US20010026918A1 - Reduction of nonspecific hybridization by using novel base-pairing schemes - Google Patents

Reduction of nonspecific hybridization by using novel base-pairing schemes Download PDF

Info

Publication number
US20010026918A1
US20010026918A1 US09/752,213 US75221300A US2001026918A1 US 20010026918 A1 US20010026918 A1 US 20010026918A1 US 75221300 A US75221300 A US 75221300A US 2001026918 A1 US2001026918 A1 US 2001026918A1
Authority
US
United States
Prior art keywords
label
assay
complex
oligonucleotide
hybrid
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
US09/752,213
Inventor
Mark Collins
Thomas Horn
Patrick Sheridan
Brian Warner
Michael Urdea
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 US09/752,213 priority Critical patent/US20010026918A1/en
Publication of US20010026918A1 publication Critical patent/US20010026918A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • 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
    • 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
    • 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
    • C12Q1/6813Hybridisation assays
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • C12Q1/682Signal amplification
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6832Enhancement of hybridisation reaction
    • 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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/336Modified G

Definitions

  • This invention relates generally to nucleic acid chemistry and hybridization assays. More particularly, the invention relates to methods for generating a more target-dependent signal in nucleic acid hybridization assays by minimizing background noise deriving primarily from nonspecific hybridization. The invention also has applications in antisense and aptamer therapeutics and drug discovery.
  • Nucleic acid hybridization assays are commonly used in genetic research, biomedical research and clinical diagnostics.
  • a basic nucleic acid hybridization assay single-stranded analyte nucleic acid is hybridized to a labeled single-stranded nucleic acid probe and resulting labeled duplexes are detected.
  • Variations of this basic scheme have been developed to enhance accuracy, facilitate the separation of the duplexes to be detected from extraneous materials, and/or amplify the signal that is detected.
  • the present invention is directed to a method of reducing background noise encountered in any nucleic acid hybridization assay.
  • the background noise which is addressed by way of the presently disclosed techniques results from undesirable interaction of various polynucleotide components that are used in a given assay, i.e., interaction which gives rise to a signal which does not correspond to the presence or quantity of analyte.
  • the invention is useful in conjunction with any number of assay formats wherein multiple hybridization steps are carried out to produce a detectable signal which correlates with the presence or quantity of a polynucleotide analyte.
  • That assay involves the use of a two-part capturing system designed to bind the polynucleotide analyte to a solid support, and a two-part labeling system designed to bind a detectable label to the polynucleotide analyte to be detected or quantitated.
  • the two-part capture system involves the use of capture probes bound to a solid support and capture extender molecules which hybridize both to a segment of the capture probes and to a segment of the polynucleotide analyte.
  • the two-part labelling system involves the use of label extender molecules which hybridize to a segment of the polynucleotide analyte, and labeled probes which hybridize to the label extender molecules and contain or bind to a detectable label.
  • An advantage of such a system is that a plurality of hybridization steps must occur in order for label to be detected in a manner that correlates with the presence of the analyte, insofar as two distinct hybridization reactions must occur for analyte “capture,” and, similarly, two distinct hybridization reactions must occur for analyte labelling.
  • a detectable signal can be generated in a manner which does not correspond to the presence or quantity of analyte, and these will be discussed in detail below.
  • an assay with which the present invention is useful is a signal amplification method which is described in commonly assigned U.S. Pat. No. 5,124,246 to Urdea et al., the disclosure of which is incorporated herein by reference.
  • the signal is amplified through the use of amplification multimers, polynucleotides which are constructed so as to contain a first segment that hybridizes specifically to the label extenders, and a multiplicity of identical second segments that hybridize specifically to a labeled probe.
  • the degree of amplification is theoretically proportional to the number of iterations of the second segment.
  • the multimers may be either linear or branched. Branched multimers may be in the shape of a fork or a comb, with comb-type multimers preferred.
  • the present invention is also designed to increase the accuracy of detection and quantitation of polynucleotide analytes in hybridization assays.
  • the invention increases both the sensitivity and specificity of such assays, by reducing the incidence of signal generation that occurs in the absence of target, and does not involve an increase in either time or cost relative to currently used assay configurations.
  • nucleoside variants that form base pairs by virtue of “non-natural” hydrogen bonding patterns.
  • a “non-natural” base pair is one formed between nucleotidic units other than adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U).
  • A adenosine
  • T thymidine
  • C cytidine
  • G guanosine
  • U uridine
  • One such non-natural nucleoside base pair is formed between isocytosine (isoC) and isoguanine (isoG).
  • IsoC and isoG can form a base pair with a standard geometry (i.e., a “Watson-Crick base pair”) but involving hydrogen bonding other than that involved in the bonding of cytosine (C) to guanine (G), as shown below:
  • the present invention provides methods and kits for detecting nucleic acid analytes in a sample.
  • the methods represent improvements in nucleic acid hybridization assays, such as in situ hybridization assays, Southerns, Northerns, dot blots and polymerase chain reaction assays.
  • the methods represent improvements on solution phase sandwich hybridization assays which involve binding the analyte to a solid support, labelling the analyte, and detecting the presence of label on the support.
  • Preferred methods involve the use of amplification multimers which enable the binding of significantly more label in the analyte-probe complex, enhancing assay sensitivity and specificity.
  • an assay in which one or more nucleotidic units which are capable of forming base pairs and which are other than adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U), are incorporated into non-target hybridizing oligonucleotide segments, i.e., “universal” segments, of nucleic acid hybridization assay components.
  • A adenosine
  • T thymidine
  • C cytidine
  • G guanosine
  • U uridine
  • an assay in which at least one first nucleotidic unit other than A, T, C, G, or U capable of forming a base pair with a second nucleotidic unit other than A, T, C, G, or U, is incorporated into nucleic acid sequences of assay components which are complementary to nucleic acid sequences present in assay components other than the target molecule.
  • first nucleotidic unit other than A, T, C, G, or U capable of forming a base pair with a second nucleotidic unit other than A, T, C, G, or U
  • R represents a backbone which will allow the bases to form a base pair with a complementary nucleotidic unit when incorporated into a polynucleotide
  • R′ is, for example, hydrogen, methyl, ⁇ - or ⁇ -propynyl, bromine, fluorine, iodine, or the like.
  • the first and second nucleotidic units interchangeably consist of isocytidine and isoguanosine, as shown in Formula (I).
  • an assay in which the melt temperature T m1 of the complex formed between the analyte and the support-bound capture probes, mediated by one or more distinct capture extender molecules, and/or the label extender and amplifier or preamplifiers, is significantly lower than the melt temperature T m1 of the complex formed between the labeled probes and the amplifier.
  • the assay is carried out under conditions which initially favor the formation of all hybrid complexes. The conditions are then altered during the course of the assay so as to destabilize the T m1 hybrid complexes.
  • the invention additionally encompasses a method for carrying out a hybridization assay in which each of the aforementioned techniques are combined, i.e., in which nucleotidic units other than A, T, G, C, or U are incorporated into universal segments of assay components and in which the melt temperature of T m1 hybrid complexes is significantly lower than the melt temperature of T m2 hybrid complexes.
  • the invention encompasses a novel method for synthesizing isoguanosine or 2′-deoxy-isoguanosine.
  • FIG. 1 diagrams a solution phase sandwich hybridization assay of the prior art with heavy lines indicating the universal sequences.
  • FIG. 2 portrays a method for binding probes to double-stranded DNA with heavy lines indicating the universal sequences.
  • FIG. 3 depicts the use of non-natural nucleotide-containing probes and competimers to block non-specific hybridization.
  • polynucleotide and “oligonucleotide” shall be generic to polydeoxyribonucleotides (containing 2-deoxy-D-ribose), to polyribonucleotides (containing D-ribose), to any other type of polynucleotide which is an N- or C-glycoside of a purine or pyrimidine base, and to other polymers containing non-nucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids (PNAs)) and polymorpholino (commercially available from the Anti-Virals, Inc., Corvallis, Oregon, as NeugeneTM polymers), and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base pairing and base stacking, such as is found in DNA and RNA.
  • PNAs peptide nucleic acids
  • polynucleotide and “oligonucleotide,” and these terms will be used interchangeably. These terms refer only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA, DNA:RNA hybrids, and hybrids between PNAs and DNA or RNA, and also include known types of modifications, for example, labels which are known in the art, methylation, “caps,” substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), with negatively charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), and with positively charged linkages (e.g., aminoalklyphosphoramidates, aminoalkylphosphotries
  • nucleoside and nucleotide will include those moieties which contain not only the known purine and pyrimidine bases, but also other heterocyclic bases which have been modified. Such modifications include methylated purines or pyrimidines, acylated purines or pyrimidines, or other heterocycles. Modified nucleosides or nucleotides will also include modifications on the sugar moiety, e.g., wherein one or more of the hydroxyl groups are replaced with halogen, aliphatic groups, or are functionalized as ethers, amines, or the like.
  • the term “nucleotidic unit” is intended to encompass nucleosides and nucleotides.
  • modifications to nucleotidic units include rearranging, appending, substituting for or otherwise altering functional groups on the purine or pyrimidine base which form hydrogen bonds to a respective complementary pyrimidine or purine.
  • the resultant modified nucleotidic unit may form a base pair with other such modified nucleotidic units but not with A, T, C, G or U.
  • Standard A-T and G-C base pairs form under conditions which allow the formation of hydrogen bonds between the N 3 -H and C 4 -oxy of thymidine and the N 1 and C 6 -NH 2 , respectively, of adenosine and between the C 2 -oxy, N 3 and C 4 -NH 2 , of cytidine and the C 2 -NH 2 , N 1 -H and C 6 -oxy, respectively, of guanosine.
  • guanosine (2-amino-6-oxy-9- ⁇ -D-ribofuranosyl-purine) may be modified to form isoguanosine (2-oxy-6-amino-9-, ⁇ -D-ribofuranosyl-purine).
  • polynucleotide analytel refers to a single- or double-stranded nucleic acid molecule which contains a target nucleotide sequence.
  • the analyte nucleic acids may be from a variety of sources, e.g., biological fluids or solids, food stuffs, environmental materials, etc., and may be prepared for the hybridization analysis by a variety of means, e.g., proteinase K/SDS, chaotropic salts, or the like.
  • polynucleotide analyte is used interchangeably herein with the terms “analyte,” “analyte nucleic acid,” “target” and “target molecule.”
  • target region or “target nucleotide sequence” refers to a probe binding region contained within the target molecule.
  • target sequence refers to a sequence with which a probe will form a stable hybrid under desired conditions.
  • probe refers to a structure comprised of a polynucleotide, as defined above, which contains a nucleic acid sequence complementary to a nucleic acid sequence present in the target molecule.
  • the polynucleotide regions of probes may be composed of DNA, and/or RNA, and/or synthetic nucleotide analogs.
  • binding sequences need not have perfect complementarity to provide stable hybrids. In many situations, stable hybrids will form where fewer than about 10% of the bases are mismatches, ignoring loops of four or more nucleotides. Accordingly, as used herein the term “complementary” refers to an oligonucleotide that forms a stable duplex with its “complement” under assay conditions, generally where there is about 90% or greater homology.
  • nucleic acid multimer or “amplification multimer” are used herein to refer to a linear or branched polymer of the same repeating single-stranded oligonucleotide segment or different single-stranded polynucleotide segments, each of which contains a region where a labeled probe can bind, i.e., contains a nucleic acid sequence complementary to a nucleic acid sequence contained within a labeled probe; the oligonucleotide segments may be composed of RNA, DNA, modified nucleotides or combinations thereof.
  • At least one of the segments has a sequence, length, and composition that permits it to bind specifically to a labeled probe; additionally, at least one of the segments has a sequence, length, and composition that permits it to bind specifically to a label extender or preamplifier.
  • such segments will contain approximately 15 to 50, preferably 15 to 30, nucleotides, and will have a GC content in the range of about 20% to about 80%.
  • the total number of oligonucleotide segments in the multimer will usually be in the range of about 3 to 1000, more typically in the range of about 10 to 100, and most typically about 50.
  • the oligonucleotide segments of the multimer may be covalently linked directly to each other through phosphodiester bonds or through interposed linking agents such as nucleic acid, amino acid, carbohydrate or polyol bridges, or through other cross-linking agents that are capable of cross-linking nucleic acid or modified nucleic acid strands.
  • the multimer may be comprised of oligonucleotide segments which are not covalently attached, but are bonded in some other manner, e.g., through hybridization. Such a multimer is described, for example, in U.S. Pat. No. 5,175,270 to Nilsen et al.
  • the site(s) of linkage may be at the ends of the segment (in either normal, 3′-5′ orientation or randomly oriented) and/or at one or more internal nucleotides in the strand.
  • linear multimers the individual segments are linked end-to-end to form a linear polymer.
  • three or more oligonucleotide segments emanate from a point of origin to form a branched structure.
  • the point of origin may be another nucleotide segment or a multifunctional molecule to which at least three segments can be covalently bound.
  • multimers are “fork-like,” “comb-like” or combination “fork-” and “comb-like” in structure, wherein “comb-like” multimers, the preferred multimers herein, are polynucleotides having a linear backbone with a multiplicity of sidechains extending from the backbone.
  • the pendant segments will normally depend from a modified nucleotide or other organic moiety having appropriate functional groups to which oligonucleotides may be conjugated or otherwise attached.
  • the multimer may be totally linear, totally branched, or a combination of linear and branched portions. Typically, there will be at least two branch points in the multimer, more preferably at least three, more preferably in the range of about 5 to 30, although in some embodiments there may be more.
  • the multimer may include one or more segments of double-stranded sequences. Further information concerning multimer synthesis and specific multimer structures may be found in commonly owned U.S. Pat. No. 5,124,246 to Urdea et al.
  • a “preamplifier” molecule may also be used, which serves as a bridging moiety between the label extender molecules and the amplification multimers. In this way, more amplifier and thus more label is bound in any given target-probe complex.
  • Preamplifier molecules may be either linear or branched, and typically contain in the range of about 30 to about 3000 nucleotides.
  • the preamplifier molecule binds to at least two different label extender molecules, such that the overall accuracy of the assay is increased (i.e., because, again, a plurality of hybridization events are required for the probe-target complex to form).
  • a “biological sample” refers to a sample of tissue or fluid isolated from an individual, including but not limited to, for example, plasma, serum, spinal fluid, semen, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs, and also samples of in vitro cell culture constituents (including but not limited to conditioned medium resulting from the growth of cells in cell culture medium, putatively virally infected cells, recombinant cells, and cell components).
  • Preferred uses of the present method are in detecting and/or quantitating nucleic acids as follows: (a) viral nucleic acids, such as from hepatitis B virus (“HBV”), hepatitis C virus (“HCV”), hepatitis D virus (“HDV”), human immunodeficiency virus (“HIV”), and the herpes family of viruses, including herpes zoster (chicken pox), herpes simplex virus I & II, cytomegalovirus, Epstein-Barr virus, and the recently isolated Herpes VI virus; (b) bacterial nucleic acids, such as Chlamydia, Mycobacterium tuberculosis, etc.; and (c) numerous human sequences of interest.
  • viral nucleic acids such as from hepatitis B virus (“HBV”), hepatitis C virus (“HCV”), hepatitis D virus (“HDV”), human immunodeficiency virus (“HIV”)
  • herpes family of viruses including herpe
  • nonspecific hybridization is used to refer to those occurrences in which a segment of a first polynucleotide which is intended to hybridize to a segment of a selected second polynucleotide also hybridizes to a third polynucleotide, triggering an erroneous result, i.e., giving rise to a situation where label may be detected in the absence of target molecule.
  • hybridizes is not meant to exclude non-Watson-Crick base pairing.
  • nonspecific binding is used to refer to those occurrences in which a polynucleotide binds to the solid support, or other assay component, through an interaction--which may be either direct or indirect--that does not involve hydrogen bonding to support-bound polynucleotides.
  • Label extender molecules also referred to herein as “label extenders,” contain regions of complementarity vis-à-vis the analyte polynucleotide and to the amplifying multimer (“AMP”). If a preamplifier is used (not shown in the figure), the label extender molecules will bind to this intermediate species rather than directly to the amplifier multimer. If neither preamplifier or amplifier is used, the label extender molecules will bind directly to a sequence in the labeled probe (“LP”).
  • AMP amplifying multimer
  • label extender molecules are single-stranded polynucleotide chains having a first nucleic acid sequence L- 1 complementary to a sequence of the analyte polynucleotide, and a second universal region having a multimer recognition sequence L- 2 complementary to a segment M- 1 of label probe, amplifier multimer or preamplifier.
  • LPs Labeled probes
  • L- 3 complementary to a nucleic acid sequence M- 2 present within the repeating oligonucleotide units of the multimer and are bound to, or structured so as to bind to, a label which provides, directly or indirectly, a detectable signal.
  • capture extender molecules also referred to herein as “capture extenders,” bind to the analyte polynucleotide and to capture probes, which are in turn bound to a solid support.
  • capture extender molecules are single-stranded polynucleotide chains having a first polynucleotide sequence region containing a nucleic acid sequence C- 1 which is-complementary to a sequence of the analyte, and a second, noncomplementary region having a capture probe recognition sequence C- 2 .
  • the sequences C- 1 and L- 1 are nonidentical, noncomplementary sequences that are each complementary to physically distinct sequences of the analyte.
  • Capture probes bind to the capture extenders and to a solid support.
  • capture probes have a nucleic acid sequence C- 3 complementary to C- 2 and are covalently bound to (or capable of being covalently bound to) a solid support.
  • solution phase hybridization assays carried out using the system illustrated in FIG. 1 proceed as follows.
  • Single-stranded analyte nucleic acid is incubated under hybridization conditions with the capture extenders and label extenders.
  • the resulting product is a nucleic acid complex of the analyte polynucleotide bound to the capture extenders and to the label extenders.
  • This complex may be subsequently added under hybridizing conditions to a solid phase having the capture probes bound to the surface thereof; however, in a preferred embodiment, the initial incubation is carried out in the presence of the support-bound capture probes.
  • the resulting product comprises the complex bound to the solid phase via the capture extender molecules and capture probes.
  • the solid phase with bound complex is then separated from unbound materials.
  • An amplification multimer preferably a comb-type multimer as described above, is then optionally added to the solid phase-analyte-probe complex under hybridization conditions to permit the multimer to hybridize to the LEs; if preamplifier probes are used, the solid phase-analyte-probe complex is incubated with the preamplifier probes either along with the amplifier multimer or, preferably, prior to incubation with the amplifier multimer. The resulting solid phase complex is then separated from any unbound preamplifier and/or multimer by washing. The labeled probes are then added under conditions which permit hybridization to LEs, or, if an amplification multimer was used, to the repeating oligonucleotide segments of the multimer.
  • the resulting solid phase labeled nucleic acid complex is then washed to remove unbound labeled oligonucleotide, and read.
  • the components represented in FIG. 1 are not necessarily drawn to scale, and that the amplification multimers, if used, contain a far greater number of repeating oligonucleotide segments than shown (as explained above), each of which is designed to bind a labeled probe.
  • the primary focus of the present method is on eliminating the sources of background noise, by minimizing the interaction of capture probes and capture extender molecules with the labeled probes, label extender molecules and amplifiers, reducing the likelihood that incorrect moieties will bind to the support-bound capture probes.
  • Hybridization between complementary oligonucleotide sequences is premised on the ability of the purine and pyrimidine nucleotides contained therein to form stable base pairs.
  • the five naturally occurring nucleotides adenosine (A), guanosine (G), thymidine (T), cytidine (C) and uridine (U) form the purine-pyrimidine base pairs G-C and A-T(U).
  • the binding energy of the G-C base pair is greater than that of the A-T base pair due to the presence of three hydrogen-bonding moieties in the former compared with two in the latter, as shown below:
  • oligonucleotide molecules are designed to contain nucleic acid sequences which are complementary to and, therefore, hybridize with nucleic acid sequences in other assay components or in the target molecule, as explained in detail above.
  • the method of the invention reduces nonspecific hybridization by incorporating non-natural nucleotidic units into universal oligonucleotide segments of assay components which are capable of forming unique base pairs. Furthermore, the method of the invention reduces the contribution of nonspecific binding of assay components by separating detectably labelled assay components which are associated with the presence and/or quantity of a target analyte from those which are nonspecifically bound and contribute to assay background noise.
  • a hybridization assay in which nucleotidic units other than A, T, C, G and U which are capable of forming unique base pairs are incorporated into hybridizing oligonucleotide segments of assay components which are not target analyte specific and thus will be less likely to form stable hybrids with target-specific probe sequences or with extraneous non-target nucleic acid sequences.
  • nucleotidic units may be incorporated in complementary nucleic acid sequences C- 2 /C- 3 , L- 2 /M- 1 and L- 3 /M- 2 .
  • the hybridizing oligonucleotide segments of assay components which are complementary to nucleic acid segments of the target molecule are constructed from naturally occurring nucleotides (i.e., A, T, C, G or U). Oligonucleotide segments which contain nucleotidic units may be constructed by replacing from about 15% to about 100% of the naturally occurring nucleotides with the nucleotidic unit counterpart. Preferably, every third or fourth base in an oligonucleotide will be replaced with a nucleotidic unit capable of forming a unique base pair. It will be apparent to those skilled in the art that as the percent of replacement nucleotidic units is increased, nonspecific hybridization is decreased concomitantly. However, complete replacement will require at least two new base pairs in order to maintain sufficient sequence diversity to preclude nonspecific hybridization among the universal sequences.
  • the phenomenon of target-independent signal generation is addressed by providing a hybridization assay which is configured such that the melt temperature T m1 of the C- 2 /C- 3 hybrid or the L- 2 /M- 1 hybrid is significantly lower than the melt temperature T m2 of the L- 3 /M- 2 hybrid.
  • This method is premised on the design and construction of hybrid complexes such that the melt temperature T m1 is at least about 5° C. lower than, preferably at least about 10° C. lower than, more preferably at least about 20° C. lower than the melt temperature T m2 .
  • Preferred stringency controls are pH and salt concentration: one assay step is conducted at a pH or salt concentration which destabilizes the hybrid complex formed between capture probe/capture extender or destabilizes the hybrid formed between label extender/amplifier (or preamplifier).
  • a preferred step at which stringency is exercised is the addition of substrate.
  • the hybridization assay is conducted under conditions which favors the stability of hybrid complexes formed between all assay components and thereafter, with the addition of label substrate, the stringency is altered to destabilize hybrid complexes such as the capture probe/capture extender, or label extender/amplifier (preamplifier), and the like, with the proviso that the labeled probe is not released from the label extender or amplifier.
  • Another embodiment of the invention represents one means by which the above embodiment of the invention may be effected is by configuring the hybridization assay such that the complementary nucleotide sequences which form T m1 hybrid complexes are shorter than those which form T m2 hybrid complexes. It will be appreciated by those of skill in the art that, with shorter complementary nucleotide sequences, the opportunity for sequence diversity therein decreases. This diversity may be maintained, however, by incorporating into the complementary sequences a non-natural base pair, e.g., an isoC-isoG base pair.
