ts   Gs   As   Cs   Gs   Gs   Ts   moeGs
    moeGs    moeTs    moeCs
    32 C08 200 moeCs   moeAs   moeGs   moeGs   Ts
    Cs  Gs  Ts   Cs   Ts   Ts   Cs   Cs   Cs   moeGs
    moeTs    moeGs    moeGs
    33 C09 200 moeCs   moeTs   moeGs   moeTs   Gs
    Gs  Ts  As   Gs   As   Cs   Gs   Ts   Gs   moeGs
    moeAs    moeCs    moeAs
    34 C10 200 moeCs   moeTs   moeAs   moeAs   Cs
    Gs  As  Ts   Gs   Ts   Cs   Cs   Cs   Cs   moeAs
    moeAs    moeAs    moeGs
    35 C11 200 moeCs   moeTs   moeGs   moeTs   Ts
    Cs  Gs  As   Cs   As   Cs   Ts   Cs   Ts   moeGs
    moeGs    moeTs    moeTs
    36 C12 200 moeCs   moeTs   moeGs   moeGs   As
    Cs  Cs  As   As   Cs   As   Cs   Gs   Ts   moeTs
    moeGs    moeTs    moeCs
    37 D01 200 moeCs   moeCs   moeGs   moeTs   Cs
    Cs  Gs  Ts   Gs   Ts   Ts   Ts   Gs   Ts   moeTs
    moeCs    moeTs    moeGs
    38 D02 200 moeCs   moeTs   moeGs   moeAs   Cs
    Ts  As  Cs   As   As   Cs   As   Gs   As   moeCs
    moeAs    moeCs    moeCs
    39 D03 200 moeCs   moeAs   moeAs   moeCs   As
    Gs  As  Cs   As   Cs   Cs   As   Gs   Gs   moeGs
    moeGs    moeTs    moeCs
    40 D04 200 moeCs   moeAs   moeGs   moeGs   Gs
    Gs  Ts  Cs   Cs   Ts   As   Gs   Cs   Cs   moeGs
    moeAs    moeCs    moeTs
    41 D05 200 moeCs   moeTs   moeCs   moeTs   As
    Gs  Ts  Ts   As   As   As   As   Gs   Gs   moeGs
    moeCs    moeTs    moeGs
    42 D06 200 moeCs   moeTs   moeGs   moeCs   Ts
    As  Gs  As   As   Gs   Gs   As   Cs   Cs   moeGs
    moeAs    moeGs    moeGs
    43 D07 200 moeCs   moeTs   moeGs   moeAs   As
    As  Ts  Gs   Ts   As   Cs   Cs   Ts   As   moeCs
    moeGs    moeGs    moeTs
    44 D08 200 moeCs   moeAs   moeCs   moeCs   Cs
    Gs  Ts  Ts   Ts   Gs   Ts   Cs   Cs   Gs   moeTs
    moeCs    moeAs    moeAs
    45 D09 200 moeCs   moeTs   moeCs   moeGs   As
    Ts  As  Cs   Gs   Gs   Gs   Ts   Cs   As   moeGs
    moeTs    moeCs    moeAs
    46 D10 200 moeGs   moeGs   moeTs   moeAs   Gs
    Gs  Ts  Cs   Ts   Ts   Gs   Gs   Ts   Gs   moeGs
    moeGs    moeTs    moeGs
    47 D11 200 moeGs   moeAs   moeCs   moeTs   Ts
    Ts  Gs  Cs   Cs   Ts   Ts   As   Cs   Gs   moeGs
    moeAs    moeAs    moeGs
    48 D12 200 moeGs   moeTs   moeGs   moeGs   As
    Gs  Ts  Cs   Ts   Ts   Ts   Gs   Ts   Cs   moeTs
    moeGs    moeTs    moeGs
    49 E01 200 moeGs   moeGs   moeAs   moeGs   Ts
    Cs  Ts  Ts   Ts   Gs   Ts   Cs   Ts   Gs   moeTs
    moeGs    moeGs    moeTs
    50 E02 200 moeGs   moeGs   moeAs   moeCs   As
    Cs  Ts  Cs   Ts   Cs   Gs   As   Cs   As   moeCs
    moeAs    moeGs    moeGs
    51 E03 200 moeGs   moeAs   moeCs   moeAs   Cs
    As  Gs  Gs   As   Cs   Gs   Ts   Gs   Gs   moeCs
    moeGs    moeAs    moeGs
    52 E04 200 moeGs   moeAs   moeGs   moeTs   As
    Cs  Gs  As   Gs   Cs   Gs   Gs   Gs   Cs   moeCs
    moeGs    moeAs    moeAs
    53 E05 200 moeGs   moeAs   moeCs   moeTs   As
    Ts  Gs  Gs   Ts   As   Gs   As   Cs   Gs   moeCs
    moeTs    moeCs    moeGs
    54 E06 200 moeGs   moeAs   moeAs   moeGs   As
    Gs  Gs  Ts   Ts   As   Cs   As   Cs   As   moeGs
    moeTs    moeAs    moeGs
    55 E07 200 moeGs   moeAs   moeGs   moeGs   Ts
    Ts  As  Cs   As   Cs   As   Gs   Ts   As   moeGs
    moeAs    moeCs    moeGs
    56 E08 200 moeGs   moeTs   moeTs   moeGs   Ts
    Cs  Cs  Gs   Ts   Cs   Cs   Gs   Ts   Gs   moeTs
    moeTs    moeTs    moeGs
    57 E09 200 moeGs   moeAs   moeCs   moeTs   Cs
    Ts  Cs  Gs   Gs   Gs   As   Cs   Cs   As   moeCs
    moeCs    moeAs    moeCs
    58 E10 200 moeGs   moeTs   moeAs   moeGs   Gs
    As  Gs  As   As   Cs   Cs   As   Cs   Gs   moeAs
    moeCs    moeCs    moeAs
    59 E11 200 moeGs   moeGs   moeTs   moeTs   Cs
    Ts  Ts  Cs   Gs   Gs   Ts   Ts   Gs   Gs   moeTs
    moeTs    moeAs    moeTs
    60 E12 200 moeGs   moeTs   moeGs   moeGs   Gs
    Gs  Ts  Ts   Cs   Gs   Ts   Cs   Cs   Ts   moeTs
    moeGs    moeGs    moeGs
    61 F01 200 moeGs   moeTs   moeCs   moeAs   Cs
    Gs  Ts  Cs   Cs   Ts   Cs   Ts   Gs   As   moeAs
    moeAs    moeTs    moeGs
    62 F02 200 moeGs   moeTs   moeCs   moeCs   Ts
    Cs  Cs  Ts   As   Cs   Cs   Gs   Ts   Ts   moeTs
    moeCs    moeTs    moeCs
    63 F03 200 moeGs   moeTs   moeCs   moeCs   Cs
    Cs  As  Cs   Gs   Ts   Cs   Cs   Gs   Ts   moeCs
    moeTs    moeTs    moeCs
    64 F04 200 moeTs   moeCs   moeAs   moeCs   Cs
    As  Gs  Gs   As   Cs   Gs   Gs   Cs   Gs   moeGs
    moeAs    moeCs    moeCs
    65 F05 200 moeTs   moeAs   moeCs   moeCs   As
    As  Gs  Cs   As   Gs   As   Cs   Gs   Gs   moeAs
    moeGs    moeAs    moeCs
    66 F06 200 moeTs   moeCs   moeCs   moeTs   Gs
    Ts  Cs  Ts   Ts   Ts   Gs   As   Cs   Cs   moeAs
    moeCs    moeTs    moeCs
    67 F07 200 moeTs   moeGs   moeTs   moeCs   Ts
    Ts  Ts  Gs   As   Cs   Cs   As   Cs   Ts   moeCs
    moeAs    moeCs    moeTs
    68 F08 200 moeTs   moeGs   moeAs   moeCs   Cs
    As  Cs  Ts   Cs   As   Cs   Ts   Gs   As   moeCs
    moeGs    moeTs    moeGs
    69 F09 200 moeTs   moeGs   moeAs   moeCs   Gs
    Ts  Gs  Ts   Cs   Ts   Cs   As   As   Gs   moeTs
    moeGs    moeAs    moeCs
    70 F10 200 moeTs   moeCs   moeAs   moeAs   Gs
    Ts  Gs  As   Cs   Ts   Ts   Ts   Gs   Cs   moeCs
    moeTs    moeTs    moeAs
    71 F11 200 moeTs   moeGs   moeTs   moeTs   Ts
    As  Ts  Gs   As   Cs   Gs   Cs   Ts   Gs   moeGs
    moeGs    moeGs    moeTs
    72 F12 200 moeTs   moeTs   moeAs   moeTs   Gs
    As  Cs  Gs   Cs   Ts   Gs   Gs   Gs   Gs   moeTs
    moeTs    moeGs    moeGs
    73 G01 200 moeTs   moeGs   moeAs   moeCs   Gs
    Cs  Ts  Gs   Gs   Gs   Gs   Ts   Ts   Gs   moeGs
    moeAs    moeTs    moeCs
    74 G02 200 moeTs   moeCs   moeGs   moeTs   Cs
    Ts  Ts  Cs   Cs   Cs   Gs   Ts   Gs   Gs   moeAs
    moeGs    moeTs    moeCs
    75 G03 200 moeTs   moeGs   moeGs   moeTs   As
    Gs  As  Cs   Gs   Ts   Gs   Gs   As   Cs   moeAs
    moeCs    moeTs    moeTs
    76 G04 200 moeTs   moeTs   moeCs   moeTs   Ts
    Cs  Cs  Gs   As   Cs   Cs   Gs   Ts   Gs   moeAs
    moeCs    moeAs    moeTs
    77 G05 200 moeTs   moeGs   moeGs   moeTs   As
    Gs  As  Cs   Gs   Cs   Ts   Cs   Gs   Gs   moeGs
    moeAs    moeCs    moeGs
    78 G06 200 moeTs   moeAs   moeGs   moeAs   Cs
    Gs  Cs  Ts   Cs   Gs   Gs   Gs   As   Cs   moeGs
    moeGs    moeGs    moeTs
    79 G07 200 moeTs   moeTs   moeTs   moeTs   As
    Cs  As  Gs   Ts   Gs   Gs   Gs   As   As   moeCs
    moeCs    moeTs    moeGs
    80 G08 200 moeTs   moeGs   moeGs   moeGs   As
    As  Cs  Cs   Ts   Gs   Ts   Ts   Cs   Gs   moeAs
    moeCs    moeAs    moeCs
    81 G09 200 moeTs   moeCs   moeGs   moeGs   Gs
    As  Cs  Cs   As   Cs   Cs   As   Cs   Ts   moeAs
    moeGs    moeGs    moeGs
    82 G10 200 moeTs   moeAs   moeGs   moeGs   As
    Cs  As  As   As   Cs   Gs   Gs   Ts   As   moeGs
    moeGs    moeAs    moeGs
    83 G11 200 moeTs   moeGs   moeCs   moeTs   As
    Gs  As  As   Gs   Gs   As   Cs   CsGs   moeAs
    moeGs    moeGs    moeTs
    84 G12 200 moeTs   moeCs   moeTs   moeGs   Ts
    Cs  As  Cs   Ts   Cs   Cs   Gs   As   Cs   moeGs
    moeTs    moeGs    moeGs
  • Reagent File (.tab File) [0255]
  • Table 5 is a tab for reagents necessary for synthesizing an oligonucleotides having both 2′-O-(methoxy-ethy)nucleosides and 2′-deoxy nucleosides located therein. [0256]
    TABLE 5
    Identity of columns: GroupName, Bottle ID, ReagentName,
    FlowRate, Concentration. Wherein reagent name is
    identified using base identifier, ‘moe’ indicated a 2′-O-
    (methoxyethy) 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
     0 moeG moeGcpg 100 1
     0 moe5meC moe5meCcpg 100 1
     0 moeA moeAcpg 100 1
     0 moeT moeTcpg 100 1
    END
    DEBLOCK BEGIN
    70 TCA TCA 100 1
    END
    WASH BEGIN
    65 ACN ACN 190 1
    END
    OXIDIZERS BEGIN
    68 BEAU BEAUCAGE 320 1
    END
    CAPPING BEGIN
    66 CAP_B CAP_B 220 1
    67 CAP_A CAP_A 230 1
    END
    DEOXY THIOATE BEGIN
    31, 32 Gs deoxyG 270 1
    39, 40 5meCs 5methyldeoxyC 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 moe5meCs 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 multi 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-[0257] one 1,1 dioxide (Beaucage Reagent) in anhydrous acetonitrile. Standard base-protected beta-cyanoethyldiisopropyl 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.
  • Oligonucleotides were cleaved from support and deprotected with concentrated NH[0258] 4OH 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 oligo-nucleotides are alternately synthesized on an automated DNA synthesizer (Applied Biosystems model 380B) using standard phosphoramidite chemistry with oxidation by iodine. [0259]
  • 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-[0260] 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 NaCl solution.
  • Phosphinate oligonucleotides are prepared as described in U.S. Pat. No. 5,508,270, herein incorporated by reference. [0261]
  • Alkyl phosphonate oligonucleotides are prepared as described in U.S. Pat. No. 4,469,863, herein incorporated by reference. [0262]
  • 3′-Deoxy-3′-methylene phosphonate oligonucleotides are prepared as described in U.S. Pat. Nos. 5,610,289 or 5,625,050, herein incorporated by reference. [0263]
  • Phosphoramidite oligonucleotides are prepared as described in U.S. Pat. No. 5,256,775 or U.S. Pat. No. 5,366,878, hereby incorporated by reference. [0264]
  • Alkylphosphonothioate oligonucleotides are prepared as described in published PCT applications PCT/US94/00902 and PCT/US93/06976 (published as WO 94/17093 and WO 94/02499, respectively). [0265]
  • 3′,-Deoxy-3′-amino phosphoramidate oligonucleotides are prepared as described in U.S. Pat. No. 5,476,925, herein incorporated by reference. [0266]
  • Phosphotriester oligonucleotides are prepared as described in U.S. Pat. No. 5,023,243, herein incorporated by reference. [0267]
  • Boranophosphate oligonucleotides are prepared as described in U.S. Pat. Nos. 5,130,302 and 5,177,198, both herein incorporated by reference. [0268]
  • Methylenemethylimino linked oligonucleosides, also identified as MMI linked oligonucleosides, methylenedi-methylhydrazo 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 oligonucleo-sides, as well as mixed backbone compounds having, for instance, alternating MMI and PO or PS linkages are prepared as described in U.S. Pat. Nos. 5,378,825; 5,386,023; 5,489,677; 5,602,240 and 5,610,289, all of which are herein incorporated by reference. [0269]
  • Formacetal and thioformacetal linked oligonucleosides are prepared as described in U.S. Pat. Nos. 5,264,562 and 5,264,564, herein incorporated by reference. [0270]
  • Ethylene oxide linked oligonucleosides are prepared as described in U.S. Pat. No. 5,223,618, herein incorporated by reference. [0271]
  • 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. Pat. Nos. 5,539,082; 5,700,922, and 5,719,262, herein incorporated by reference. [0272]
  • 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.’[0273]
  • A. [2′-O-Me]—[2′-deoxy]—[2′-O-Me] Chimeric Phosphorothioate Oligonucleotides [0274]
  • Chimeric oligonucleotides having 2′-0-alkyl phosphorothioate and 2′-deoxy phosphorothioate oligo-nucleotide segments are synthesized using 2′-deoxy-5′-dimethoxytrityl-3′-O-phosphoramidites for the DNA portion and 5′-dimethoxytrityl-2′-O-methyl-3′-0-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. [0275]
  • B. [2′-O-(2-Methoxyethyl)]—[2′-deoxy]—[2′-O-(Methoxyethyl)] Chimeric Phosphorothioate Oligonucleotides [0276]
  • [2′-O-(2-methoxyethyl)]—[2′-deoxy]—[-2′-O-(methoxy-ethyl)] chimeric phosphorothioate oligonucleotides are prepared as per the procedure above for the 2′-O-methyl chimeric oligonucleotide, with the substitution of 2′-O-(methoxyethyl) amidites for the 2′-O-methyl amidites. [0277]
  • C. [2′-O-(2-Methoxyethyl)Phosphodiester]—[2′-deoxy Phosphorothioate]—[2′-O-(2-Methoxyethyl) Phosphodiester] Chimeric Oligonucleotide [0278]
  • [2′-0-(2-methoxyethyl phosphodiester]—[2′-deoxy phosphorothioate]—[2′-O-(methoxyethyl) phosphodiester] chimeric oligonucleotides are prepared as per the above procedure for the 2′-O-methyl chimeric oligonucleotide with the substitution of 2′-O-(methoxyethyl) amidites for the 2′-O-methyl amidites in the wing portions. Sulfurization utilizing 3, H-1,2 benzodithiole-3-[0279] 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 oligo-nucleosides and mixed chimeric oligonucleotides/oligo-nucleosides are synthesized according to U.S. Pat. No. 5,623,065, herein incorporated by reference. [0280]
  • 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 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-(methoxyethyl)nucleotides on either side of a center gap region of 2′-deoxy nucleotides. 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. [0281]
  • For illustrative purposes Tables 6-a and 6-b show the sequences of an intial first library, i.e., a library of phosphoroethioate oligonucleotides targeted to a CD40 target. The compounds of Table 6-a shows the members of this listed in compliance with the established rule for listing SEQ ID NO:, i.e., in numerical SEQ ID NO: order. [0282]
    TABLE 6-a
    Sequences of Oligonucleotides Targeted to CD40 by
    SEQ ID NO.:
    SEQ ID
    NUCLEOBASE SEQUENCE NO.
    CCAGGCGGCAGGACCACT 1
    GACCAGGCGGCAGGACCA 2
    AGGTGAGACCAGGCGGCA 3
    CAGAGGCAGACGAACCAT 4
    GCAGAGGCAGACGAACCA 5
    GCAAGCAGCCCCAGAGGA 6
    GGTCAGCAAGCAGCcCCA 7
    GACAGCGGTCAGCAAGCA 8
    GATGGACAGCGGTCAGCA 9
    TCTGGATGGACAGCGGTC 10
    GGTGGTTCTGGATGGACA 11
    GTGGGTGGTTCTGGATGG 12
    GCAGTGGGTGGTTCTGGA 13
    CACAAAGAACAGCACTGA 14
    CTGGCACAAAGAACAGCA 15
    TCCTGGCTGGCACAAAGA 16
    CTGTCCTGGCTGGCACAA 17
    CTCACCAGTTTCTGTCCT 18
    TCACTCACCAGTTTCTGT 19
    GTGCAGTCACTCACCAGT 20
    ACTCTGTGCAGTCACTCA 21
    CAGTGAACTCTGTGCAGT 22
    ATTCCGTTTCAGTGAACT 23
    GAAGGCATTCCGTTTCAG 24
    TTCACCGCAAGGAAGGCA 25
    CTCTGTTCCAGGTGTCTA 26
    CTGGTGGCAGTGTGTCTC 27
    TGGGGTCGCAGTATTTGT 28
    GGTTGGGGTCGCAGTATT 29
    CTAGGTTGGGGTCGCAGT 30
    GGTGCCCTTCTGCTGGAC 31
    CTGAGGTGCCCTTCTGCT 32
    GTGTCTGTTTCTGAGGTG 33
    TGGTGTCTGTTTCTGAGG 34
    ACAGGTGCAGATGGTGTC 35
    TTCACAGGTGCAGATGGT 36
    GTGCCAGCCTTCTTCACA 37
    TACAGTGCCAGCCTTCTT 38
    GGACACAGCTCTCACAGG 39
    TGCAGGACACAGCTCTCA 40
    GAGCGGTGCAGGACACAG 41
    AAGCCGGGCGAGCATGAG 42
    AATCTGCTTGACCCCAAA 43
    GAAACCCCTGTAGCAATC 44
    GTATCAGAAACCCCTGTA 45
    GCTCGCAGATGGTATCAG 46
    GCAGGGCTCGCAGATGGT 47
    TGGGCAGGGCTCGCAGAT 48
    GACTGGGCAGGGCTCGCA 49
    CATTGGAGAAGAAGCCGA 50
    GATGACACATTGGAGAAG 51
    GCAGATGACACATTGGAG 52
    TCGAAAGCAGATGACACA 53
    GTCCAAGGGTGACATTTT 54
    CACAGCTTGTCCAAGGGT 55
    TTGGTCTCACAGCTTGTC 56
    CAGGTCTTTGGTCTCACA 57
    CTGTTGCACAACCAGGTC 58
    GTTTGTGCCTGCCTGTTG 59
    GTCTTGTTTGTGCCTGCC 60
    CCACAGACAACATCAGTC 61
    CTGGGGACCACAGACAAC 62
    TCAGCCGATCCTGGGGAC 63
    CACCACCAGGGCTCTCAG 64
    GGGATCACCACCAGGGCT 65
    GAGGATGGCAAACAGGAT 66
    ACCAGCACCAAGAGGATG 67
    TTTTGATAAAGACCAGCA 68
    TATTGGTTGGCTTCTTGG 69
    GGGTTCCTGCTTGGGGTG 70
    GTCGGGAAAATTGATCTC 71
    GATCGTCGGGAAAATTGA 72
    GGAGCCAGGAAGATCGTC 73
    TGGAGCCAGGAAGATCGT 74
    TGGAGCAGCAGTGTTGGA 75
    GTAAAGTCTCCTGCACTG 76
    TGGCATCCATGTAAAGTC 77
    CGGTTGGCATCCATGTAA 78
    CTCTTTGCCATCCTCCTG 79
    CTGTCTCTCCTGCACTGA 80
    GGTGCAGCCTCACTGTCT 81
    AACTGCCTGTTTGCCCAC 82
    CTTCTGCCTGCACCCCTG 83
    ACTGACTGGGCATAGCTC 84
  • 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. [0283]
  • 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, [0284] column 8.
  • In Table 6-b below, the oligonucleotide 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. [0285]
    TABLE 6-b
    Sequences of Oligonucleotides Targeted to CD40
    Order by Synthesis Well No.
    Well SEQ ID
    A01 GACCAGGCGGCAGGACCA 2
    A02 AGGTGAGACCAGGCGGCA 3
    A03 GCAGAGGCAGACGAACCA 5
    A04 GCAAGCAGCCCCAGAGGA 6
    A05 GGTCAGCAAGCAGCCCCA 7
    A06 GACAGCGGTCAGCAAGCA 8
    A07 GATGGACAGCGGTCAGCA 9
    A08 GGTGGTTCTGGATGGACA 11
    A09 GCAGTGGGTGGTTCTGGA 13
    A10 CACAAAGAACAGCACTGA 14
    A11 CTGGCACAAAGAACAGCA 15
    A12 TCCTGGCTGGCACAAAGA 16
    B01 CTGTCCTGGCTGGCAcAA 17
    B02 ACTCTGTGCAGTCACTCA 21
    B03 TTCACCGCAAGGAAGGCA 25
    B04 CTCTGTTCCAGGTGTCTA 26
    B05 GTGCCAGCCTTCTTCACA 37
    B06 TGCAGGACACAGCTCTCA 40
    B07 AATCTGCTTGACCCCAAA 43
    B08 GTATCAGAAACCCCTGTA 45
    B09 GACTGGGCAGGGCTCGCA 49
    B10 CATTGGAGAAGAAGCCGA 50
    B11 TCGAAAGCAGATGACACA 53
    B12 CAGGTCTTTGGTCTCACA 57
    C01 TTTTGATAAAGACCAGCA 68
    C02 GATCGTCGGGAAAATTGA 72
    C03 TGGAGCAGCAGTGTTGGA 75
    C04 CGGTTGGCATCCATGTAA 78
    C05 CTGTCTCTCCTGCACTGA 80
    C06 TCTGGATGGACAGCGGTC 10
    C07 CTGGTGGCAGTGTGTCTC 27
    C08 GGTGCCCTTCTGCTGGAC 31
    C09 ACAGGTGCAGATGGTGTC 35
    C10 GAAACCCCTGTAGCAATC 44
    C11 TTGGTCTCACAGCTTGTC 56
    C12 CTGTTGCACAACCAGGTC 58
    D01 GTCTTGTTTGTGCCTGCC 60
    D02 CCACAGACAACATCAGTC 61
    D03 CTGGGGACCACAGACAAC 62
    D04 TCAGCCGATCCTGGGGAC 63
    D05 GTCGGGAAAATTGATCTC 71
    D06 GGAGCCAGGAAGATCGTC 73
    D07 TGGCATCCATGTAAAGTC 77
    D08 AACTGCCTGTTTGCCCAC 82
    D09 ACTGACTGGGCATAGCTC 84
    D10 GTGGGTGGTTCTGGATGG 12
    D11 GAAGGCATTCCGTTTCAG 24
    D12 GTGTCTGTTTCTGAGGTG 33
    E01 TGGTGTCTGTTTCTGAGG 34
    E02 GGACACAGCTCTCACAGG 39
    E03 GAGCGGTGCAGGACACAG 41
    E04 AAGCCGGGCGAGCATGAG 42
    E05 GCTCGCAGATGGTATCAG 46
    E06 GATGACACATTGGAGAAG 51
    E07 GCAGATGACACATTGGAG 52
    E08 GTTTGTGCCTGCCTGTTG 59
    E09 CACCACCAGGGCTCTCAG 64
    E10 ACCAGCACCAAGAGGATG 67
    E11 TATTGGTTGGCTTCTTGG 69
    E12 GGGTTCCTGCTTGGGGTG 70
    F01 GTAAAGTCTCCTGCACTG 76
    F02 CTCTTTGCCATCCTCCTG 79
    F03 CTTCTGCCTGCACCCCTG 83
    F04 CCAGGCGGCAGGACCACT 1
    F05 CAGAGGCAGACGAACCAT 4
    F06 CTCACCAGTTTCTGTCCT 18
    F07 TCACTCACCAGTTTCTGT 19
    F08 GTGCAGTCACTCACCAGT 20
    F09 CAGTGAACTCTGTGCAGT 22
    F10 ATTCCGTTTCAGTGAACT 23
    F11 TGGGGTCGCAGTATTTGT 28
    F12 GGTTGGGGTCGCAGTATT 29
    G01 CTAGGTTGGGGTCGCAGT 30
    G02 CTGAGGTGCCCTTCTGCT 32
    G03 TTCACAGGTGCAGATGGT 36
    G04 TACAGTGCCAGCCTTCTT 38
    G05 GCAGGGCTCGCAGATGGT 47
    G06 TGGGCAGGGCTCGCAGAT 48
    G07 GTCCAAGGGTGACATTTT 54
    G08 CACAGCTTGTCCAAGGGT 55
    G09 GGGATCACCACCAGGGCT 65
    G10 GAGGATGGCAAACAGGAT 66
    G11 TGGAGCCAGGAAGATCGT 74
    G12 GGTGCAGCCTCACTGTCT 81
  • Example 9 Oligonucleotide Analysis
  • A. Oligonucleotide Analysis—96 Well Plate Format [0286]
  • 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 multi-channel robotic pipettors. [0287]
  • B. Alternative Oligonucleotide Analysis [0288]
  • 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 NaCl with 2.5 volumes ethanol. Synthesized oligonucleotides are analyzed by polyacrylamide gel electrophoresis on denaturing gels. Oligonucleotide purity is checked by [0289] 31P nuclear 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 Oligonucleotides
  • A. Poly(A)+ mRNA Isolation. [0290]
  • Poly(A)+ mRNA was isolated according to Miura et al. ([0291] 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 μl cold PBS. 60 μl lysis buffer (10 mM Tris-HCl, pH 7.6, 1 mM EDTA, 0.5 M NaCl, 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 μl of lysate was transferred to Oligo d(T) coated 96 well plates (AGCT Inc., Irvine, Calif.). Plates were incubated for 60 minutes at room temperature, washed 3 times with 200 μl of wash buffer (10 mM Tris-HCl pH 7.6, 1 mM EDTA, 0.3 M NaCl). After the final wash, the plate was blotted on paper towels to remove excess wash buffer and then air-dried for 5 minutes. 60 μl 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. RT-PCR Analysis of CD40 mRNA Levels [0292]
  • Quantitation of CD40 mRNA levels was determined by reverse transcriptase polymerase chain reaction (RT-PCR) using the ABI PRISM™[0293] 7700 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions. This is a closed-tube, non-gel-based, fluorescence detection system which allows high-throughput quantitation of polymerase 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, Calif.) is attached to the 5′ end of the probe and a quencher dye (e.g., TAMRA, PE-Applied Biosystems, Foster City, Calif.) 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 polymerase. During the extension phase of the PCR amplification cycle, cleavage of the probe by Taq polymerase releases the reporter dye from the remainder of the probe (and hence from the quencher moiety) and a sequence-specific fluorescent signal is generated. [0294]
  • 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 PRISM™[0295] 7700 Sequence Detection System. In each assay, a series of parallel reactions containing serial dilutions of mRNA from 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, Calif. RT-PCR reactions were carried out by adding 25 μl PCR cocktail (1× Taqman™ buffer A, 5.5 mM MgCl[0296] 2, 300 uM each of DATP, dCTP and dGTP, 600 uM of dUTP, 100 nM each of forward primer, reverse primer, and probe, 20 U RNAse inhibitor, 1.25 units AmpliTaq Gold™, and 12.5 U MuLV reverse transcriptase) to 96 well plates containing 25 μl poly(A) mRNA solution. The RT reaction was carried out by incubation for 30 minutes at 48° C. following a 10 minute incubation at 95° C. to activate the AmpliTaq Gold™, 40 cycles of a two-step PCR protocol were carried out: 95° C. for 15 seconds (denaturation) followed by 60° C. for 1.5 minutes (annealing/extension).
  • For CD40, the PCR primers were: [0297]
  • forward primer: [0298]
    forward primer:
    CAGAGTTCACTGAAACGGAATGC (SEQ ID NO:86)
    reverse primer:
    GGTGGCAGTGTGTCTCTCTGTTC (SEQ ID NO:87)
  • reverse primer: [0299]
  • and the PCR probe was: FAM-TTCCTTGCGGTGAAAGCGAATTCCT-TAMRA [0300]
  • (SEQ ID NO:88) where FAM (PE-Applied Biosystems, Foster City, Calif.) is the fluorescent reporter dye and TAMRA (PE-Applied Biosystems, Foster City, Calif.) is the quencher dye. [0301]
  • For GAPDH the PCR primers were: [0302]
  • forward primer: [0303]
  • reverse primer: [0304]
    GAAGGTGAAGGTCGGAGTC (SEQ ID NO:89)
    GAAGATGGTGATGGGATTTC, (SEQ ID NO:90)
  • and the PCR probe was: 5′ JOE-CAAGCTTCCCGTTCTCAGCC-[0305] TAMRA 3′ (SEQ ID No. 91) where JOE (PE-Applied Biosystems, Foster City, Calif.) is the fluorescent reporter dye and TAMRA (PE-Applied Biosystems, Foster City, Calif.) 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 CD40 mRNA, using published sequences (GenBank accession number X60592, incorporated herein 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. [0306]
    TABLE 7
    Inhibition of CD40 mRNA Levels by Phosphorothioate
    Oligodeoxynucleotides
    TARGET SEQ ID
    ISIS# SITE SEQUENCE % INHIB. NO.
    18623 18 CCAGGCGGCAGGACCACT 30.71 1
    18624 20 GACCAGGCGGCAGGACCA 28.09 2
    18625 26 AGGTGAGACCAGGCGGCA 21.89 3
    18626 48 CAGAGGCAGACGAACCAT 0.00 4
    18627 49 GCAGAGGCAGACGAACCA 0.00 5
    18628 73 GCAAGCAGCCCCAGAGGA 0.00 6
    18629 78 GGTCAGCAAGCAGCCCCA 29.96 7
    18630 84 GACAGCGGTCAGCAAGCA 0.00 8
    18631 88 GATGGACAGCGGTCAGCA 0.00 9
    18632 92 TCTGGATGGACAGCGGTC 0.00 10
    18633 98 GGTGGTTCTGGATGGACA 0.00 11
    18634 101 GTGGGTGGTTCTGGATGG 0.00 12
    18635 104 GCAGTGGGTGGTTCTGGA 0.00 13
    18636 152 CACAAAGAACAGCACTGA 0.00 14
    18637 156 CTGGCACAAAGAACAGCA 0.00 15
    18638 162 TCCTGGCTGGCACAAAGA 0.00 16
    18639 165 CTGTCCTGGCTGGCACAA 4.99 17
    18640 176 CTCACCAGTTTCTGTCCT 0.00 18
    18641 179 TCACTCACCAGTTTCTGT 0.00 19
    18642 185 GTGCAGTCACTCACCAGT 0.00 20
    18643 190 ACTCTGTGCAGTCACTcA 0.00 21
    18644 196 CAGTGAACTCTGTGCAGT 5.30 22
    18645 205 ATTCCGTTTCAGTGAACT 0.00 23
    18646 211 GAAGGCATTCCGTTTCAG 9.00 24
    18647 222 TTCACCGCAAGGAAGGCA 0.00 25
    18648 250 CTCTGTTCCAGGTGTCTA 0.00 26
    18649 267 CTGGTGGCAGTGTGTCTC 0.00 27
    18650 286 TGGGGTCGCAGTATTTGT 0.00 28
    18651 289 GGTTGGGGTCGCAGTATT 0.00 29
    18652 292 CTAGGTTGGGGTCGCAGT 0.00 30
    18653 318 GGTGCCCTTCTGCTGGAC 19.67 31
    18654 322 CTGAGGTGCCCTTCTGCT 15.63 32
    18655 332 GTGTCTGTTTCTGAGGTG 0.00 33
    18656 334 TGGTGTCTGTTTCTGAGG 0.00 34
    18657 345 ACAGGTGCAGATGGTGTC 0.00 35
    18658 348 TTCACAGGTGCAGATGGT 0.00 36
    18659 360 GTGCCAGCCTTCTTCACA 5.67 37
    18660 364 TACAGTGCCAGCCTTCTT 7.80 38
    18661 391 GGACACAGCTCTCACAGG 0.00 39
    18662 395 TGCAGGACACAGCTCTCA 0.00 40
    18663 401 GAGCGGTGCAGGACACAG 0.00 41
    18664 416 AAGCCGGGCGAGCATGAG 0.00 42
    18665 432 AATCTGCTTGACCCCAAA 5.59 43
    18666 446 GAAACCCCTGTAGCAATC 0.10 44
    18667 452 GTATCAGAAACCCCTGTA 0.00 45
    18668 463 GCTCGCAGATGGTATCAG 0.00 46
    18669 468 GCAGGGCTCGCAGATGGT 34.05 47
    18670 471 TGGGCAGGGCTCGCAGAT 0.00 48
    18671 474 GACTGGGCAGGGCTCGCA 2.71 49
    18672 490 CATTGGAGAAGAAGCCGA 0.00 50
    18673 497 GATGACACATTGGAGAAG 0.00 51
    18674 500 GCAGATGACACATTGGAG 0.00 52
    18675 506 TCGAAAGCAGATGACACA 0.00 53
    1.8676 524 GTCCAAGGGTGACATTTT 8.01 54
    18677 532 CACAGCTTGTCCAAGGGT 0.00 55
    18678 539 TTGGTCTCACAGCTTGTC 0.00 56
    18679 546 CAGGTCTTTGGTCTCACA 6.98 57
    18680 558 CTGTTGCACAACCAGGTC 18.76 58
    18681 570 GTTTGTGCCTGCCTGTTG 2.43 59
    18682 575 GTCTTGTTTGTGCCTGCC 0.00 60
    18683 590 CCACAGACAACATCAGTC 0.00 61
    18684 597 CTGGGGACCACAGACAAC 0.00 62
    18685 607 TCAGCCGATCCTGGGGAC 0.00 63
    18686 621 CACCACCAGGGCTCTCAG 23.31 64
    18687 626 GGGATCACCACCAGGGCT 0.00 65
    18688 657 GAGGATGGCAAACAGGAT 0.00 66
    18689 668 ACCAGCACCAAGAGGATG 0.00 67
    18690 679 TTTTGATAAAGACCAGCA 0.00 68
    18691 703 TATTGGTTGGCTTCTTGG 0.00 69
    18692 729 GGGTTCCTGCTTGGGGTG 0.00 70
    18693 750 GTCGGGAAAATTGATCTC 0.00 71
    18694 754 GATCGTCGGGAAAATTGA 0.00 72
    18695 765 GGAGCCAGGAAGATCGTC 0.00 73
    18696 766 TGGAGCCAGGAAGATCGT 0.00 74
    18697 780 TGGAGCAGCAGTGTTGGA 0.00 75
    18698 796 GTAAAGTCTCCTGCACTG 0.00 76
    18699 806 TGGCATCCATGTAAAGTC 0.00 77
    18700 810 CGGTTGGCATCCATGTAA 0.00 78
    18701 834 CTCTTTGCCATCCTCCTG 4.38 79
    18702 861 CTGTCTCTCCTGCACTGA 0.00 80
    18703 873 GGTGCAGCCTCACTGTCT 0.00 81
    18704 910 AACTGCCTGTTTGCCCAC 33.89 82
    18705 954 CTTCTGCCTGCACCCCTG 0.00 83
    18706 976 ACTGACTGGGCATAGCTC 0.00 84
  • Example 12 Inhibition of CD40 Expression by Phosphoro-Thioate 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 CD40 mRNA, 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. [0307]
  • 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′-methoxyethyl (2′-MOE) nucleotides. The intersugar (backbone) linkages are phosphorothioate (P═S) throughout the oligonucleotide. Cytidine residues in the 2′-MOE wings are 5-methylcytidines. [0308]
  • Data are averaged from three experiments. [0309]
    TABLE 8
    Inhibition of CD40 mRNA Levels by Chimeric
    Phosphorothioate Oligonucleotides
    TARGET SEQ ID
    ISIS# SITE SEQUENCE % Inhibition NO.
    19211 18 CCAGGCGGCAGGACCACT 75.71 1
    19212 20 GACCAGGCGGCAGGACCA 77.23 2
    19213 26 AGGTGAGACCAGGCGGCA 80.82 3
    19214 48 CAGAGGCAGACGAACCAT 23.68 4
    19215 49 GCAGAGGCAGACGAACCA 45.97 5
    19216 73 GCAAGCAGCCCCAGAGGA 65.80 6
    19217 78 GGTCAGCAAGCAGCCCCA 74.73 7
    19218 84 GACAGCGGTCAGCAAGCA 67.21 8
    19219 88 GATGGACAGCGGTCAGCA 65.14 9
    19220 92 TCTGGATGGACAGCGGTC 78.71 10
    19221 98 GGTGGTTCTGGATGGACA 81.33 11
    19222 101 GTGGGTGGTTCTGGATGG 57.79 12
    19223 104 GCAGTGGGTGGTTCTGGA 73.70 13
    19224 152 CACAAAGAACAGCACTGA 40.25 14
    19225 156 CTGGCACAAAGAACAGCA 60.11 15
    19226 162 TCCTGGCTGGCACAAAGA 10.18 16
    19227 165 CTGTCCTGGCTGGCACAA 24.37 17
    19228 176 CTCACCAGTTTCTGTCCT 22.30 18
    19229 179 TCACTCACCAGTTTCTGT 40.64 19
    19230 185 GTGCAGTCACTCACCAGT 82.04 20
    19231 190 ACTCTGTGCAGTCACTCA 37.59 21
    19232 196 CAGTGAACTCTGTGCAGT 40.26 22
    19233 205 ATTCCGTTTCAGTGAACT 56.03 23
    19234 211 GAAGGCATTCCGTTTCAG 32.21 24
    19235 222 TTCACCGCAAGGAAGGCA 61.03 25
    19236 250 CTCTGTTCCAGGTGTCTA 62.19 26
    19237 267 CTGGTGGCAGTGTGTCTC 70.32 27
    19238 286 TGGGGTCGCAGTATTTGT 0.00 28
    19239 289 GGTTGGGGTCGCAGTATT 19.40 29
    19240 292 CTAGGTTGGGGTCGCAGT 36.32 30
    19241 318 GGTGCCCTTCTGCTGGAC 78.91 31
    19242 322 CTGAGGTGCCCTTCTGCT 69.84 32
    19243 332 GTGTCTGTTTCTGAGGTG 63.32 33
    19244 334 TGGTGTCTGTTTCTGAGG 42.83 34
    19245 345 ACAGGTGCAGATGGTGTC 73.31 35
    19246 348 TTCACAGGTGCAGATGGT 47.72 36
    19247 360 GTGCCAGCCTTCTTCACA 61.32 37
    19248 364 TACAGTGCCAGCCTTCTT 46.82 38
    19249 391 GGACACAGCTCTCACAGG 0.00 39
    19250 395 TGCAGGACACAGCTCTCA 52.05 40
    19251 401 GAGCGGTGCAGGACACAG 50.15 41
    19252 416 AAGCCGGGCGAGCATGAG 32.36 42
    19253 432 AATCTGCTTGACCCCAAA 0.00 43
    19254 446 GAAACCCCTGTAGCAATC 0.00 44
    19255 452 GTATCAGAAACCCCTGTA 36.13 45
    19256 463 GCTCGCAGATGGTATCAG 64.65 46
    19257 468 GCAGGGCTCGCAGATGGT 74.95 47
    19258 471 TGGGCAGGGCTCGCAGAT 0.00 48
    19259 474 GACTGGGCAGGGCTCGCA 82.00 49
    19260 490 CATTGGAGAAGAAGCCGA 41.31 50
    19261 497 GATGACACATTGGAGAAG 13.81 51
    19262 500 GCAGATGACACATTGGAG 78.48 52
    19263 506 TCGAAAGCAGATGACACA 59.28 53
    19264 524 GTCCAAGGGTGACATTTT 70.99 54
    19265 532 CACAGCTTGTCCAAGGGT 0.00 55
    19266 539 TTGGTCTCACAGCTTGTC 45.92 56
    19267 546 CAGGTCTTTGGTCTCACA 63.95 57
    19268 558 CTGTTGCACAACCAGGTC 82.32 58
    19269 570 GTTTGTGCCTGCCTGTTG 70.10 59
    19270 575 GTCTTGTTTGTGCCTGCC 68.95 60
    19271 590 CCACAGACAACATCAGTC 11.22 61
    19272 597 CTGGGGACCACAGACAAC 9.04 62
    19273 607 TCAGCCGATCCTGGGGAC 0.00 63
    19274 621 CACCACCAGGGCTCTCAG 23.08 64
    19275 626 GGGATCACCACCAGGGCT 57.94 65
    19276 657 GAGGATGGCAAACAGGAT 49.14 66
    19277 668 ACCAGCACCAAGAGGATG 3.48 67
    19278 679 TTTTGATAAAGACCAGCA 30.58 68
    19279 703 TATTGGTTGGCTTCTTGG 49.26 69
    19280 729 GGGTTCCTGCTTGGGGTG 13.95 70
    19281 750 GTCGGGAAAATTGATCTC 54.78 71
    19282 754 GATCGTCGGGAAAATTGA 0.00 72
    19283 765 GGAGCCAGGAAGATCGTC 69.47 73
    19284 766 TGGAGCCAGGAAGATCGT 54.48 74
    19285 780 TGGAGCAGCAGTGTTGGA 15.17 75
    19286 796 GTAAAGTCTCCTGCACTG 30.62 76
    19287 806 TGGCATCCATGTAAAGTC 65.03 77
    19288 810 CGGTTGGCATCCATGTAA 34.49 78
    19289 834 CTCTTTGCCATCCTCCTG 41.84 79
    19290 861 CTGTCTCTCCTGCACTGA 25.68 80
    19291 873 GGTGCAGCCTCACTGTCT 76.27 81
    19292 910 AACTGCCTGTTTGCCCAC 63.34 82
    19293 954 CTTCTGCCTGCACCCCTG 0.00 83
    19294 976 ACTGACTGGGCATAGCTC 11.55 84
  • 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. [0310]
  • 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. [0311]
  • 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 NO:92: [0312]
    5′- AGTGGTCCTGCCGCCTGGTC -3′ SEQ ID NO:92
        ||||||||||||||||||||
        TCACCAGGACGGCGGACC   -5′ SEQ ID NO:1
          ACCAGGACGGCGGACCAG -5′ SEQ ID NO: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 and disorders. [0313]
  • Example 14 Site Selection Program
  • In a preferred embodiment of the invention, illustrated in FIG. 20, an application is deployed which facilitates the selection process for determining the target positions of 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 interface allows users to skip from process to process at will, in any order. [0314]
  • Before running the site picking program, the target must have all relevant properties computed as described previously and indicated in [0315] 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. 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 properties. The user selects one of the properties to view plotted versus target position at [0316] 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 [0317] 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 finding 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 [0318] process step 2230. This property indicates the nearness of the site two 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 he 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 FIG. 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. [0319]
  • To begin using the oligonucleotide chemistry assignment program, the user launches it at [0320] 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. [0321]
  • 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. [0322]