  • a plurality of capture probes are affixed to a solid surface. Most often, the surface area available for non-specific binding is controlled by incubating the surface with DNA from, e.g., salmon sperm or calf thymus. However, the presence of this DNA increases the potential for nonspecific hybridization of assay components to the solid support and, therefore, increased background noise. Replacement of these natural DNAs with synthetic DNAs containing non-natural bases will minimize the non-specific hybridization and the non-specific binding.
  • these polynucleotides will be prepared by 3′ tailing short oligonucleotides with mixtures of nucleotides by methods well known in the art.
  • short, nearly random-sequence oligonucleotides containing non-natural nucleotides can be joined together to form polynucleotides.
  • Branched DNAs can be conveniently used for this purpose.
  • the block sequence -TNVN-F-TNVN-J-TNVN-, wherein F is isoC and J is isoG can be prepared and chemically joined to form a polymer.
  • the advantage of using this approach over using the enzymatic 3′ tailing approach is the elimination of homopolymer/homooligomer sequences.
  • W091/11535, W091/09865, W091/04753, W090/13641, W091/13080 and, WO 91/06629 are oligonucleotides that bind to and disable or prevent the production of the mRNA responsible for generating a particular protein.
  • Conventional antisense molecules are generally capable of reacting with a variety of oligonucleotide species. However, due to their length (generally oligonucleotide sequences of up to 30 nucleotidic units), such antisense molecules present problems associated with nonspecific hybridization with non-target species.
  • dimerization domains may be designed to have tails with complementary sequences containing non-natural nucleotidic units and thereby provide highly efficient and specific binding to the target molecule without increasing non-specific hybridization to non-target molecules.
  • FIG. 2 The idea is illustrated in FIG. 2 with a double-stranded DNA target.
  • strand displacement may be used to pry apart double-stranded DNA.
  • AT-rich promoter sequences under superhelical stress which are S 1 nuclease-sensitive and are thus already partially single-stranded, are a particularly preferred site for this type of antigene application.
  • Short oligonucleotides would be used to maximize specificity; their binding energy to the target would be enhanced by joining them together to form a network of oligonucleotides.
  • the short universal sequences which will not form stable base-pairs in the absence of target, contain isoC and isoG to limit nonspecific hybridization of the probes with the human sequences.
  • the universal sequences Upon binding of probes 1 , 2 and 3 to the target, the universal sequences will be in sufficiently close proximity that their effective concentration will be significantly increased. The universal sequences will then pair, resulting in a further increase in the strength of the binding.
  • RNA targets may also be used in conjunction with this approach.
  • Target molecules as distinct from “target analytes,” include polymers such as proteins, polysaccharides, oligonucletides or other macromolecules, and small molecules such as drugs, metabolites, toxins, or the like, to which an aptamer is designed to bind.
  • an oligonucleotide is constructed wherein an n-mer, preferably a randomized sequence of nucleotides thereby forming a “randomer pool” of oligonucleotides, is flanked by two polymerase chain reaction (PCR) primers.
  • PCR polymerase chain reaction
  • oligonucleotides which bind the target molecule are: (a) separated from those oligonucleotides which do not bind the target molecule using conventional methods such as filtration, centrifugation, chromatography, or the like; (b) dissociated from the target molecule; and (c) amplified using conventional PCR technology to form a ligand-enriched pool of oligonucleotides. Further rounds of binding, separation, dissociation and amplification are performed until an aptamer with the desired binding affinity, specificity or both is achieved. The final aptamer sequence identified can then be prepared chemically or by in vitro transcription. When preparing such aptamers, selected base pairs are replaced with nonnatural base pairs to reduce the likelihood of the aptamers hybridizing to human nucleic acids.
  • isodG and isodc can be included among the sequences in the randomer DNA sequence pool.
  • the number of possible randomeric structures that may recognize proteins or other important biomolecules is increased by synthesizing strands of DNA out of six or more nucleotides, rather than the conventional four nucleotide A, T, G and C. This is turn improves the chances of identifying a sequence which binds with greater affinity and/or specificity to the target molecule.
  • the conserved oligonucleotide sequences selected for may have unwanted hybridization to cellular sequences. This nonspecific hybridization can be reduced using nonnatural bases in the selection process. Nucleotides that are not recognized by human RNA and DNA polymerases but which are recognized by certain phage or bacterial polymerases are particularly useful in this application.
  • a second use for the instant invention in the SELEX process is in the preparation of a final aptamer construct with minimized nonspecific hybridization.
  • aptamers which display predetermined binding affinity, specificity or other target molecule recognition characteristics are selected from a pool of RNA or DNA sequences using the SELEX process. These target molecule recognition characteristics are determined by the secondary structure of the aptamer which is maintained, in part, by the formation of intramolecular oligonucleotide hybrid complexes.
  • base pairs in certain intramolecular hybrid complexes are highly preferred for maintaining the secondary structure and, therefore, the target molecule recognition and binding characteristics of the aptamer, i.e., there will base pairs which are preferably G-C or A-T.
  • base pairs which are preferably G-C or A-T.
  • base pairs in these intramolecular hybrid complexes for example, in the base-pairing portion of the stem loop, which may be replaced by any pair of complementary nucleotides, referred to herein as N-N′ base pairs, without altering the secondary structure of the aptamer.
  • a simple replacement of selected N-N′ base pairs and G-C and C-G base pairs in the final aptamer construct with isoG-isoC or isoC-isoG will reduce nonspecific hybridization to nontarget oligonucleotide sequences. Since the isoC-isoG base pair is isoenergetic with the C-G base pair, the basic shape of the molecule and the strength of the hairpins will be very similar. A base pair isoenergetic with A-U would be desirable for replacing base pairs where the winning sequences show a strong preference for A-U or U-A over C-G. These substitutions have the effect of making the aptamers more specific for the target molecule by limiting their potential for unwanted hybridization to cellular RNA and DNA-sequences.
  • isoC-isoG base pairs can comprise ribonucleotides or deoxyribonucleotides.
  • a chimeric aptamer (composed of both ribonucleotides and deoxyribonucleotides) molecule can be made chemically.
  • the ribo-isoGTP and ribo-isoCTP (with suitable 2′ protection) can be used to prepare the aptamer by in vitro transcription of DNA templates containing isoC and isoG.
  • In situ hybridization lacks sufficient sensitivity to detect a single molecule of target analyte.
  • In situ PCR (see, e.g., Bagasra et al. (1993) J. Immunological Methods 158:131-145) has been developed to meet this sensitivity need; however, quantitation is not as precise with the PCR method.
  • An alternative would use multiple label extender probes to bind the target analyte.
  • the label extenders would bind either preamplifiers or amplifiers. If used, preamplifiers would-bridge label extenders and amplifiers.
  • the amplifiers would bind labeled probes, which would preferably be detected by luminescence (fluorescence if the sensitivity is high enough).
  • L- 2 /M- 1 and M- 2 /L- 3 would consist of short oligonucleotides containing optimally between 15-30% isoC and isoG to reduce unwanted hybridization to human sequences.
  • a fourth base-pair could be used to further reduce the representation of the natural bases in these sequences.
  • nonspecific binding as well as nonspecific hybridization can be reduced by using nonnatural base pairs. Random polymers or nearly random block copolymers consisting of 6-8 different nucleotides could be used to reduce nonspecific binding of the amplifier and labeled probes to the cellular constituents that have high affinity for polynucleotides. Thus nonspecific binding will be reduced without risking an increase in nonspecific hybridization by introducing natural sequences from calf or salmon, as is commonly done.
  • the present invention also finds several uses in PCR and other exponential amplification technologies. For example, in nested PCR, after the target analyte is initially amplified and then diluted several thousand-fold, it is common to use a 5′ overhang on one primer for capture and a 5′ overhang on the other primer for labeling. A spacer that cannot be read by the polymerase is inserted so that the overhangs remain single-stranded (see, e.g., Newton et al. (1993) Nucl. Acids Res. 21:1155-1162). The generic sequences in these 5′ overhangs can be prepared to contain modified base-pairs to reduce the frequency of priming on nontargets.
  • isodC or isodG in the first base of the 5′ overhang can be used in place of the currently used spacers; the polymerase cannot read isodC or isodG because it will have no isodGTP or isodCTP to put in place of it. Because the polymerase may put T into the polymer at a low frequency when it detects isodG in what was the primer, it is preferable to use isoC as the first base in the 5′ overhang.
  • 2′-deoxy-isoguanosine has been synthesized: 1) from 2′-deoxyadenosine via 2′-deoxyadenosine-(N 1 -oxide) by direct photolysis under basic conditions (Switzer et al. (1993), supra); 2) from 2-chloro-2′-deoxyadenosine by direct photolysis under basic conditions (Seela et al. (1992) Helv. Chim.
  • Tetrahedron Letters 31:319-322 described a convenient synthesis of O 6 -phenyl-2′-deoxyguanosine using a procedure involving treatment of 2′-deoxyguanosine with trifluoroacetic anhydride/pyridine followed by in situ displacement with phenol.
  • O 6 -phenyl moieties into 2′-deoxyguanosine has been described by Reese et al. (1984) J. Chem. Soc., Perkin Trans.
  • An isoguanosine-like compound was generated from 2-(methylmercapto)-6-amino-pyrazolopyrimidine ribonucleoside by S-oxidation, producing 2-(methylsulfonyl)-6-amino-pyrazolopyrimidine ribonucleoside, followed by displacement with NaOH to give the isoguanosine analogue (Cottam et al. (1983) Nucleic Acids Research 11:871-882).
  • R 1 is selected from the group consisting of hydrogen, hydroxyl, sulfhydryl, halogeno, amino, alkyl, allyl and -OR 2 , where R 2 is alkyl, allyl, silyl or phosphate, comprises:
  • step (b) reacting the product of step (a) with a reagent suitable to convert the O 6 -oxy moiety into a functional group which is susceptible to nucleophilic displacement, thereby producing a functionalized O 6 moiety;
  • step (c) reacting the product of step (c) with a nucleophilic reagent to displace the functionalized O 6 moiety;
  • step (d) reacting the product of step (d) with a reagent suitable to deprotect the protected 3′ and 5′ hydroxyl groups.
  • the conversion of guanosine or 2′-deoxyguanosine to isoguanosine or 2′-deoxy-isoguanosine, respectively, may be effected by protecting the hydroxyl groups on the sugar moiety using a suitable reagent, e.g., TBDMS, benzoyl chloride, acetic anhydride, or the like.
  • a suitable reagent e.g., TBDMS, benzoyl chloride, acetic anhydride, or the like.
  • one or more of the hydroxyl groups on the sugar moiety may be replaced by halogen, aliphatic groups, or may be functionalized as ethers, amines, or the like.
  • the product is isolated and the O 6 is modified such that it can be displaced by a suitable nucleophile.
  • displaceable groups examples include, for example, CH 3 -S-C 6 H 4 -O 6 -, C 6 H 5 -SO 2 -O 6 -, C 6 H 5 -O 6 -, 4-nitro-C 6 H 4 -O 6 -, 2,4,6-trinitro-C 6 H 2 -O 6 -, or the like.
  • the 2-amino group is then transformed to the oxy function using an alkyl nitrite, or other suitable agent as known in the art (see, Nair et al. (1982), supra; Trevidi (1991), supra; or Steinbrecher et al. (1993), supra).
  • the product is reacted with a suitable nucleophile, e.g., NH 4 OH, or other aminoalkyl, aminoaryl, aminoheteroalkyl, aminoheteroaryl containing a terminal -NH 2 , -SH, -COOH, or the like, thereby displacing the modified O 6 leaving group.
  • a suitable nucleophile e.g., NH 4 OH, or other aminoalkyl, aminoaryl, aminoheteroalkyl, aminoheteroaryl containing a terminal -NH 2 , -SH, -COOH, or the like.
  • Deprotection of the protected hydroxyl groups may be effected by treatment with, for example, base or fluoride.
  • O 6 -(4-methylthiophenyl) will serve as an exemplary displaceable group.
  • its use is for the purpose of describing particular embodiments only and is not intended to be limiting.
  • N 6 -alkylated isoguanosine derivatives can be readily synthesized by using an alkyl amine as the nucleophile.
  • hexanediamine may be used to displace the O 6 -(4-methylthiophenyl) to form N 6 -(6-aminohexyl)-isoguanosine. Protection of the aminohexyl group (e.g., as the trifluoroacetamido derivative) and subsequent conversion into a phosphoramidite reagent provides a functionalizable isoguanosine analog which may be incorporated in any desired position in an oligonucleotide for further post-synthesis derivatization.
  • O 2 -(4-methylthiophenyl)-2′-deoxyxantosine in its fully protected phosphoramidite form (O 2 -(4-methylthiophenyl)-5′-O-DMT-3′O-(BCE-diisopropylphosphoramidite)-2′-deoxyxantosine) may be used as a convertible derivative following incorporation into an oligonucleotide.
  • Post-synthesis displacement of the O 2 -(4-methylthiophenyl) from the O 2 -(4-methylthiophenyl)-2′-deoxyxantosine with an alkyldiamine, or other functionalized alkyl amine produces N 6 -(aminoalkyl)-2′-deoxy-isoguanosine-containing oligonucleotides.
  • the derivatized isoguanosine can serve as a site for introduction of a label or other reporter molecule specifically at the functionalized isoguanosine residue.
  • Derivatives of isocytidine or 2′-deoxy-isocytidine may be prepared in which the glycosidic bond is stabilized against exposure to dilute acid during oligonucleotide synthesis.
  • N 2 -amidine derivatives have been described for 2′-deoxyadenosine by, for example, McBride et al. (1986) J. Am. Chem. Soc. 108:2040-2048, Froehler et al. (1983) Nucleic Acids Res. 11:8031-8036 and Pudlo et al. (1994) Biorg. Med. Chem. Lett. 4:1025-1028.
  • N 2 -(N,N-di(X)formamidino)-2′-deoxy-isocytidine was synthesized by the following procedure.
  • X is n-butyl.
  • X may be C 2 -C 10 alkyl, aryl, heteroalkyl, heteroalkyl, or the like.
  • N-di-n-butylformamide dimethylacetal was synthesized by transamination of N,N-methylformamide dimethylacetal with di-n-butylamine as described in McBride et al. (1986), supra, Froehler et al. (1983), supra, and Pudlo et al. (1994), supra. Ten mmole of 2′-deoxy-5-methyl-isocytidine was suspended in 100 ml methanol and 10 mmole of N,N-di-n-butylformamide dimethylacetal was added. After 2 hours at room temperature with stirring, a clear solution resulted.
  • isocytidine derivatives may be prepared which provide functionalizable substituents by which detectable labels may be incorporated into a specific position of an oligonucleotide.
  • 5-alkylated 2′-deoxyuridine derivatives have been described, e.g., 5-[N-(6-trifluoroacetylaminohexyl)-3-(E)acrylamido]-2′-deoxyuridine, by Ruth (1991) Oligodeoxynucleotides with Reporter Groups Attached to the Base , in Eckstein (ed.) Oligonucleotides and Analogues , IRL press, p. 255-282.
  • Such 5-position derivatives have been found not to obstruct base pair hybridization patterns.
  • the chemistry described by Ruth can be used to synthesize 5-[N-(6-trifluoroacetylaminohexyl)-3-(E)acrylamido]-2′-deoxy-isocytidine, thereby providing a functionalized isocytidine which may be detectably labelled at selected isoguanosine*isocytidine base pairs.
  • Kits for carrying out nucleic acid hybridization assays will comprise in packaged combination at least one hybridizing oligonucleotide probe, a segment of which is capable of forming a hybrid complex with the analyte, and a means for detecting the hybrid complex, wherein the at least one hybridizing oligonucleotide probe comprises a first nucleotidic unit which, under conditions in which A-T and G-C base pairs are formed, will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U).
  • the reagents will typically be in separate containers in the kit.
  • the kit may also include a denaturation reagent for denaturing the analyte, hybridization buffers, wash solutions, enzyme substrates, negative and positive controls and written instructions for carrying out the assay.
  • polynucleotides of the invention may be assembled using a combination of solid phase direct oligonucleotide synthesis, enzymatic ligation methods, and solution phase chemical synthesis as described in detail in commonly assigned U.S. patent application Ser. No. 07/813,588.
  • a space separates the 3′nontarget binding region from the target-binding region of each probe.
  • the assay was run essentially as described in copending application 08/164,388 (Urdea et al). Briefly, after overnight hybridization at 63° C. in microtiter wells containing capture probes complementary to the nontarget binding region of the capture extenders, the plates were cooled at room temperature for 10 min, washed twice with a buffer containing 0.1x SSC (15 mM NaCl; 1.5 mM sodium citrate; pH 7 . 0 ), 0.1% sodium dodecyl sulfate.
  • a 15 ⁇ 3 (15 “arms” each with 3 alkaline phosphatase probe binding sites) branched DNA amplifier (100 fm) complementary to the 3′ nontarget binding region of the label extender was added to the wells and the incubation was continued for 30 min at 53° C. after which the plates were cooled and washed as above. After the addition of an alkaline phosphatase probe (200 fm) to the wells and a further incubation for 15 min at 53° C., the plates were again cooled and washed as above. Three additional washes were done with a 0.1x SSC buffer.
  • pool B capture extenders does not increase the net signal, but does increase the noise about one hundred-fold
  • Computer analysis of the sequences involved showed that capture extender # 8 of pool B has extensive homology with the T 20 —LLA 2 sequence of the branched DNA amplifier (including a 9mer oligo(dA)--oligo(dT)), while capture extender # 9 of pool B has extensive homology with the BLA3c sequence of the branched DNA amplifier.
  • nucleotide sequences are constructed which are interrupted by nucleotides that do not form stable base pairs with “natural” nucleobases, thereby inhibiting the hybridization of such sequences with natural sequences.
  • every third or fourth base in the universal sequence would be a modified nucleotide that does not pair with A,C, G, or T(U).
  • the data also show the specificity of the isoC*isoG base-pair.
  • the isoC*G and isoG*C pairs behave as mismatches.
  • the destabilization in degrees C is approximated by the percent mismatching.
  • T m the percent mismatching.
  • the observed 8° C. change when the C*G or isoC*isoG matches are compared with the mismatches is similar to the change which would occur in an average mismatch with A, T, C, and G code.
  • IsoG exists in at least two tautomeric forms, the keto and the enol.
  • the keto form is favored in aqueous solvents and the enol is favored in organic solvents (Sepiol et al. (1976) Zeitschrift fuer Naturforschung 31C:361-370).
  • the isoG enol tautomer can, in principle, form two hydrogen bonds to dT, making it analogous to the A*T base pair. If the enol tautomer were present at significant levels in the hybridization buffer, the specificity of isoC*isoG base pair would be limited. However, the observed T m in the isoG*T mismatch was 53° C., essentially the same as the other mismatches.
  • oligonucleotide hybrids are stable at pH 5 and pH 10 . Below pH 5 , C and A become protonated, while above pH 10 , G and T begin to lose their imino protons. Thus, below pH 5 and above pH 10 , nucleic acid hybrids show reduced stability.
  • the data of Table 2 show that the isoC*isoG base pair has normal acid stability. However, both the isoG*isoC hybrid and the G*C hybrid show an unusual ⁇ 9° C. change in T m over a 1.6 unit pH increase. This is probably due to their very short length.
  • the substrate buffer used is typically pH 9 . 5 to 10 . 5 .
  • the target With a capture probe with the proper base lability, the target will come off the surface and could be detected in another well. The background will be left behind.
  • Minimization of capture extender binding to the support by the methods disclosed in copending application 08/164,388 (Urdea et al.) will reduce background noise caused by release of molecules nonspecifically bound to capture probes through capture extenders.
  • the capture probe-capture extender hybrids would be selected to have considerably more base lability (i.e., higher T m at a given pH) than the amplifier and labeled probe and the amplifier and label extender hybrids.
  • L- 2 /M- 2 hybrid of FIG. 1 could be the base-labile hybrid. In either instance, the M- 2 /L- 3 hybrid must be the most stable; otherwise, labeled probe hybridized to nonspecifically bound amplifier would be released.
  • the specific release of the target could be coupled with masking the background on the surface. In this case, the transfer to another support would be unnecessary.
  • the surface of the solid support could be coated with inhibitors of the labeled probe and/or various luminescence inhibitors, absorbers, or quenchers.
  • One surface coating currently in use is poly(phe-lys).
  • Phenylalanine is a known inhibitor of alkaline phosphatase, a particularly preferred enzyme label.
  • One could include in the polymeric peptide coating other inhibitors of alkaline phosphatase such as tryptophan and cysteine.
  • luminescent inhibitors include compounds with low quantum yields, i.e., any compound that preferentially gives off heat rather than light after being excited by collision with a dephosphorylated dioxetane.
  • the target-associated signals can be read in solution by making the solid phase inaccessible to visualization reagents or by masking signal generating reactions which occur on the solid support. Isolating the solid phase from subsequent visualization steps could be done by adding a heavier-than-water immiscible oil to the reaction vessel. This oil would cover the bottom of the vessel while allowing the solution of interest to float to the top. For simple calorimetric detection by visual or by reflectance measurement, an opaque substance could be added to the oil to serve as a neutral background for visualization.
  • the oil could be filled with an optically opaque substance. If a white solid such as titanium dioxide were used, light emitted from the floating aqueous layer would be reflected upward out of the container for detection. A dark solid or dye molecule dissolved in the oil could also be used to mask the stationary phase. Even if the oil solution does not completely isolate the solid phase from visualization reagents, the suspended solids or dissolved dyes would block the transmission of this light from the surface.
  • a stationary phase could be colored with a dye that would block emission of light from reactions that occur near its surface. This would be particularly convenient with a colored bead as a solid phase contained within an opaque well.
  • T m analysis was conducted of the oligonucleotides provided in Example 2.
  • the SELEX procedure could be used to find DNA or RNA sequences that show enhanced salt-independence in their melting at any selected pH.
  • F isoC
  • J isoG
  • ALK. PHOS. alkaline phosphatase
  • X a spacer sequence containing an amine for attachment to the solid support.
  • Labelled probe 32 the alkaline phosphatase oligonucleotide conjugate, was made as described (Urdea et al. (1988) Nucl. Acids Res. 16:4937-4955). Labelled probe 32 was hybridized with control probe 30 to create the alk. phos.-probe 30 * 32 . Labelled probe 32 was hybridized with modified probe 31 to create the isoC,isoG-alk. phos.-probe 31 * 32 .
  • Probe 35 the capture probe, was bound to microtiter wells as described (PCT Publication No. W091/813,338, the disclosure of which is incorporated by reference herein) to create a solid support for hybridization.
  • Probe 34 a capture extender, was hybridized to probe 35 .
  • This capture extender is complementary to the alk. phos.-probe 30 * 32 and partially complementary to the alk. phos.-probe 31 * 32 .
  • Probe 33 is a “competimer” that can bind to the capture extender and block the binding of either alkaline phosphatase probe.
  • probe 33 the competimer, can form 21 base pairs with the capture extender and in theory can block both alkaline phosphatase probes from binding.
  • the modified probe*labelled probe ( 31 * 32 ) can hybridize to the capture extender, forming 11 base pairs and two mismatches (e.g., G*isoC,isoG*T).
  • the control probe*labelled probe ( 30 * 32 ) can form 13 base pairs with the capture extender.
  • the two mismatches accounted for a 40-fold change in hybridization. This is as expected for 2 mismatches, each of which destabilizes the T m by 7-8° C. (cf.
  • Step 1 2′-Deoxyguanosine monohydrate (50 mmole) and imidazole (200 mmole) were dried by coevaporation with 500 mL dimethylformamide (DMF) and the residue dissolved in 500 mL DMF. To this solution was added t-butyldimethylsilyl chloride (150 mmole), and the reaction mixture was left stirring at room temperature for 18 hours. Methanol (30 mL) was added and after 25 minutes the solvents were removed in vacuo.
  • DMF dimethylformamide
  • Step 2 3′, 5′-TBDMS 2 -2′-deoxyguanosine (12 mmole) was suspended in 125 mL CH 2 Cl 2 containing triethylamine (150 mmole) and N,N-dimethylaminopyridine (100 mg). 4-Toluenesulfonyl chloride (40 mmole) was at 0° C., and the reaction mixture stirred at room temperature for 20 hours. At that time all solid material had dissolved resulting in a slightly yellow solution. The reaction was quenched with 50 mL 5% NaHCO 3 with stirring for 1 hour.
  • Step 3 Twelve grams of O 6 -(4-toluenesulfonyl)-3′,5′-O-TBDMS 2 -2′-deoxyguanosine (17 mmole) was suspended in 300 mL CH 3 CN. Then methylpyrrolidine (17 mL) was added and the suspension stirred for one hour to produce a clear solution. TLC analysis showed that all starting material had been converted to base line material. Eleven grams of 4-(methylthio)phenol (85 mmole) was added and the solution stirred for 60 hours. After evaporation to a small volume 600 mL ethyl acetate was added.
  • Step 4 Four grams of O 6 -(4-(methyl-thio) phenyl)-3′,5′-O-TBDMS 2 -2′-deoxyguanosine (6.5 mmole) was dissolved in 65 mL CH 2 Cl 2 at 0° C. and 6.5 mL of tert-butyl nitrite was added dropwise. The solution was allowed to warm to room temperature and gas evolved from the mixture (N 2 ). After 40 minutes, when TLC analysis showed complete consumption of starting material and emergence of a new, slower migrating spot, excess t-butyl nitrite was removed by coevaporation with 2 ⁇ 100 mL toluene in vacuo.
  • Step 5 All of the purified 2.75 grams of O 6 -(4-(methylthio) phenyl)-3′,5′-O-TBDMS 2 -2′-deoxyxantosine (4.45 mmole) was dissolved in 50 mL of methanol. Concentrated aqueous ammonium hydroxide (50 mL) was added and the mixture heated in a tightly sealed bomb at 100° C. for 4 hours. After cooling the solvents were removed by coevaporation with ethanol in vacuo to give 1.8 grams of crude product (3.6 mmole).