  • Once the user has created a template, or selected an existing one, the software applies the template at [0323] 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 [0324] 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 [0325] 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 [0326] process step 2324 to the 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 [0327] process step 2328 with the new IDs 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 than 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:15 (5′-CTGGCACAAAGAACAGCA. In effecting this gene walk, the following parameters are used: [0328]
    Gene Walk Parameter Entered value
    Oligonucleotide Sequence ID: 15
    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: [0329]
    TABLE 8
    Oligonucleotide Generated By Gene Walk
    SEQ ID Sequence
    93 GAACAGCACTGACTGTTT
    94 AGAACAGCACTGACTGTT
    95 AAGAACAGCACTGACTGT
    96 AAAGAACAGCACTGACTG
    97 CAAAGAACAGCACTGACT
    98 ACAAAGAACAGCACTGAC
    99 CACAAAGAACAGCACTGA
    100 GCACAAAGAACAGCACTG
    101 GGCACAAAGAACAGCACT
    102 TGGCACAAAGAACAGCAC
    15 CTGGCACAAAGAACAGCA
    103 GCTGGCACAAAGAACAGC
    104 GGCTGGCACAAAGAACAG
    105 TGGCTGGCACAAAGAACA
    106 CTGGCTGGCACAAAGAAC
    107 CCTGGCTGGCACAAAGAA
    108 TCCTGGCTGGCACAAAGA
    109 GTCCTGGCTGGCACAAAG
    110 TGTCCTGGCTGGCACAAA
    111 CTGTCCTGGCTGGCACAA
    112 TCTGTCCTGGCTGGCACA
  • 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 at which the 5 ′ terminus of each oligonucleotide hybridizes. This, the first ten oligonucleotides are single-base frame shift sequences directed against the CD40 sequence upstream of compound SEQ ID NO: 15 and the latter ten are single-base frame shift sequences directed against the CD40 sequence downstream of compound SEQ ID NO: 15. [0330]
  • 1 112 1 18 DNA Artificial Sequence Novel Sequence 1 ccaggcggca ggaccact 18 2 18 DNA Artificial Sequence Novel Sequence 2 gaccaggcgg caggacca 18 3 18 DNA Artificial Sequence Novel Sequence 3 aggtgagacc aggcggca 18 4 18 DNA Artificial Sequence Novel Sequence 4 cagaggcaga cgaaccat 18 5 18 DNA Artificial Sequence Novel Sequence 5 gcagaggcag acgaacca 18 6 18 DNA Artificial Sequence Novel Sequence 6 gcaagcagcc ccagagga 18 7 18 DNA Artificial Sequence Novel Sequence 7 ggtcagcaag cagcccca 18 8 18 DNA Artificial Sequence Novel Sequence 8 gacagcggtc agcaagca 18 9 18 DNA Artificial Sequence Novel Sequence 9 gatggacagc ggtcagca 18 10 18 DNA Artificial Sequence Novel Sequence 10 tctggatgga cagcggtc 18 11 18 DNA Artificial Sequence Novel Sequence 11 ggtggttctg gatggaca 18 12 18 DNA Artificial Sequence Novel Sequence 12 gtgggtggtt ctggatgg 18 13 18 DNA Artificial Sequence Novel Sequence 13 gcagtgggtg gttctgga 18 14 18 DNA Artificial Sequence Novel Sequence 14 cacaaagaac agcactga 18 15 18 DNA Artificial Sequence Novel Sequence 15 ctggcacaaa gaacagca 18 16 18 DNA Artificial Sequence Novel Sequence 16 tcctggctgg cacaaaga 18 17 18 DNA Artificial Sequence Novel Sequence 17 ctgtcctggc tggcacaa 18 18 18 DNA Artificial Sequence Novel Sequence 18 ctcaccagtt tctgtcct 18 19 18 DNA Artificial Sequence Novel Sequence 19 tcactcacca gtttctgt 18 20 18 DNA Artificial Sequence Novel Sequence 20 gtgcagtcac tcaccagt 18 21 18 DNA Artificial Sequence Novel Sequence 21 actctgtgca gtcactca 18 22 18 DNA Artificial Sequence Novel Sequence 22 cagtgaactc tgtgcagt 18 23 18 DNA Artificial Sequence Novel Sequence 23 attccgtttc agtgaact 18 24 18 DNA Artificial Sequence Novel Sequence 24 gaaggcattc cgtttcag 18 25 18 DNA Artificial Sequence Novel Sequence 25 ttcaccgcaa ggaaggca 18 26 18 DNA Artificial Sequence Novel Sequence 26 ctctgttcca ggtgtcta 18 27 18 DNA Artificial Sequence Novel Sequence 27 ctggtggcag tgtgtctc 18 28 18 DNA Artificial Sequence Novel Sequence 28 tggggtcgca gtatttgt 18 29 18 DNA Artificial Sequence Novel Sequence 29 ggttggggtc gcagtatt 18 30 18 DNA Artificial Sequence Novel Sequence 30 ctaggttggg gtcgcagt 18 31 18 DNA Artificial Sequence Novel Sequence 31 ggtgcccttc tgctggac 18 32 18 DNA Artificial Sequence Novel Sequence 32 ctgaggtgcc cttctgct 18 33 18 DNA Artificial Sequence Novel Sequence 33 gtgtctgttt ctgaggtg 18 34 18 DNA Artificial Sequence Novel Sequence 34 tggtgtctgt ttctgagg 18 35 18 DNA Artificial Sequence Novel Sequence 35 acaggtgcag atggtgtc 18 36 18 DNA Artificial Sequence Novel Sequence 36 ttcacaggtg cagatggt 18 37 18 DNA Artificial Sequence Novel Sequence 37 gtgccagcct tcttcaca 18 38 18 DNA Artificial Sequence Novel Sequence 38 tacagtgcca gccttctt 18 39 18 DNA Artificial Sequence Novel Sequence 39 ggacacagct ctcacagg 18 40 18 DNA Artificial Sequence Novel Sequence 40 tgcaggacac agctctca 18 41 18 DNA Artificial Sequence Novel Sequence 41 gagcggtgca ggacacag 18 42 18 DNA Artificial Sequence Novel Sequence 42 aagccgggcg agcatgag 18 43 18 DNA Artificial Sequence Novel Sequence 43 aatctgcttg accccaaa 18 44 18 DNA Artificial Sequence Novel Sequence 44 gaaacccctg tagcaatc 18 45 18 DNA Artificial Sequence Novel Sequence 45 gtatcagaaa cccctgta 18 46 18 DNA Artificial Sequence Novel Sequence 46 gctcgcagat ggtatcag 18 47 18 DNA Artificial Sequence Novel Sequence 47 gcagggctcg cagatggt 18 48 18 DNA Artificial Sequence Novel Sequence 48 tgggcagggc tcgcagat 18 49 18 DNA Artificial Sequence Novel Sequence 49 gactgggcag ggctcgca 18 50 18 DNA Artificial Sequence Novel Sequence 50 cattggagaa gaagccga 18 51 18 DNA Artificial Sequence Novel Sequence 51 gatgacacat tggagaag 18 52 18 DNA Artificial Sequence Novel Sequence 52 gcagatgaca cattggag 18 53 18 DNA Artificial Sequence Novel Sequence 53 tcgaaagcag atgacaca 18 54 18 DNA Artificial Sequence Novel Sequence 54 gtccaagggt gacatttt 18 55 18 DNA Artificial Sequence Novel Sequence 55 cacagcttgt ccaagggt 18 56 18 DNA Artificial Sequence Novel Sequence 56 ttggtctcac agcttgtc 18 57 18 DNA Artificial Sequence Novel Sequence 57 caggtctttg gtctcaca 18 58 18 DNA Artificial Sequence Novel Sequence 58 ctgttgcaca accaggtc 18 59 18 DNA Artificial Sequence Novel Sequence 59 gtttgtgcct gcctgttg 18 60 18 DNA Artificial Sequence Novel Sequence 60 gtcttgtttg tgcctgcc 18 61 18 DNA Artificial Sequence Novel Sequence 61 ccacagacaa catcagtc 18 62 18 DNA Artificial Sequence Novel Sequence 62 ctggggacca cagacaac 18 63 18 DNA Artificial Sequence Novel Sequence 63 tcagccgatc ctggggac 18 64 18 DNA Artificial Sequence Novel Sequence 64 caccaccagg gctctcag 18 65 18 DNA Artificial Sequence Novel Sequence 65 gggatcacca ccagggct 18 66 18 DNA Artificial Sequence Novel Sequence 66 gaggatggca aacaggat 18 67 18 DNA Artificial Sequence Novel Sequence 67 accagcacca agaggatg 18 68 18 DNA Artificial Sequence Novel Sequence 68 ttttgataaa gaccagca 18 69 18 DNA Artificial Sequence Novel Sequence 69 tattggttgg cttcttgg 18 70 18 DNA Artificial Sequence Novel Sequence 70 gggttcctgc ttggggtg 18 71 18 DNA Artificial Sequence Novel Sequence 71 gtcgggaaaa ttgatctc 18 72 18 DNA Artificial Sequence Novel Sequence 72 gatcgtcggg aaaattga 18 73 18 DNA Artificial Sequence Novel Sequence 73 ggagccagga agatcgtc 18 74 18 DNA Artificial Sequence Novel Sequence 74 tggagccagg aagatcgt 18 75 18 DNA Artificial Sequence Novel Sequence 75 tggagcagca gtgttgga 18 76 18 DNA Artificial Sequence Novel Sequence 76 gtaaagtctc ctgcactg 18 77 18 DNA Artificial Sequence Novel Sequence 77 tggcatccat gtaaagtc 18 78 18 DNA Artificial Sequence Novel Sequence 78 cggttggcat ccatgtaa 18 79 18 DNA Artificial Sequence Novel Sequence 79 ctctttgcca tcctcctg 18 80 18 DNA Artificial Sequence Novel Sequence 80 ctgtctctcc tgcactga 18 81 18 DNA Artificial Sequence Novel Sequence 81 ggtgcagcct cactgtct 18 82 18 DNA Artificial Sequence Novel Sequence 82 aactgcctgt ttgcccac 18 83 18 DNA Artificial Sequence Novel Sequence 83 cttctgcctg cacccctg 18 84 18 DNA Artificial Sequence Novel Sequence 84 actgactggg catagctc 18 85 1004 DNA Artificial Sequence Novel Sequence 85 gcctcgctcg ggcgcccagt ggtcctgccg cctggtctca cctcgccatg gttcgtctgc 60 ctctgcagtg cgtcctctgg ggctgcttgc tgaccgctgt ccatccagaa ccacccactg 120 catgcagaga aaaacagtac ctaataaaca gtcagtgctg ttctttgtgc cagccaggac 180 agaaactggt gagtgactgc acagagttca ctgaaacgga atgccttcct tgcggtgaaa 240 gcgaattcct agacacctgg aacagagaga cacactgcca ccagcacaaa tactgcgacc 300 ccaacctagg gcttcgggtc cagcagaagg gcacctcaga aacagacacc atctgcacct 360 gtgaagaagg ctggcactgt acgagtgagg cctgtgagag ctgtgtcctg caccgctcat 420 gctcgcccgg ctttggggtc aagcagattg ctacaggggt ttctgatacc atctgcgagc 480 cctgcccagt cggcttcttc tccaatgtgt catctgcttt cgaaaaatgt cacccttgga 540 caagctgtga gaccaaagac ctggttgtgc aacaggcagg cacaaacaag actgatgttg 600 tctgtggtcc ccaggatcgg ctgagagccc tggtggtgat ccccatcatc ttcgggatcc 660 tgtttgccat cctcttggtg ctggtcttta tcaaaaaggt ggccaagaag ccaaccaata 720 aggcccccca ccccaagcag gaaccccagg agatcaattt tcccgacgat cttcctggct 780 ccaacactgc tgctccagtg caggagactt tacatggatg ccaaccggtc acccaggagg 840 atggcaaaga gagtcgcatc tcagtgcagg agagacagtg aggctgcacc cacccaggag 900 tgtggccacg tgggcaaaca ggcagttggc cagagagcct ggtgctgctg ctgcaggggt 960 gcaggcagaa gcggggagct atgcccagtc agtgccagcc cctc 1004 86 23 DNA Artificial Sequence Novel Sequence 86 cagagttcac tgaaacggaa tgc 23 87 23 DNA Artificial Sequence Novel Sequence 87 ggtggcagtg tgtctctctg ttc 23 88 25 DNA Artificial Sequence Novel Sequence 88 ttccttgcgg tgaaagcgaa ttcct 25 89 19 DNA Artificial Sequence Novel Sequence 89 gaaggtgaag gtcggagtc 19 90 20 DNA Artificial Sequence Novel Sequence 90 gaagatggtg atgggatttc 20 91 20 DNA Artificial Sequence Novel Sequence 91 caagcttccc gttctcagcc 20 92 20 DNA Artificial Sequence Novel Sequence 92 agtggtcctg ccgcctggtc 20 93 18 DNA Artificial Sequence Novel Sequence 93 gaacagcact gactgttt 18 94 18 DNA Artificial Sequence Novel Sequence 94 agaacagcac tgactgtt 18 95 18 DNA Artificial Sequence Novel Sequence 95 aagaacagca ctgactgt 18 96 18 DNA Artificial Sequence Novel Sequence 96 aaagaacagc actgactg 18 97 18 DNA Artificial Sequence Novel Sequence 97 caaagaacag cactgact 18 98 18 DNA Artificial Sequence Novel Sequence 98 acaaagaaca gcactgac 18 99 18 DNA Artificial Sequence Novel Sequence 99 cacaaagaac agcactga 18 100 18 DNA Artificial Sequence Novel Sequence 100 gcacaaagaa cagcactg 18 101 18 DNA Artificial Sequence Novel Sequence 101 ggcacaaaga acagcact 18 102 18 DNA Artificial Sequence Novel Sequence 102 tggcacaaag aacagcac 18 103 18 DNA Artificial Sequence Novel Sequence 103 gctggcacaa agaacagc 18 104 18 DNA Artificial Sequence Novel Sequence 104 ggctggcaca aagaacag 18 105 18 DNA Artificial Sequence Novel Sequence 105 tggctggcac aaagaaca 18 106 18 DNA Artificial Sequence Novel Sequence 106 ctggctggca caaagaac 18 107 18 DNA Artificial Sequence Novel Sequence 107 cctggctggc acaaagaa 18 108 18 DNA Artificial Sequence Novel Sequence 108 tcctggctgg cacaaaga 18 109 18 DNA Artificial Sequence Novel Sequence 109 gtcctggctg gcacaaag 18 110 18 DNA Artificial Sequence Novel Sequence 110 tgtcctggct ggcacaaa 18 111 18 DNA Artificial Sequence Novel Sequence 111 ctgtcctggc tggcacaa 18 112 18 DNA Artificial Sequence Novel Sequence 112 tctgtcctgg ctggcaca 18

Claims (46)

What is claimed is:
1. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising generating a library of nucleobase sequences in silico according to defined criteria.
2. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria.
3. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising robotically synthesizing a plurality of defined oligonucleotide compounds.
4. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
5. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising generating a library of nucleobase sequences in silico 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 generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria and robotically synthesizing a plurality of oligonucleotide compounds corresponding to said plurality of virtual oligonucleodites.
7. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria and robotically assaying a plurality of oligonucleotide compounds corresponding to said virtual oligonucleotides for one or more desired physical, chemical or biological properties.
8. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically synthesizing a plurality of oligonucleotide compounds having said nucleobase sequences.
9. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising robotically synthesizing a plurality of oligonucleotide compounds and robotically assaying said plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
10. A method of generating a set of compounds that modulate the expression of a target nucleic acid sequence comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically assaying a plurality of oligonucleotide compounds having said nucleobase sequences for one or more desired physical, chemical or biological properties.
11. A method of generating a set of compounds that modulate the expression of a 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 oligonucleotide compounds.
12. A method of generating a set of compounds that modulate the expression of a 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 oligonucleotide compounds for one or more desired physical, chemical or biological properties.
13. A method of generating a set of compounds that modulate the expression of a 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 oligonucleotide compounds; and
(c) robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
14. A method of generating a set of compounds that modulate the expression of a 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 oligonucleotide compounds; and
(c) robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
15. A method of generating a set of compounds that modulate the expression of a 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 oligonucleotide compounds; and
(d) robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
16. A method of generating a set of compounds that modulate the expression of a 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 oligonucleotide compounds having said nucleobase sequences of step (a) and said oligonucleotide chemistry of step (b);
(d) robotically assaying said set of oligonucleotide compounds of step (c) for a physical, chemical or biological activity; and
(e) selecting a subset of said set of oligonucleotide compounds 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 compounds that modulate the expression of a 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 oligonucleotide compounds having said preferred nucleobase sequences of step (c) and said oligonucleotide chemistry of step (b);
(e) robotically assaying said set of oligonucleotide compounds of step (d) for a physical, chemical or biological activity; and
(f) selecting a subset of said set of oligonucleotide compounds of step (d) having a desired level of physical, chemical or biological activity in order to generate said set of compounds.
18. The method of claim 4, wherein said step of robotically assaying said plurality of 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 1, 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 1, 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 modulation comprising generating a library of antisense nucleobase sequences in silico according to defined criteria.
22. A method of generating a set of compounds that modulate the expression of a 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 modulation comprising robotically synthesizing a plurality of antisense compounds.
24. A method of identifying one or more nucleic acid sequences amenable to antisense modulation comprising robotically assaying a plurality of 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 modulation comprising generating a library of nucleobase sequences in silico 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 modulation comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria and robotically synthesizing a plurality of oligonucleotide compounds.
27. A method of identifying one or more nucleic acid sequences amenable to antisense modulation comprising evaluating in silico a plurality of virtual oligonucleotides according to defined criteria and robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
28. A method of identifying one or more nucleic acid sequences amenable to antisense modulation comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically synthesizing a plurality of oligonucleotide compounds having said nucleobase sequences.
29. A method of identifying one or more nucleic acid sequences amenable to antisense modulation comprising robotically synthesizing a plurality of oligonucleotide compounds and robotically assaying said plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
30. A method of identifying one or more nucleic acid sequences amenable to antisense modulation comprising generating a library of nucleobase sequences in silico according to defined criteria and robotically assaying a plurality of oligonucleotide compounds 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 modulation 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 oligonucleotide compounds.
32. A method of identifying one or more nucleic acid sequences amenable to antisense modulation, 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 oligonucleotide compounds for one or more desired physical, chemical or biological properties.
33. A method of identifying one or more nucleic acid sequences amenable to antisense modulation, comprising the steps of:
(a) generating a library of nucleobase sequences in silico according to defined criteria;
(b) robotically synthesizing a plurality of oligonucleotide compounds; and
(c) robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
34. A method of identifying one or more nucleic acid sequences amenable to antisense modulation, comprising the steps of:
(a) evaluating in silico a plurality of virtual oligonucleotides according to defined criteria;
(b) robotically synthesizing a plurality of oligonucleotide compounds; and
(c) robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
35. A method of identifying one or more nucleic acid sequences amenable to antisense modulation, 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 oligonucleotide compounds; and
(d) robotically assaying a plurality of oligonucleotide compounds for one or more desired physical, chemical or biological properties.
36. A method of identifying one or more nucleic acid sequences amenable to antisense modulation, 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 oligonucleotide compounds having said nucleobase sequences of step (a) and said oligonucleotide chemistry of step (b);
(d) robotically assaying said set of oligonucleotide compounds of step (c) for a physical, chemical or biological activity; and
(e) selecting a subset of said set of oligonucleotide compounds of step (c) having a desired level of physical, chemical or biological activity in order to generate said set of compounds.
37. A method of identifying one or more nucleic acid sequences amenable to antisense modulation, 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 oligonucleotide compounds having said preferred nucleobase sequences of step (b) and said oligonucleotide chemistry of step (c);
(e) robotically assaying said set of oligonucleotide compounds of step (d) for a physical, chemical or biological activity; and
(f) selecting a subset of said set of oligonucleotide compounds of step (d) having a desired level of physical, chemical or biological activity in order to generate said set of compounds.
38. The method of claim 24, wherein said step of robotically assaying said plurality of nucleic acid sequences is performed by computer-controlled real-time polymerase chain reaction or by computer-controlled enzyme-linked immunosorbent assay.
39. The method of claim 21, 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 21, wherein said nucleic acid sequence is a human nucleic acid.
41. A computer formatted medium comprising computer readable instructions for identifying active compounds.
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 modulation.
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 modulation in computer readable form.
46. A computer formatted medium comprising one or more nucleic acid sequences amenable to antisense modulation in computer readable form, wherein said one or more nucleic acid sequences is identified according to the method of any one of claims 21, 22 or 24-40.
US10/116,325 1998-04-13 2002-04-04 System of components for preparing oligonucleotides Abandoned US20030113739A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/116,325 US20030113739A1 (en) 1998-04-13 2002-04-04 System of components for preparing oligonucleotides