Abstract

Methods are provided for substantially reducing background signals encountered in nucleic acid hybridization assays. The method is premised on the elimination or significant reduction of the phenomenon of nonspecific hybridization, so as to provide a detectable signal which is produced only in the presence the target polynucleotide of interest. In addition, a novel method for the chemical synthesis of isoguanosine or 2′-deoxy-isoguanosine is provided. The invention also has applications in antisense and aptamer therapeutics and drug discovery.

Description

    TECHNICAL FIELD
  • This invention relates generally to nucleic acid chemistry and hybridization assays. More particularly, the invention relates to methods for generating a more target-dependent signal in nucleic acid hybridization assays by minimizing background noise deriving primarily from nonspecific hybridization. The invention also has applications in antisense and aptamer therapeutics and drug discovery. [0001]
  • BACKGROUND
  • Nucleic acid hybridization assays are commonly used in genetic research, biomedical research and clinical diagnostics. In a basic nucleic acid hybridization assay, single-stranded analyte nucleic acid is hybridized to a labeled single-stranded nucleic acid probe and resulting labeled duplexes are detected. Variations of this basic scheme have been developed to enhance accuracy, facilitate the separation of the duplexes to be detected from extraneous materials, and/or amplify the signal that is detected. [0002]
  • The present invention is directed to a method of reducing background noise encountered in any nucleic acid hybridization assay. Generally, the background noise which is addressed by way of the presently disclosed techniques results from undesirable interaction of various polynucleotide components that are used in a given assay, i.e., interaction which gives rise to a signal which does not correspond to the presence or quantity of analyte. The invention is useful in conjunction with any number of assay formats wherein multiple hybridization steps are carried out to produce a detectable signal which correlates with the presence or quantity of a polynucleotide analyte. [0003]
  • One such assay is described in detail in commonly assigned U.S. Pat. No. 4,868,105 to Urdea et al., the disclosure of which is incorporated herein by reference. That assay involves the use of a two-part capturing system designed to bind the polynucleotide analyte to a solid support, and a two-part labeling system designed to bind a detectable label to the polynucleotide analyte to be detected or quantitated. The two-part capture system involves the use of capture probes bound to a solid support and capture extender molecules which hybridize both to a segment of the capture probes and to a segment of the polynucleotide analyte. The two-part labelling system involves the use of label extender molecules which hybridize to a segment of the polynucleotide analyte, and labeled probes which hybridize to the label extender molecules and contain or bind to a detectable label. An advantage of such a system is that a plurality of hybridization steps must occur in order for label to be detected in a manner that correlates with the presence of the analyte, insofar as two distinct hybridization reactions must occur for analyte “capture,” and, similarly, two distinct hybridization reactions must occur for analyte labelling. However, there remain a number of ways in which a detectable signal can be generated in a manner which does not correspond to the presence or quantity of analyte, and these will be discussed in detail below. [0004]
  • Another example of an assay with which the present invention is useful is a signal amplification method which is described in commonly assigned U.S. Pat. No. 5,124,246 to Urdea et al., the disclosure of which is incorporated herein by reference. In that method, the signal is amplified through the use of amplification multimers, polynucleotides which are constructed so as to contain a first segment that hybridizes specifically to the label extenders, and a multiplicity of identical second segments that hybridize specifically to a labeled probe. The degree of amplification is theoretically proportional to the number of iterations of the second segment. The multimers may be either linear or branched. Branched multimers may be in the shape of a fork or a comb, with comb-type multimers preferred. [0005]
  • One approach to solving the problem of interfering background signals in nucleic acid hybridization assays is provided in commonly assigned U.S. patent application Ser. No. 08/164,388 to Urdea et al. in which at least two capture extenders and/or two or more label extenders must bind to the analyte in order to trigger a detectable signal. To further reduce background noise, the assay is conducted under conditions which favor the formation of multicomponent complexes. [0006]
  • Another approach which has been proposed to increase the target dependence of the signal in a hybridization assay is described in European Patent Publication No. 70,685, inventors Heller et al. That reference describes a homogeneous hybridization assay in which a nonradiative transfer of energy occurs between proximal probes; two distinct events must occur for a target-generated signal to be produced, enhancing the accuracy of detection. [0007]
  • The present invention is also designed to increase the accuracy of detection and quantitation of polynucleotide analytes in hybridization assays. The invention increases both the sensitivity and specificity of such assays, by reducing the incidence of signal generation that occurs in the absence of target, and does not involve an increase in either time or cost relative to currently used assay configurations. [0008]
  • The goals of the present invention, namely to reduce background noise and to increase accuracy of detection and quantitation of analytes in nucleic acid hybridization assays have been achieved, in part, by the use of nucleoside variants that form base pairs by virtue of “non-natural” hydrogen bonding patterns. As used herein, a “non-natural” base pair is one formed between nucleotidic units other than adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U). One such non-natural nucleoside base pair is formed between isocytosine (isoC) and isoguanine (isoG). IsoC and isoG can form a base pair with a standard geometry (i.e., a “Watson-Crick base pair”) but involving hydrogen bonding other than that involved in the bonding of cytosine (C) to guanine (G), as shown below: [0009]
    Figure US20010026918A1-20011004-C00001
    Figure US20010026918A1-20011004-C00002
  • Leach et al. (1992) [0010] J. Am. Chem. Soc. 114:3675-3683 applied molecular mechanics, molecular dynamics and free energy perturbation calculations to study the structure and stability of the isoC*isoG base pair. Tor et al. (1993) J. Am. Chem. Soc. 115:4461-4467 describe a method whereby a modified isoC in a DNA template will direct the incorporation of an isoG analog into the transcribed RNA product. Switzer et al. (1993) Biochemistry 32:10489-10496 studied the conditions under which the base pair formed between isoC and isoG might be incorporated into DNA and RNA by DNA and RNA polymerases.
  • Introduction of a new base pair into DNA oligomers offers the potential of allowing more precise control over hybridization. [0011]
  • SUMMARY OF THE INVENTION
  • The present invention provides methods and kits for detecting nucleic acid analytes in a sample. In general, the methods represent improvements in nucleic acid hybridization assays, such as in situ hybridization assays, Southerns, Northerns, dot blots and polymerase chain reaction assays. In particular, the methods represent improvements on solution phase sandwich hybridization assays which involve binding the analyte to a solid support, labelling the analyte, and detecting the presence of label on the support. Preferred methods involve the use of amplification multimers which enable the binding of significantly more label in the analyte-probe complex, enhancing assay sensitivity and specificity. [0012]
  • In a first aspect of the invention, an assay is provided in which one or more nucleotidic units which are capable of forming base pairs and which are other than adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U), are incorporated into non-target hybridizing oligonucleotide segments, i.e., “universal” segments, of nucleic acid hybridization assay components. This use of such nucleotidic units gives rise to unique base-pairing schemes which result in enhanced binding specificity between universal segments. [0013]
  • In a related aspect of the invention, an assay is provided in which at least one first nucleotidic unit other than A, T, C, G, or U capable of forming a base pair with a second nucleotidic unit other than A, T, C, G, or U, is incorporated into nucleic acid sequences of assay components which are complementary to nucleic acid sequences present in assay components other than the target molecule. Examples of base pairs formed between two such nucleotidic units are given in the following structures (I) to (IV): [0014]
    Figure US20010026918A1-20011004-C00003
    Figure US20010026918A1-20011004-C00004
  • wherein R represents a backbone which will allow the bases to form a base pair with a complementary nucleotidic unit when incorporated into a polynucleotide, and R′ is, for example, hydrogen, methyl, α- or β-propynyl, bromine, fluorine, iodine, or the like. By incorporating such nucleotidic units into such so-called “universal” sequences, i.e., sequences not involved in hybridization to the target analyte, the potential for nonspecific hybridization is greatly reduced. In one preferred embodiment, the first and second nucleotidic units interchangeably consist of isocytidine and isoguanosine, as shown in Formula (I). [0015]
  • In a related aspect of the invention, an assay is provided in which the melt temperature T[0016] m1 of the complex formed between the analyte and the support-bound capture probes, mediated by one or more distinct capture extender molecules, and/or the label extender and amplifier or preamplifiers, is significantly lower than the melt temperature Tm1 of the complex formed between the labeled probes and the amplifier. In this aspect, the assay is carried out under conditions which initially favor the formation of all hybrid complexes. The conditions are then altered during the course of the assay so as to destabilize the Tm1 hybrid complexes.
  • The invention additionally encompasses a method for carrying out a hybridization assay in which each of the aforementioned techniques are combined, i.e., in which nucleotidic units other than A, T, G, C, or U are incorporated into universal segments of assay components and in which the melt temperature of T[0017] m1 hybrid complexes is significantly lower than the melt temperature of Tm2 hybrid complexes.
  • In a further aspect, the invention encompasses a novel method for synthesizing isoguanosine or 2′-deoxy-isoguanosine. [0018]
  • Finally, the invention encompasses kits containing the reagents necessary to carry out the assays described and claimed herein.[0019]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. FIG. 1 diagrams a solution phase sandwich hybridization assay of the prior art with heavy lines indicating the universal sequences. [0020]
  • FIG. 2. FIG. 2 portrays a method for binding probes to double-stranded DNA with heavy lines indicating the universal sequences. [0021]
  • FIG. 3. FIG. 3 depicts the use of non-natural nucleotide-containing probes and competimers to block non-specific hybridization.[0022]
  • DETAILED DESCRIPTION OF THE INVENTION Definitions and nomenclature:
  • Before the present invention is disclosed and described in detail, it is to be understood that this invention is not limited to specific assay formats, materials or reagents, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. [0023]
  • In this specification and in the claims which follow, reference will be made to a number of terms which shall be defined to have the following meanings: [0024]
  • As used herein, the terms “polynucleotide” and “oligonucleotide” shall be generic to polydeoxyribonucleotides (containing 2-deoxy-D-ribose), to polyribonucleotides (containing D-ribose), to any other type of polynucleotide which is an N- or C-glycoside of a purine or pyrimidine base, and to other polymers containing non-nucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids (PNAs)) and polymorpholino (commercially available from the Anti-Virals, Inc., Corvallis, Oregon, as Neugene™ polymers), and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base pairing and base stacking, such as is found in DNA and RNA. There is no intended distinction in length between the term “polynucleotide” and “oligonucleotide,” and these terms will be used interchangeably. These terms refer only to the primary structure of the molecule. Thus, these terms include double- and single-stranded DNA, as well as double- and single-stranded RNA, DNA:RNA hybrids, and hybrids between PNAs and DNA or RNA, and also include known types of modifications, for example, labels which are known in the art, methylation, “caps,” substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), with negatively charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), and with positively charged linkages (e.g., aminoalklyphosphoramidates, aminoalkylphosphotriesters), those containing pendant moieties, such as, for example, proteins (including nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide or oligonucleotide. [0025]
  • It will be appreciated that, as used herein, the terms “nucleoside” and “nucleotide” will include those moieties which contain not only the known purine and pyrimidine bases, but also other heterocyclic bases which have been modified. Such modifications include methylated purines or pyrimidines, acylated purines or pyrimidines, or other heterocycles. Modified nucleosides or nucleotides will also include modifications on the sugar moiety, e.g., wherein one or more of the hydroxyl groups are replaced with halogen, aliphatic groups, or are functionalized as ethers, amines, or the like. The term “nucleotidic unit” is intended to encompass nucleosides and nucleotides. [0026]
  • Furthermore, modifications to nucleotidic units include rearranging, appending, substituting for or otherwise altering functional groups on the purine or pyrimidine base which form hydrogen bonds to a respective complementary pyrimidine or purine. The resultant modified nucleotidic unit may form a base pair with other such modified nucleotidic units but not with A, T, C, G or U. Standard A-T and G-C base pairs form under conditions which allow the formation of hydrogen bonds between the N[0027] 3-H and C4-oxy of thymidine and the N1 and C6-NH2, respectively, of adenosine and between the C2-oxy, N3 and C4-NH2, of cytidine and the C2-NH2, N1-H and C6-oxy, respectively, of guanosine. Thus, for example, guanosine (2-amino-6-oxy-9-β-D-ribofuranosyl-purine) may be modified to form isoguanosine (2-oxy-6-amino-9-,β-D-ribofuranosyl-purine). Such modification results in a nucleoside base which will no longer effectively form a standard base pair with cytosine. However, modification of cytosine (1-β-D-ribofuranosyl-2-oxy-4-amino-pyrimidine) to form isocytosine (1-β-D-ribofuranosyl-2-amino-4-oxy-pyrimidine) results in a modified nucleotide which will not effectively base pair with guanosine but will form a base pair with isoguanosine. Isocytosine is available from Sigma Chemical Co. (St. Louis, Mo.); isocytidine may be prepared by the method described by Switzer et al. (1993), supra, and references cited therein; 2′-deoxy-5-methyl-isocytidine may be prepared by the method of Tor et al. (1993), supra, and references cited therein; and isoguanine nucleotides may be prepared using the method described by Switzer et al., supra, and Mantsch et al. (1993) Biochem. 14:5593-5601, or by the method described in detail below. The non-natural base pairs depicted in structure (II), referred to as κ and π, may be synthesized by the method described in Piccirilli et al. (1990) Nature 343:33-37 for the synthesis of 2,6-diamino-pyrimidine and its complement (1-methylpyrazolo[4,3]-pyrimidine-5,7-(4H,6H)-dione. Other such modified nucleotidic units which form unique base pairs have been described in Leach et al. (1992) J. Am. Chem. Soc. 114:3675-3683 and Switzer et al., supra, or will be apparent to those of ordinary skill in the art.
  • The term “polynucleotide analytel” refers to a single- or double-stranded nucleic acid molecule which contains a target nucleotide sequence. The analyte nucleic acids may be from a variety of sources, e.g., biological fluids or solids, food stuffs, environmental materials, etc., and may be prepared for the hybridization analysis by a variety of means, e.g., proteinase K/SDS, chaotropic salts, or the like. The term “polynucleotide analyte” is used interchangeably herein with the terms “analyte,” “analyte nucleic acid,” “target” and “target molecule.” As used herein, the term “target region” or “target nucleotide sequence” refers to a probe binding region contained within the target molecule. The term “target sequence” refers to a sequence with which a probe will form a stable hybrid under desired conditions. [0028]
  • As used herein, the term “probe” refers to a structure comprised of a polynucleotide, as defined above, which contains a nucleic acid sequence complementary to a nucleic acid sequence present in the target molecule. The polynucleotide regions of probes may be composed of DNA, and/or RNA, and/or synthetic nucleotide analogs. [0029]
  • It will be appreciated that the binding sequences need not have perfect complementarity to provide stable hybrids. In many situations, stable hybrids will form where fewer than about 10% of the bases are mismatches, ignoring loops of four or more nucleotides. Accordingly, as used herein the term “complementary” refers to an oligonucleotide that forms a stable duplex with its “complement” under assay conditions, generally where there is about 90% or greater homology. [0030]
  • The terms “nucleic acid multimer” or “amplification multimer” are used herein to refer to a linear or branched polymer of the same repeating single-stranded oligonucleotide segment or different single-stranded polynucleotide segments, each of which contains a region where a labeled probe can bind, i.e., contains a nucleic acid sequence complementary to a nucleic acid sequence contained within a labeled probe; the oligonucleotide segments may be composed of RNA, DNA, modified nucleotides or combinations thereof. At least one of the segments has a sequence, length, and composition that permits it to bind specifically to a labeled probe; additionally, at least one of the segments has a sequence, length, and composition that permits it to bind specifically to a label extender or preamplifier. Typically, such segments will contain approximately 15 to 50, preferably 15 to 30, nucleotides, and will have a GC content in the range of about 20% to about 80%. The total number of oligonucleotide segments in the multimer will usually be in the range of about 3 to 1000, more typically in the range of about 10 to 100, and most typically about 50. The oligonucleotide segments of the multimer may be covalently linked directly to each other through phosphodiester bonds or through interposed linking agents such as nucleic acid, amino acid, carbohydrate or polyol bridges, or through other cross-linking agents that are capable of cross-linking nucleic acid or modified nucleic acid strands. Alternatively, the multimer may be comprised of oligonucleotide segments which are not covalently attached, but are bonded in some other manner, e.g., through hybridization. Such a multimer is described, for example, in U.S. Pat. No. 5,175,270 to Nilsen et al. The site(s) of linkage may be at the ends of the segment (in either normal, 3′-5′ orientation or randomly oriented) and/or at one or more internal nucleotides in the strand. In linear multimers the individual segments are linked end-to-end to form a linear polymer. In one type of branched multimer three or more oligonucleotide segments emanate from a point of origin to form a branched structure. The point of origin may be another nucleotide segment or a multifunctional molecule to which at least three segments can be covalently bound. In another type, there is an oligonucleotide segment backbone with one or more pendant oligonucleotide segments. These latter-type multimers are “fork-like,” “comb-like” or combination “fork-” and “comb-like” in structure, wherein “comb-like” multimers, the preferred multimers herein, are polynucleotides having a linear backbone with a multiplicity of sidechains extending from the backbone. The pendant segments will normally depend from a modified nucleotide or other organic moiety having appropriate functional groups to which oligonucleotides may be conjugated or otherwise attached. The multimer may be totally linear, totally branched, or a combination of linear and branched portions. Typically, there will be at least two branch points in the multimer, more preferably at least three, more preferably in the range of about 5 to 30, although in some embodiments there may be more. The multimer may include one or more segments of double-stranded sequences. Further information concerning multimer synthesis and specific multimer structures may be found in commonly owned U.S. Pat. No. 5,124,246 to Urdea et al. [0031]
  • Commonly assigned U.S. patent application Ser. No. 07/813,588 and European Patent Publication No. 541,693 describe the comb-type branched multimers which are particularly preferred in conjunction with the present method, and which are composed of a linear backbone and pendant sidechains; the backbone includes a segment that provides a specific hybridization site for analyte nucleic acid or nucleic acid bound to the analyte, whereas the pendant sidechains include iterations of a segment that provide specific hybridization sites for a labeled probe. [0032]
  • As noted above, a “preamplifier” molecule may also be used, which serves as a bridging moiety between the label extender molecules and the amplification multimers. In this way, more amplifier and thus more label is bound in any given target-probe complex. Preamplifier molecules may be either linear or branched, and typically contain in the range of about 30 to about 3000 nucleotides. In the preferred embodiment herein, the preamplifier molecule binds to at least two different label extender molecules, such that the overall accuracy of the assay is increased (i.e., because, again, a plurality of hybridization events are required for the probe-target complex to form). [0033]
  • As used herein, a “biological sample” refers to a sample of tissue or fluid isolated from an individual, including but not limited to, for example, plasma, serum, spinal fluid, semen, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs, and also samples of in vitro cell culture constituents (including but not limited to conditioned medium resulting from the growth of cells in cell culture medium, putatively virally infected cells, recombinant cells, and cell components). Preferred uses of the present method are in detecting and/or quantitating nucleic acids as follows: (a) viral nucleic acids, such as from hepatitis B virus (“HBV”), hepatitis C virus (“HCV”), hepatitis D virus (“HDV”), human immunodeficiency virus (“HIV”), and the herpes family of viruses, including herpes zoster (chicken pox), herpes simplex virus I & II, cytomegalovirus, Epstein-Barr virus, and the recently isolated Herpes VI virus; (b) bacterial nucleic acids, such as Chlamydia, Mycobacterium tuberculosis, etc.; and (c) numerous human sequences of interest. [0034]
  • As used herein, the term “nonspecific hybridization” is used to refer to those occurrences in which a segment of a first polynucleotide which is intended to hybridize to a segment of a selected second polynucleotide also hybridizes to a third polynucleotide, triggering an erroneous result, i.e., giving rise to a situation where label may be detected in the absence of target molecule. The use of the term “hybridizes” is not meant to exclude non-Watson-Crick base pairing. [0035]
  • As used herein, the term “nonspecific binding” is used to refer to those occurrences in which a polynucleotide binds to the solid support, or other assay component, through an interaction--which may be either direct or indirect--that does not involve hydrogen bonding to support-bound polynucleotides. [0036]
  • Referring now to the preferred embodiment represented in FIG. 1, the following terms apply to the hybridization assay depicted therein. Note that, in FIG. 1, the universal sequences are indicated by heavy lines for clarity. [0037]
  • “Label extender molecules (LEs),” also referred to herein as “label extenders,” contain regions of complementarity vis-à-vis the analyte polynucleotide and to the amplifying multimer (“AMP”). If a preamplifier is used (not shown in the figure), the label extender molecules will bind to this intermediate species rather than directly to the amplifier multimer. If neither preamplifier or amplifier is used, the label extender molecules will bind directly to a sequence in the labeled probe (“LP”). Thus, label extender molecules are single-stranded polynucleotide chains having a first nucleic acid sequence L-[0038] 1 complementary to a sequence of the analyte polynucleotide, and a second universal region having a multimer recognition sequence L-2 complementary to a segment M-1 of label probe, amplifier multimer or preamplifier.
  • “Labeled probes (LPs)” are designed to bind either to the label extender, or, if an amplifier multimer is employed in the assay, to the repeating oligonucleotide segments of the multimer. LPs either contain a label or are structured so as to bind to a label. Thus, LPs contain a nucleic acid sequence L-[0039] 3 complementary to a nucleic acid sequence M-2 present within the repeating oligonucleotide units of the multimer and are bound to, or structured so as to bind to, a label which provides, directly or indirectly, a detectable signal.
  • “Capture extender molecules (CEs),” also referred to herein as “capture extenders,” bind to the analyte polynucleotide and to capture probes, which are in turn bound to a solid support. Thus, capture extender molecules are single-stranded polynucleotide chains having a first polynucleotide sequence region containing a nucleic acid sequence C-[0040] 1 which is-complementary to a sequence of the analyte, and a second, noncomplementary region having a capture probe recognition sequence C-2. The sequences C-1 and L-1 are nonidentical, noncomplementary sequences that are each complementary to physically distinct sequences of the analyte.
  • “Capture probes (CPs)” bind to the capture extenders and to a solid support. Thus, as illustrated in FIG. 1, capture probes have a nucleic acid sequence C-[0041] 3 complementary to C-2 and are covalently bound to (or capable of being covalently bound to) a solid support.
  • Generally, solution phase hybridization assays carried out using the system illustrated in FIG. 1 proceed as follows. Single-stranded analyte nucleic acid is incubated under hybridization conditions with the capture extenders and label extenders. The resulting product is a nucleic acid complex of the analyte polynucleotide bound to the capture extenders and to the label extenders. This complex may be subsequently added under hybridizing conditions to a solid phase having the capture probes bound to the surface thereof; however, in a preferred embodiment, the initial incubation is carried out in the presence of the support-bound capture probes. The resulting product comprises the complex bound to the solid phase via the capture extender molecules and capture probes. The solid phase with bound complex is then separated from unbound materials. An amplification multimer, preferably a comb-type multimer as described above, is then optionally added to the solid phase-analyte-probe complex under hybridization conditions to permit the multimer to hybridize to the LEs; if preamplifier probes are used, the solid phase-analyte-probe complex is incubated with the preamplifier probes either along with the amplifier multimer or, preferably, prior to incubation with the amplifier multimer. The resulting solid phase complex is then separated from any unbound preamplifier and/or multimer by washing. The labeled probes are then added under conditions which permit hybridization to LEs, or, if an amplification multimer was used, to the repeating oligonucleotide segments of the multimer. The resulting solid phase labeled nucleic acid complex is then washed to remove unbound labeled oligonucleotide, and read. It should be noted that the components represented in FIG. 1 are not necessarily drawn to scale, and that the amplification multimers, if used, contain a far greater number of repeating oligonucleotide segments than shown (as explained above), each of which is designed to bind a labeled probe. [0042]
  • The primary focus of the present method is on eliminating the sources of background noise, by minimizing the interaction of capture probes and capture extender molecules with the labeled probes, label extender molecules and amplifiers, reducing the likelihood that incorrect moieties will bind to the support-bound capture probes. [0043]
  • Hybridization between complementary oligonucleotide sequences is premised on the ability of the purine and pyrimidine nucleotides contained therein to form stable base pairs. The five naturally occurring nucleotides adenosine (A), guanosine (G), thymidine (T), cytidine (C) and uridine (U) form the purine-pyrimidine base pairs G-C and A-T(U). The binding energy of the G-C base pair is greater than that of the A-T base pair due to the presence of three hydrogen-bonding moieties in the former compared with two in the latter, as shown below: [0044]
    Figure US20010026918A1-20011004-C00005
    Figure US20010026918A1-20011004-C00006