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US8148398P 1998-04-13 1998-04-13
US09/067,638 US7321828B2 (en) 1998-04-13 1998-04-28 System of components for preparing oligonucleotides
US10/116,325 US20030113739A1 (en) 1998-04-13 2002-04-04 System of components for preparing oligonucleotides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/067,638 Continuation US7321828B2 (en) 1998-04-13 1998-04-28 System of components for preparing oligonucleotides

Publications (1)

Publication Number Publication Date
US20030113739A1 true US20030113739A1 (en) 2003-06-19

Family

ID=22077363

Family Applications (5)

Application Number Title Priority Date Filing Date
US09/067,638 Expired - Fee Related US7321828B2 (en) 1998-04-13 1998-04-28 System of components for preparing oligonucleotides
US10/116,325 Abandoned US20030113739A1 (en) 1998-04-13 2002-04-04 System of components for preparing oligonucleotides
US10/649,467 Abandoned US20050033524A1 (en) 1998-04-13 2003-08-27 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US10/830,475 Abandoned US20040197814A1 (en) 1998-04-13 2004-04-21 System of components for preparing oligonucleotides
US11/226,882 Abandoned US20060069518A1 (en) 1998-04-13 2005-09-14 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/067,638 Expired - Fee Related US7321828B2 (en) 1998-04-13 1998-04-28 System of components for preparing oligonucleotides

Family Applications After (3)

Application Number Title Priority Date Filing Date
US10/649,467 Abandoned US20050033524A1 (en) 1998-04-13 2003-08-27 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US10/830,475 Abandoned US20040197814A1 (en) 1998-04-13 2004-04-21 System of components for preparing oligonucleotides
US11/226,882 Abandoned US20060069518A1 (en) 1998-04-13 2005-09-14 Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation

Country Status (2)

Country Link
US (5) US7321828B2 (en)
WO (1) WO1999053101A1 (en)

Families Citing this family (128)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5958771A (en) * 1998-12-03 1999-09-28 Isis Pharmaceuticals, Inc. Antisense modulation of cellular inhibitor of Apoptosis-2 expression
US6197584B1 (en) * 1998-05-01 2001-03-06 Isis Pharmaceuticals, Inc. Antisense modulation of CD40 expression
WO2001031579A2 (en) * 1999-10-27 2001-05-03 Barnhill Technologies, Llc Methods and devices for identifying patterns in biological patterns
EP2357582A1 (en) * 1999-10-27 2011-08-17 Health Discovery Corporation Methods and devices for identifying patterns in biological systems
US6629038B1 (en) * 2000-09-05 2003-09-30 Yeda Research And Development Co. Ltd. Method and system for identifying commercially distributed organisms
US6766082B2 (en) * 2000-10-18 2004-07-20 Nippon Telegraph And Telephone Corporation Waveguide-type optical device and manufacturing method therefor
EP1385950B1 (en) 2001-01-19 2008-07-02 Centocor, Inc. Computer-directed assembly of a polynucleotide encoding a target polypeptide
US20030027135A1 (en) * 2001-03-02 2003-02-06 Ecker David J. Method for rapid detection and identification of bioagents
US7226739B2 (en) 2001-03-02 2007-06-05 Isis Pharmaceuticals, Inc Methods for rapid detection and identification of bioagents in epidemiological and forensic investigations
US20040121313A1 (en) * 2002-12-06 2004-06-24 Ecker David J. Methods for rapid detection and identification of bioagents in organs for transplantation
US7666588B2 (en) 2001-03-02 2010-02-23 Ibis Biosciences, Inc. Methods for rapid forensic analysis of mitochondrial DNA and characterization of mitochondrial DNA heteroplasmy
WO2004060278A2 (en) 2002-12-06 2004-07-22 Isis Pharmaceuticals, Inc. Methods for rapid identification of pathogens in humans and animals
US7718354B2 (en) 2001-03-02 2010-05-18 Ibis Biosciences, Inc. Methods for rapid identification of pathogens in humans and animals
US8073627B2 (en) * 2001-06-26 2011-12-06 Ibis Biosciences, Inc. System for indentification of pathogens
US7217510B2 (en) 2001-06-26 2007-05-15 Isis Pharmaceuticals, Inc. Methods for providing bacterial bioagent characterizing information
EP1414841B1 (en) 2001-07-10 2012-11-14 Lakewood-Amedex, Inc Oligonucleotide-containing pharmacological compositions and their use
EP1461674A4 (en) * 2001-12-03 2007-10-24 Dyax Corp Library screening
JP2003331168A (en) * 2002-05-15 2003-11-21 Fujitsu Ltd Method for ordering primer synthesis for gene amplification, method for ordering reagent kit for gene amplification factor detection, and device and program for ordering primer synthesis and reagent kit
DE10238298A1 (en) * 2002-08-21 2004-03-04 Beiersdorf Ag Use of antisense oligonucleotides for the treatment of degenerative skin symptoms
US20040077569A1 (en) * 2002-10-16 2004-04-22 Isis Pharmaceuticals Inc. Antisense modulation of Notch (Drosophila) homolog 4 expression
DE10254214A1 (en) * 2002-11-20 2004-06-09 Beiersdorf Ag Oligoribonucleotides for the treatment of degenerative skin symptoms by RNA interference
US8046171B2 (en) * 2003-04-18 2011-10-25 Ibis Biosciences, Inc. Methods and apparatus for genetic evaluation
US8057993B2 (en) 2003-04-26 2011-11-15 Ibis Biosciences, Inc. Methods for identification of coronaviruses
WO2005009202A2 (en) * 2003-05-12 2005-02-03 Isis Pharmaceuticals, Inc. Automatic identification of bioagents
US7964343B2 (en) * 2003-05-13 2011-06-21 Ibis Biosciences, Inc. Method for rapid purification of nucleic acids for subsequent analysis by mass spectrometry by solution capture
US8158354B2 (en) 2003-05-13 2012-04-17 Ibis Biosciences, Inc. Methods for rapid purification of nucleic acids for subsequent analysis by mass spectrometry by solution capture
US8097416B2 (en) 2003-09-11 2012-01-17 Ibis Biosciences, Inc. Methods for identification of sepsis-causing bacteria
US8546082B2 (en) 2003-09-11 2013-10-01 Ibis Biosciences, Inc. Methods for identification of sepsis-causing bacteria
US20100035239A1 (en) * 2003-09-11 2010-02-11 Isis Pharmaceuticals, Inc. Compositions for use in identification of bacteria
US20080138808A1 (en) * 2003-09-11 2008-06-12 Hall Thomas A Methods for identification of sepsis-causing bacteria
US20120122103A1 (en) 2003-09-11 2012-05-17 Rangarajan Sampath Compositions for use in identification of bacteria
US20060240412A1 (en) * 2003-09-11 2006-10-26 Hall Thomas A Compositions for use in identification of adenoviruses
US7480382B2 (en) * 2003-09-30 2009-01-20 Microsoft Corporation Image file container
US8163895B2 (en) 2003-12-05 2012-04-24 Ibis Biosciences, Inc. Compositions for use in identification of orthopoxviruses
US7666592B2 (en) 2004-02-18 2010-02-23 Ibis Biosciences, Inc. Methods for concurrent identification and quantification of an unknown bioagent
US8119336B2 (en) * 2004-03-03 2012-02-21 Ibis Biosciences, Inc. Compositions for use in identification of alphaviruses
EP2458619B1 (en) * 2004-05-24 2017-08-02 Ibis Biosciences, Inc. Mass spectrometry with selective ion filtration by digital thresholding
US20100152280A1 (en) * 2004-05-24 2010-06-17 Isis Pharmaceuticals, Inc. Modulation of sid-1 expression
US20050266411A1 (en) * 2004-05-25 2005-12-01 Hofstadler Steven A Methods for rapid forensic analysis of mitochondrial DNA
EP1771250B1 (en) * 2004-06-04 2011-01-26 Abacus Diagnostica OY Temperature control of reaction vessel, system with reaction vessel and use of system
US7811753B2 (en) 2004-07-14 2010-10-12 Ibis Biosciences, Inc. Methods for repairing degraded DNA
US8084207B2 (en) * 2005-03-03 2011-12-27 Ibis Bioscience, Inc. Compositions for use in identification of papillomavirus
CA2600184A1 (en) * 2005-03-03 2006-09-08 Isis Pharmaceuticals, Inc. Compositions for use in identification of adventitious viruses
US8026084B2 (en) * 2005-07-21 2011-09-27 Ibis Biosciences, Inc. Methods for rapid identification and quantitation of nucleic acid variants
EP1976567B1 (en) * 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
KR100794705B1 (en) * 2006-06-13 2008-01-14 (주)바이오니아 Method of Inhibiting Expression of Target mRNA Using siRNA Considering Alternative Splicing of Genes
US9149473B2 (en) * 2006-09-14 2015-10-06 Ibis Biosciences, Inc. Targeted whole genome amplification method for identification of pathogens
WO2008104002A2 (en) * 2007-02-23 2008-08-28 Ibis Biosciences, Inc. Methods for rapid forensic dna analysis
US20100204266A1 (en) * 2007-03-23 2010-08-12 Ibis Biosciences, INC Compositions for use in identification of mixed populations of bioagents
WO2008151023A2 (en) 2007-06-01 2008-12-11 Ibis Biosciences, Inc. Methods and compositions for multiple displacement amplification of nucleic acids
USRE47320E1 (en) 2007-11-20 2019-03-26 Ionis Pharmaceuticals, Inc. Modulation of CD40 expression
EP2349549B1 (en) 2008-09-16 2012-07-18 Ibis Biosciences, Inc. Mixing cartridges, mixing stations, and related kits, and system
US8148163B2 (en) 2008-09-16 2012-04-03 Ibis Biosciences, Inc. Sample processing units, systems, and related methods
US8534447B2 (en) 2008-09-16 2013-09-17 Ibis Biosciences, Inc. Microplate handling systems and related computer program products and methods
MX339820B (en) * 2008-10-03 2016-06-13 Curna Inc Treatment of apolipoprotein-a1 related diseases by inhibition of natural antisense transcript to apolipoprotein-a1.
JP5971948B2 (en) * 2008-12-04 2016-08-17 クルナ・インコーポレーテッド Treatment of vascular endothelial growth factor (VEGF) -related diseases by suppression of natural antisense transcripts against VEGF
RU2746478C2 (en) 2008-12-04 2021-04-14 КьюРНА, Инк. Treatment of tumors of diseases related to the genom-suppressor by therapy of natural transcript inhibition in anti-significant orientation regarding this gene
KR101829469B1 (en) * 2008-12-04 2018-03-30 큐알엔에이, 인크. Treatment of erythropoietin (epo) related diseases by inhibition of natural antisense transcript to epo
WO2010093943A1 (en) 2009-02-12 2010-08-19 Ibis Biosciences, Inc. Ionization probe assemblies
KR101682735B1 (en) 2009-02-12 2016-12-06 큐알엔에이, 인크. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
ES2656290T3 (en) 2009-03-16 2018-02-26 Curna, Inc. Treatment of diseases related to nuclear factor (derived from erythroid 2) similar to 2 (NRF2) by inhibition of natural antisense transcript to NRF2
EP2408920B1 (en) 2009-03-17 2017-03-08 CuRNA, Inc. Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
JP5883782B2 (en) 2009-05-06 2016-03-15 クルナ・インコーポレーテッド Treatment of lipid transport metabolism gene-related diseases by suppression of natural antisense transcripts on lipid transport metabolism genes
WO2010129746A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
KR101742334B1 (en) 2009-05-08 2017-06-01 큐알엔에이, 인크. Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family
DK2432881T3 (en) 2009-05-18 2018-02-26 Curna Inc TREATMENT OF REPROGRAMMING FACTOR-RELATED DISEASES BY INHIBITING NATURAL ANTISENSE TRANSCRIPTS TO A REPROGRAMMING FACTOR
KR101703695B1 (en) 2009-05-22 2017-02-08 큐알엔에이, 인크. Treatment of transcription factor e3 (tfe3) and insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to tfe3
CN103221541B (en) 2009-05-28 2017-03-01 库尔纳公司 Antiviral gene relevant disease is treated by the natural antisense transcript suppressing antiviral gene
WO2010148050A2 (en) 2009-06-16 2010-12-23 Curna, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
JP6128846B2 (en) 2009-06-16 2017-05-17 クルナ・インコーポレーテッド Treatment of PON1 gene-related diseases by suppression of natural antisense transcripts against paraoxonase (PON1)
US8859515B2 (en) 2009-06-24 2014-10-14 Curna, Inc. Treatment of tumor necrosis factor receptor 2 (TNFR2) related diseases by inhibition of natural antisense transcript to TNFR2
EP2446037B1 (en) 2009-06-26 2016-04-20 CuRNA, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
EP2454000A4 (en) 2009-07-17 2016-08-10 Ibis Biosciences Inc Systems for bioagent identification
US8950604B2 (en) * 2009-07-17 2015-02-10 Ibis Biosciences, Inc. Lift and mount apparatus
US20120252869A1 (en) 2009-07-24 2012-10-04 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CA2769665A1 (en) 2009-08-05 2011-02-10 Opko Curna, Llc Treatment of insulin gene (ins) related diseases by inhibition of natural antisense transcript to an insulin gene (ins)
CN102625841A (en) 2009-08-11 2012-08-01 欧科库尔纳有限责任公司 Treatment of Adiponectin (ADIPOQ) related diseases by inhibition of natural antisense transcript to an Adiponectin (ADIPOQ)
US8791087B2 (en) 2009-08-21 2014-07-29 Curna, Inc. Treatment of ‘C terminus of HSP70-interacting protein’ (CHIP)related diseases by inhibition of natural antisense transcript to CHIP
CN102482671B (en) 2009-08-25 2017-12-01 库尔纳公司 IQGAP relevant diseases are treated by suppressing the natural antisense transcript of ' gtpase activating protein containing IQ die bodys ' (IQGAP)
ES2664591T3 (en) 2009-09-25 2018-04-20 Curna, Inc. Treatment of phylagrin-related diseases (flg) by modulating the expression and activity of the FLG gene
WO2011041695A1 (en) * 2009-10-02 2011-04-07 Ibis Biosciences, Inc. Determination of methylation status of polynucleotides
ES2628739T3 (en) * 2009-10-15 2017-08-03 Ibis Biosciences, Inc. Multiple displacement amplification
WO2011084455A2 (en) 2009-12-16 2011-07-14 Opko Curna, Llc. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
RU2619185C2 (en) 2009-12-23 2017-05-12 Курна, Инк. Treatment of diseases associated with uncoupling proteins 2 (ucp2), by inhibiting of natural antisense transcript to ucp2
KR101891352B1 (en) 2009-12-23 2018-08-24 큐알엔에이, 인크. Treatment of hepatocyte growth factor (hgf) related diseases by inhibition of natural antisense transcript to hgf
JP5982288B2 (en) 2009-12-29 2016-08-31 カッパーアールエヌエー,インコーポレイテッド Treatment of tumor protein 63-related diseases by inhibition of natural antisense transcripts against tumor protein 63 (p63)
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
US20120289583A1 (en) 2009-12-31 2012-11-15 Curna, Inc. Treatment of insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to irs2 and transcription factor e3 (tfe3)
NO2521784T3 (en) 2010-01-04 2018-05-05
EP2521785B1 (en) 2010-01-06 2022-03-09 CuRNA, Inc. Inhibition of natural antisense transcript to a pancreatic developmental gene for use in a treatment of pancreatic developmental gene related diseases
ES2664866T3 (en) 2010-01-11 2018-04-23 Curna, Inc. Treatment of diseases related to sex hormone binding globulin (shbg) by inhibition of the natural antisense transcript to shbg
CA2786568A1 (en) 2010-01-25 2011-07-28 Curna, Inc. Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
JP5976548B2 (en) 2010-02-22 2016-08-23 カッパーアールエヌエー,インコーポレイテッド Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcripts against PYCR1
WO2011123745A2 (en) 2010-04-02 2011-10-06 Opko Curna Llc Treatment of colony-stimulating factor 3 (csf3) related diseases by inhibition of natural antisense transcript to csf3
WO2011127337A2 (en) 2010-04-09 2011-10-13 Opko Curna Llc Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
WO2011139387A1 (en) 2010-05-03 2011-11-10 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
RU2620978C2 (en) 2010-05-26 2017-05-30 Курна, Инк. Treatment of diseases associated with methionine sulfoxide reductase a (msra), by msra natural antisense transcript inhibition
RU2585229C2 (en) 2010-05-26 2016-05-27 Курна, Инк. Treatment of diseases associated with atonal homolog 1 (aton1) by inhibiting natural antisense transcript of gene aton1
ES2863526T3 (en) 2010-06-23 2021-10-11 Curna Inc Treatment of voltage-controlled sodium channel alpha subunit (SCNA) -related diseases by inhibiting natural antisense transcription to SCNA
RU2611190C2 (en) 2010-07-14 2017-02-21 Курна, Инк. Treatment of diseases related with gene dlg by inhibition of natural antisense transcript of dlg gene
TWI541024B (en) 2010-09-01 2016-07-11 日本新藥股份有限公司 Antisense nucleic acid
ES2640755T3 (en) 2010-10-06 2017-11-06 Curna, Inc. Treatment of diseases related to Sialidase 4 (neu4) by inhibition of the natural antisense transcript to the neu4 gene
WO2012054723A2 (en) 2010-10-22 2012-04-26 Opko Curna Llc Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
US10000752B2 (en) 2010-11-18 2018-06-19 Curna, Inc. Antagonat compositions and methods of use
CA2818824A1 (en) 2010-11-23 2012-05-31 Joseph Collard Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
ES2653247T3 (en) 2011-06-09 2018-02-06 Curna, Inc. Treatment of frataxin-related diseases (FXN) by inhibiting the natural antisense transcript to the FXN gene
JP6125505B2 (en) 2011-09-06 2017-05-10 カッパーアールエヌエー,インコーポレイテッド Treatment of voltage-gated sodium channel alpha subunit related diseases with small molecules
KR101762295B1 (en) * 2012-02-10 2017-08-04 (주)바이오니아 Automatic analysis apparatus and method of biological samples
CA2867262C (en) 2012-03-15 2021-03-16 Curna, Inc. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
WO2013138159A1 (en) * 2012-03-15 2013-09-19 Amgen Inc. Methods of determining exposure to uv light
TWM445178U (en) * 2012-06-25 2013-01-11 Chin-Feng Wan Microfluidic chip automatic system with optical platform
KR102423377B1 (en) 2013-08-05 2022-07-25 트위스트 바이오사이언스 코포레이션 De novo synthesized gene libraries
US10040048B1 (en) 2014-09-25 2018-08-07 Synthego Corporation Automated modular system and method for production of biopolymers
CA2975852A1 (en) 2015-02-04 2016-08-11 Twist Bioscience Corporation Methods and devices for de novo oligonucleic acid assembly
WO2016172377A1 (en) 2015-04-21 2016-10-27 Twist Bioscience Corporation Devices and methods for oligonucleic acid library synthesis
CN108368482A (en) 2015-09-18 2018-08-03 特韦斯特生物科学公司 Oligonucleotide Mutant libraries and its synthesis
WO2017053450A1 (en) 2015-09-22 2017-03-30 Twist Bioscience Corporation Flexible substrates for nucleic acid synthesis
US10417457B2 (en) 2016-09-21 2019-09-17 Twist Bioscience Corporation Nucleic acid based data storage
CA3054303A1 (en) 2017-02-22 2018-08-30 Twist Bioscience Corporation Nucleic acid based data storage
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
AU2018328847A1 (en) 2017-09-11 2020-04-23 Twist Bioscience Corporation GPCR binding proteins and synthesis thereof
KR102637566B1 (en) 2017-10-20 2024-02-16 트위스트 바이오사이언스 코포레이션 Heated nanowells for polynucleotide synthesis
EP3814497A4 (en) 2018-05-18 2022-03-02 Twist Bioscience Corporation Polynucleotides, reagents, and methods for nucleic acid hybridization
SG11202109283UA (en) 2019-02-26 2021-09-29 Twist Bioscience Corp Variant nucleic acid libraries for antibody optimization
JP2022550497A (en) 2019-06-21 2022-12-02 ツイスト バイオサイエンス コーポレーション Barcode-based nucleic acid sequence assembly
CN114144110B (en) 2019-07-30 2023-02-03 Emd密理博公司 Method for synthesizing compound

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4800159A (en) * 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US5352775A (en) * 1991-01-16 1994-10-04 The Johns Hopkins Univ. APC gene and nucleic acid probes derived therefrom
US5407796A (en) * 1991-01-04 1995-04-18 The Johns Hopkins University Cystic fibrosis mutation cluster
US5436327A (en) * 1988-09-21 1995-07-25 Isis Innovation Limited Support-bound oligonucleotides
US5463564A (en) * 1994-09-16 1995-10-31 3-Dimensional Pharmaceuticals, Inc. System and method of automatically generating chemical compounds with desired properties
US5463567A (en) * 1993-10-15 1995-10-31 Caterpillar Inc. Apparatus and method for providing historical data regarding machine operating parameters
US5541313A (en) * 1983-02-22 1996-07-30 Molecular Biosystems, Inc. Single-stranded labelled oligonucleotides of preselected sequence
US5571639A (en) * 1994-05-24 1996-11-05 Affymax Technologies N.V. Computer-aided engineering system for design of sequence arrays and lithographic masks
US5612455A (en) * 1994-07-05 1997-03-18 Tularik, Inc. Nuclear factors and binding assay
US5639603A (en) * 1991-09-18 1997-06-17 Affymax Technologies N.V. Synthesizing and screening molecular diversity
US5650122A (en) * 1986-10-31 1997-07-22 Pasteur Sanofi Diagnostics Automated patient sample analysis instrument having tubes and reaction wells washing apparatus
US5700637A (en) * 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5707796A (en) * 1990-06-11 1998-01-13 Nexstar Pharmaceuticals, Inc. Method for selecting nucleic acids on the basis of structure
US5720923A (en) * 1993-07-28 1998-02-24 The Perkin-Elmer Corporation Nucleic acid amplification reaction apparatus
US6016348A (en) * 1996-11-27 2000-01-18 Thomson Consumer Electronics, Inc. Decoding system and data format for processing and storing encrypted broadcast, cable or satellite video data
US6127191A (en) * 1996-12-03 2000-10-03 3-Dimensional Pharmaceuticals, Inc. Aminobenzenedicarboxylic acid-based combinatorial libraries
US6295514B1 (en) * 1996-11-04 2001-09-25 3-Dimensional Pharmaceuticals, Inc. Method, system, and computer program product for representing similarity/dissimilarity between chemical compounds
US6453246B1 (en) * 1996-11-04 2002-09-17 3-Dimensional Pharmaceuticals, Inc. System, method, and computer program product for representing proximity data in a multi-dimensional space
US6506784B1 (en) * 1999-07-01 2003-01-14 3-Dimensional Pharmaceuticals, Inc. Use of 1,3-substituted pyrazol-5-yl sulfonates as pesticides
US6518266B1 (en) * 1999-07-22 2003-02-11 3-Dimensional Pharmaceuticals 1- Aryl-3-thioalkyl pyrazoles, the synthesis thereof and the use thereof as insecticides
US6571227B1 (en) * 1996-11-04 2003-05-27 3-Dimensional Pharmaceuticals, Inc. Method, system and computer program product for non-linear mapping of multi-dimensional data