  • Thus, in a conventional solution phase nucleic acid sandwich assay, oligonucleotide molecules are designed to contain nucleic acid sequences which are complementary to and, therefore, hybridize with nucleic acid sequences in other assay components or in the target molecule, as explained in detail above. The method of the invention reduces nonspecific hybridization by incorporating non-natural nucleotidic units into universal oligonucleotide segments of assay components which are capable of forming unique base pairs. Furthermore, the method of the invention reduces the contribution of nonspecific binding of assay components by separating detectably labelled assay components which are associated with the presence and/or quantity of a target analyte from those which are nonspecifically bound and contribute to assay background noise. [0045]
  • In a first embodiment of the invention, a hybridization assay is provided in which nucleotidic units other than A, T, C, G and U which are capable of forming unique base pairs are incorporated into hybridizing oligonucleotide segments of assay components which are not target analyte specific and thus will be less likely to form stable hybrids with target-specific probe sequences or with extraneous non-target nucleic acid sequences. Thus, as shown in FIG. 1, for example, such nucleotidic units may be incorporated in complementary nucleic acid sequences C-[0046] 2/C-3, L-2/M-1 and L-3/M-2. The hybridizing oligonucleotide segments of assay components which are complementary to nucleic acid segments of the target molecule are constructed from naturally occurring nucleotides (i.e., A, T, C, G or U). Oligonucleotide segments which contain nucleotidic units may be constructed by replacing from about 15% to about 100% of the naturally occurring nucleotides with the nucleotidic unit counterpart. Preferably, every third or fourth base in an oligonucleotide will be replaced with a nucleotidic unit capable of forming a unique base pair. It will be apparent to those skilled in the art that as the percent of replacement nucleotidic units is increased, nonspecific hybridization is decreased concomitantly. However, complete replacement will require at least two new base pairs in order to maintain sufficient sequence diversity to preclude nonspecific hybridization among the universal sequences.
  • In another embodiment of the invention, the phenomenon of target-independent signal generation is addressed by providing a hybridization assay which is configured such that the melt temperature T[0047] m1 of the C-2/C-3 hybrid or the L-2/M-1 hybrid is significantly lower than the melt temperature Tm2 of the L-3/M-2 hybrid. This method is premised on the design and construction of hybrid complexes such that the melt temperature Tm1 is at least about 5° C. lower than, preferably at least about 10° C. lower than, more preferably at least about 20° C. lower than the melt temperature Tm2.
  • This stability difference is exploited by conducting the assay under stringency conditions which initially favor the formation of T[0048] m1 and Tm2 hybrid complexes. The stringency is altered at a subsequent step of the assay which thereby affords the physical separation of the target molecule from the capture probes or the physical separation of the amplifier-bound labeled probes from the target. Stringency can be controlled by altering a parameter which is a thermodynamic variable. Such variables are well known in the art, and include formamide concentration, salt concentration, chaotropic salt concentration, pH (hydrogen ion concentration), organic solvent content, and temperature. Preferred stringency controls are pH and salt concentration: one assay step is conducted at a pH or salt concentration which destabilizes the hybrid complex formed between capture probe/capture extender or destabilizes the hybrid formed between label extender/amplifier (or preamplifier). A preferred step at which stringency is exercised is the addition of substrate. Thus, in a preferred embodiment, the hybridization assay is conducted under conditions which favors the stability of hybrid complexes formed between all assay components and thereafter, with the addition of label substrate, the stringency is altered to destabilize hybrid complexes such as the capture probe/capture extender, or label extender/amplifier (preamplifier), and the like, with the proviso that the labeled probe is not released from the label extender or amplifier.
  • Another embodiment of the invention represents one means by which the above embodiment of the invention may be effected is by configuring the hybridization assay such that the complementary nucleotide sequences which form T[0049] m1 hybrid complexes are shorter than those which form Tm2 hybrid complexes. It will be appreciated by those of skill in the art that, with shorter complementary nucleotide sequences, the opportunity for sequence diversity therein decreases. This diversity may be maintained, however, by incorporating into the complementary sequences a non-natural base pair, e.g., an isoC-isoG base pair.
  • It will be readily apparent to one skilled in the art that the greater the temperature difference between T[0050] m1 and Tm2, the greater the “efficiency” of this technique in removing background noise. Thus, one skilled in the art will recognize that temperature differentials of less than 10° C., even less than 5° C., would also permit reduction of background noise, albeit to a lesser extent.
  • The method of the disclosed invention, whereby non-natural nucleotidic units are incorporated into hybridizing oligonucleotide sequences to increase the specificity of the hybridization with a target molecule, finds utility in a variety of applications. [0051]
  • In the basic or amplified solution phase nucleic acid sandwich assay, a plurality of capture probes are affixed to a solid surface. Most often, the surface area available for non-specific binding is controlled by incubating the surface with DNA from, e.g., salmon sperm or calf thymus. However, the presence of this DNA increases the potential for nonspecific hybridization of assay components to the solid support and, therefore, increased background noise. Replacement of these natural DNAs with synthetic DNAs containing non-natural bases will minimize the non-specific hybridization and the non-specific binding. [0052]
  • Preferably, these polynucleotides will be prepared by 3′ tailing short oligonucleotides with mixtures of nucleotides by methods well known in the art. Alteratively, short, nearly random-sequence oligonucleotides containing non-natural nucleotides can be joined together to form polynucleotides. Branched DNAs can be conveniently used for this purpose. For example, the block sequence -TNVN-F-TNVN-J-TNVN-, wherein F is isoC and J is isoG, can be prepared and chemically joined to form a polymer. The advantage of using this approach over using the enzymatic 3′ tailing approach is the elimination of homopolymer/homooligomer sequences. [0053]
  • Another application in which the construction of hybridizing oligonucleotides containing non-natural nucleotidic units finds utility is in the design of antisense compounds. Antisense compounds, as explained, for example, in Ching et al. (1989) [0054] Proc. Natl. Acad. Sci. U.S.A. 86:10006-10010, Broder et al. (1990) Ann. Int. Med. 111:604-618, Loreau et al. (1990) FEBS Letters 274:53-56, and PCT Publication Nos. W091/11535, W091/09865, W091/04753, W090/13641, W091/13080 and, WO 91/06629, are oligonucleotides that bind to and disable or prevent the production of the mRNA responsible for generating a particular protein. Conventional antisense molecules are generally capable of reacting with a variety of oligonucleotide species. However, due to their length (generally oligonucleotide sequences of up to 30 nucleotidic units), such antisense molecules present problems associated with nonspecific hybridization with non-target species. One solution is to use short regions of hybridization between multiple probes and the target; to strengthen the overall complex, short “dimerization domains” between the probes are used, as described by Distefano et al. (1992) J. Am. Chem. Soc. 114:1006-1007. The dimerization domains may be designed to have tails with complementary sequences containing non-natural nucleotidic units and thereby provide highly efficient and specific binding to the target molecule without increasing non-specific hybridization to non-target molecules. The idea is illustrated in FIG. 2 with a double-stranded DNA target.
  • As illustrated in FIG. 2, strand displacement may be used to pry apart double-stranded DNA. AT-rich promoter sequences under superhelical stress, which are S[0055] 1 nuclease-sensitive and are thus already partially single-stranded, are a particularly preferred site for this type of antigene application. Short oligonucleotides would be used to maximize specificity; their binding energy to the target would be enhanced by joining them together to form a network of oligonucleotides.
  • In this construct, the short universal sequences, which will not form stable base-pairs in the absence of target, contain isoC and isoG to limit nonspecific hybridization of the probes with the human sequences. Upon binding of [0056] probes 1, 2 and 3 to the target, the universal sequences will be in sufficiently close proximity that their effective concentration will be significantly increased. The universal sequences will then pair, resulting in a further increase in the strength of the binding. RNA targets may also be used in conjunction with this approach.
  • The SELEX procedure, described in U.S. Pat. No. 5,270,163 to Gold et al., Tuerk et al. (1990) [0057] Science 249:505-510, Szostak et al. (1990) Nature 346:818-822 and Joyce (1989) Gene 82:83-87, can be used to select for RNA or DNA sequences that recognize and bind to a desired target molecule by virtue of their shape. The term “aptamer” (or nucleic acid antibody) is used herein to refer to such a single- or double-stranded DNA or a single-stranded RNA molecule. See, e.g., PCT Publication Nos. W092/14843, W091/19813, and W092/05285, the disclosures of which are incorporated by reference herein. “Target molecules,” as distinct from “target analytes,” include polymers such as proteins, polysaccharides, oligonucletides or other macromolecules, and small molecules such as drugs, metabolites, toxins, or the like, to which an aptamer is designed to bind.
  • In the SELEX procedure, an oligonucleotide is constructed wherein an n-mer, preferably a randomized sequence of nucleotides thereby forming a “randomer pool” of oligonucleotides, is flanked by two polymerase chain reaction (PCR) primers. The construct is then contacted with a target molecule under conditions which favor binding of the oligonucleotides to the target molecule. Those oligonucleotides which bind the target molecule are: (a) separated from those oligonucleotides which do not bind the target molecule using conventional methods such as filtration, centrifugation, chromatography, or the like; (b) dissociated from the target molecule; and (c) amplified using conventional PCR technology to form a ligand-enriched pool of oligonucleotides. Further rounds of binding, separation, dissociation and amplification are performed until an aptamer with the desired binding affinity, specificity or both is achieved. The final aptamer sequence identified can then be prepared chemically or by in vitro transcription. When preparing such aptamers, selected base pairs are replaced with nonnatural base pairs to reduce the likelihood of the aptamers hybridizing to human nucleic acids. [0058]
  • One can use the present invention in at least two general ways in SELEX. First, isodG and isodc can be included among the sequences in the randomer DNA sequence pool. The number of possible randomeric structures that may recognize proteins or other important biomolecules is increased by synthesizing strands of DNA out of six or more nucleotides, rather than the conventional four nucleotide A, T, G and C. This is turn improves the chances of identifying a sequence which binds with greater affinity and/or specificity to the target molecule. [0059]
  • In SELEX, the conserved oligonucleotide sequences selected for may have unwanted hybridization to cellular sequences. This nonspecific hybridization can be reduced using nonnatural bases in the selection process. Nucleotides that are not recognized by human RNA and DNA polymerases but which are recognized by certain phage or bacterial polymerases are particularly useful in this application. [0060]
  • A second use for the instant invention in the SELEX process is in the preparation of a final aptamer construct with minimized nonspecific hybridization. For example, aptamers which display predetermined binding affinity, specificity or other target molecule recognition characteristics are selected from a pool of RNA or DNA sequences using the SELEX process. These target molecule recognition characteristics are determined by the secondary structure of the aptamer which is maintained, in part, by the formation of intramolecular oligonucleotide hybrid complexes. Upon elucidation of the secondary structure of the aptamer, it will be apparent to one of ordinary skill in the art that the specificity of base pairs in certain intramolecular hybrid complexes is highly preferred for maintaining the secondary structure and, therefore, the target molecule recognition and binding characteristics of the aptamer, i.e., there will base pairs which are preferably G-C or A-T. There will be other base pairs in these intramolecular hybrid complexes, for example, in the base-pairing portion of the stem loop, which may be replaced by any pair of complementary nucleotides, referred to herein as N-N′ base pairs, without altering the secondary structure of the aptamer. [0061]
  • A simple replacement of selected N-N′ base pairs and G-C and C-G base pairs in the final aptamer construct with isoG-isoC or isoC-isoG will reduce nonspecific hybridization to nontarget oligonucleotide sequences. Since the isoC-isoG base pair is isoenergetic with the C-G base pair, the basic shape of the molecule and the strength of the hairpins will be very similar. A base pair isoenergetic with A-U would be desirable for replacing base pairs where the winning sequences show a strong preference for A-U or U-A over C-G. These substitutions have the effect of making the aptamers more specific for the target molecule by limiting their potential for unwanted hybridization to cellular RNA and DNA-sequences. [0062]
  • In the basic process, selected base pairs are replaced with isoC-isoG or isoG-isoC base pairs. In the final construct, isoC-isoG base pairs can comprise ribonucleotides or deoxyribonucleotides. A chimeric aptamer (composed of both ribonucleotides and deoxyribonucleotides) molecule can be made chemically. Alternatively, the ribo-isoGTP and ribo-isoCTP (with suitable 2′ protection) can be used to prepare the aptamer by in vitro transcription of DNA templates containing isoC and isoG. [0063]
  • Other applications in which the present invention may find utility include in situ hybridizations, in reducing of nonspecific binding in hybridization assays and in polymerase chain reaction (PCR) assays. [0064]
  • In situ hybridization lacks sufficient sensitivity to detect a single molecule of target analyte. In situ PCR (see, e.g., Bagasra et al. (1993) [0065] J. Immunological Methods 158:131-145) has been developed to meet this sensitivity need; however, quantitation is not as precise with the PCR method. An alternative would use multiple label extender probes to bind the target analyte. The label extenders would bind either preamplifiers or amplifiers. If used, preamplifiers would-bridge label extenders and amplifiers. The amplifiers would bind labeled probes, which would preferably be detected by luminescence (fluorescence if the sensitivity is high enough). As before, the universal sequences, L-2/M-1 and M-2/L-3 would consist of short oligonucleotides containing optimally between 15-30% isoC and isoG to reduce unwanted hybridization to human sequences. A fourth base-pair could be used to further reduce the representation of the natural bases in these sequences.
  • As noted earlier, nonspecific binding as well as nonspecific hybridization can be reduced by using nonnatural base pairs. Random polymers or nearly random block copolymers consisting of 6-8 different nucleotides could be used to reduce nonspecific binding of the amplifier and labeled probes to the cellular constituents that have high affinity for polynucleotides. Thus nonspecific binding will be reduced without risking an increase in nonspecific hybridization by introducing natural sequences from calf or salmon, as is commonly done. [0066]
  • One skilled in the art will recognize that the same strategy could be applied to blot assays, such as dot blots, Southerns, and Northerns, to reduce nonspecific hybridization and nonspecific binding of the probes to the solid supports. [0067]
  • The present invention also finds several uses in PCR and other exponential amplification technologies. For example, in nested PCR, after the target analyte is initially amplified and then diluted several thousand-fold, it is common to use a 5′ overhang on one primer for capture and a 5′ overhang on the other primer for labeling. A spacer that cannot be read by the polymerase is inserted so that the overhangs remain single-stranded (see, e.g., Newton et al. (1993) [0068] Nucl. Acids Res. 21:1155-1162). The generic sequences in these 5′ overhangs can be prepared to contain modified base-pairs to reduce the frequency of priming on nontargets. Indeed, the presence of isodC or isodG in the first base of the 5′ overhang can be used in place of the currently used spacers; the polymerase cannot read isodC or isodG because it will have no isodGTP or isodCTP to put in place of it. Because the polymerase may put T into the polymer at a low frequency when it detects isodG in what was the primer, it is preferable to use isoC as the first base in the 5′ overhang.
  • Experimental
  • The practice of the present invention will employ, unless otherwise indicated, conventional techniques of synthetic organic chemistry, biochemistry, molecular biology, and the like, which are within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Sambrook, Fritsch & Maniatis, [0069] Molecular Cloning: A Laboratory Manual, Second Edition (1989); Oligonucleotide Synthesis (M. J. Gait, ed., 1984); Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins, eds., 1984); and the series, Methods in Enzymology (Academic Press, Inc.).
  • All patents, patent applications, and publications mentioned herein, both supra and infra, are hereby incorporated by reference. [0070]
  • It is to be understood that while the invention has been described in conjunction with the preferred specific embodiments thereof, that the description above as well as the examples which follow are intended to illustrate and not limit the scope of the invention. Other aspects, advantages and modifications within the scope of the invention will be apparent to those skilled in the art to which the invention pertains. [0071]
  • In the following examples, efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental error and deviation should be accounted for. Temperature is always given in degrees C and, unless otherwise indicated, pressure is at or near atmospheric. [0072]
  • Synthesis of Isoguanosine or 2′-Deoxy-isoguanosine
  • Few procedures have been reported for the synthesis of isoguanosine or 2′-deoxy-isoguanosine. For example, 2′-deoxy-isoguanosine has been synthesized: 1) from 2′-deoxyadenosine via 2′-deoxyadenosine-(N[0073] 1-oxide) by direct photolysis under basic conditions (Switzer et al. (1993), supra); 2) from 2-chloro-2′-deoxyadenosine by direct photolysis under basic conditions (Seela et al. (1992) Helv. Chim. Acta 75:2298-2306); and 3) by a chemical route from 6-amino-1-(2′-deoxy-beta-D-erythropentofuranosyl)-1H-imidazole-4-carbonitrile [AICA 2′-deoxynucleoside], which was reacted with benzoyl isocyanate followed by treatment with ammonia to affect annealation of the pyrimidine ring (Kazimierczuk et al. (1991) Helv. Chim. Acta 74:1742-1748).
  • However, because the photolytic conversion of 2′-deoxyadenosine-(N[0074] 1-oxide) to 2′-deoxy-iso-guanosine does not lend itself readily to scale-up, a convenient, chemical route to 2′-deoxy-isoguanosine from readily available 2′-deoxyribonucleoside starting materials was developed.
  • Several procedures for the conversion of 2′-deoxyguanosine into 2,6-diaminopurine nucleoside and N[0075] 6-alkyl-2,6-diaminopurine nucleoside via special “convertible” 2′-deoxyguanosine derivatives, such as O6-phenyl-2′-deoxyguanosine, have been described (MacMillan et al. (1991) Tetrahedron 47:2603-2616; Gao et al. (1992) J. Org. Chem. 57:6954-6959; and Xu et al. (1992) Tetrahedron 48:1729-1740). Further, Fathi et al. (1990) Tetrahedron Letters 31:319-322 described a convenient synthesis of O6-phenyl-2′-deoxyguanosine using a procedure involving treatment of 2′-deoxyguanosine with trifluoroacetic anhydride/pyridine followed by in situ displacement with phenol. Alternatively, the introduction of O6-phenyl moieties into 2′-deoxyguanosine has been described by Reese et al. (1984) J. Chem. Soc., Perkin Trans. I, 1263-1271, where the intermediate O6-(4-toluenesulfonyl)-2′-deoxyguanosine was treated with trimethylamine followed by phenol to affect displacement of O6-(4-toluenesulfonyl) to give O6-phenyl-2′-deoxyguanosine. An isoguanosine-like compound was generated from 2-(methylmercapto)-6-amino-pyrazolopyrimidine ribonucleoside by S-oxidation, producing 2-(methylsulfonyl)-6-amino-pyrazolopyrimidine ribonucleoside, followed by displacement with NaOH to give the isoguanosine analogue (Cottam et al. (1983) Nucleic Acids Research 11:871-882).
  • Transformation of the 2-amino group in guanosine and 2′-deoxyguanosine using alkyl nitrites have been described. These include conversion to 2-halo (Nair et al.(1982) [0076] Synthesis 670-672), and 2-(methylmercapto)-6-chloro-purine ribonucleoside (Trivedi (1991) in Nucleic Acid Chemistry, Townsend et al. (eds.) Wiley Inter-Science, Part 4, 269-273), in radical-reactions. Oxidation of O6-(p-nitrophenylethyl)-3′, 5′-O-di-t-butyl-dimethyl silane-2′-deoxyguanosine with neat pentyl nitrite to yield O6-(p-nitrophenylethyl)-3′, 5′-O-di-TBDMS-2′-deoxyxantosine has been reported (Steinbrecher et al. (1993) Angew. Chem. Int. Ed. Engl. 32:404-406).
  • A procedure for the synthesis of 2′-deoxy-iso-guanosine has been described in Seela et al. (1994) [0077] Helv. Chim. Acta 77:622-30. In a first step, 2′-deoxyguanosine was converted to 2-amino-2′-deoxyadenosine. In a second step, 2-amino-2′-deoxyadenosine was deaminated by diazotization of the 2-amino group with sodium nitrite to give 2′-deoxy-isoguanosine.
  • The method disclosed and claimed herein for synthesizing a compound having the structural formula [0078]
    Figure US20010026918A1-20011004-C00007
  • wherein R[0079] 1 is selected from the group consisting of hydrogen, hydroxyl, sulfhydryl, halogeno, amino, alkyl, allyl and -OR2, where R2 is alkyl, allyl, silyl or phosphate, comprises:
  • a) reacting a compound having the structural formula [0080]
    Figure US20010026918A1-20011004-C00008
  • with a reagent suitable to protect both the 3′ and 5′ hydroxyl groups; [0081]
  • b) reacting the product of step (a) with a reagent suitable to convert the O[0082] 6-oxy moiety into a functional group which is susceptible to nucleophilic displacement, thereby producing a functionalized O6 moiety;
  • c) oxidizing the 2-amino group of the product of step (b); [0083]
  • d) reacting the product of step (c) with a nucleophilic reagent to displace the functionalized O[0084] 6 moiety; and
  • e) reacting the product of step (d) with a reagent suitable to deprotect the protected 3′ and 5′ hydroxyl groups. [0085]
  • The conversion of guanosine or 2′-deoxyguanosine to isoguanosine or 2′-deoxy-isoguanosine, respectively, may be effected by protecting the hydroxyl groups on the sugar moiety using a suitable reagent, e.g., TBDMS, benzoyl chloride, acetic anhydride, or the like. As previously noted, one or more of the hydroxyl groups on the sugar moiety may be replaced by halogen, aliphatic groups, or may be functionalized as ethers, amines, or the like. The product is isolated and the O[0086] 6 is modified such that it can be displaced by a suitable nucleophile. Examples of such displaceable groups include, for example, CH3-S-C6H4-O6-, C6H5-SO2-O6-, C6H5-O6-, 4-nitro-C6H4-O6-, 2,4,6-trinitro-C6H2-O6-, or the like. The 2-amino group is then transformed to the oxy function using an alkyl nitrite, or other suitable agent as known in the art (see, Nair et al. (1982), supra; Trevidi (1991), supra; or Steinbrecher et al. (1993), supra). The product is reacted with a suitable nucleophile, e.g., NH4OH, or other aminoalkyl, aminoaryl, aminoheteroalkyl, aminoheteroaryl containing a terminal -NH2, -SH, -COOH, or the like, thereby displacing the modified O6 leaving group. Deprotection of the protected hydroxyl groups may be effected by treatment with, for example, base or fluoride.
  • In the following discussion, O[0087] 6-(4-methylthiophenyl) will serve as an exemplary displaceable group. However, its use is for the purpose of describing particular embodiments only and is not intended to be limiting.
  • N[0088] 6-alkylated isoguanosine derivatives can be readily synthesized by using an alkyl amine as the nucleophile. For example, hexanediamine may be used to displace the O6-(4-methylthiophenyl) to form N6-(6-aminohexyl)-isoguanosine. Protection of the aminohexyl group (e.g., as the trifluoroacetamido derivative) and subsequent conversion into a phosphoramidite reagent provides a functionalizable isoguanosine analog which may be incorporated in any desired position in an oligonucleotide for further post-synthesis derivatization. Thus, it would be possible to label specifically the isoguanosine moiety of selected isoguanosine/isocytidine base pairs. It would also be possible to synthesize a series of N6-derivatives of isoguanosine which carry any desired functionality simply by displacing the O6-(4-methylthiophenyl) group with a suitably terminated nucleophile, e.g., -COOH, -SH, -NH2, or the like, derivatives can be readily prepared.
  • Furthermore, O[0089] 2-(4-methylthiophenyl)-2′-deoxyxantosine, in its fully protected phosphoramidite form (O2-(4-methylthiophenyl)-5′-O-DMT-3′O-(BCE-diisopropylphosphoramidite)-2′-deoxyxantosine) may be used as a convertible derivative following incorporation into an oligonucleotide. Post-synthesis displacement of the O2-(4-methylthiophenyl) from the O2-(4-methylthiophenyl)-2′-deoxyxantosine with an alkyldiamine, or other functionalized alkyl amine, produces N6-(aminoalkyl)-2′-deoxy-isoguanosine-containing oligonucleotides. The derivatized isoguanosine can serve as a site for introduction of a label or other reporter molecule specifically at the functionalized isoguanosine residue.
  • Outline of Synthetic Approach
  • As depicted in [0090] Scheme 1, the synthesis of 2′-deoxy-isoguanosine was accomplished in five steps from 2′-deoxyguanosine as follows:
  • 1) conversion of 2′-deoxyguanosine to 3′, 5′-O-(t-butyldimethylsilyl)[0091] 2-2′-deoxyguanosine (Ogilvie et al. (1973) Can. J. Chem. 51:3799-3807), with purification by recrystallization;
  • 2) conversion to O[0092] 6-(4-toluenesulfonyl)-3′, 5′-O-TBDMS2-2′-deoxyguanosine;
  • 3) displacement of 4-toluenesulfonyl group at O[0093] 6 with a suitable phenol, e.g., 4-(methylthio)phenol or phentachlorophenyl, using Reese's procedure to give O6-(4-(methylthio) phenyl)-3′, 5′-O-TBDMS2-2′-deoxyguanosine (Reese et al. (1984), supra);
  • 4) oxidation of the 2-amino group to the oxy function with tert-butyl nitrite under neutral conditions to give O[0094] 6-(4-(methylthio)phenyl)-3′, 5′-O-TBDMS2-2′-deoxyxantosine (Steinbrecher et al. (1993), supra); and
  • 5) displacement of O[0095] 2-(4-methylthiophenyl) group with ammonium hydroxide at elevated temperature to give 3′, 5′-O-TBDMS2-2′-deoxy-isoguanosine.
  • The synthesis of isoguanosine from guanosine may be effected using a similar reaction scheme. [0096]
    Figure US20010026918A1-20011004-C00009
  • The material obtained was identical in every respect (TLC, HPLC, UV, and NMR) to an authentic sample prepared by a published, photolytic route (Switzer et al. (1993), supra). [0097]
  • Preparation of Isocytidine or 2′-Deoxy-isocytidine Derivatives
  • Derivatives of isocytidine or 2′-deoxy-isocytidine may be prepared in which the glycosidic bond is stabilized against exposure to dilute acid during oligonucleotide synthesis. N[0098] 2-amidine derivatives have been described for 2′-deoxyadenosine by, for example, McBride et al. (1986) J. Am. Chem. Soc. 108:2040-2048, Froehler et al. (1983) Nucleic Acids Res. 11:8031-8036 and Pudlo et al. (1994) Biorg. Med. Chem. Lett. 4:1025-1028. N2-(N,N-di(X)formamidino)-2′-deoxy-isocytidine was synthesized by the following procedure. As exemplified herein, X is n-butyl. However, X may be C2-C10 alkyl, aryl, heteroalkyl, heteroalkyl, or the like.
  • N-di-n-butylformamide dimethylacetal was synthesized by transamination of N,N-methylformamide dimethylacetal with di-n-butylamine as described in McBride et al. (1986), supra, Froehler et al. (1983), supra, and Pudlo et al. (1994), supra. Ten mmole of 2′-deoxy-5-methyl-isocytidine was suspended in 100 ml methanol and 10 mmole of N,N-di-n-butylformamide dimethylacetal was added. After 2 hours at room temperature with stirring, a clear solution resulted. Thin layer chromatography analysis on silica [0099] 60H developed using 10% methanol in methylene chloride indicated that the starting material was completely consumed. Water (10 ml) was added to destroy excess reagent, and the solvents were removed in vacuo to give 3.8 grams of crude N2-(N,N-dibutylformamidino)-2′-deoxy-isocytidine. This derivative can be directly converted to 5′-O-DMT-N2-(N,N-dibutylformamidino)-2′-deoxy-isocytidine for incorporation into oligonucleotides.
  • Other isocytidine derivatives may be prepared which provide functionalizable substituents by which detectable labels may be incorporated into a specific position of an oligonucleotide. For example, 5-alkylated 2′-deoxyuridine derivatives have been described, e.g., 5-[N-(6-trifluoroacetylaminohexyl)-3-(E)acrylamido]-2′-deoxyuridine, by Ruth (1991) [0100] Oligodeoxynucleotides with Reporter Groups Attached to the Base, in Eckstein (ed.) Oligonucleotides and Analogues, IRL press, p. 255-282. Such 5-position derivatives have been found not to obstruct base pair hybridization patterns. The chemistry described by Ruth can be used to synthesize 5-[N-(6-trifluoroacetylaminohexyl)-3-(E)acrylamido]-2′-deoxy-isocytidine, thereby providing a functionalized isocytidine which may be detectably labelled at selected isoguanosine*isocytidine base pairs.
  • These and other 5-position derivatives of isocytidine and 2′-deoxy-isocytidine provide additional stabilization for base pair formation. Such derivatives include: 5-β-propynyl (see, Froehler et al. (1993) [0101] Tetrahedron Lett. 34:1003-1006), 5-β-propenyl or other 5-alkyl isocytidine or 2′-deoxy-isocytidine derivatives.
  • Kits for carrying out nucleic acid hybridization assays according to the invention will comprise in packaged combination at least one hybridizing oligonucleotide probe, a segment of which is capable of forming a hybrid complex with the analyte, and a means for detecting the hybrid complex, wherein the at least one hybridizing oligonucleotide probe comprises a first nucleotidic unit which, under conditions in which A-T and G-C base pairs are formed, will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U). The reagents will typically be in separate containers in the kit. The kit may also include a denaturation reagent for denaturing the analyte, hybridization buffers, wash solutions, enzyme substrates, negative and positive controls and written instructions for carrying out the assay. [0102]
  • The polynucleotides of the invention may be assembled using a combination of solid phase direct oligonucleotide synthesis, enzymatic ligation methods, and solution phase chemical synthesis as described in detail in commonly assigned U.S. patent application Ser. No. 07/813,588. [0103]
  • All chemical syntheses of oligonucleotides can be performed on an automatic DNA synthesizer (Perkin Elmer/Applied Biosystems Division model 380 B). Phosphoramidite chemistry of the β-cyanoethyl type was used including 5′-phosphorylation which employed PHOSTEL™ reagent (DMT-O-CH[0104] 2CH2-(SO2) -CH2CH2-O-P(N(iPr)2) (-O-CH2CH2CN) wherein DMT is dimethoxytrityl and iPr is isopropyl). Standard manufacturer's protocols were used unless otherwise indicated.
  • Example 1 Assay Background Noise Caused by Nonspecific Hybridization of Target-Specific Extender Sequences with Generic Assay Components
  • In order to determine how assay background noise can be caused by cross-hybridization of target-specific extender sequences with generic assay components, an amplified DNA hybridization assay was performed to quantitate M[0105] 13 phage using the pools of capture extenders and label extenders as shown in Tables 1, 2 and 3.
    TABLE 1
    Capture Extender Pool A
    SEQ
    ID
    NO:
    1 ATTGCGAATAATAATTTTTTCACGTTGAAAATC
    TTCTCTTGGAAAGAAAGTGAT
    2 GAATTTCTTAAACAGCTTGATACCGATAGTTG
    TTCTCTTGGAAAGAAAGTGAT
    3 ATTGTATCGGTTTATCAGCTTGCTTTCGAGGT
    TTCTCTTGGAAAGAAAGTGAT
    4 CCGCTTTTGCGGGATCGTCACCTTCTC
    TTGGAAAGAAAGTGAT
    5 GCTGAGGCTTGCAGGGAGTTAAAGG
    TTCTCTTGGAAAGAAAGTGAT
    6 ATGAGGAAGTTTCCATTAAACGGGT
    TTCTCTTGGAAAGAAAGTGAT
    7 TCGCCTGATAAATTGTGTCGAAATCC
    TTCTCTTGGAAAGAAAGTGAT
  • [0106]
    TABLE 2
    Capture Extender Pool B
    SEQ
    ID
    NO:
    8 TCCAAAAAAAAAGGCTCCAAAAGGAGCCTTTA
    TTCTCTTGGAAAGAAAGTGAT
    9 CGCCGACAATGACAACAACCATCGC
    TTCTCTTGGAAAGAAAGTGAT
  • [0107]
    TABLE 3
    Label Extender Pool
    SEQ
    ID
    NO:
    10 ATGAGGAAGTTTCCATTAAACGGGT
    TTAGGCATAGGACCCGTGTCT
    11 GAGGCTTTGAGGACTAAAGACTTTTTC
    TTAGGCATAGGACCCGTGTCT
    12 CCCAGCGATTATACCAAGCGCG
    TTAGGCATAGGACCCGTGTCT
    13 AAGAATACACTAAAACACTCATCTTTGACC
    TTAGGCATAGGACCCGTGTCT
    14 CTTTGAAAGAGGACAGATGAACGGTG
    TTAGGCATAGGACCCGTGTCT
    15 GGAACGAGGCGCAGACGGTCA
    TTAGGCATAGGACCCGTGTCT
    16 ACGAGGGTAGCAACGGCTACA
    TTAGGCATAGGACCCGTGTCT
    17 GCGACCTGCTCCATGTTACTTAGCC
    TTAGGCATAGGACCCGTGTCT
    18 CTCAGCAGCGAAAGACAGCATCGGA
    TTAGGCATAGGACCCGTGTCT
    19 ATCATAAGGGAACCGAACTGACCAA
    TTAGGCATAGGACCCGTGTCT
    20 CCACGCATAACCGATATATTCGGTC
    TTAGGCATAGGACCCGTGTCT
    21 TACAGACCAGGCGCATAGGCTGGC
    TTAGGCATAGGACCCGTGTCT
    22 AAACAAAGTACAACGGAGATTTGTATCA
    TTAGGCATAGGACCCGTGTCT
    23 CACCAACCTAAAACGAAAGAGGCGA
    TTAGGCATAGGACCCGTGTCT
    24 AAAATACGTAATGCCACTACGAAGG
    TTAGGCATAGGACCCGTGTCT
  • For the purpose of illustration, a space separates the 3′nontarget binding region from the target-binding region of each probe. [0108]
  • The assay was run essentially as described in copending application 08/164,388 (Urdea et al). Briefly, after overnight hybridization at 63° C. in microtiter wells containing capture probes complementary to the nontarget binding region of the capture extenders, the plates were cooled at room temperature for 10 min, washed twice with a buffer containing 0.1x SSC (15 mM NaCl; 1.5 mM sodium citrate; pH [0109] 7.0), 0.1% sodium dodecyl sulfate. A 15×3 (15 “arms” each with 3 alkaline phosphatase probe binding sites) branched DNA amplifier (100 fm) complementary to the 3′ nontarget binding region of the label extender was added to the wells and the incubation was continued for 30 min at 53° C. after which the plates were cooled and washed as above. After the addition of an alkaline phosphatase probe (200 fm) to the wells and a further incubation for 15 min at 53° C., the plates were again cooled and washed as above. Three additional washes were done with a 0.1x SSC buffer. The signals were detected in a Chiron luminometer after 20 min in the dioxetane phosphate substrate solution Lumiphos 530 (Lumigen). The results are shown in Table 4.
    TABLE 4
    Nonspecific Binding Assay Background Noise
    Signal Noise
    Capture Extender Pool (+ M13 phage) (− M13 phage)
    Pool A alone 293, 306, 337, 359 1.1, 0.9, 1.1, 2.0
    Pool A + Pool B 390, 393, 379, 376 103, 130, 436, 172
  • The addition of the pool B capture extenders does not increase the net signal, but does increase the noise about one hundred-fold Computer analysis of the sequences involved showed that capture extender #[0110] 8 of pool B has extensive homology with the T20—LLA2 sequence of the branched DNA amplifier (including a 9mer oligo(dA)--oligo(dT)), while capture extender #9 of pool B has extensive homology with the BLA3c sequence of the branched DNA amplifier.
  • The present invention addresses the problem of hybridization-dependent assay background noise. Nucleotide sequences are constructed which are interrupted by nucleotides that do not form stable base pairs with “natural” nucleobases, thereby inhibiting the hybridization of such sequences with natural sequences. Ideally, every third or fourth base in the universal sequence would be a modified nucleotide that does not pair with A,C, G, or T(U). By using base pairs isoenergetic with the C*G base pair, one can also reduce the length of the universal sequences. Statistical arguments show that this should also reduce the frequency of undesirable cross-hybridization among universal sequences and between universal sequences and nontarget sequences in the sample and between universal sequences and the target-specific sequences in the extender probes. By relying on multidentate binding to form stable hybrids, the lengths of the universal sequences can be further reduced (see copending application 08/164,338). All universal sequences would be designed with at least 6 and preferably 8 nucleotides: capture probe, capture extender tails, label extender tails, amplifiers, labeled probes, and preamplifiers (when applicable). [0111]
  • Example 2 Specificity and Strength of isoC-isoG Base Pairs
  • In order to determine the specificity and strength of the-isoC-isoG base pair, thermal melt analysis was done on the following oligonucleotides: [0112]
    1) 5′ (L) CA CCA CTT TCT CC (T) 3′ [SEQ ID NO: 25];
    2) 5′ (L) CA CFA CTT TCT CC (T) 3′ [SEQ ID NO: 26]
    3) 3′ (T) GT GGT GAA AGA GG 5′ [SEQ ID NO: 27];
    4) 3′ (T) GT GJT GAA AGA GG 5′ [SEQ ID NO: 28]; and
    5) 5′ CA CTA CTT TCT CC (T) 3′ [SEQ ID NO: 29].
  • The core hybrid of these oligonucleotides consists of thirteen nucleotides. Nucleotides not involved in the base-pairing are indicated in parentheses. L=a primary amine, F=isoc, J=isoG. Thermal melt analysis was done on a Cary [0113] 3E Spectrophotometer in 3x SSC (0.45 M NaCl, 0.045 M sodium citrate), pH 7.9. Each of the two oligonucleotides incubated together was present at approximately 1.5 μM. The Tm was calculated as the maximum in a plot of dA260/dT vs temperature. The results shown in Table 4 indicate that the isoC*isoG base pair is isoenergetic with the natural C*G base pair.
    TABLE 4
    Tm Analysis of Specificity of isoC*isoG Base-pairing
    Avg
    Match/Mismatch, Paired Oligonucleotides Tm1 Tm2 Tm
    C * G match, 1*3 60 60 60
    isoC * isoG match, 2*4 60 61 60
    isoC * G mismatch, 2*3 52 52 52
    isoG * C mismatch, 1*4 52 52 52
    G * T mismatch, 3*5 50 49 49
    isoG * T mismatch, 4*5 53 53 53
  • Accordingly, universal sequences containing approximately equimolar C, G, isoC, isoG, A, and T, can be shorter than sequences containing only A, T, C, G in approximately equal ratios. This limits the potential for cross-reactivity with natural nontarget sequences in the sample and with LE and CE target-binding sequences that are more or less constrained to be composed of A, T(U), C, and G. [0114]
  • The data also show the specificity of the isoC*isoG base-pair. The isoC*G and isoG*C pairs behave as mismatches. Classically, the destabilization in degrees C is approximated by the percent mismatching. Thus, about a 7.5° C. change in T[0115] m would be predicted to occur for 1 mismatch in 13 nucleotides (7.5% mismatch). The observed 8° C. change when the C*G or isoC*isoG matches are compared with the mismatches is similar to the change which would occur in an average mismatch with A, T, C, and G code.
  • IsoG exists in at least two tautomeric forms, the keto and the enol. The keto form is favored in aqueous solvents and the enol is favored in organic solvents (Sepiol et al. (1976) [0116] Zeitschrift fuer Naturforschung 31C:361-370). The isoG enol tautomer can, in principle, form two hydrogen bonds to dT, making it analogous to the A*T base pair. If the enol tautomer were present at significant levels in the hybridization buffer, the specificity of isoC*isoG base pair would be limited. However, the observed Tm in the isoG*T mismatch was 53° C., essentially the same as the other mismatches.
  • These data support the conclusion that the enol tautomer is present at very low concentration in 3X SSC at pH [0117] 7.9 or, if present, it still forms a hybrid with 7-8° C. lower Tm than the isoC-isoG hybrid. The control with a G*T mismatch had a Tm of about 49° C. This is somewhat lower than expected for the average G*T mispair, but is close to the isoG-T mispair.
  • One skilled in the art will appreciate that having still another base-pairing combination (i.e., 8 bases, 4 pairs), whether isoenergetic with C*G or not, would further improve the specificity of the base-pairing among universal sequences. In this case, one could nearly eliminate A, T, C, and G from the universal sequences. However, having a small representation of these bases adds to the diversity of the library of possible universal sequences, which enables one to design universal sequences that are as noninteracting as possible among themselves. [0118]
  • For example, with a 4 base code one can design only two pairs of universal 15mers that do not have even a single 3mer cross hybrid. That is, with the addition of a third pair of 15mer sequences, there must be at least some 3 nucleotide cross hybrids. With a six base code, one can design 8 pairs of 15mer sequences without even one 3mer Watson-Crick type of cross-hybrid. With an eight base code, one can design 19 such pairs of 15mers. [0119]
  • Example 3 The Effect of pH on isoC*IsoG Base Pairing
  • In order to examine the behavior of the isoC*isoG base pair as a function of pH, T[0120] m analysis was conducted on the oligonucleotides provided in Example 2.
  • The effect of pH on the T[0121] m of the oligonucleotides containing the complementary isoC*isoG base pair (sequences 2 and 4, respectively) and C*G base pair ( sequences 1 and 3, respectively) was determined (n=2 or 3) at 0.5 M salt and approximately 1.5 μM oligonucleotide, and the results are shown in Table 5.
    TABLE 5
    Tm Analysis of pH-sensitivity of isoC*isoG Base Pair
    Avg
    Hybrid, Paired Oligonucleotides pH Tm1 Tm2 Tm3 Tm
    isoG*isoC, 2*4 7.9 62 60 62 61
    isoG*isoC, 2*4 5.1 60 59 60 60
    isoG*isoC, 2*4 9.5 53 51 52 52
    G*C, 1*3 9.5 52 52 52
  • Generally, oligonucleotide hybrids are stable at pH [0122] 5 and pH 10. Below pH 5, C and A become protonated, while above pH 10, G and T begin to lose their imino protons. Thus, below pH 5 and above pH 10, nucleic acid hybrids show reduced stability. The data of Table 2 show that the isoC*isoG base pair has normal acid stability. However, both the isoG*isoC hybrid and the G*C hybrid show an unusual −9° C. change in Tm over a 1.6 unit pH increase. This is probably due to their very short length.
  • Theoretically, one could select hybrids with still greater pH-sensitivity using the SELEX protocol, described in U.S. Pat. No. 5,270,163 to Gold et al., Tuerk et al. (1990) [0123] Science 249:505-510, Szostak et al. (1990) Nature 346:818-822 and Joyce (1989) Gene 82:83-87, in which a population of DNA or RNA randomers would be selected for binding at neutral pH and for dissociation from the target sequence at mildly alkaline or mildly acid pH. Following amplification, the selection process would be iteratively repeated. After the final iteration, those oligomers which show the desired pH sensitivity would be cloned and sequenced. Those sequences would be synthesized and the best performers selected in a direct competition assay.
  • Lability in mild base can be exploited in the current amplified DNA assay format to reduce assay background noise. In the final step, the substrate buffer used is typically pH [0124] 9.5 to 10.5. With a capture probe with the proper base lability, the target will come off the surface and could be detected in another well. The background will be left behind. Minimization of capture extender binding to the support by the methods disclosed in copending application 08/164,388 (Urdea et al.) will reduce background noise caused by release of molecules nonspecifically bound to capture probes through capture extenders.
  • Since one would not want to release alkaline phosphatase probes hybridized to nonspecifically bound amplifiers, preferably the capture probe-capture extender hybrids would be selected to have considerably more base lability (i.e., higher T[0125] m at a given pH) than the amplifier and labeled probe and the amplifier and label extender hybrids. Alternatively, L-2/M-2 hybrid of FIG. 1 could be the base-labile hybrid. In either instance, the M-2/L-3 hybrid must be the most stable; otherwise, labeled probe hybridized to nonspecifically bound amplifier would be released.
  • As noted above, one could also conceivably transfer the released target to fresh wells for reading. However, it would be preferable to read the released solution in the well where it was generated. This would avoid additional pipetting steps and eliminate imprecision associated with additional liquid transfer steps. There are several methods by which well transfers may be avoided, as described below. [0126]
  • To further enhance the specificity of the assay, the specific release of the target could be coupled with masking the background on the surface. In this case, the transfer to another support would be unnecessary. For example, the surface of the solid support could be coated with inhibitors of the labeled probe and/or various luminescence inhibitors, absorbers, or quenchers. One surface coating currently in use is poly(phe-lys). Phenylalanine is a known inhibitor of alkaline phosphatase, a particularly preferred enzyme label. One could include in the polymeric peptide coating other inhibitors of alkaline phosphatase such as tryptophan and cysteine. Examples of luminescent inhibitors include compounds with low quantum yields, i.e., any compound that preferentially gives off heat rather than light after being excited by collision with a dephosphorylated dioxetane. [0127]
  • There are at least two other convenient ways to make detection of the released solution more selective to avoid transfer of the released target to another well. The target-associated signals can be read in solution by making the solid phase inaccessible to visualization reagents or by masking signal generating reactions which occur on the solid support. Isolating the solid phase from subsequent visualization steps could be done by adding a heavier-than-water immiscible oil to the reaction vessel. This oil would cover the bottom of the vessel while allowing the solution of interest to float to the top. For simple calorimetric detection by visual or by reflectance measurement, an opaque substance could be added to the oil to serve as a neutral background for visualization. [0128]
  • For chemiluminescent detection the oil could be filled with an optically opaque substance. If a white solid such as titanium dioxide were used, light emitted from the floating aqueous layer would be reflected upward out of the container for detection. A dark solid or dye molecule dissolved in the oil could also be used to mask the stationary phase. Even if the oil solution does not completely isolate the solid phase from visualization reagents, the suspended solids or dissolved dyes would block the transmission of this light from the surface. [0129]
  • It is also possible that a stationary phase could be colored with a dye that would block emission of light from reactions that occur near its surface. This would be particularly convenient with a colored bead as a solid phase contained within an opaque well. [0130]
  • Example 4 The Effect of Salt on isoC*isoG Base Pair At Neutral and Alkaline pH
  • In order to examine the behavior of the isoC*isoG base pair as a function of salt concentration, T[0131] m analysis was conducted of the oligonucleotides provided in Example 2. The effect of salt concentration on the Tm of the oligonucleotides containing the complementary isoC*isoG base pair (sequences 2 and 4, respectively) and C*G base pair ( sequences 1 and 3, respectively) was determined (n=3) at pH 7.9 or 9.5 and approximately 1.5 μM oligonucleotide, and the results are shown in Table 6.
  • Classically, polynucleotides show a change of approximately 16-17° C. in T[0132] m for each log change in salt concentration. Oligonucleotides often show somewhat reduced salt dependence. The 10-11° C. change in Tm per log change in salt at pH 7.9 calculated for the isoC*isoG hybrid approximates what would be expected for a 13mer. However, the change at pH 9.5 of only about 3° C. for the isoC*isoG hybrid and 5 degrees for the C*G hybrid per log change in salt was surprisingly low.
  • This can be also exploited in a specific release of target. Generally, low salt is used for specific release of target. Unfortunately, often a significant fraction of the background is also released. [0133]
    TABLE 6
    IsoC*IsOG Stability as a Function of Salt Concentration
    AVG
    Salt Tm dTm
    Hybrid, Paired Oligonucleotide (M) pH (° C.) dlog [Na+]
    isoC*isoG, 2*4 0.5 7.9 61
    isoC*isoG, 2*4 0.17 7.9 56 10-11
    IsoC*isoG, 2*4 0.5 9.5 52
    isoC*isoG, 2*4 0.17 9.5 50 3
    C*G, 1*3 0.5 9.5 52
    C*G, 1*3 0.1 9.5 48.5 5
  • Because of the salt independence of the melt of the isoC*isoG base pair at mildly alkaline pH, there is no additional advantage gained from lowering the salt as well as increasing the pH. Thus one can use high salt (which is also preferred for alkaline phosphatase) for the release and minimize the release of the background. [0134]
  • As explained in Example 3, the SELEX procedure could be used to find DNA or RNA sequences that show enhanced salt-independence in their melting at any selected pH. [0135]
  • Example 5 The Effect of Base Pair Mismatching on Hybridization
  • The previous examples showed that an oligomer with isoG base pairs specifically with its complement containing isoC. The isOG-containing oligomer is destabilized by about 7-8° C. when hybridized to another oligomer containing a single isoG*T or isoG*C mismatch. Typically, there is about a tenfold decrease in binding for each 10° C. degree change in T[0136] m.
  • The effect of mismatching two bases on binding of a 13mer hybrid was assessed using the probes shown in Table 7. [0137]
    TABLE 7
    SEQ
    ID
    NO: SEQUENCE1
    30 5′ GATGTGGTTGTCGTACTTTTTTTGACACTCCACCAT
    31 5′ GATGTGGTTGTCGTACTTTTTTTGACAFTCCJCCAT
    32 ALK. PHOS.-- CTACACCAACAGCATGAA 5′
    33 3′ TCACTAAGTACCACCTCACAG
    34 5′ AGTGATTCATGGTGGAGTGTCTCTCTTGGAAAGAAAGTGAT
    35 3′ GAGAACCTTTCTTTCACTX
  • [0138] 1 F=isoC, J=isoG, ALK. PHOS.=alkaline phosphatase, and X=a spacer sequence containing an amine for attachment to the solid support.
  • Labelled probe [0139] 32, the alkaline phosphatase oligonucleotide conjugate, was made as described (Urdea et al. (1988) Nucl. Acids Res. 16:4937-4955). Labelled probe 32 was hybridized with control probe 30 to create the alk. phos.-probe 30*32. Labelled probe 32 was hybridized with modified probe 31 to create the isoC,isoG-alk. phos.-probe 31*32.
  • Probe [0140] 35, the capture probe, was bound to microtiter wells as described (PCT Publication No. W091/813,338, the disclosure of which is incorporated by reference herein) to create a solid support for hybridization. Probe 34, a capture extender, was hybridized to probe 35. This capture extender is complementary to the alk. phos.-probe 30*32 and partially complementary to the alk. phos.-probe 31*32. Probe 33 is a “competimer” that can bind to the capture extender and block the binding of either alkaline phosphatase probe.
  • The following incubations were done for 30 min at 53° C. in approximately 1.0 M NaCl: [0141]
  • (1) 250 fmoles probe [0142] 34 in wells containing 1 pmole of immobilized probe 35;
  • (2) 250 fmoles probe [0143] 34+5 pmoles probe 33 in wells containing 1 pmole of immobilized probe 35;
  • (3) 5 pmoles probe [0144] 33 in wells containing 1 pmole of immobilized probe 35; and
  • (4) buffer only. [0145]
  • After 2 washes with 0.1x SSC, 0.1% SDS, as defined in Example 1, each of the above first incubations was exposed to a second, 15 min. incubation under the same conditions with each of the following: [0146]
  • (1) 25 fmoles probe [0147] 30+500 attomoles probe 32;
  • (2) 25 fmoles probe [0148] 31+500 attomoles probe 32;
  • (3) 500 attomoles probe [0149] 32; and
  • (4) buffer only. [0150]
  • The plates were washed twice as above and three times with the same buffer supplemented with 10 mM MgCl[0151] 2, 1 mM ZnCl2, 0.1% Brij-35. After a 25 min. incubation with Lumiphos Plus (Lumigen), the plates were read on a Chiron luminometer.
  • The hybrids that can form are depicted in FIG. 3, wherein Z, exemplified herein by isoC and isoG, represents a nonnatural nucleotide. Probe [0152] 33, the competimer, can form 21 base pairs with the capture extender and in theory can block both alkaline phosphatase probes from binding. The modified probe*labelled probe (31*32) can hybridize to the capture extender, forming 11 base pairs and two mismatches (e.g., G*isoC,isoG*T). The control probe*labelled probe (30*32) can form 13 base pairs with the capture extender.
  • As shown in Table 8, the capture extender ([0153] 34) forms a strong hybrid with the control probe*labelled probe (30*32) (Sample 1=399 Relative Light Units (RLU)). Preincubation of the capture extender with a 20-fold molar excess of competimer, sample 2, reduced this background noise about tenfold (30 RLU). The modified probe*labelled probe (31*32) shows 40-fold less hybridization (sample 3=9 RLU) to the capture extender than control probe*labelled probe (30*32). The two mismatches accounted for a 40-fold change in hybridization. This is as expected for 2 mismatches, each of which destabilizes the Tm by 7-8° C. (cf. 7×8=56-fold). The use of the competimer and the mismatched alk. phos. probe (sample 4=0.4 RLU), reduced the background noise about 1000-fold. Sample 5 is a control and has essentially no background noise (0.1 RLU). This is as expected since the labelled probe 32 has no detectable homology with the capture extender.
    TABLE 8
    The Effect of Base Pair Mismatching on Hybridization
    AVG
    Sample First Second RLU1 %
    No. Hybridization Hybridization (n = 6) CV 2
    1 34 + 35 30 + 32 399 7
    2 33 + 34 + 35 30 + 32 30 9
    3 34 + 35 31 + 32 9 6
    4 33 + 34 + 35 31 + 32 0.4 4
    5 34 + 35 32 0.1 11 
  • In hybridization assays, the use of competimers for all the capture extenders is impractical since there are typically 5-10 capture extenders per assay. In addition, this example shows that preincubation with the competimer was not as efficient as simply using 15% base substitution (with isoC, isoG), e.g., 2 bases out of 13, in the universal sequences. The use of 30% base substitution (3 out of 10) would be expected to reduce nonspecific hybridization of an otherwise perfectly base-paired complement by about 1000-fold (30% mismatch equals approximately 30° C. change in T[0154] m; there is about a tenfold decrease in binding for each 10° C. change in Tm).
  • Example 6 Chemical Synthesis of 2′-deoxy-isoguanosine.
  • The synthesis of 2′-deoxy-isoguanosine from 2′-deoxyguanosine was accomplished by the following procedure. [0155]
  • [0156] Step 1. 2′-Deoxyguanosine monohydrate (50 mmole) and imidazole (200 mmole) were dried by coevaporation with 500 mL dimethylformamide (DMF) and the residue dissolved in 500 mL DMF. To this solution was added t-butyldimethylsilyl chloride (150 mmole), and the reaction mixture was left stirring at room temperature for 18 hours. Methanol (30 mL) was added and after 25 minutes the solvents were removed in vacuo. The solvents were removed by evaporation, the residue dissolved in 1L CH2Cl1, washed with 1L 5% NaHCO3 and 1L 80% saturated NaCl, the organic phase dried over Na2SO4, filtered and evaporated to dryness yielded crude product (30 grams) which was directly dissolved in 2L hot ethanol. Slow cooling to 20° C. followed by storage at 4° C. for 20 hours produced pure 3′, 5′-TBDMS2-2′-deoxyguanosine (65% yield).
  • [0157] Step 2. 3′, 5′-TBDMS2-2′-deoxyguanosine (12 mmole) was suspended in 125 mL CH2Cl2 containing triethylamine (150 mmole) and N,N-dimethylaminopyridine (100 mg). 4-Toluenesulfonyl chloride (40 mmole) was at 0° C., and the reaction mixture stirred at room temperature for 20 hours. At that time all solid material had dissolved resulting in a slightly yellow solution. The reaction was quenched with 50 mL 5% NaHCO3 with stirring for 1 hour. The reaction mixture was diluted with 300 mL CH2Cl2, washed with 300 mL 5% NaHCO3 and 300 mL 80% saturated NaCl, the organic phase- dried over Na2SO4, filtered and evaporated to dryness yielded crude product (8.9 grams). Silica gel flash chromatography using a 1% to 4% methanol/CH2Cl2 gradient yielded 7.95 grams of pure O6-(4-toluenesulfonyl)-3′,5′-O-TBDMS2-2′-deoxyguanosine (11 mmole).
  • [0158] Step 3. Twelve grams of O6-(4-toluenesulfonyl)-3′,5′-O-TBDMS2-2′-deoxyguanosine (17 mmole) was suspended in 300 mL CH3CN. Then methylpyrrolidine (17 mL) was added and the suspension stirred for one hour to produce a clear solution. TLC analysis showed that all starting material had been converted to base line material. Eleven grams of 4-(methylthio)phenol (85 mmole) was added and the solution stirred for 60 hours. After evaporation to a small volume 600 mL ethyl acetate was added. This solution was extracted with 3×400 mL of 0.3 M NaOH and 400 mL 80% saturated NaCl, the organic phase dried over Na2SO4, filtered and evaporated to dryness to yield 11.55 grams crude product. Silica gel flash chromatography using a 4% to 5% methanol/CH2Cl2 gradient yielded 8.16 grams of O6-(4-(methylthio)phenyl)-3′,5′-O-TBDMS2-2′-deoxy-guanosine (11 mmole).
  • Step 4. Four grams of O[0159] 6-(4-(methyl-thio) phenyl)-3′,5′-O-TBDMS2-2′-deoxyguanosine (6.5 mmole) was dissolved in 65 mL CH2Cl2 at 0° C. and 6.5 mL of tert-butyl nitrite was added dropwise. The solution was allowed to warm to room temperature and gas evolved from the mixture (N2). After 40 minutes, when TLC analysis showed complete consumption of starting material and emergence of a new, slower migrating spot, excess t-butyl nitrite was removed by coevaporation with 2×100 mL toluene in vacuo. The residue of crude product was purified by silica gel flash chromatography using a 4% to 5% methanol/CH2Cl2 gradient to yield 2.75 grams of O6 (4-(methylthio) phenyl)-3′,5′-O-TBDMS2-2′-deoxyxantosine (4.45 mmole).
  • Step 5. All of the purified 2.75 grams of O[0160] 6-(4-(methylthio) phenyl)-3′,5′-O-TBDMS2-2′-deoxyxantosine (4.45 mmole) was dissolved in 50 mL of methanol. Concentrated aqueous ammonium hydroxide (50 mL) was added and the mixture heated in a tightly sealed bomb at 100° C. for 4 hours. After cooling the solvents were removed by coevaporation with ethanol in vacuo to give 1.8 grams of crude product (3.6 mmole). Purification by recrystallization from hot ethanol yielded a sample of pure 3′,5′-O-TBDMS2-2′-deoxy-isoguanosine. This material was in every respect (UV, TLC, NMR and MS) identical to a sample prepared by the published, photolytic route (Switzer et al. (1993), supra).
  • Thus, novel methods for generating a more target-dependent signal in solution phase sandwich hybridization assays have been disclosed. In addition, a novel method for synthesizing 2′-deoxy-isoguanosine has been disclosed. Although preferred embodiments of the subject invention have been described in some detail, it is to be understood that obvious variations can be made without departing from the spirit and the scope of the invention as defined by the appended claims. [0161]

Claims (27)

We claim:
1. In a nucleic acid hybridization assay for detecting a nucleic acid analyte in a sample using a plurality of assay components each of which comprises at least one hybridizing oligonucleotide segment, the improvement which comprises incorporating into at least one hybridizing oligonucleotide segment a first nucleotidic unit which will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U) under conditions in which A-T and G-C base pairs are formed.
2. The method of
claim 1
, wherein the first nucleotidic unit is capable of forming a base pair with a second, complementary nucleotidic unit.
3. The method of
claim 2
, wherein the first and second nucleotidic units are incorporated into hybridizing oligonucleotide segments of assay components which are not complementary to nucleic acid segments of the nucleic acid analyte.
4. The method of
claim 3
, wherein the first and second nucleotidic units are interchangeably selected from the group of complementary base pairs consisting of:
Figure US20010026918A1-20011004-C00010
wherein R is a backbone which will allow the first and second nucleotidic units to form a base pair with a complementary nucleotidic unit when incorporated into a polynucleotide, and R′ is hydrogen, methyl, α- or β-propynyl, bromine, fluorine or iodine.
5. The method of
claim 4
, wherein the first and second nucleotidic units have the structure (I)
Figure US20010026918A1-20011004-C00011
6. The method of
claim 1
, wherein the nucleic acid hybridization assay is a solution phase sandwich hybridization assay comprising (a) binding the analyte directly or indirectly to a solid support, (b) labelling the analyte, and (c) detecting the presence of analyte-associated label,
7. In a nucleic acid hybridization assay for detecting a nucleic acid analyte in a sample using a plurality of assay components each of which comprises at least one hybridizing oligonucleotide segment,
the improvement which comprises incorporating Tm1 hybrid complexes and Tm2 hybrid complexes such that assay stringency can be controlled to selectively destabilize the Tm1 hybrid complexes.
8. The assay of
claim 7
, wherein the Tm1 hybrid complexes comprise at least one of the hybrid complexes selected from the group consisting of a capture probe/capture extender complex, a label extender/amplifier complex and a label extender/preamplifier complex, and wherein the label comprises a member of a Tm2 hybrid complex.
9. The assay of
claim 7
, wherein the Tm2 hybrid complexes comprise one or more of the complexes selected from the group consisting of a label extender/amplifier complex, a label extender/preamplifier complex and a label/amplifier complex, with the proviso that the label comprises a member of a Tm2 complex.
10. The assay of
claim 7
, wherein the assay stringency is controlled by altering formamide concentration, salt concentration, chaotropic salt concentration, pH, organic solvent content or temperature.
11. The assay of
claim 10
, wherein the assay stringency is controlled by altering the pH.
12. The assay of
claim 10
, wherein the assay stringency is controlled by altering the salt concentration.
13. The assay of
claim 11
, further comprising inhibiting the detection of nonspecifically bound label.
14. The assay of
claim 13
, wherein the step of inhibiting the detection of nonspecifically bound label is effected by coating the solid support with an opaque substance, a label inhibitor or a luminescence inhibitor, absorber or quencher, providing a layer of oil over the nonspecifically bound label, or transferring the target-associated label to another vessel for label detection.
15. The assay of
claim 6
, which further comprises incorporating Tm1 hybrid complexes and Tm2 hybrid complexes such that assay stringency can be controlled to selectively destabilize the Tm1 hybrid complexes.
16. In a solution phase sandwich hybridization assay for detecting a nucleic acid analyte in a sample using a plurality of assay components each of which comprises at least one hybridizing oligonucleotide segment, comprising (a) binding the analyte directly or indirectly to a solid support, (b) labelling the analyte, and (c) detecting the presence of analyte-associated label,
the improvement which comprises incorporating into at least one hybridizing oligonucleotide segment a first nucleotidic unit a first nucleotidic unit which will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U) under conditions in which A-T and G-C base pairs are formed.
17. In a solution phase sandwich hybridization assay for detecting a nucleic acid analyte in a sample using a plurality of assay components each of which comprises at least one hybridizing oligonucleotide segment, comprising (a) binding the analyte directly or indirectly to a solid support, (b) labelling the analyte, and (c) detecting the presence of analyte-associated label,
the improvement which comprises incorporating Tm1 hybrid complexes and Tm2 hybrid complexes such that assay stringency can be controlled to selectively destabilize the Tm1 hybrid complexes.
18. A method for synthesizing a compound having the structural formula
Figure US20010026918A1-20011004-C00012
wherein R′ is selected from the group consisting of hydrogen, hydroxyl, sulfhydryl, halogeno, amino, alkyl, allyl and -OR2, where R2is alkyl, allyl, silyl or phosphate, comprising:
a) reacting a compound having the structural formula
Figure US20010026918A1-20011004-C00013
with a reagent suitable to protect both the 3′ and 5′ hydroxyl groups;
b) reacting the product of step (a) with a reagent suitable to convert the O6-oxy moiety into a functional group which is susceptible to nucleophilic displacement, thereby producing a functionalized O6 moiety;
c) oxidizing the 2-amino group of the product of step (b);
d) reacting the product of step (c) with a nucleophilic reagent to displace the functionalized O6 moiety; and
e) reacting the product of step (d) with a reagent suitable to deprotect the protected 3′ and 5′ hydroxyl groups.
19. A method for synthesizing 2′-deoxy-iso-guanosine comprising:
a) converting 2′-deoxyguanosine to 3′, 5′-O-(t-butyldimethylsilyl)2-2′-deoxyguanosine by reacting 2′-deoxyguanosine with t-butyldimethylsilyl (TBDMS) chloride;
b) converting 3′, 5′-O-TBDMS2-2′-deoxyguanosine to O6 (4-toluenesulfonyl)-3′,5′-O-TBDMS2-2′-deoxyguanosine by reacting 3′,5′-O-TBDMS2-2′-deoxyguanosine with 4-toluenesulfonyl chloride;
c) displacing the O6-(4-toluenesulfonyl) group by treating O6-(4-toluenesulfonyl)-3′,5′-O-TBDMS2-2′-deoxyguanosine with a phenol to give O6-(4-(methylthio)phenyl)-3′, 5′-O-TBDMS2-2′-deoxyguanosine;
d) oxidizing the 2-amino group of O6-(4-(methylthio)phenyl)-3′, 5′-O-TBDMS2-2′-deoxyguanosine to the oxy function by treating O6-(4-(methylthio)phenyl)-3′,5′-O-TBDMS2-2′-deoxyguanosine with t-butyl nitrite under neutral conditions to give O6-(4-(methylthio)-phenyl)-3′, 5′-O-TBDMS2-2′-deoxyxantosine; and
e) displacing the O2-(4-(methylthio)phenyl) group of O6-(4-(methylthio)phenyl)-3′,5′-O-TBDMS2-2′-deoxyxantosine with ammonium hydroxide at elevated temperature to give 3′, 5′-O-TBDMS2-2′-deoxy-isoguanosine.
20. A kit for detecting a nucleic acid analyte in a sample, comprising at least one hybridizing oligonucleotide probe, a segment of which is capable of forming a hybrid complex with the analyte, and a means for detecting the hybrid complex, wherein the at least one hybridizing oligonucleotide probe comprises a first nucleotidic unit which will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U) under conditions in which A-T and G-C base pairs are formed.
21. The kit of
claim 20
comprising:
(a) a set of capture probes, wherein said capture probes comprise a first nucleotidic unit which will not effectively base pair with A, T, C, G or U under conditions in which A-T and G-C base pairs are formed;
(b) a set of capture extender molecules comprising first and second hybridizing oligonucleotide segments, wherein the first hybridizing oligonucleotide segment is capable of forming hybrid complexes with the capture probes and the second hybridizing oligonucleotide segment is capable of forming hybrid complexes with predetermined segments of the nucleic acid analyte;
(c) label extender molecules comprising third and fourth hybridizing oligonucleotide segments, wherein the third hybridizing oligonucleotide segment is capable of forming hybrid complexes with segments of the nucleic acid analyte other than those to which the set of capture extender molecules bind;
(d) an optional preamplifier molecule comprising fifth and sixth hybridizing oligonucleotide segments, wherein the hybridizing oligonucleotide segments comprise a first nucleotidic unit which will not effectively base pair with A, T, C, G or U under conditions in which A-T and G-C base pairs are formed, and wherein the preamplifier molecule is capable of forming hybrid complexes with the label extender molecules and a plurality of amplification multimers;
(e) an amplification multimer comprising seventh and eighth hybridizing oligonucleotide segments, wherein the hybridizing oligonucleotide segments comprise a first nucleotidic unit which will not effectively base pair with A, T, C, G or U under conditions in which A-T and G-C base pairs are formed, and wherein the amplification multimer is capable of forming hybrid complexes with the label extender molecules or to the preamplifier molecules, and a plurality of identical oligonucleotide subunits; and
(f) label probes comprising a label, which are designed to form hybrid complexes with the identical oligonucleotide subunits and which provide, directly or indirectly, a detectable signal.
22. The kit of
claim 21
, wherein the hybrid complexes are selected from the group consisting of Tm1 hybrid complexes and Tm2 hybrid complexes such that assay stringency can be controlled to selectively destabilize the Tm1 hybrid complexes with the proviso that the label comprises a member of a Tm2 complex.
23. The kit of
claim 22
, wherein the Tm1 hybrid complexes comprise at least one of the hybrid complexes selected from the group consisting of a capture probe/capture extender complex, a label extender/amplifier complex and a label extender/preamplifier complex, and wherein the label comprises a member of a Tm2 hybrid complex.
24. The kit of
claim 23
, wherein the Tm2 hybrid complexes comprise one or more of the complexes selected from the group consisting of a label extender/amplifier complex, a label extender/preamplifier complex and a label/amplifier complex, with the proviso that the label comprises a member of a Tm2 complex.
25. An oligonucleotide useful as an aptamer, comprising an intramolecular oligonucleotide hybrid complex containing a plurality of complementary base pairs at least one of which comprises complementary nonnatural nucleotidic units that will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U) under conditions in which A-T and G-C base pairs are normally formed, and wherein the nonnatural nucleotidic unit is contained within an oligonucleotide segment in which specificity of the base pairs is not required for maintaining secondary structure of the aptamer.
26. A method for preparing an aptamer comprising:
(a) providing a target molecule;
(b) contacting the target molecule with a randomer pool of oligonucleotides under conditions which favor binding of the oligonucleotides to the target molecule;
(c) separating the oligonucleotides which bind to the target molecule and form an oligonucleotide-target complex from the oligonucleotides which do not bind to the target molecule;
(d) dissociating the oligonucleotide from the oligonucleotide-target complex;
(e) amplifying the oligonucleotide using a polymerase chain reaction;
(f) repeating steps (b) through (e) at least once to form a final aptamer construct; and
(g) replacing one or more nucleotidic units in the final aptamer construct with nonnatural nucleotidic units that will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U) under conditions in which A-T and G-C base pairs are normally formed.
27. An antisense molecule comprising first and second hybridizing segments, wherein the first hybridizing segment is capable of forming a hybrid complex with a target oligonucleotide and the second segment comprises at least one nucleotidic unit which will not effectively base pair with adenosine (A), thymidine (T), cytidine (C), guanosine (G) or uridine (U) under conditions in which A-T and G-C base pairs are formed, and is capable of forming a hybrid complex with a second antisense molecule.
US09/752,213 1994-08-30 2000-12-28 Reduction of nonspecific hybridization by using novel base-pairing schemes Abandoned US20010026918A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US09/752,213 US20010026918A1 (en) 1994-08-30 2000-12-28 Reduction of nonspecific hybridization by using novel base-pairing schemes

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US08/298,073 US5681702A (en) 1994-08-30 1994-08-30 Reduction of nonspecific hybridization by using novel base-pairing schemes
US43554795A 1995-05-05 1995-05-05
US08/794,153 US5780610A (en) 1994-08-30 1997-02-03 Reduction of nonspecific hybridization by using novel base-pairing schemes
US09/115,566 US6232462B1 (en) 1994-08-30 1998-07-14 Reduction of nonspecific hybridization by using novel base-pairing schemes
US09/752,213 US20010026918A1 (en) 1994-08-30 2000-12-28 Reduction of nonspecific hybridization by using novel base-pairing schemes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/115,566 Division US6232462B1 (en) 1994-08-30 1998-07-14 Reduction of nonspecific hybridization by using novel base-pairing schemes

Publications (1)

Publication Number Publication Date
US20010026918A1 true US20010026918A1 (en) 2001-10-04

Family

ID=23148897

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/298,073 Expired - Lifetime US5681702A (en) 1994-08-30 1994-08-30 Reduction of nonspecific hybridization by using novel base-pairing schemes
US08/794,153 Expired - Lifetime US5780610A (en) 1994-08-30 1997-02-03 Reduction of nonspecific hybridization by using novel base-pairing schemes
US09/115,566 Expired - Lifetime US6232462B1 (en) 1994-08-30 1998-07-14 Reduction of nonspecific hybridization by using novel base-pairing schemes
US09/752,213 Abandoned US20010026918A1 (en) 1994-08-30 2000-12-28 Reduction of nonspecific hybridization by using novel base-pairing schemes

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/298,073 Expired - Lifetime US5681702A (en) 1994-08-30 1994-08-30 Reduction of nonspecific hybridization by using novel base-pairing schemes
US08/794,153 Expired - Lifetime US5780610A (en) 1994-08-30 1997-02-03 Reduction of nonspecific hybridization by using novel base-pairing schemes
US09/115,566 Expired - Lifetime US6232462B1 (en) 1994-08-30 1998-07-14 Reduction of nonspecific hybridization by using novel base-pairing schemes

Country Status (21)

Country Link
US (4) US5681702A (en)
EP (2) EP0778898B1 (en)
JP (4) JP4461278B2 (en)
KR (1) KR100218113B1 (en)
CN (1) CN100335649C (en)
AT (2) ATE227778T1 (en)
AU (1) AU708194B2 (en)
BG (1) BG101246A (en)
BR (1) BR9508674A (en)
CA (1) CA2197901A1 (en)
CZ (1) CZ58997A3 (en)
DE (2) DE69528839T2 (en)
ES (2) ES2187571T3 (en)
FI (1) FI970803A (en)
HU (1) HUT77754A (en)
MX (1) MX9701419A (en)
NO (1) NO970884L (en)
NZ (1) NZ292451A (en)
PL (1) PL318933A1 (en)
SK (1) SK25497A3 (en)
WO (1) WO1996006950A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040132061A1 (en) * 2002-09-20 2004-07-08 Quinn John J. Dual-purpose primers and probes for providing enhanced hybridization assays by disruption of secondary structure formation
US20050221319A1 (en) * 2002-03-14 2005-10-06 Gnothis Holding Sa Use of capturing probes for identifying nucleic acids
US20120071357A1 (en) * 2006-04-24 2012-03-22 Jacob Kitzman Methods and systems for uniform enrichment of genomic regions
WO2017151680A3 (en) * 2016-02-29 2017-10-12 Dodo Omnidata, Inc. Methods, compositions, and devices for information storage
US10438662B2 (en) 2016-02-29 2019-10-08 Iridia, Inc. Methods, compositions, and devices for information storage
US10457980B2 (en) 2013-04-30 2019-10-29 California Institute Of Technology Multiplex labeling of molecules by sequential hybridization barcoding
US10510435B2 (en) 2013-04-30 2019-12-17 California Institute Of Technology Error correction of multiplex imaging analysis by sequential hybridization
US10640822B2 (en) 2016-02-29 2020-05-05 Iridia, Inc. Systems and methods for writing, reading, and controlling data stored in a polymer
US10859562B2 (en) 2016-02-29 2020-12-08 Iridia, Inc. Methods, compositions, and devices for information storage
US11214825B2 (en) 2016-07-05 2022-01-04 California Institute Of Technology Fractional initiator hybridization chain reaction
WO2022029159A1 (en) * 2020-08-04 2022-02-10 Theraxen S.A. Xeno-nucleotide based branched dna assay for the detection of nucleic acids
US11655465B1 (en) 2019-05-02 2023-05-23 Iridia, Inc. Enzymes and systems for synthesizing DNA
US11837302B1 (en) 2020-08-07 2023-12-05 Iridia, Inc. Systems and methods for writing and reading data stored in a polymer using nano-channels
US11873485B2 (en) 2021-01-26 2024-01-16 California Institute Of Technology Allosteric conditional guide RNAs for cell-selective regulation of CRISPR/Cas

Families Citing this family (281)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4140463A1 (en) * 1991-12-09 1993-06-17 Boehringer Mannheim Gmbh 2'-DESOXY-ISOGUANOSINE, THE ISOSTERAL ANALOGS AND THE APPLICATION THEREOF
CA2206451C (en) * 1994-12-09 2002-11-26 Wakunaga Seiyaku Kabushiki Kaisha Method for suppressing nonspecific hybridization in primer extension method
US5631146A (en) * 1995-01-19 1997-05-20 The General Hospital Corporation DNA aptamers and catalysts that bind adenosine or adenosine-5'-phosphates and methods for isolation thereof
WO1997018334A2 (en) 1995-11-15 1997-05-22 Gen-Probe Incorporated Nucleic acid probes complementary to human papillomavirus nucleic acid and related methods and kits
WO1998002580A2 (en) * 1996-07-12 1998-01-22 Tm Technologies, Inc. Signal amplification method
WO1998002582A2 (en) * 1996-07-16 1998-01-22 Gen-Probe Incorporated Methods for detecting and amplifying nucleic acid sequences using modified oligonucleotides having increased target specific t¿m?
US7070925B1 (en) 1996-07-16 2006-07-04 Gen-Probe Incorporated Method for determining the presence of an RNA analyte in a sample using a modified oligonucleotide probe
US6009480A (en) * 1997-09-12 1999-12-28 Telxon Corporation Integrated device driver wherein the peripheral downloads the device driver via an I/O device after it is determined that the I/O device has the resources to support the peripheral device
US20030165888A1 (en) * 2001-07-18 2003-09-04 Brown Bob D. Oligonucleotide probes and primers comprising universal bases for diagnostic purposes
US6518017B1 (en) * 1997-10-02 2003-02-11 Oasis Biosciences Incorporated Combinatorial antisense library
DE69840416D1 (en) * 1997-10-28 2009-02-12 Los Alamos Nat Security Llc Identification of DNA polymorphisms by flow cytometry
EP1038022B1 (en) * 1997-12-12 2005-07-20 Digene Corporation Assessment of human papilloma virus-related disease
US20070166741A1 (en) * 1998-12-14 2007-07-19 Somalogic, Incorporated Multiplexed analyses of test samples
US6242246B1 (en) * 1997-12-15 2001-06-05 Somalogic, Inc. Nucleic acid ligand diagnostic Biochip
US20060057573A1 (en) 2002-02-15 2006-03-16 Somalogic, Inc Methods and reagents for detecting target binding by nucleic acid ligands
US6686150B1 (en) 1998-01-27 2004-02-03 Clinical Micro Sensors, Inc. Amplification of nucleic acids with electronic detection
US7090804B2 (en) 1998-01-27 2006-08-15 Clinical Mirco Sensors, Inc. Amplification of nucleic acids with electronic detection
US6458559B1 (en) * 1998-04-22 2002-10-01 Cornell Research Foundation, Inc. Multivalent RNA aptamers and their expression in multicellular organisms
AU775179B2 (en) * 1999-03-15 2004-07-22 Applied Biosystems, Llc Probe/mobility modifier complexes for multiplex nucleic acid detection
EP1173614A4 (en) * 1999-04-08 2003-10-29 Oasis Biosciences Inc Antisense oligonucleotides comprising universal and/or degenerate bases
US8114850B2 (en) * 1999-04-08 2012-02-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US20080318889A1 (en) * 1999-04-08 2008-12-25 Antisoma Research Limited Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
ATE375358T1 (en) * 1999-04-08 2007-10-15 Antisoma Res Ltd ANTIPROLIFERATIVE ACTIVITY OF G-RICH OLIGONUCLEOTIDES AND METHOD FOR USING THEM TO BIND TO NUCLEOTIN
US20080318890A1 (en) * 1999-04-08 2008-12-25 Antisoma Research Limited Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US7960540B2 (en) * 1999-04-08 2011-06-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
AU4476900A (en) 1999-04-20 2000-11-02 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US8481268B2 (en) 1999-05-21 2013-07-09 Illumina, Inc. Use of microfluidic systems in the detection of target analytes using microsphere arrays
US8080380B2 (en) 1999-05-21 2011-12-20 Illumina, Inc. Use of microfluidic systems in the detection of target analytes using microsphere arrays
US20020045182A1 (en) * 1999-07-16 2002-04-18 Lynx Therapeutics, Inc. Multiplexed differential displacement for nucleic acid determinations
US7078167B2 (en) * 1999-09-17 2006-07-18 Agilent Technologies, Inc. Arrays having background features and methods for using the same
US7122303B2 (en) 1999-09-17 2006-10-17 Agilent Technologies, Inc. Arrays comprising background features that provide for a measure of a non-specific binding and methods for using the same
US6428957B1 (en) * 1999-11-08 2002-08-06 Agilent Technologies, Inc. Systems tools and methods of assaying biological materials using spatially-addressable arrays
US20020009728A1 (en) * 2000-01-18 2002-01-24 Quantum Dot Corporation Oligonucleotide-tagged semiconductor nanocrystals for microarray and fluorescence in situ hybridization
US6235483B1 (en) 2000-01-31 2001-05-22 Agilent Technologies, Inc. Methods and kits for indirect labeling of nucleic acids
US20020006617A1 (en) 2000-02-07 2002-01-17 Jian-Bing Fan Nucleic acid detection methods using universal priming
EP1967595A3 (en) 2000-02-16 2008-12-03 Illumina, Inc. Parallel genotyping of multiple patient samples
US6500622B2 (en) 2000-03-22 2002-12-31 Quantum Dot Corporation Methods of using semiconductor nanocrystals in bead-based nucleic acid assays
US7422850B2 (en) * 2000-05-19 2008-09-09 Eragen Biosciences, Inc. Materials and methods for detection of nucleic acids
JP2003535600A (en) * 2000-06-02 2003-12-02 バイエル コーポレイション Sensitive gene detection and localization using in situ branched DNA hybridization
JP2003535599A (en) 2000-06-06 2003-12-02 ティーエム バイオサイエンス コーポレイション Nucleic acid capture unit and its use
US7439016B1 (en) 2000-06-15 2008-10-21 Digene Corporation Detection of nucleic acids by type-specific hybrid capture method
US7601497B2 (en) 2000-06-15 2009-10-13 Qiagen Gaithersburg, Inc. Detection of nucleic acids by target-specific hybrid capture method
US6548251B1 (en) 2000-09-05 2003-04-15 Fidelity Systems, Inc. Inhibition of molecular and biological processes using modified oligonucleotides
US20020115058A1 (en) * 2000-09-22 2002-08-22 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of Pik3r1
US20020164576A1 (en) * 2000-09-22 2002-11-07 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of Nrf2
US20030044803A1 (en) * 2000-09-22 2003-03-06 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of JAK1
EP2351855A1 (en) * 2000-09-26 2011-08-03 Duke University RNA aptamers and methods for identifying the same
US20050059031A1 (en) 2000-10-06 2005-03-17 Quantum Dot Corporation Method for enhancing transport of semiconductor nanocrystals across biological membranes
ATE536550T1 (en) 2000-10-06 2011-12-15 Life Technologies Corp TRANSFECTABLE MICELLARS CONTAINING SEMICONDUCTOR NANOCRYSTALS
EP1325457A4 (en) * 2000-10-10 2007-10-24 Genencor Int Information rich libraries
JP4499987B2 (en) 2000-10-14 2010-07-14 エラジェン バイオサイエンシズ インコーポレイテッド Solid support assay systems and methods using non-standard bases
US7374882B2 (en) * 2000-11-28 2008-05-20 Riken Method for base sequencing and biologically active nucleic acids
US20030087252A1 (en) * 2000-12-22 2003-05-08 Morris David W. Novel compositions and methods in cancer associated with altered expression of PRDM11
US20030099963A1 (en) * 2000-12-22 2003-05-29 Morris David W. Novel compositions and methods in cancer associated with altered expression of TBX21
US20030165878A1 (en) * 2000-12-22 2003-09-04 Morris David W. Novel compositions and methods in cancer associated with altered expression of MCM3AP
US7892730B2 (en) * 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US20030232334A1 (en) * 2000-12-22 2003-12-18 Morris David W. Novel compositions and methods for cancer
US7820447B2 (en) 2000-12-22 2010-10-26 Sagres Discovery Inc. Compositions and methods for cancer
US7645441B2 (en) * 2000-12-22 2010-01-12 Sagres Discovery Inc. Compositions and methods in cancer associated with altered expression of PRLR
US7700274B2 (en) * 2000-12-22 2010-04-20 Sagres Discovery, Inc. Compositions and methods in cancer associated with altered expression of KCNJ9
GB0101397D0 (en) * 2001-01-19 2001-03-07 Amersham Pharm Biotech Uk Ltd Suppression of non-specific nucleic acid amplication
US20030092157A1 (en) * 2001-03-16 2003-05-15 Hayden Michael R. Compositions, screening systems and methods for modulating HDL cholesterol and triglyceride levels
US20030191073A1 (en) 2001-11-07 2003-10-09 Challita-Eid Pia M. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
AU2002312059B2 (en) 2001-05-25 2009-01-15 Duke University Modulators of pharmacological agents
AU2002305019A1 (en) * 2001-05-25 2002-12-09 University Of British Columbia Diagnostic methods for cardiovascular disease, low hdl-cholesterol levels, and high triglyceride levels
CA2348042A1 (en) * 2001-06-04 2002-12-04 Ann Huletsky Sequences for detection and identification of methicillin-resistant staphylococcus aureus
AU2002327236A1 (en) 2001-07-12 2003-01-29 Illumina, Inc. Multiplex nucleic acid reactions
US20030129626A1 (en) 2001-09-24 2003-07-10 Nielsen Kirsten Vang Methods, kits and compositions pertaining to the suppression of detectable probe binding to randomly distributed repeat sequences in genomic nucleic acid
US20070098728A1 (en) * 2001-09-24 2007-05-03 Pedersen Finn S Novel compositions and methods in cancer
US7070933B2 (en) * 2001-09-28 2006-07-04 Gen-Probe Incorporated Inversion probes
US20040166490A1 (en) * 2002-12-17 2004-08-26 Morris David W. Novel therapeutic targets in cancer
US20040126762A1 (en) * 2002-12-17 2004-07-01 Morris David W. Novel compositions and methods in cancer
WO2003040395A2 (en) * 2001-11-07 2003-05-15 Applera Corporation Universal nucleotides for nucleic acid analysis
US20040197778A1 (en) * 2002-12-26 2004-10-07 Sagres Discovery, Inc. Novel compositions and methods in cancer
US20040180344A1 (en) * 2003-03-14 2004-09-16 Morris David W. Novel therapeutic targets in cancer
US20060040262A1 (en) * 2002-12-27 2006-02-23 Morris David W Novel compositions and methods in cancer
CA2470774A1 (en) * 2001-12-19 2003-06-26 Hvidovre Hospital A method and a kit for determination of a microbial count
US20070184474A1 (en) * 2002-01-10 2007-08-09 Nichirei Corporation Process for amplifying DNA
US20030194705A1 (en) * 2002-03-01 2003-10-16 Schroth Gary P. Methods of using unnatural nucleobases for decoding
EP1487343B1 (en) 2002-03-05 2008-12-31 Board of Regents, The University of Texas System Biospecific contrast agents
CA2479730A1 (en) * 2002-03-21 2003-10-02 Sagres Discovery, Inc. Novel compositions and methods in cancer
US20040009506A1 (en) * 2002-03-29 2004-01-15 Genentech, Inc. Methods and compositions for detection and quantitation of nucleic acid analytes
US7094537B2 (en) * 2002-04-30 2006-08-22 Agilent Technologies, Inc. Micro arrays with structured and unstructured probes
EP1534857B1 (en) 2002-06-20 2010-08-04 The Regents of The University of California Methods and compositions for detection and analysis of polynucleotides using light harvesting multichromophores
US7144950B2 (en) 2003-09-17 2006-12-05 The Regents Of The University Of California Conformationally flexible cationic conjugated polymers
WO2004001065A2 (en) * 2002-06-24 2003-12-31 Cornell Research Foundation, Inc. Exhaustive selection or rna aptamers against complex targets
WO2004016733A2 (en) 2002-08-16 2004-02-26 Agensys, Inc. Nucleic acid and corresponding protein entitled 251p5g2 useful in treatment and detection of cancer
EA007653B1 (en) 2002-08-26 2006-12-29 Дзе Риджентс Оф Дзе Юниверсити Оф Калифорния Methods and compositions for detection and analysis of polynucleotides using light harvesting multichromophores
US20040157238A1 (en) * 2002-09-20 2004-08-12 Quinn John J. Method for detection of multiple nucleic acid sequence variations
DE602004018270D1 (en) * 2003-01-17 2009-01-22 Eragen Biosciences Inc NUCLEIC ACID AMPLIFICATION WITH NON-STANDARD BASES
WO2004072263A2 (en) 2003-02-10 2004-08-26 Agensys, Inc. Nucleic acid and corresponding protein named 158p1d7 useful in the treatment and detection of bladder and other cancers
EP2351856A1 (en) 2003-02-13 2011-08-03 The Regents Of the University of California Methods and compositions for detection and analysis of polynucleotide-binding protein interactions using light harvesting multichromophores
US20070218071A1 (en) * 2003-09-15 2007-09-20 Morris David W Novel therapeutic targets in cancer
US7767387B2 (en) * 2003-06-13 2010-08-03 Sagres Discovery, Inc. Therapeutic targets in cancer
US20040170982A1 (en) 2003-02-14 2004-09-02 Morris David W. Novel therapeutic targets in cancer
JP2007524362A (en) 2003-02-14 2007-08-30 サイグレス ディスカバリー, インコーポレイテッド Therapeutic GPCR targets in cancer
US7820808B2 (en) 2003-04-01 2010-10-26 Eragen Biosciences, Inc. Polymerase inhibitor and method of using same
US20040241661A1 (en) * 2003-05-29 2004-12-02 Arindam Bhattacharjee Pseudo single color method for array assays
NZ578591A (en) 2003-05-30 2011-01-28 Agensys Inc Prostate stem cell antigen (PSCA) variants and subsequences thereof, namely SEQ ID NO: 6554
WO2005001129A2 (en) * 2003-06-06 2005-01-06 Applera Corporation Mobility cassettes
US20040259100A1 (en) 2003-06-20 2004-12-23 Illumina, Inc. Methods and compositions for whole genome amplification and genotyping
DE20310332U1 (en) * 2003-07-04 2004-11-11 Mwg-Biotech Ag Device for automatic opening and closing of reaction vessels
EP1939779A3 (en) 2003-08-01 2009-04-01 Dna Twopointo Inc. Systems and methods for biopolymer engineering
US20080050357A1 (en) 2003-08-01 2008-02-28 Claes Gustafsson Systems and Methods for Antibody Engineering
US20090163375A1 (en) 2003-09-09 2009-06-25 Bowman Christopher N Use of Photopolymerization for Amplification and Detection of a Molecular Recognition Event
US7354706B2 (en) * 2003-09-09 2008-04-08 The Regents Of The University Of Colorado, A Body Corporate Use of photopolymerization for amplification and detection of a molecular recognition event
US20070281896A1 (en) * 2003-09-30 2007-12-06 Morris David W Novel compositions and methods in cancer
US20050136414A1 (en) * 2003-12-23 2005-06-23 Kevin Gunderson Methods and compositions for making locus-specific arrays
WO2005081776A2 (en) * 2004-01-30 2005-09-09 Eragen Biosciences, Inc. Materials and methods for the detection of sars
US7803931B2 (en) 2004-02-12 2010-09-28 Archemix Corp. Aptamer therapeutics useful in the treatment of complement-related disorders
EP1727567B1 (en) * 2004-02-12 2013-07-03 Archemix LLC Aptamer therapeutics useful in the treatment of complement-related disorders
US20050266432A1 (en) * 2004-02-26 2005-12-01 Illumina, Inc. Haplotype markers for diagnosing susceptibility to immunological conditions
WO2005106035A2 (en) * 2004-04-09 2005-11-10 Cornell Research Foundation, Inc. Modular design and construction of nucleic acid molecules, aptamer-derived nucleic acid constructs, rna scaffolds, their expression, and methods of use
PT1745062E (en) * 2004-04-22 2014-07-11 Regado Biosciences Inc Improved modulators of coagulation factors
US20090317873A1 (en) 2004-05-04 2009-12-24 Sridhar Govindarajan Design, synthesis and assembly of synthetic nucleic acids
EP2327798A1 (en) 2004-05-14 2011-06-01 Rosetta Genomics Ltd MicroRNAs and uses thereof
EP1784501B1 (en) 2004-05-14 2015-11-18 Rosetta Genomics Ltd VIRAL AND VIRUS ASSOCIATED MicroRNAS AND USES THEREOF
WO2006076027A2 (en) * 2004-05-17 2006-07-20 Cambrios Technology Corp. Biofabrication of transistors including field effect transistors
US7473551B2 (en) 2004-05-21 2009-01-06 Atonomics A/S Nano-mechanic microsensors and methods for detecting target analytes
NZ551627A (en) 2004-05-28 2010-02-26 Agensys Inc Antibodies and related molecules that bind to PSCA proteins
US7827558B2 (en) * 2004-06-30 2010-11-02 Devicevm, Inc. Mechanism for enabling a program to be executed while the execution of an operating system is suspended
KR100663992B1 (en) 2004-07-05 2007-01-02 (주)바이오메드랩 The method selecting highly specific probes for HPV genotype analysis and the probes thereof
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
EP1838752B1 (en) 2005-01-10 2017-10-04 The Regents of The University of California Cationic conjugated polymers suitable for strand-specific polynucleotide detection in homogeneous and solid state assays
US8063196B2 (en) * 2005-02-01 2011-11-22 Siemens Healthcare Diagnostics Inc. Highly orthogonal universal sequences for use in nucleic acid assays
EP1853722B1 (en) * 2005-03-03 2010-02-24 National Research Council of Canada Methods and compositions for the detection and analysis of nucleic acids by signal amplification
US7498136B2 (en) * 2005-03-18 2009-03-03 Eragen Biosciences, Inc. Methods for detecting multiple species and subspecies of Neisseria
EP2444099A1 (en) 2005-03-31 2012-04-25 Agensys, Inc. Antibodies and related molecules that bind to 161P2F10B proteins
US20090214536A1 (en) 2005-04-07 2009-08-27 Guoying Yu CACNA1E in Cancer Diagnosis, Detection and Treatment
EP1871911A2 (en) 2005-04-07 2008-01-02 Chiron Corporation Cancer-related genes (prlr)
US8628918B2 (en) * 2005-05-09 2014-01-14 Affymetrix, Inc. Multiplex capture of nucleic acids
US8632970B2 (en) 2005-05-09 2014-01-21 Affymetrix, Inc. Multiplex capture of nucleic acids
US7803541B2 (en) 2005-05-12 2010-09-28 Panomics, Inc. Multiplex branched-chain DNA assays
CA3027954C (en) 2005-06-07 2021-11-02 Luminex Corporation Methods for detection and typing of nucleic acids
US20070087360A1 (en) * 2005-06-20 2007-04-19 Boyd Victoria L Methods and compositions for detecting nucleotides
CN104673903B (en) 2005-06-20 2018-11-13 领先细胞医疗诊断有限公司 The method for detecting the nucleic acid in individual cells and identifying rare cells in heterogeneous maxicell group
EP1910575B1 (en) * 2005-07-01 2014-12-10 Dako Denmark A/S Immunohistochemistry detection method
WO2007044427A2 (en) * 2005-10-05 2007-04-19 Panomics, Inc. Detection of nucleic acids from whole blood
US7838221B2 (en) * 2005-10-11 2010-11-23 Geneohm Sciences, Inc. Sequences for detection and identification of methicillin-resistant Staphylococcus aureus (MRSA)
US11834720B2 (en) 2005-10-11 2023-12-05 Geneohm Sciences, Inc. Sequences for detection and identification of methicillin-resistant Staphylococcus aureus (MRSA) of MREJ types xi to xx
EP1951905B1 (en) * 2005-11-10 2012-04-11 Affymetrix, Inc. Detection of nucleic acids through amplification of surrogate nucleic acids
TWI507528B (en) * 2006-01-17 2015-11-11 Somalogic Inc Multiplexed analyses of test samples
US20070264694A1 (en) * 2006-04-07 2007-11-15 Eragen Biosciences, Inc. Use of non-standard bases and proximity effects for gene assembly and conversion of non-standard bases to standard bases during dna synthesis
US7833716B2 (en) 2006-06-06 2010-11-16 Gen-Probe Incorporated Tagged oligonucleotides and their use in nucleic acid amplification methods
EP2038432B1 (en) 2006-06-30 2017-02-08 Rosetta Genomics Ltd Method for detecting and quantifying a target nucleic acid generated by rt-pcr of mirna
JP2008043332A (en) 2006-08-17 2008-02-28 Panomics Inc Quantitative determination of nucleic acid from tissue slide
US8481506B2 (en) * 2006-12-05 2013-07-09 Rosetta Genomics, Ltd. Nucleic acids involved in viral infection
ES2426289T3 (en) 2006-12-19 2013-10-22 Becton Dickinson Infusion Therapy Systems Inc. Staphylococcus aureus detection and identification of methicillin-resistant Staphylococcus aureus
US7855054B2 (en) * 2007-01-16 2010-12-21 Somalogic, Inc. Multiplexed analyses of test samples
US20110136099A1 (en) * 2007-01-16 2011-06-09 Somalogic, Inc. Multiplexed Analyses of Test Samples
US8975026B2 (en) 2007-01-16 2015-03-10 Somalogic, Inc. Method for generating aptamers with improved off-rates
US7964356B2 (en) * 2007-01-16 2011-06-21 Somalogic, Inc. Method for generating aptamers with improved off-rates
US7947447B2 (en) 2007-01-16 2011-05-24 Somalogic, Inc. Method for generating aptamers with improved off-rates
US20080241140A1 (en) 2007-02-12 2008-10-02 Medical College Of Georgia Gene amplification of coactivator coaa and uses thereof
US8273746B2 (en) 2007-03-23 2012-09-25 The Board Of Regents Of The University Of Texas System Methods involving aldose reductase inhibitors
US8114681B2 (en) * 2007-10-05 2012-02-14 Affymetrix, Inc. Highly multiplexed particle-based assays
US8906700B2 (en) 2007-11-06 2014-12-09 Ambergen, Inc. Methods and compositions for phototransfer
US20090137405A1 (en) * 2007-11-16 2009-05-28 Christopher Bowman Detection of nucleic acid biomarkers using polymerization-based amplification
US20090131351A1 (en) * 2007-11-16 2009-05-21 Antisoma Research Limited Methods, compositions, and kits for modulating tumor cell proliferation
EP2268664B1 (en) 2007-12-03 2017-05-24 The Government of the United States of America as represented by the Secretary of the Department of Health and Human Services Doc1 compositions and methods for treating cancer
ES2916498T3 (en) 2007-12-06 2022-07-01 Genalyte Inc Method for identifying a nucleotide sequence in an unknown species of nucleic acid; and device for carrying out the method
US20090215050A1 (en) * 2008-02-22 2009-08-27 Robert Delmar Jenison Systems and methods for point-of-care amplification and detection of polynucleotides
US9249455B2 (en) * 2008-04-18 2016-02-02 Luminex Corporation Methods for detection and quantification of small RNA
WO2009142214A1 (en) * 2008-05-19 2009-11-26 株式会社セリッシュエフディー Rna in situ hybridization
US20090298709A1 (en) * 2008-05-28 2009-12-03 Affymetrix, Inc. Assays for determining telomere length and repeated sequence copy number
US8703416B2 (en) 2008-07-17 2014-04-22 Somalogic, Inc. Method for purification and identification of sperm cells
WO2010062556A1 (en) 2008-10-27 2010-06-03 Qiagen Gaithersburg Inc. Fast results hybrid capture assay and system
EP2347247B1 (en) 2008-10-27 2019-06-26 Genalyte, Inc. Biosensors based on optical probing and sensing
EP2367959B1 (en) * 2008-12-18 2016-12-07 Siemens Healthcare Diagnostics Inc. Methods for shortening incubation times in hybridization assays
ES2644516T3 (en) * 2009-01-28 2017-11-29 Qiagen Gaithersburg, Inc. Method and test for sample preparation of large sequence specific volume
CN102388150A (en) 2009-02-19 2012-03-21 吉恩奥姆科技公司 Methods for the detection and identification of extended spectrum beta lactamases
EP3021120A1 (en) 2009-02-20 2016-05-18 Michael P. Lisanti Diagnosis, prognosis, therapeutics and methods for treating neoplastic deiseases comprising determining the level of caveolin-1 in a stromal cell sample
JP5419138B2 (en) * 2009-03-27 2014-02-19 Necソフト株式会社 Detection method of detection object and detection kit used therefor
US20120010091A1 (en) 2009-03-30 2012-01-12 Illumina, Inc. Gene expression analysis in single cells
CN102414327B (en) 2009-05-01 2015-04-08 奇亚根盖瑟斯堡股份有限公司 A non-target amplification method for detection of RNA splice-forms in a sample
US20100291706A1 (en) 2009-05-15 2010-11-18 Millipore Corporation Dye conjugates and methods of use
WO2011032124A1 (en) 2009-09-14 2011-03-17 Qiagen Gaithersburg, Inc. Compositions and methods for recovery of nucleic acids or proteins from tissue samples fixed in cytology media
WO2011038403A1 (en) 2009-09-28 2011-03-31 Yuling Luo Methods of detecting nucleic acid sequences with high specificity
JP5938348B2 (en) * 2009-10-23 2016-06-22 ルミネックス コーポレーション Amplification primers containing non-standard bases for increased reaction specificity
WO2011094528A2 (en) 2010-01-29 2011-08-04 Qiagen Gaithersburg, Inc. Method of determining and confirming the presence of an hpv in a sample
JP6103941B2 (en) * 2010-01-29 2017-03-29 キアジェン ゲイサーズバーグ インコーポレイテッド Methods and compositions for sequence-specific purification and multiplex analysis of nucleic acids
US8951940B2 (en) 2010-04-01 2015-02-10 Illumina, Inc. Solid-phase clonal amplification and related methods
EP2567213B1 (en) 2010-05-05 2018-01-24 The Governing Council of the Universtiy of Toronto Method of processing dried samples using digital microfluidic device
WO2011146629A2 (en) 2010-05-19 2011-11-24 Qiagen Gaithersburg Inc. Methods and compositions for sequence-specific purification and multiplex analysis of nucleic acids
US20120003648A1 (en) * 2010-07-01 2012-01-05 Affymetrix, Inc. Signal Multiplexing and Signal Amplification
EP2601307A4 (en) 2010-08-06 2014-01-01 Capitalbio Corp Microarray-based assay integrated with particles for analyzing molecular interactions
EP2612146A2 (en) * 2010-09-02 2013-07-10 Heed Diagnostics ApS Electrochemical detection of analyte
ES2648564T3 (en) 2010-10-21 2018-01-04 Advanced Cell Diagnostics, Inc. Ultrasensitive method for in situ detection of nucleic acids
JP5872569B2 (en) 2010-10-27 2016-03-01 キャピタルバイオ コーポレーションCapitalBio Corporation Molecules labeled with luminophores coupled to particles for microarray-based assays
CA3060724A1 (en) 2010-11-05 2012-05-10 Genalyte, Inc. Optical analyte detection systems and methods of use
WO2012106081A2 (en) 2011-01-31 2012-08-09 Illumina, Inc. Methods for reducing nucleic acid damage
EP2481812A1 (en) 2011-01-31 2012-08-01 Westfälische Wilhelms-Universität Münster Molecular sexing of avian subjects
US9885092B2 (en) 2011-02-24 2018-02-06 Qiagen Gaithersburg Inc. Materials and methods for detection of HPV nucleic acids
WO2012129363A2 (en) 2011-03-24 2012-09-27 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
WO2013067202A1 (en) 2011-11-04 2013-05-10 Handylab, Inc. Polynucleotide sample preparation device
US9340567B2 (en) 2011-11-04 2016-05-17 Ohmx Corporation Chemistry used in biosensors
SG11201402402SA (en) * 2011-11-18 2014-09-26 Tagcyx Biotechnologies Nucleic acid fragment binding to target protein
EP2783016A1 (en) 2011-11-22 2014-10-01 Intermune, Inc. Methods of diagnosing and treating idiopathic pulmonary fibrosis
US10085987B2 (en) 2012-01-27 2018-10-02 Thomas Jefferson University MCT protein inhibitor-related prognostic and therapeutic methods
EP2825885B1 (en) 2012-03-12 2021-05-12 The Board of Trustees of the University of Illinois Optical analyte detection systems with magnetic enhancement
WO2014018093A1 (en) 2012-07-26 2014-01-30 Illumina, Inc. Compositions and methods for the amplification of nucleic acids
US9783841B2 (en) 2012-10-04 2017-10-10 The Board Of Trustees Of The Leland Stanford Junior University Detection of target nucleic acids in a cellular sample
US20140322706A1 (en) 2012-10-24 2014-10-30 Jon Faiz Kayyem Integrated multipelx target analysis
EP2912432B1 (en) 2012-10-24 2018-07-04 Genmark Diagnostics Inc. Integrated multiplex target analysis
WO2014074785A1 (en) 2012-11-08 2014-05-15 Ludwig Institute For Cancer Research Ltd. Methods of predicting outcome and treating breast cancer
US20140274811A1 (en) * 2013-03-14 2014-09-18 Lyle J. Arnold Methods for Amplifying a Complete Genome or Transcriptome
US9273349B2 (en) 2013-03-14 2016-03-01 Affymetrix, Inc. Detection of nucleic acids
US20140272959A1 (en) * 2013-03-14 2014-09-18 President And Fellows Of Harvard College Methods of Hybridizing Probes to Genomic DNA
EP3520895A1 (en) 2013-03-15 2019-08-07 Genmark Diagnostics Inc. Fluid container with cantilevered lance
WO2014143637A1 (en) 2013-03-15 2014-09-18 The Board Of Trustees Of The University Of Illinois Methods and compositions for enhancing immunoassays
WO2014172627A1 (en) 2013-04-19 2014-10-23 Thomas Jefferson University Caveolin-1 related methods for treating glioblastoma with temozolomide
USD881409S1 (en) 2013-10-24 2020-04-14 Genmark Diagnostics, Inc. Biochip cartridge
US9498778B2 (en) 2014-11-11 2016-11-22 Genmark Diagnostics, Inc. Instrument for processing cartridge for performing assays in a closed sample preparation and reaction system
DK3129487T3 (en) 2014-04-09 2020-11-30 Dna Twopointo Inc IMPROVED NUCLEIC ACID CONSTRUCTIONS FOR EUKARYOT GENEPRESSION
EP4324473A2 (en) 2014-11-10 2024-02-21 ModernaTX, Inc. Multiparametric nucleic acid optimization
EP3218108B1 (en) 2014-11-11 2020-09-02 Genmark Diagnostics Inc. Fluid sample processing cartridge and use thereof
US9598722B2 (en) 2014-11-11 2017-03-21 Genmark Diagnostics, Inc. Cartridge for performing assays in a closed sample preparation and reaction system
US10005080B2 (en) 2014-11-11 2018-06-26 Genmark Diagnostics, Inc. Instrument and cartridge for performing assays in a closed sample preparation and reaction system employing electrowetting fluid manipulation
US10464067B2 (en) 2015-06-05 2019-11-05 Miroculus Inc. Air-matrix digital microfluidics apparatuses and methods for limiting evaporation and surface fouling
CN208562324U (en) 2015-06-05 2019-03-01 米罗库鲁斯公司 Digital microcurrent-controlled (DMF) device of air matrix
JP2018527947A (en) 2015-07-17 2018-09-27 プレジデント アンド フェローズ オブ ハーバード カレッジ Nucleic acid sequence amplification method
ES2893100T3 (en) 2015-10-12 2022-02-08 Advanced Cell Diagnostics Inc In situ detection of nucleotide variants in samples with a high level of noise, and related compositions and methods
PT3394093T (en) 2015-12-23 2022-05-30 Modernatx Inc Methods of using ox40 ligand encoding polynucleotides
MA43587A (en) 2016-01-10 2018-11-14 Modernatx Inc THERAPEUTIC RNA CODING FOR ANTI-CTLA-4 ANTIBODIES
WO2017180917A2 (en) 2016-04-13 2017-10-19 Modernatx, Inc. Lipid compositions and their uses for intratumoral polynucleotide delivery
SG11201810256XA (en) 2016-05-18 2018-12-28 Modernatx Inc Polynucleotides encoding relaxin
LT3458474T (en) 2016-05-18 2022-10-10 Modernatx, Inc. Combinations of mrnas encoding immune modulating polypeptides and uses thereof
EP3458092A1 (en) 2016-05-18 2019-03-27 Modernatx, Inc. Mrna combination therapy for the treatment of cancer
EP3463388A4 (en) 2016-05-24 2020-01-01 Emory University Particles with rna cleaving nucleobase polymers and uses for managing inflammatory disorders
EP3500660A4 (en) 2016-08-22 2020-03-04 Miroculus Inc. Feedback system for parallel droplet control in a digital microfluidic device
EP3516401A1 (en) 2016-09-19 2019-07-31 Genmark Diagnostics Inc. Instrument for processing cartridge for performing assays in a closed sample preparation and reaction system
WO2018098363A2 (en) 2016-11-23 2018-05-31 Bioverativ Therapeutics Inc. Bispecific antibodies binding to coagulation factor ix and coagulation factor x
WO2018126082A1 (en) 2016-12-28 2018-07-05 Miroculis Inc. Digital microfluidic devices and methods
US11623219B2 (en) 2017-04-04 2023-04-11 Miroculus Inc. Digital microfluidics apparatuses and methods for manipulating and processing encapsulated droplets
CA3063723A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
EP3658908A4 (en) 2017-07-24 2021-04-07 Miroculus Inc. Digital microfluidics systems and methods with integrated plasma collection device
EP3676009A4 (en) 2017-09-01 2021-06-16 Miroculus Inc. Digital microfluidics devices and methods of using them
MA50802A (en) 2017-11-22 2020-09-30 Modernatx Inc POLYNUCLEOTIDES CODING FOR ALPHA AND BETA SUB-UNITS OF PROPIONYL-COA CARBOXYLASE FOR THE TREATMENT OF PROPIONIC ACIDEMIA
CA3079428A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
MA50801A (en) 2017-11-22 2020-09-30 Modernatx Inc POLYNUCLEOTIDES CODING FOR PHENYLALANINE HYDROXYLASE FOR THE TREATMENT OF PHENYLKETONURIS
CN110577981B (en) * 2018-06-07 2023-04-07 北京福安华生物科技有限公司 Molecular beacon based on artificial simulation nucleic acid
US20220184185A1 (en) 2018-07-25 2022-06-16 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
MA53609A (en) 2018-09-13 2021-07-21 Modernatx Inc POLYNUCLEOTIDES ENCODED GLUCOSE-6-PHOSPHATASE FOR THE TREATMENT OF GLYCOGENOSIS
US20220243182A1 (en) 2018-09-13 2022-08-04 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
AU2019339430A1 (en) 2018-09-14 2021-04-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide A1 for the treatment of Crigler-Najjar Syndrome
US20220152225A1 (en) 2018-09-27 2022-05-19 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
JP2022511596A (en) 2018-11-14 2022-02-01 ウロバント サイエンシズ ゲーエムベーハー Compositions and Methods for the Treatment of Smooth Muscle Dysfunction
CN112639126A (en) 2018-12-14 2021-04-09 伊卢米纳剑桥有限公司 Reducing phasing with unlabeled nucleotides during sequencing
SG11202012758WA (en) 2018-12-17 2021-01-28 Illumina Cambridge Ltd Primer oligonucleotide for sequencing
AU2019411266A1 (en) 2018-12-17 2021-01-07 Illumina Cambridge Limited Compositions for use in polyunucleotide sequencing
CN111378746A (en) * 2018-12-27 2020-07-07 北京福安华生物科技有限公司 Artificial mimic nucleic acid molecular beacon and kit for detecting rs12415607 site polymorphism of CASP7 gene
CN114206499A (en) 2019-04-08 2022-03-18 米罗库鲁斯公司 Multi-cartridge digital microfluidic devices and methods of use
JP2022532078A (en) 2019-05-08 2022-07-13 アストラゼネカ アクチボラグ Compositions for skin and wounds and methods of their use
US11524298B2 (en) 2019-07-25 2022-12-13 Miroculus Inc. Digital microfluidics devices and methods of use thereof
EP3859012A1 (en) 2020-02-03 2021-08-04 Albert-Ludwigs-Universität Freiburg Methods for amplification of genomic dna and preparation of sequencing libraries
EP4153189A1 (en) * 2020-05-22 2023-03-29 Merck Sharp & Dohme LLC Synthesis of fluorinated nucleotides
JP2023527875A (en) 2020-06-01 2023-06-30 モダーナティエックス・インコーポレイテッド Phenylalanine hydroxylase variants and uses thereof
US20230285310A1 (en) 2020-08-06 2023-09-14 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
JP2024512026A (en) 2021-03-24 2024-03-18 モデルナティエックス インコーポレイテッド Lipid nanoparticles and polynucleotide encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
CA3213037A1 (en) 2021-03-31 2022-10-06 Colin Brown Blocking oligonucleotides for the selective depletion of non-desirable fragments from amplified libraries
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
TW202320737A (en) 2021-07-26 2023-06-01 美商現代公司 Processes for preparing lipid nanoparticle compositions for the delivery of payload molecules to airway epithelium
WO2023009421A1 (en) 2021-07-26 2023-02-02 Modernatx, Inc. Processes for preparing lipid nanoparticle compositions
US20230096386A1 (en) 2021-09-30 2023-03-30 Illumina Cambridge Limited Polynucleotide sequencing
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
TW202332467A (en) 2021-10-29 2023-08-16 美商現代公司 Lipid amines
WO2023076605A1 (en) 2021-10-29 2023-05-04 Modernatx, Inc. Lipid amines
WO2023086465A1 (en) 2021-11-12 2023-05-19 Modernatx, Inc. Compositions for the delivery of payload molecules to airway epithelium
AU2022424380A1 (en) 2021-12-29 2024-01-18 Illumina, Inc. Methods of nucleic acid sequencing using surface-bound primers
US11772093B2 (en) 2022-01-12 2023-10-03 Miroculus Inc. Methods of mechanical microfluidic manipulation
TW202345863A (en) 2022-02-09 2023-12-01 美商現代公司 Mucosal administration methods and formulations
WO2023196399A1 (en) 2022-04-06 2023-10-12 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding argininosuccinate lyase for the treatment of argininosuccinic aciduria
CN115060912B (en) * 2022-06-23 2024-02-02 南京浦光生物科技有限公司 Reagent combination, kit, detection system and detection method for detecting target antibody

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1190838A (en) * 1981-07-17 1985-07-23 Cavit Akin Homogeneous nucleic acid hybridization diagnostics by non-radiative energy transfer
US4868105A (en) * 1985-12-11 1989-09-19 Chiron Corporation Solution phase nucleic acid sandwich assay
US5124246A (en) * 1987-10-15 1992-06-23 Chiron Corporation Nucleic acid multimers and amplified nucleic acid hybridization assays using same
US5270163A (en) * 1990-06-11 1993-12-14 University Research Corporation Methods for identifying nucleic acid ligands
US5135917A (en) * 1990-07-12 1992-08-04 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
CA2104698A1 (en) * 1991-02-21 1992-08-22 John J. Toole Aptamers specific for biomolecules and methods of making
DE4140463A1 (en) * 1991-12-09 1993-06-17 Boehringer Mannheim Gmbh 2'-DESOXY-ISOGUANOSINE, THE ISOSTERAL ANALOGS AND THE APPLICATION THEREOF
US5681697A (en) * 1993-12-08 1997-10-28 Chiron Corporation Solution phase nucleic acid sandwich assays having reduced background noise and kits therefor

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050221319A1 (en) * 2002-03-14 2005-10-06 Gnothis Holding Sa Use of capturing probes for identifying nucleic acids
US8394944B2 (en) 2002-09-20 2013-03-12 Siemens Healthcare Diagnostics Inc. Dual-purpose primers and probes for providing enhanced hybridization assays by disruption of secondary structure formation
US20040132061A1 (en) * 2002-09-20 2004-07-08 Quinn John J. Dual-purpose primers and probes for providing enhanced hybridization assays by disruption of secondary structure formation
US20120071357A1 (en) * 2006-04-24 2012-03-22 Jacob Kitzman Methods and systems for uniform enrichment of genomic regions
US10457980B2 (en) 2013-04-30 2019-10-29 California Institute Of Technology Multiplex labeling of molecules by sequential hybridization barcoding
US10510435B2 (en) 2013-04-30 2019-12-17 California Institute Of Technology Error correction of multiplex imaging analysis by sequential hybridization
US10640822B2 (en) 2016-02-29 2020-05-05 Iridia, Inc. Systems and methods for writing, reading, and controlling data stored in a polymer
US10438662B2 (en) 2016-02-29 2019-10-08 Iridia, Inc. Methods, compositions, and devices for information storage
WO2017151680A3 (en) * 2016-02-29 2017-10-12 Dodo Omnidata, Inc. Methods, compositions, and devices for information storage
US10714178B2 (en) 2016-02-29 2020-07-14 Iridia, Inc. Methods, compositions, and devices for information storage
US10859562B2 (en) 2016-02-29 2020-12-08 Iridia, Inc. Methods, compositions, and devices for information storage
US10995373B2 (en) 2016-02-29 2021-05-04 Iridia, Inc. Systems and methods for writing, reading, and controlling data stored in a polymer
US11505825B2 (en) 2016-02-29 2022-11-22 Iridia, Inc. Methods of synthesizing DNA
US11549140B2 (en) 2016-02-29 2023-01-10 Iridia, Inc. Systems and methods for writing, reading, and controlling data stored in a polymer
US11214825B2 (en) 2016-07-05 2022-01-04 California Institute Of Technology Fractional initiator hybridization chain reaction
US11655465B1 (en) 2019-05-02 2023-05-23 Iridia, Inc. Enzymes and systems for synthesizing DNA
WO2022029159A1 (en) * 2020-08-04 2022-02-10 Theraxen S.A. Xeno-nucleotide based branched dna assay for the detection of nucleic acids
US11837302B1 (en) 2020-08-07 2023-12-05 Iridia, Inc. Systems and methods for writing and reading data stored in a polymer using nano-channels
US11873485B2 (en) 2021-01-26 2024-01-16 California Institute Of Technology Allosteric conditional guide RNAs for cell-selective regulation of CRISPR/Cas

Also Published As

Publication number Publication date
BG101246A (en) 1998-03-31
KR970705644A (en) 1997-10-09
CN100335649C (en) 2007-09-05
DE69528839D1 (en) 2002-12-19
NO970884L (en) 1997-04-28
JPH10506270A (en) 1998-06-23
AU708194B2 (en) 1999-07-29
CZ58997A3 (en) 1997-07-16
ATE259374T1 (en) 2004-02-15
DE69532565T2 (en) 2004-12-16
NO970884D0 (en) 1997-02-27
ES2215797T3 (en) 2004-10-16
JP2008048741A (en) 2008-03-06
CA2197901A1 (en) 1996-03-07
BR9508674A (en) 1997-11-18
ES2187571T3 (en) 2003-06-16
DE69528839T2 (en) 2003-03-27
AU3463195A (en) 1996-03-22
US5681702A (en) 1997-10-28
SK25497A3 (en) 1997-12-10
FI970803A (en) 1997-04-23
US5780610A (en) 1998-07-14
KR100218113B1 (en) 1999-10-01
MX9701419A (en) 1997-05-31
DE69532565D1 (en) 2004-03-18
JP2008043341A (en) 2008-02-28
EP1097939B1 (en) 2004-02-11
EP1097939A3 (en) 2001-05-30
JP2006217925A (en) 2006-08-24
EP0778898B1 (en) 2002-11-13
WO1996006950A1 (en) 1996-03-07
JP4461278B2 (en) 2010-05-12
CN1164260A (en) 1997-11-05
US6232462B1 (en) 2001-05-15
EP1097939A2 (en) 2001-05-09
HUT77754A (en) 1998-07-28
PL318933A1 (en) 1997-07-21
FI970803A0 (en) 1997-02-26
EP0778898A1 (en) 1997-06-18
NZ292451A (en) 1999-09-29
ATE227778T1 (en) 2002-11-15

Similar Documents

Publication Publication Date Title
US5780610A (en) Reduction of nonspecific hybridization by using novel base-pairing schemes
WO1996006950A9 (en) Reduction of nonspecific hybridization by using novel base-pairing schemes
EP0731848B1 (en) Solution phase nucleic acid sandwich assays having reduced background noise
EP1009852A2 (en) Oligonucleotide probes bearing quenchable fluorescent labels, and methods of use thereof
JPH06128284A (en) Synthetic catalyst oligonucleotide structure, method of cleaving target sequence of nucleic acid, antiviral therapeutic medicine, its preparation, diagnostic agent and its preparation
WO2012102616A1 (en) Heterodirectional polynucleotides
WO1993012135A1 (en) Nuclease stable and binding competent oligomers and methods for their use
Efimov et al. Convenient approaches to the synthesis of oligonucleotide macrocycles containing non-nucleotide linkers
WO2001032924A1 (en) Compositions and methods of synthesis and use of novel nucleic acid structures

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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