Family Cites Families (144)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023A (en) * 1847-03-20 Stocking-loom
US552538A (en) * 1896-01-07 Potato sorter and cleaner
US360966A (en) * 1887-04-12 Folding umbrella
US2122177A (en) * 1937-05-19 1938-06-28 Lincoln Eng Co Injector
US2155250A (en) * 1938-01-17 1939-04-18 Lincoln Eng Co Injector
US2162898A (en) * 1938-01-17 1939-06-20 Lincoln Eng Co Relief device for lubricating apparatus
US2283638A (en) * 1940-08-12 1942-05-19 Lincoln Eng Co Injector
US2637413A (en) * 1950-04-22 1953-05-05 Lincoln Eng Co Lubricant injector
US2627320A (en) * 1950-09-07 1953-02-03 Lincoln Eng Co Lubricating system
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
JPS4833493Y1 (en) * 1969-10-15 1973-10-11
US3664462A (en) * 1970-05-27 1972-05-23 William O Smith Sr Lubricant metering valve
US3715013A (en) * 1971-08-19 1973-02-06 Eaton Corp Lubricant injector
US3958725A (en) * 1974-10-15 1976-05-25 Auto Research Corporation Metering valve for lubrication injector
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
US4494913A (en) * 1982-07-08 1985-01-22 Bijur Lubricating Corp. Positive displacement injector
JPS5927900A (en) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk Oligonucleotide derivative and its preparation
FR2540122B1 (en) 1983-01-27 1985-11-29 Centre Nat Rech Scient NOVEL COMPOUNDS COMPRISING A SEQUENCE OF OLIGONUCLEOTIDE LINKED TO AN INTERCALATION AGENT, THEIR SYNTHESIS PROCESS AND THEIR APPLICATION
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4520902A (en) * 1983-04-19 1985-06-04 Lubriquip-Houdaille, Inc. Lubricant applying system and injector means
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US4668476A (en) * 1984-03-23 1987-05-26 Applied Biosystems, Inc. Automated polypeptide synthesis apparatus
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
IT1196307B (en) * 1984-10-22 1988-11-16 Chiesi Farma Spa AQUEOUS PHARMACEUTICAL FORMULATIONS OF PIROXICAM MONOHYDRATE
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
US4806463A (en) 1986-05-23 1989-02-21 Worcester Foundation For Experimental Biology Inhibition of HTLV-III by exogenous oligonucleotides
JPS638396A (en) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd Poly-labeled oligonucleotide derivative
DE3788914T2 (en) 1986-09-08 1994-08-25 Ajinomoto Kk Compounds for cleaving RNA at a specific position, oligomers used in the preparation of these compounds and starting materials for the synthesis of these oligomers.
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US5525465A (en) * 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (en) 1987-11-12 1989-05-24 Max Planck Gesellschaft MODIFIED OLIGONUCLEOTIDS
US5403711A (en) 1987-11-30 1995-04-04 University Of Iowa Research Foundation Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved
EP0348458B1 (en) 1987-11-30 1997-04-09 University Of Iowa Research Foundation Dna molecules stabilized by modifications of the 3'-terminal phosphodiester linkage and their use as nucleic acid probes and as therapeutic agents to block the expression of specifically targeted genes
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
GB8810400D0 (en) * 1988-05-03 1988-06-08 Southern E Analysing polynucleotide sequences
JPH03503894A (en) * 1988-03-25 1991-08-29 ユニバーシィティ オブ バージニア アランミ パテンツ ファウンデイション Oligonucleotide N-alkylphosphoramidate
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5599923A (en) * 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5591722A (en) * 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
ATE190981T1 (en) 1989-10-24 2000-04-15 Isis Pharmaceuticals Inc 2'-MODIFIED NUCLEOTIDES
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5955589A (en) * 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
US5859221A (en) * 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5623065A (en) * 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US5220007A (en) 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
AU7579991A (en) 1990-02-20 1991-09-18 Gilead Sciences, Inc. Pseudonucleosides and pseudonucleotides and their polymers
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
ATE167523T1 (en) * 1990-05-11 1998-07-15 Microprobe Corp IMMERSIBLE TEST STRIPS FOR NUCLEIC ACID HYBRIDIZATION ASSAY AND METHOD FOR COVALENT IMMOBILIZATION OF OLIGONUCLEOTIDES
US5623070A (en) * 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
ATE154246T1 (en) * 1990-07-27 1997-06-15 Isis Pharmaceuticals Inc NUCLEASE RESISTANT PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECTE AND MODULATE GENE EXPRESSION
US5608046A (en) * 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5610289A (en) * 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5618704A (en) * 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5602240A (en) * 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
JPH06505704A (en) * 1990-09-20 1994-06-30 ギリアド サイエンシズ,インコーポレイテッド Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
KR930702373A (en) 1990-11-08 1993-09-08 안토니 제이. 페이네 Addition of Multiple Reporter Groups to Synthetic Oligonucleotides
AU1999092A (en) * 1991-05-24 1992-12-30 Walter Gilbert Method and apparatus for rapid nucleic acid sequencing
US5714331A (en) * 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5719262A (en) * 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
WO1993000446A1 (en) * 1991-06-27 1993-01-07 Genelabs Technologies, Inc. Screening assay for the detection of dna-binding molecules
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US5594121A (en) * 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5595726A (en) * 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
FR2687679B1 (en) * 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US5633360A (en) * 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (en) * 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US5293959A (en) * 1992-09-21 1994-03-15 Ingersoll-Rand Company Low pressure system lubrication for a jackhammer
IL107150A0 (en) * 1992-09-29 1993-12-28 Isis Pharmaceuticals Inc Oligonucleotides having a conserved g4 core sequence
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5472672A (en) * 1993-10-22 1995-12-05 The Board Of Trustees Of The Leland Stanford Junior University Apparatus and method for polymer synthesis using arrays
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5463657A (en) 1994-02-15 1995-10-31 Lockheed Missiles & Space Company, Inc. Detection of a multi-sequence spread spectrum signal
US5596091A (en) * 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) * 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) * 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5507796A (en) 1994-04-28 1996-04-16 Hasson; Harrith M. Method of suspending a pelvic organ and instrument for performing the method
US5525711A (en) * 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5597696A (en) * 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5580731A (en) * 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) * 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
HUP9801871A3 (en) * 1995-04-24 1999-12-28 Chromaxome Corp San Diego Methods for generating and screening novel metabolic pathways
US5783431A (en) * 1996-04-24 1998-07-21 Chromaxome Corporation Methods for generating and screening novel metabolic pathways
US6015667A (en) * 1996-06-03 2000-01-18 The Perkin-Emer Corporation Multicomponent analysis method including the determination of a statistical confidence interval

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5541313A (en) * 1983-02-22 1996-07-30 Molecular Biosystems, Inc. Single-stranded labelled oligonucleotides of preselected sequence
US4800159A (en) * 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US5650122A (en) * 1986-10-31 1997-07-22 Pasteur Sanofi Diagnostics Automated patient sample analysis instrument having tubes and reaction wells washing apparatus
US5700637A (en) * 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5436327A (en) * 1988-09-21 1995-07-25 Isis Innovation Limited Support-bound oligonucleotides
US5707796A (en) * 1990-06-11 1998-01-13 Nexstar Pharmaceuticals, Inc. Method for selecting nucleic acids on the basis of structure
US5407796A (en) * 1991-01-04 1995-04-18 The Johns Hopkins University Cystic fibrosis mutation cluster
US5352775A (en) * 1991-01-16 1994-10-04 The Johns Hopkins Univ. APC gene and nucleic acid probes derived therefrom
US5639603A (en) * 1991-09-18 1997-06-17 Affymax Technologies N.V. Synthesizing and screening molecular diversity
US5720923A (en) * 1993-07-28 1998-02-24 The Perkin-Elmer Corporation Nucleic acid amplification reaction apparatus
US5463567A (en) * 1993-10-15 1995-10-31 Caterpillar Inc. Apparatus and method for providing historical data regarding machine operating parameters
US5571639A (en) * 1994-05-24 1996-11-05 Affymax Technologies N.V. Computer-aided engineering system for design of sequence arrays and lithographic masks
US5612455A (en) * 1994-07-05 1997-03-18 Tularik, Inc. Nuclear factors and binding assay
US5684711A (en) * 1994-09-16 1997-11-04 3-Dimensional Pharmaceuticals, Inc. System, method, and computer program for at least partially automatically generating chemical compounds having desired properties
US5574656A (en) * 1994-09-16 1996-11-12 3-Dimensional Pharmaceuticals, Inc. System and method of automatically generating chemical compounds with desired properties
US5463564A (en) * 1994-09-16 1995-10-31 3-Dimensional Pharmaceuticals, Inc. System and method of automatically generating chemical compounds with desired properties
US5901069A (en) * 1994-09-16 1999-05-04 3-Dimensional Pharmaceuticals, Inc. System, method, and computer program product for at least partially automatically generating chemical compounds with desired properties from a list of potential chemical compounds to synthesize
US6434490B1 (en) * 1994-09-16 2002-08-13 3-Dimensional Pharmaceuticals, Inc. Method of generating chemical compounds having desired properties
US6295514B1 (en) * 1996-11-04 2001-09-25 3-Dimensional Pharmaceuticals, Inc. Method, system, and computer program product for representing similarity/dissimilarity between chemical compounds
US6421612B1 (en) * 1996-11-04 2002-07-16 3-Dimensional Pharmaceuticals Inc. System, method and computer program product for identifying chemical compounds having desired properties
US6453246B1 (en) * 1996-11-04 2002-09-17 3-Dimensional Pharmaceuticals, Inc. System, method, and computer program product for representing proximity data in a multi-dimensional space
US6571227B1 (en) * 1996-11-04 2003-05-27 3-Dimensional Pharmaceuticals, Inc. Method, system and computer program product for non-linear mapping of multi-dimensional data
US6016348A (en) * 1996-11-27 2000-01-18 Thomson Consumer Electronics, Inc. Decoding system and data format for processing and storing encrypted broadcast, cable or satellite video data
US6127191A (en) * 1996-12-03 2000-10-03 3-Dimensional Pharmaceuticals, Inc. Aminobenzenedicarboxylic acid-based combinatorial libraries
US6506784B1 (en) * 1999-07-01 2003-01-14 3-Dimensional Pharmaceuticals, Inc. Use of 1,3-substituted pyrazol-5-yl sulfonates as pesticides
US6518266B1 (en) * 1999-07-22 2003-02-11 3-Dimensional Pharmaceuticals 1- Aryl-3-thioalkyl pyrazoles, the synthesis thereof and the use thereof as insecticides

Also Published As

Publication number Publication date
WO1999053101A1 (en) 1999-10-21
US20050033524A1 (en) 2005-02-10
US7321828B2 (en) 2008-01-22
US20040197814A1 (en) 2004-10-07
US20060069518A1 (en) 2006-03-30
US20020028923A1 (en) 2002-03-07

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
Desmet et al. Human Splicing Finder: an online bioinformatics tool to predict splicing signals
Molina et al. Genome wide analysis of Arabidopsis core promoters
Kurella et al. DNA microarray analysis of complex biologic processes
Oliver et al. Functional genomics: high-throughput mRNA, protein, and metabolite analyses
US20030228597A1 (en) Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
Watson et al. Gene chips and arrays revealed: a primer on their power and their uses
US20090082975A1 (en) Method of selecting an active oligonucleotide predictive model
US20020045175A1 (en) Gene recombination and hybrid protein development
Chatterjee et al. A guide for designing and analyzing RNA-Seq data
Blank Next-generation analysis of deep sequencing data: bringing light into the black box of SELEX experiments
US20030200033A1 (en) High-throughput alignment methods for extension and discovery
Cuperlovic-Culf et al. Microarray analysis of alternative splicing
US20040110227A1 (en) Methods and systems for identifying putative fusion transcripts, polypeptides encoded therefrom and polynucleotide sequences related thereto and methods and kits utilizing same
Cortese The array of today
US20030032059A1 (en) Gene recombination and hybrid protein development
Faccioli et al. From single genes to co-expression networks: extracting knowledge from barley functional genomics
US20030017483A1 (en) Modulation of molecular interaction sites on RNA and other biomolecules
Bertucci et al. DNA arrays: technological aspects and applications
Kolchanov et al. GenExpress: A Computer System for Description, Analysis and Recognition of Regulatory Sequences in Eukaryotic Genome.
Levanon et al. The importance of alternative splicing in the drug discovery process
Lambert et al. Relevance and limitations of public databases for microarray design: a critical approach to gene predictions

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION