WO1991009073A1 - Uncharged morpholino-based polymers having achiral intersubunit linkages - Google Patents

Uncharged morpholino-based polymers having achiral intersubunit linkages Download PDF

Info

Publication number
WO1991009073A1
WO1991009073A1 PCT/US1990/007565 US9007565W WO9109073A1 WO 1991009073 A1 WO1991009073 A1 WO 1991009073A1 US 9007565 W US9007565 W US 9007565W WO 9109073 A1 WO9109073 A1 WO 9109073A1
Authority
WO
WIPO (PCT)
Prior art keywords
subunit
polymer
morpholino
base
composition
Prior art date
Application number
PCT/US1990/007565
Other languages
French (fr)
Inventor
James E. Summerton
Dwight D. Weller
Original Assignee
Anti-Gene Development Group
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 Anti-Gene Development Group filed Critical Anti-Gene Development Group
Priority to DE69032167T priority Critical patent/DE69032167T2/en
Priority to EP91902401A priority patent/EP0506845B1/en
Priority to KR1019920701480A priority patent/KR0154317B1/en
Priority to JP50289391A priority patent/JP3172174B2/en
Priority to AU71587/91A priority patent/AU654473B2/en
Publication of WO1991009073A1 publication Critical patent/WO1991009073A1/en

Links

Classifications

    • 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
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G75/00Macromolecular compounds obtained by reactions forming a linkage containing sulfur with or without nitrogen, oxygen, or carbon in the main chain of the macromolecule
    • C08G75/18Polysulfoxides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/548Phosphates or phosphonates, e.g. bone-seeking
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/001Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
    • C07K14/003Peptide-nucleic acids (PNAs)
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G75/00Macromolecular compounds obtained by reactions forming a linkage containing sulfur with or without nitrogen, oxygen, or carbon in the main chain of the macromolecule
    • C08G75/20Polysulfones
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08GMACROMOLECULAR COMPOUNDS OBTAINED OTHERWISE THAN BY REACTIONS ONLY INVOLVING UNSATURATED CARBON-TO-CARBON BONDS
    • C08G75/00Macromolecular compounds obtained by reactions forming a linkage containing sulfur with or without nitrogen, oxygen, or carbon in the main chain of the macromolecule
    • C08G75/24Polysulfonates
    • 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
    • C12Q1/6839Triple helix formation or other higher order conformations in hybridisation assays
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J2219/00Chemical, physical or physico-chemical processes in general; Their relevant apparatus
    • B01J2219/00274Sequential or parallel reactions; Apparatus and devices for combinatorial chemistry or for making arrays; Chemical library technology
    • B01J2219/00718Type of compounds synthesised
    • B01J2219/0072Organic compounds

Definitions

  • the present invention relates to morpholino-based polymers.
  • Polymers which are designed for base-specific bind ⁇ ing to polynucleotides have significant potential both for in vitro detection of specific genetic sequences characteristic of pathogens (Ler an) and for in vivo inactivation of genetic sequences causing many diseases—particularly viral diseases (Belikova, Summerton) .
  • Standard ribo- and deoxyribonucleotide polymers have been widely used both for detection of complementary genetic sequences, and more recently, for inactivating targeted genetic sequences.
  • standard polynucle- otides suffer from a number of limitations when used for base-specific binding to target oligonucleotides. These limitations include (i) restricted passage across biolo ⁇ gical membranes, (ii) nuclease sensitivity, (iii) target binding which is sensitive to ionic concentration, and (iv) susceptibility to cellular strand-separating mecha ⁇ nisms.
  • polymers cannot be used for base-specific binding to selected target sequence ⁇ olynucleotide analogs containing uncharged, but stereoisomeric, methylphospho- nate linkages between the deoxyribonucleoside subunits have been reported (Miller, 1979, 1980; Jayaraman; Murakami; Blake, 1985a, 1985b; Smith) . More recently a variety of analogous uncharged phosphoramidate-linked oligonucleotide analogs have also been reported (Froehler, 1988) . These polymers comprise deoxynucleo- sides linked by the 3'OH group of one subunit and the 5'OH group of another subunit via an uncharged chiral phosphorous-containing group.
  • deoxyribonucleotide analogs having un- charged and achiral intersubunit linkages have been constructed (Stirchak 1987) . Since these polymers are stereoregular, all polymers having a given subunit se ⁇ quence will have the same Tm value for a given target nucleotide sequence, thus avoiding some of the limita- tions inherent in chirally-linked polymers. These un ⁇ charged, achiral deoxyribonucleoside-derived analogs, however, are limited by relatively high cost of starting materials.
  • the invention includes a polymer composition con ⁇ taining morpholino ring structures of the form:
  • the ring structures are linked together by uncharged, achiral linkages, one to three atoms long, joining the morpholino nitrogen of one ring structure to the 5' exocyclic carbon of an adjacent ring structure.
  • Each ring structure includes a purine or pyrimidine base-pairing moiety P i which is effective to bind by base-specific hydrogen bonding to a base in a target sequence in a polynucleotide.
  • Figure 1 shows a basic ⁇ -morpholino ring structure which is linked through uncharged, achiral linkages to form the polymer of the present invention.
  • P is a purine or pyrimidine base pairing moiety.
  • Figure 3 shows several preferred subunits having 5- atom (A) , six-atom (B and C) and seven-atom (D-G) linkin groups suitable for forming polymers.
  • Y 0 or S.
  • X 1 O or S
  • X 2 O, S, CH 2 , or NR
  • X 3 O, S, CH 2 , or NR 2 .
  • X A 0, S, or NR,.
  • X 5 0, S, CH 2 , NR 2 , or S0 2 .
  • When n 1, X 5 is CH 2 or S0 2 .
  • FIG. 1 shows a repeating subunit segment of exemplary morpholino-based polymers, designated A-A through G-G, constructed using subunits A-G, respectively, of Figure 3.
  • Y, X,, X 2 , X 3 , X 4 , X 5 , n, R and R 2 are as in Figure 3.
  • Figure 5 shows the steps in the synthesis of several types of morpholino subunits from a ribonucleoside.
  • Figure 6 shows an alternative synthesis of the basic morpholino subunit.
  • Figure 7 shows the steps in the synthesis of a mor- pholino subunit designed for construction of polymers with seven-atom repeating-unit backbones.
  • Figure 8 shows the binding mode for 2-amine- containing purines to polar major-groove sites of respective target base-pairs (Figure 8a) and a representative base sequence of a duplex-binding polymer (Figure 8b) .
  • a Adenine
  • c cytosine
  • g guanine
  • t thymine
  • u uracil
  • D 2,6-Diaminopurine or 2-aminopurine
  • G Guanine or thioguanine
  • high specificity hydrogen bonding
  • and : low specificity hydrogen bonding.
  • Figure 9 shows the steps in linking two morpholino subunits through a carbamate linkage.
  • Figure 10 shows the activation of sulfamic acid and coupling to form a sulfamide linkage.
  • Figure 11 shows the activation of sulfonic acid and coupling to form a sulfonamide linkage.
  • Figure 12 shows the steps in linking two morpholino subunits through a sulfamate linkage.
  • Figure 13 shows the steps in linking two morpholino subunits through an amide linkage.
  • Figure 14 shows a subunit coupling procedure which simultaneously generates the morpholino ring structure.
  • Figure 15 shows thermal denaturation plots for poly(dC)/poly(dG) and. poly(C morpholino)/poly(dG) duplexes where the poly(C morpholino) was constructed according to the present invention.
  • Figure 16 illustrates a diagnostic solid-support particle employing polymers of the present invention for use in a probe-diagnostic assay.
  • the present invention includes a morpholino- based polymer which is designed for base-specific binding to a target sequence of a polynucleotide.
  • the polymer is composed of morpholino-based ring structures which are linked together by uncharged, achiral linkages, one to three atoms long, joining the morpholino nitrogen of one structure to the 5' exocyclic carbon of an adjacent structure.
  • Figure 1 shows the ⁇ -morpholino ring structures on which the polymer subunits are based, where the morpho ⁇ lino carbon atoms are numbered as in the parent ribose.
  • the ring structure contains a 5' methylene attached to the A ' carbon in the ⁇ -orientation.
  • Each ring structure includes a purine or pyrimidine or related hydrogen-bonding moiety, P A , attached to the backbone morpholine moiety through a linkage in the ⁇ - orientation.
  • the purine hydrogen-bonding moieties or bases in ⁇ clude purines as well as purine-like planar ring struc ⁇ tures having a 5-6 fused ring in which one or more of the atoms, such as N3, N7, or N9 is replaced by a suitable atom, such as carbon.
  • the pyrimidine moieties likewise include pyrimidines as well as pyrimidine-like planar 6- membered rings in which one or more of the atoms, such as NI, is replaced by a suitable atom, such as carbon.
  • Preferred hydrogen-bonding moieties in the invention include the set of purines and pyrimidines shown in Figure 2.
  • Each base includes at least two hydrogen- bonding sites specific for a polynucleotide base or base- pair.
  • the purine 8 structures 1-3 are designed to bind to thymine or uracil bases; structures 7-8, to guanine bases; structures 4-6, to cytosine bases; and structure 9, to adenine bases.
  • the polymers of the invention are also effective to bind to hydrogen-bonding sites accessible through the major-groove in duplex polynucleotides having mostly purine bases in one strand and mostly pyrimidine bases in the complementary strand, as discussed below.
  • the H-bonding moiety of a duplex-binding polymer must hydrogen-bond to the N7 of its target base-pair in order to uniquely recognize a given base-pair in a target genetic duplex.
  • the hydrogen-bonding moieties of the polymer preferably contain purines having an amine at the 2 position since that amine is suitably positioned for H- bonding to the N7 of the target base-pair.
  • Structures 2 and 3 of Figure 2 provide for specific binding to a TA or UA base-pair, and Structures 4 and 6 provide for specific binding to a CG base-pair.
  • Two bases which are parti ⁇ cularly useful in a duplex-binding polymer are 2,6-diami- nopurine (Structure 3) and guanine (Structure 4) .
  • Figure 8 illustrates the binding of these two bases to the polar major-groove sites of their respective target base-pairs in duplex nucleic acids.
  • the morpholino subunits of the instant invention are combined to form polymers by linking the subunits through stable, achiral, uncharged linkages.
  • the linking group of a subunit usually includes a carbonyl or sulfonyl electrophile for reaction with a nucleophile of the subunit to which it is to be linked.
  • duplex DNA and RNA duplexes are constructed according to parameters deter- mined by x-ray diffraction of oligodeoxyribonucleotides in the B-form and oligoribonucleotide-containing duplexes in the A-form.
  • each of these constructed duplexes one of the two sugar phosphate backbones is removed, and the pro- spective backbone, including the morpholino ring and intersubunit linkage, is replaced, if possible, on the sites of the bases from which the original sugar-phos ⁇ phate backbone has been removed.
  • Each resulting poly- nucleotide/polymer duplex is then examined for coplana- rity of the Watson/Crick base pairs, torsional and angle strain in the prospective binding polymer backbone, degree of distortion imposed on the nucleic acid strand, and interstrand and intrastrand nonbonded interactions.
  • the linkages in the five-atom, six-atom, and seven-atom repeating-unit backbone contribute one, two, and three atoms to the backbone length, respectively.
  • the linkage between the ring structures is (a) uncharged, (b) achi- ral, (c) stable, and (d) must permit adoption of a con ⁇ formation suitable for binding to the target polynucleo ⁇ tide.
  • Figure 3 shows several preferred ⁇ -morpholino sub- unit types, including linkage groups, which meet the constraints and requirements outlined above. It will be appreciated that a polymer may contain more than one linkage type.
  • Subunit A in Figure 3 contains a 1-atom sulfonyl linkage which forms the five atom repeating-unit backbone shown at A-A in Figure 4, where the morpholino rings are linked by a 1-atom sulfonamide linkage. It is noted here that the corresponding amide linkage (substituting a carbonyl for sulfonyl linkage) is not acceptable due to lack of rotational freedom about the carbon-nitrogen 11 tertiary amide bond.
  • Subunits B and C in Figure 3 are designed for 6-atom repeating-unit backbones, as shown at B-B and C-C, re ⁇ spectively, in Figure 4.
  • the atom X linking the 5' morpholino carbon to the carbonyl group may be oxygen or sulfur, but not nitrogen or carbon, due to lack of free rotation about the resultant intersubunit linkage.
  • the nitrogen may be secondary (NH) , or tertiary (NR) , where R is a methyl or other group which does not interfere with polymer binding to the target polynucleotide (as can be easily determined from molecular modeling studies such as those outlined above) .
  • Subunits D-G in Figure 3 are designed for 7-atom repeating-unit backbones, as shown at D-D through G-G, respectively, in Figure 4.
  • the X can be a secondary nitrogen (NH) , or a tertiary nitrogen (NR) where R is a is a methyl or other group which does not interfere with polymer binding to the target polynucleo ⁇ tide, as can be determined from molecular modeling stu- dies.
  • X in Structure E can be an oxygen since the 5'' methylene in such morpholino structures is surprisingly resistant to nucleophilic attack.
  • morpholino-subunits are generally ribon- ucleosides.
  • ribonucleosides containing hydro ⁇ gen-bonding moieties or bases are transformed to their morpholino derivatives to provide a complete set of subunits for polymer synthesis.
  • a 1-haloribose or, preferably, a l ⁇ -bromoglucose derivative can be linked to a suitable base and this nucleoside analog then converted to the desired ⁇ -morpholino structure via peri- odate cleavage, and closing the resultant dialdehyde on a suitable amine.
  • Example 1 Methods for base protecting a number of the more common ribonucleosides are given in Example 1.
  • the methods detailed in the example are generally applicable for forming nucleosides with amine-protective groups.
  • Standard base- protective groups used for nucleic acid chemistry are often suitable including the following groups: benzoyl for the N4 of cytosine (C) ; benzoyl or p-nitrobenzoyl for the N6 of adenine (A) ; acetyl, phenyl- acetyl or isobutyryl for the N2 of guanine (G) ; and N2,N6-bisisobutyryl for 2, 6-diaminopurine residues.
  • These protective groups can be removed after polymer assembly by treatment with ammonium hydroxide.
  • Suitable base protective groups removable by a strong non-nucleophilic base via a ⁇ -elimination mechanism in ⁇ clude: 2-(4-nitrophenyl)ethoxy carbonyl or 2- (phenyl sulfonyl)ethoxycarbonyl for both the N4 of C and the N6 of A; and the 9-fluorenyl methoxycarbonyl for the N2 of G and the N2 and N6 of 2, 6-diaminopurine.
  • These groups can be removed after polymer assembly by treatment with the strong nonnucleophilic base 1,8-diazabicyclo[5.4.0]- undec-7-ene (DBU) , under stringently anhydrous condi ⁇ tions.
  • a base-protected ribonucleoside is reacted with sodium periodate to form a transient 2', 3'-dialdehyde which then closes upon ammonia to form a morpholino-ring having 2' and 3' hydroxyl groups (numbered as in the parent ribose, see Figure 1) .
  • the compound is then treated with sodium cyanoborohydride to reduce the ring hydroxyl groups.
  • the ring nitrogen is preferably protected by trityl derivatization or by a benzhydraloxycarbonyl group for subsequent subunit coupling.
  • the protective group can be added by reacting the morpholino subunit with trityl chloride or with nitrophenyl benzhydryl carbonate or by reacting the dialdehyde with a primary amine, as illustrated in Figure 6 and described in Example 3.
  • the stereochemistry of the nucleoside starting material is retained as long as the pH of the reaction mixture at the iminium stage is not allowed to go above about 10.
  • a variety of nitrogen sources can be used — including ammonia, ammo ⁇ nium hydroxide, ammonium carbonate, and ammonium bicar ⁇ bonate. Best results are obtained when the reaction solution is maintained near neutrality during the oxida ⁇ tion and morpholino ring closure reactions. This can be accomplished by continually titrating the reaction mix or, more conveniently, by using ammonium biborate as the ammonia source. When the solution is too acidic the yield of product is low and when it is too basic, side products (possibly due to epimerization of the 1' and/or 4' carbons) are produced which are difficult to separate from the desired product. It is also noted that the reducing agent can be added before, during, or after the 15 oxidation step with little noticeable effect on product yield.
  • Ribonucleosides lacking base protection groups can also be successfully oxidized, ring closed, and reduced in aqueous solution to generate the morpholino ring.
  • base protection the number and quantity of undesired side products frequently increases, par ⁇ ticularly in the case of cytidine.
  • the subunits formed by the above methods contain a 5'-OH, SH, or amine which is modified, reacted with, and/or activated, to be suitable for coupling to a second morpholino subunit (see below) .
  • Figure 5 shows the conversion of a 5'-OH of a morpholino subunit to a sulfonyl linking moiety to form a subunit (Structure 10) which is linked to form a 5-atom unit-length backbone polymer. Details of the subunit synthesis are given in Example 6.
  • the subunits are designed to include a sulfonyl or carbonyl group attached directly or in- directly to the morpholino ring nitrogen, which is cou ⁇ pled to a 5' moiety of a second morpholino subunit (Fi ⁇ gures 12 and 13) .
  • Subunits of this type are suitable for constructing morpholino polymers with 6-atom ( Figure 12) or 7-atom ( Figure 13) repeating-unit backbones.
  • An example of the synthesis of a subunit suitable for 7-atom unit-length backbones having an amine at the 5'-carbon atom, and a sulfonyl group linked to the ring nitrogen through a methylene group is detailed in Example 9 (with reference to Figure 7) .
  • Example 11 A similar synthesis, described in Example 10, is used to prepare morpholino subunits having a 5'-linked primary amine and an acetyl group linked to the ring nitrogen. This subunit is formed by coupling glycine, rather than AMSA, to the 5' ribonucleoside aldehyde 16 group. Examples 11 and 12 describe, with reference to Structure G of Figure 3, the preparation of non-morpho- lino subunits which are converted into morpholino struc ⁇ tures during polymer assembly.
  • the subunits prepared as above are coupled, in a controlled, sequential manner, generally by activating the carbonyl or sulfonyl group on one subunit (having protected nitrogen groups) and contacting this activated subunit with another subunit having an unprotected nitro ⁇ gen.
  • Different types of linkages such as those illust ⁇ rated below, may be employed in the construction of a single polymer.
  • a number of closely related variations are possible for the carbonyl-containing linkages giving six-atom backbones, corresponding to Structure B-B in Figure 4.
  • a typical activation and coupling reaction for forming a carbamate linkage (where X is 0 in Structure B-B) is illustrated in Figure 9.
  • the simplest and most obvious morpholino-type bind ⁇ ing polymers are the carbamate-linked polymers (type B-B of Figure 4) where X is oxygen.
  • the polymer has been found to effectively bind to its single-stranded DNA target sequence.
  • the polymer exhibited unusual binding, as evi ⁇ denced by a highly atypical hypochromicity profile in the 320 to 230 nm spectral range and lack of a normal thermal denaturation.
  • the sulfonyl group is activated with phosgene, and coupled to a second subunit having an unprotected ring nitrogen, as shown.
  • the polymer assembly is con ⁇ tinued by deprotecting the morpholino ring nitrogen of the dimer, and reacting the dimer with a third activated subunit.
  • the sulfamide linkage (corresponding to the linkage in structure C-C in Figure 4, where X is an amine) , is formed by sulfating the 5'-linked amine in a subunit having a protected morpholino ring nitrogen, and then activating with phosgene and reacting this subunit with a second subunit having an unprotected ring nitrogen, as illustrated in Figure 10. Details of the coupling reac ⁇ tion are given in Example 14.
  • the sulfamate linkage (corresponding to the linkage in Structure C-C in Figure 4, wherein X is 0) is produced by sulfating the morpholino ring nitrogen of a 5' protec ⁇ ted subunit, then using phosgene to generate the sulf- amoyl chloride. This activated subunit is then mixed 19 with another subunit or oligomer having a free 5'OH. Coupling of the subunits is achieved either with a cata ⁇ lyst such as silver trifluoromethanesulfonate or use of a strong base to convert the 5' hydroxyl to the anionic form. Conversion of the 5' hydroxyl to the alkoxy can be achieved by KOH and a suitable phase transfer catalyst. This sulfamate coupling is illustrated in Figure 12 and details are given in Example 15.
  • a number of 7-atom unit length backbones prepared from the morpholino-subunits allow even more flexibility in the construction of polymers which have specified distances between the base-pairing moieties.
  • distances between the mor- pholino-subunits, and consequently between the base pairing moieties can be lengthened.
  • Such lengthening of the intersubunit linkage is particularly useful when targeting duplex genetic sequences in a B conformation.
  • the 7-atom backbone polymers can be readily syn- thesized from the subunits D-F constructed as above, employing the general coupling reactions described above.
  • Structure D-D in Figure 4 can be produced by
  • Structure E-E in Figure 4 can be produced by activating the sulfonyl group with phosgene, and coup ⁇ ling the activated subunit with a second subunit having an unprotected 5'-linked amine.
  • Structure F-F in Figure 4 can be produced by a similar synthetic method in which the carboxyl group is activated with carbonyldiimidazole or a carbodiimide, and the activated compound is reacted with a second subunit 20 having an unprotected 5'-linked primary amine.
  • a novel class of linkages corresponding to Structure G-G of Figure 4 can be produced by oxidizing vicinyl hydroxyls of one ribonucleoside subunit and closing the resultant dialdehyde on a primary amine of another sub- unit followed by reduction with cyanoborohydride.
  • this same scheme could also be used to couple a secondary amine of one subunit and a mono-aldehyde of a second subunit; however, the coupling of a ribose-derived dialdehyde to a primary amine proceeds substantially faster and provides a. better yield.
  • Examples 11 and 12 describe the synthesis of ribonucleosides containing a primary amine at the 5' . Their use in formation of mor ⁇ pholino polymers is illustrated in Figure 14.
  • Example 18 outlines such a block assembly synthesis using monomers to form dimers, and dimers to form tetra ⁇ mers. The synthesis need not involve oligomers of equal size. A particular merit of this block assembly method is that each coupling product is roughly twice the length of its precursors, so purification of the product of each coupling is simplified. Example 18 details the assembly of a 4-subunit polymer formed by this method.
  • the polymers may also be synthesized by stepwise subunit addition on a solid support.
  • the op ⁇ timal synthetic approach often uses a combination of the solution and solid support assembly methods where dimers, trimers, or tetramers are synthesized by solution phase and subsequently assembled into the full-length polymer on a solid support, as described in Example 19.
  • a solid support such as glass beads derivatized with acid-stable, long-chain cleavable lin- kers, are employed as the support material, and prepared for attachment of the first subunit, or block of sub- units, as described in Example 19.
  • the glass beads are reacted with a subunit which generally has a readily cleavable protective group on a nitrogen. Whether the morpholino subunit is linked to the support via its morpholino nitrogen or a group at the 5' position depends on the direction of polymer synthesis, i.e., to which group the next subunit will be attached.
  • any un- reacted nucleophilic sites can be capped by addition of a suitable capping reagent, such as p-nitrophenyl acetate or acetic anhydride, and thereafter the support is washed.
  • a suitable capping reagent such as p-nitrophenyl acetate or acetic anhydride
  • the protecting group on the nitrogen of the terminal subunit is removed, typically by acid treatment, and after neutralization, the support is reacted with an excess of the next-in- sequence subunit (or polymer unit) which is activated by one of the methods outlined above.
  • One feature of the solid support assembly method is the need for high coupling efficiencies at each subunit addition step. This high coupling efficiency is general ⁇ ly achieved by addition of an excess of the activated subunit which maximizes the number of support-bound chains which are chain-elongated. 22
  • Chain elongation is continued in this manner, with optional capping of failure sequences after each subunit addition, until the polymer of the desired length and sequence is achieved.
  • the terminal backbone moiety may be reacted with a suitable charged or uncharged group, as described in Example 19.
  • the polymer is then cleaved from the support, e.g., by treatment with either ammonium hydroxide or a non-nucleophilic base suitable for effecting ⁇ -elimination in the linker join ⁇ ing the polymer to the. support.
  • the bases are deprotec ⁇ ted and the polymer is purified as described below and in Example 19.
  • Binding polymers assembled in solution are typically base-deprotected by suspending in DMSO or DMF and layering on the suspension an equal volume of concentrated ammonium hydroxide. The prepara- tion is mixed with shaking and incubated at 30°C for 16 hrs. Workup includes removing the ammonia under reduced pressure. If a protective group (generally trityl or a related acid-labile moiety) is present, this group is cleaved and the crude polymer preparation is suspended in the appropriate buffer for purification (Example 19) .
  • a protective group generally trityl or a related acid-labile moiety
  • Binding polymers assembled by a solid-phase method (Example 19) wherein they are linked to the support via an ester linkage can be cleaved from the support by suspending the dried support in DMSO, layering on an equal volume of concentrated NH 4 OH, capping tightly, and slowly agitating for 16 hrs at 30°C. The solid support material is removed by filtration and the filtrate is treated as described above.
  • binding polymers linked to the sup- 23 port via a ⁇ -elimination-sensitive linker can be cleaved from the support using a strong non-nucleophilic base 1,8 diazabicyclo(5.4.0.)undec-7-ene (DBU) in DMF.
  • DBU diazabicyclo(5.4.0.)undec-7-ene
  • Purification of the base-deprotected polymer is preferably carried out at pH 2.5 or pH 11, depending on the pKs of the base moieties in the polymer.
  • pH 2.5 cytosine, adenine, and 2-6-diaminopurine moieties carry a positive charge and guanine carries a partial positive charge.
  • pH 11 guanine, uracil and hypoxanthine carry a negative charge.
  • the purification can be carried out by cation ex ⁇ change on a column of S-Sepharose fast-flow (Pharmacia) developed with a shallow NaCl gradient buffered at pH 2.5.
  • the effluent is monitored at 254 nm and collected in a fraction collector.
  • the full length polymer which elutes after the shorter failure sequences, can be fur ⁇ ther purified and desalted on a column of chromatogra ⁇ phic grade polypropylene (Polysciences Inc.), eluted with an aqueous gradient of acetonitrile adjusted to pH 2.5 with formic acid, with the eluant being monitored at 254 nm.
  • the fractions containing the pure product are neutralized and dried under reduced pressure. Salts may be discarded by dissolving the polymer in trifluoroe- thanol, filtering, and evaporating the trifluoroethanol.
  • the purification may be performed on an anion exchange column of Q Sepha- rose fast-flow (Pharmacia) developed with an aqueous pH 11 gradient of NaCl.
  • the full-length polymer which 24 elutes after shorter failure sequences, is further puri ⁇ fied and desalted on a polypropylene column eluted with an aqueous pH 11 gradient of acetonitrile. Fractions containing the pure product are processed as above.
  • the purification methods described above should be carried out so that polymers containing adenine base-pairing moieties are not exposed to pH 11 for more than a few hours at room temperature, to avoid potential base lability problems.
  • the details of the purification methods are outlined in Example 19.
  • polystyrene resin In neutral, aqueous solution, longer morpholino polymers may have solubilities only in the sub-micromolar range. Therefore, it may be advantageous to enhance polymer solubility by addition of one or more hydrophilic moieties, e.g., polyethylene glycol. For most of the polymer types disclosed herein, this can be accomplished by cleaving the terminal backbone protective group from the completed polymer, and reacting the polymer, with the bases still in the protected state, with excess of car- bonyldiimidazole-activated polyethylene glycol (PEG) . Thereafter the binding polymer is treated with ammonium hydroxide to remove the base-protected groups, and the polymer is purified as above.
  • PEG car- bonyldiimidazole-activated polyethylene glycol
  • solubiliza- tion is easily adjusted through proper selection of the PEG material.
  • PEG fractions having average molecular weights of 200, 400, 600, 1,000, 1,540, 3,400, 4,000, 6,000, 7,500, and 18,500 daltons are commercially available (e.g., Polysciences, Inc.) with PEG1000 often providing the best solubilization.
  • the solubilizing moiety may be linked to the polymer through a cleavable linkage, if desired, to allow the polymer to be released from the solubilizing agent, e.g., by esterase or pep- tidase enzymes. 25
  • the polymer may be further derivatized or labeled according to known proce ⁇ dures.
  • the polymer may be radiolabeled by preparing the polymer subunits from radiolabeled ribonu- cleosides or by attaching a radiolabeled amino acid at one terminus.
  • the polymer may be readily derivatized, e.g., employing modifications of the above subunit cou ⁇ pling reactions, with enzymes, chromophoric groups, or the like, where the polymer is to be used as a diagnostic probe.
  • the polymer may be derivatized with biomolecules which serve to target the polymers to speci ⁇ fic tissues or cell types.
  • F. Structural Characterization Fully-protected binding polymers of moderate size (10 to 20 subunits) often give a strong molecular ion in FAB (Fast Atom Bombardment) mass spectroscopy, providing a key confirmation of the polymer length.
  • FAB Fluor Atom Bombardment
  • COSY-NMR two-dimensional correlated spec- troscopy
  • Mobilities on ion exchange columns also provide information on the number of C + A base-pairing moieties in a polymer when purification is carried out at pH 2.5 and information on the number of G + U residues when the purification is run at pH 11. Structural verification is easiest when the polymers have been assembled from oligo- mer blocks, such as in Examples 18, 19 and 20, since any failure sequences then differ more substantially from the full-length sequences. 26
  • the UV profiles of the polymers at pH 1, 7, and 13 can provide information about the relative nucleotide composition of the polymer.
  • comparisons can be made between the melting curve of a regular nucleic acid duplex (such as p(dC) 6 /p(dG) 6 ) and the melting curve of a hybrid duplex containing a corresponding morpholino-based polymer (such as (morpholino-based C) 6 /p(dG) 6 ).
  • Characterization of the synthetic intermediates and the full-length oligomer was achieved by proton NMR and negative ion FAB mass spec- troscopy. With these carbamates of the morpholino oligo ⁇ mers, the fragmentation of the oligomers is greatly suppressed so that little sequence information is avail ⁇ able.
  • the parent ion signal is quite strong and allows confirmation of the composition of the morpholino oligomer (see Example 20) .
  • High resolution mass spectro- metry of the morpholino-based poly C hexamer provided a satisfactory elemental analysis.
  • the length of the oligomers could be ascertained by comparing the integration of the various signals. For example, in a dimer the two 2' protons were separated by 0.5 ppm and gave a 1.02/1 ratio of integrations and for the hexamer, this ratio was 4.65/1 against the expected value of 5/1.
  • the bases of the above hexamer were deprotected by treatment with concentrated ammonia for 24 hours.
  • the 4'- terminal morpholino ring nitrogen was liberated by treat ⁇ ment of the crude oligomer with 1% formic acid in tri- fluoroethanol.
  • the hexamer was taken up in pH 2.5 buffer and puri ⁇ fied by cation exchange chromatography on S-Sepharose Fast FlowTM, eluting with potassium chloride gradients.
  • the chromatograms showed one major peak comprising over 95% of the cytosine- containing materials in the mixture, and confirming that little or no cleavage of the oligomer occurs in the deprotection of the bases and the morpho- lino-amine.
  • After neutralization the hexamer was desalted on a polypropylene column eluted with a water-acetonit- rile gradient.
  • the purified hexamer was analyzed by ** H NMR.
  • the assignment for the protons was made on the basis of a COSY plot.
  • One cytosine base has signals that were found downfield relative to the other bases (8.04 to 7.65 and 6.78 to 5.85 ppm).
  • the relative integrations of these peaks confirmed that the hexamer was deprotected and has been purified intact.
  • the 5' protons were assigned to the signal at 4.35-4.15 downfield of the 1' proton signal at 4.14-3.90 ppm.
  • the tailed hexamer was found to be freely soluble in pH 7.5 buffer in concentrations up to 2 mM. The characterization of the tailed hexamer by the 1 H NMR methods employed above was not possible. In the spectrum of the tailed hexamer there was no differentia ⁇ tion between the signals of the base protons, thus pre ⁇ cluding the assessment of the oligomer length. Addition- ally, the envelope containing the PEG tail signals ob ⁇ scured the majority of the signals of the morpholino rings. However, the ion exchange chromatography of the tailed hexamer gave one major peak indicating little or no cleavage of the oligomer during deprotection. The pattern of the chromatogram of the tailed hexamer was the same as found for the free hexamer, except that the tailed hexamer elutes faster than the free hexamer.
  • the stability of complexes of the tailed hexamer with complementary nucleic acids was investigated by thermal denaturation experiments. Difference spectra between mixed and unmixed samples of the tailed hexamer and the selected phosphodiester complement were obtained from 14°C to 85°C and over the 320 to 260 nm range (see Example 20). As a control, the duplex of p)dC) 6 with 29 p(dG) 6 was thermally denatured.
  • the difference UV spectrum of the tailed hexamer (morphC) 6 with p(dG) 6 was similar to that of the control DNA duplex, p(dC) 6 with p(dG) 6 , except that the amount of hypochromicity before denaturation of the (morphC) 6 /p(dG) 6 duplex was much greater than that of the control.
  • the thermal denatura ⁇ tion of the p(morphC) 6 /p(dG) 6 duplex gave a T m value of 62.5°C (see Example 20 and Figure 15).
  • the corresponding DNA/DNA duplex gave a Tm value of 26.5°C.
  • the target-specific polymers of the invention can be used in a variety of diagnostic assays for detection of RNA or DNA having a given target sequence.
  • the polymers are labeled with a suitable radiolabel or other detectable reporter group.
  • Target polynucleotide typically a single stranded polynucleo ⁇ tide which is bound to a solid support, is reacted with the polymer under hybridization conditions, allowed to anneal, and then the sample is examined for the presence of polymer reporter group.
  • the diagnostic assay can be carried out according to standard procedures, with suitable adjustment of the hybridization conditions to allow polymer hybridization with the target region.
  • the polymer can be designed for hybridization with the target at a higher melting temperature than the comple ⁇ mentary polynucleotide strand, since polymer binding does not entail backbone charge repulsion effects. Therefore, the polymer can bind to the target at a temperature above ⁇ the normal polynucleotide melting temperature, an impor ⁇ tant advantage of the polymer over conventional oligo ⁇ nucleotide probes. This binding at elevated temperature minimizes the problem of competition for binding to the 30 target between the probe and any corresponding single- strand oligonucleotide which may be present in the diag ⁇ nostic mixture.
  • the polymers are linked to a solid support, for capture of target RNA or DNA to the support.
  • the solid support e.g., polymeric microparticles
  • the solid support can be prepared by link ⁇ ing the polymers to the support according to the methods described above or by conventional derivatization proce- dures.
  • this support may also serve as the assay support.
  • the target polynucleotide molecules which are captured on the support by base-specific binding to the polymers can be detected on the basis of their backbone charge, since the support-bound polymers are themselves substantially uncharged.
  • the assay system may also include polycationic reporter molecules which are designed to bind to the fully charged analyte back ⁇ bone, but not the uncharged (or substantially uncharged) polymer backbone, under selected binding conditions.
  • the reporter molecules are com ⁇ posed of a polycationic moiety or tail designed to bind electrostatically to a fully charged polynucleotide, under conditions where the reporter does not bind to the less charged or uncharged binding polymer carried on the diagnostic reagent; one or more reporter groups may be attached to the tail, adapted to produce a signal by which the presence of the reporter can be detected.
  • Methods for forming polycationic molecules and for at ⁇ taching reporter molecules to cationic compounds are known in the art. 31
  • Each reporter molecule carries one or more reporter groups, and each polynucleotide can accommodate binding of typically several thousand or more reporter molecules.
  • the system has an amplification factor, in terms of reporter signal per bound analyte molecule, of several orders of magnitude.
  • the method has the advantage, noted above, that the polynucleotide binding reaction can be carried out under conditions in which binding competition with complementary nucleotide strands does not occur.
  • the design considerations applied in preparing a polynucleotide binding polymer for use as a diagnostic reagent are governed by the nature of the target analyte and the reaction conditions under which the analyte is to be assayed.
  • a first consideration there is selected a non-homopolymeric target base sequence against which the polymer is directed. This target sequence is gener ⁇ ally single-stranded and preferably unique to the ana ⁇ lyte being assayed.
  • the probability of occurrence of a given n-base target sequence is approximately (1/4) n . Accordingly, a given n-base target sequence would be expected to occur approximately once in a polymer containing 4 n bases.
  • the probability P that a 9-base target sequence will be found in a 20 kilobase polynucleotide is about 20xl0 3 /2xl0 5 or 0.08
  • the probability that a 16-base target sequence will be present is about 20xl0 3 /4.3xl0 9 or 0.0000047.
  • a polymer having 9-16 recognition moieties specific for a defined 9-16 base target sequence should have high speci ⁇ ficity for the target sequence in an assay mixture con- 32 taining only viral genomes, whose greatest complexities correspond to about 400K of unique-sequence bases.
  • a 12 to 16 subunit polymer can provide adequate specificity for a viral or bacterial target sequence in an assay mixture containing viral and bacterial genomic material only; largest geno- mic sizes about 5,000 kilobases.
  • a 16 to 22 subunit polymer can provide adequate specificity for a target sequence in a polynucleotide mixture containing mammalian genomic DNA material; genomic sizes of about 5 billion base pairs of unique-sequence DNA.
  • the polymer/analyte binding affinity, and particu ⁇ larly the temperature at which the polymer just binds with the target sequence can be selectively varied according to the following criteria: (a) number of subunits in the polymer; (b) the number of hydrogen bonds that can be formed between the base-pairing moieties and the corresponding, complemen ⁇ tary bases of the analyte target sequence; (c) unit length of the polymer backbone; (d) the particular inter ⁇ subunit linkages; and (e) concentration of denaturants, such as formamide, which reduces the temperature of melting.
  • the melting temperature of oligonucleotide duplexes in the 10 to 20 bp range in ⁇ creases roughly 3°C per additional base pair formed by two hydrogen bonds, and about 6°C per additional base pair formed by three hydrogen bonds. Therefore, the target sequence length originally selected to insure high binding specificity with the polymer may be extended to achieve a desired melting temperature under selected assay conditions.
  • the target sequence may be selected to have a high per ⁇ centage of guanine plus cytosine bases to achieve a relatively high polymer/analyte melting temperature.
  • a target sequence is selected which contains a relatively high percentage of adenine plus thymine bases.
  • S the assay reagent
  • the assay reagent is the. solid support having a number of binding polymers attached to its surface through spacer arms indicated by sawtooth lines.
  • the target DNA in single strand form is reacted with the support-bound polymers under hybridization conditions, and the solid support is then washed to remove non-hybridized nucleic acid material.
  • the washed support is then reacted with the repor ⁇ ter, under conditions which favor electrostatic binding of the reporter cationic moiety to the target DNA back ⁇ bone.
  • the reporter shown in Figure 16 is a dicationic molecule having a reporter group R.
  • the reagent After reaction with the reporter solution, typically at room temperature for 1-2 minutes, the reagent is washed to remove unbound reporter, and then the assay reagent is assessed for bound reporter.
  • One approach in determining the amount of reporter associated with the reagent, particularly in the case of fluorescent or chromophoric reporter groups, is to elute the reporter from the reagent with a high salt solution and then assess the eluate for reporterhe polymer of the invention can undergo sequence-specific binding to duplex nucleic acids via base-pair-specific hydrogen bonding sites which are accessible through the major groove of 34 the double helix.
  • the duplex binding polymer preferably includes 2-aminopurine or 2, 6-diamino ⁇ purine hydrogen bonding moieties for binding to T-A or U- A base pairs, and guanine or thioguanine hydrogen-bonding moieties for binding to C-G base pairs as illustrated in Figure 8A.
  • the polymer of the invention can be .used for diagnostic assays of the types just described, but where the target nucleic acid is in nondenatured, duplex form.
  • the polymers of the instant invention can be used in place of standard RNA or DNA oligomers for a number of standard laboratory procedures.
  • mor ⁇ pholino-based polymers can be fixed to a solid support and used to isolate complementary nucleic acid sequences, for example, purification of a specific mRNA from a poly- A fraction (Goldberg et al) .
  • the instant polymers are advantageous for such applications since they are inex ⁇ pensive and straightforward to prepare from activated subunits.
  • Morpholino-based polymers can be easily and efficiently end-labelled by the inclusion in the last step of the polymer synthesis an activated and labelled morpholino- based subunit or, preferably, an 35 S-labelled methionine.
  • the type of label to be used is dependent on the final application of the polymer, and includes radioactive ( 3 H, 1 C, 32 P, or 35 S) nucleosides and biotin. Labelled mor- 35 pholino-based oligonucleotide analogs can act as efficient probes in, for example, colony hybridization
  • the polymers of the invention also have important potential use as therapeutic agents. Recently, uncharged anti-sense nucleic acid analogs, which are nearly iso- structural with DNA, have been used as anti-viral and anti-tumor agents.
  • the polymers of the present invention provide several advantages over the more conventional anti-sense agents.
  • the morpholino polymers are substantially less expensive to synthesize than oligonucleotides. This is due in part to the fact that the morpholino subunits used in polymer synthesis are derived from ribonucleo- sides, rather than the much more expensive deoxyribonu- cleosides. Also, as noted above, the coupling reaction between a phosphorous and an amine of a second subunit occurs under relatively mild conditions, so that protec ⁇ tion steps and other precautions needed to avoid unwanted reactions are simplified.
  • the morpholino-based polymer is also more stable within the cell; the polymer backbone linkage is not susceptible to degradation by cellular nucleases.
  • the uncharged morpholino polymer is more likely to efficiently enter cells than a charged oligonucleotide.
  • the morpholino polymers of the present invention may be targeted against double-stranded genetic sequences in which one strand contains predominantly purines and the other strand contains predominantly pyrimidines (e.g., Figure 8B) .
  • morpholino binding polymers targeted to the duplex have potential for also inactivating the mRNA.
  • a polymer has the potential for inactivating key genetic sequences of a pathogen in both single-stranded and double-stranded forms.
  • oligomers and oligomer combinations can perturb 37 protein synthesis in normal intact bacteria (where D is 2,6-Diaminopurine or adenine; G is Guanine; B is 5-Bromo- uracil, other 5-Halouracil or uracil; sequences are shown with their 5' end to the left) : DGG, BDDG, DDGG; DGGD; GGDG; GDGG; DGGB; GGBG; GGAGG; GGDGG; and the combinations BDD + GGDG; DDG + GDGG; DGG + DGGB; GGD + GGBG; BDDG + GDG; DDGG + DGG; DGGD + GGB GGDG + GBG; BDD + GGDG + GBG.
  • RNA sequence which plays a key role in the metabolism of a target class of organisms but not a correspondingly important role in higher organisms should be broadly adaptable to a variety of pathogenic organisms (e.g., bacteria and fungi) having a cell wall which excludes the entrance of longer poly ⁇ mers.
  • pathogenic organisms e.g., bacteria and fungi
  • 1- ⁇ -D-ribofuranosyl)-2-pyrimidinone (2-hydroxy-pyri- midine riboside) is prepared by ..the procedure of Niedballa.
  • 2-amino-9- ⁇ -D-ribofuranosyl) -1, 6-dihydro-6hpurine-6 -thione is prepared by the procedure of Fox. Dimethoxytrityl chloride, N-6-benzoyladeno- sine, N-4-benzoylcytidine, and N-2-benzoylguanosine are obtained from Sigma Chemicals. 9-fluorenylmethoxycar- bonyl chloride (FMOC chloride) , trimethylchlorosilane, isobutyric anhydride, 4-nitrobenzoyl chloride, naphthalic anhydride, and all organic solvents for reactions and chromatography were obtained from Aldrich Chemical Co.
  • Suitable protective groups for this purpose include the naphthaloyl group (Dickshit) and the aminidine groups developed by McBride et al (1986) .
  • dihalogenated electrophiles for subunit activation generally requires that the 06 of guanine moieties is protected; this protection is achieved using the diphenylcarboamoyl group
  • N-2 9-fluorenylmethoxy- carbonyl derivative of guanosine is prepared by the procedure below which is general for the protection of nucleoside amino groups: guanosine (1 mmol) is suspended in pyridine (5 ml) and treated with tri'methyl- chlorosilane (5 mmol) .. After the solution is stirred for 15 minutes, 9-fluorenylmethoxycarbonyl chloride (5 mmol) is added and the solution is maintained at room temperature for 3 hours.
  • N-2-acetylguanosine is obtained by the method of Reese.
  • N-2-naphthaylguanosine is prepared by the method of Dikshit; this reference provides a general method for the protection of nucleoside amine groups.
  • N-6 2-(4-nitrophenyl) -ethoxycarbonyl derivative is prepared by the method of Himmelsbach. 40
  • N-6 (4-nitrobenzoyl)adenosine is prepared using the procedure above for FMOC-guanosine except that 4-nitro ⁇ benzoyl chloride is substituted for FMOC chloride.
  • N- ⁇ 2-(phenylsulfonyl) -ethoxycarbonyl derivative is prepared by the procedure for FMOC guanosine except the 2- (phenylsulfonyl) -ethyl chloroformate (Balgobin) is used as the acylating agent and N-methylimidazole or pyridine is used as the solvent.
  • N-6 naphthoyladenosine is prepared by the method of Dikshit; this reference provides a general method for the protection of nucleoside amine groups.
  • N-2,N-6-bis (9-fluorenylmethoxycarbonyl) derivative of 2,6-diaminopurine riboside is prepared by the general procedure described for guanosine.
  • N-2,N-6-bis (isobutyryl) derivative is prepared by the general procedure described for guanosine.
  • N-2 (9-fluorenylmethoxycarbonyl) derivative of thioguanosine is prepared by the general procedure described for guanosine.
  • N3 of the uracil moiety 5'0-tritylated uridine-2' ,3'-acetonide is converted to the N3 anisoyl derivative by the procedure of Kamimura et al (1983) . The product is then treated with hot 80% acetic acid or 0.1 N HC1 in THF to cleave the protective groups on the ribose moiety. 41
  • Example 2 Synthesis of 5'-OH Morpholino Subunits The steps in the method are illustrated in Figure 5, with reference to structures shown in Figure 5.
  • the base-protected ribonucleoside is oxidized with periodate to a 2'-3' dialdehyde (Structure 1).
  • the dialdehyde is closed on ammonia or primary amine (Structure 2) and the 2' and 3' hydroxyls (numbered as in the parent ribose) are removed by reduction with cyanoborohydride (Structure 3) .
  • the base-protected morpholino subunit is then pro ⁇ tected at the annular nitrogen of the morpholino ring using trityl chloride or benzyhydral nitrophenyl carbonate (Structure 4) .
  • the 5' hydroxyl 42 can be protected with a trialkylsilyl group.
  • a protection step to 2 liters of acetonitrile is added, with stirring, 0.1 mole of the tosylate salt from above followed by 0.26 mole of tri- ethylamine and 0.15 mole of trityl chloride. The mixture is covered and stirred for 1 hour at room temperature after which 100 ml methanol is added, followed by stir ⁇ ring for 15 minutes. After drying by rotovaping, 400 ml of methanol is added. After the solid is thoroughly sus- pended as a slurry, 5 liters of water is added, the mixture is stirred for 30 minutes and filtered. The solid is washed with 1 liter of water, filtered, and dried under vacuum.
  • the solid is resuspended in 500 ml of dichloromethane, filtered, and rotovaped until precipitation just begins, after which 1 liter of hexane is added and stirred for 15 minutes. The solid is removed by filtering, and dried under vacuum.
  • Example 3 Alternative Synthesis of Morpholino Subunits This example describes an alternative preparation of a morpholino subunit containing an acid-labile moiety linked to the morpholino ring nitrogen. The steps are described with respect to Figure 6.
  • the subunit is prepared by oxidizing a ribonucleoside with periodate, as in Example 2, and closing the resultant dialdehyde (Structure 1) on the primary amine 4,4'-dimethoxybenzhydrylamine (which can be prepared by the method of Greenlee, 1984) buffered with benzotriazole, or p-nitrophenol. Reduction with sodium cyanoborohydride, carried out as in Example 2, gives a 43 morpholino subunit (Structure 2) having a 4,4'- dimethoxybenzhydryl group on the morpholino nitrogen.
  • This procedure is particularly useful for preparing morpholino subunits from ribonucleosides which do not have a protective group on the base (e.g., uridine) .
  • Structure 3 of Figure 5 is silylated with t-butyldi- methlsilyl chloride to give Structure 1 of Figure 12.
  • salts of sulfamic acids e.g., Structure 7 of Figure 5, and Structure 2 of Figure 12
  • salts of sulfonic acids e.g., Structure 10 of Figure 5, and Structure 5 of Figure 7
  • silica gel using triethylamine/methanol/chloroform mixtures if the silica is first pre-eluted with 2% triethylamine in chloroform.
  • the 5'hydroxyl of the doubly-protected morpholino subunit (Structure 4, Figure 5) can be converted to the amine as follows. To 500 ml of DMSO is added 1.0 mole of 44 pyridine (Pyr) , 0.5 mole of triflouroacetic acid (TFA) , and 0.1 mole of the morpholino subunit. The mixture is stirred until dissolved, and then 0.5 mole of diisopro- pylcarbodiimide (DIC) or dicyclohexylcarbodiimide (DCC) is added. After 2 hours the reaction mixture is added to 8 liters of rapidly stirred brine, which is stirred for 30 minutes and filtered.
  • DIC diisopro- pylcarbodiimide
  • DCC dicyclohexylcarbodiimide
  • the solid is dried briefly, washed with 1 liter of ice cold hexanes, filtered, and the solid is added to 0.2 mole of sodium cyanoborohydride in 1 liter of methanol, stirred for 10 minutes, 0.4 mole of benzotriazole or p-nitrophenol is added, followed by 0.2 mole of methylamine (40% in H 2 0) and the preparation is stirred four hours at room temperature [Note: the benzotriazole or p-nitrophenol buffers the reaction mixture to prevent racemization at the 4' carbon of the subunit at the iminium stage of the reductive alkyla- tion] .
  • reaction mixture is poured into 5 liters of water, stirred until a good precipitate forms, and the solid (Structure 6, Figure 5) is collected and dried.
  • This dried product is next suspended in DMF and 4 equivalents of S0 3 /pyridine complex is added. Over a period of several hours, 8 equivalents of triethylamine is added dropwise with stirring. After an additional two hours the preparation is dumped into a large volume of brine and the solid collected by filtration and dried.
  • This sulfamic acid preparation is then purified by silica gel chromatography.
  • the morpholino nitrogen should be protected as a carbamate (e.g., Structure 4 of Figure 5) instead of with a trityl group.
  • the 5' hydroxyl of the doubly-protected morpholino subunit is converted to a sulfhydral as follows. 0.1 mole of the 5'-hydroxyl subunit (Structure 4, Figure 5) is added to 1 liter of pyridine followed by 0.12 mole of toluenesulfonylchloride, and stirred for 3 hours at room temperature to give Structure 8 of Figure 5. After removing the pyridine by rotovapping, 0.5 mole of fresh sodium hydrosulfide in 1 liter of methanol/DMF containing Nal is added and the mixture is stirred at room temperature overnight.
  • Example 7 Synthesis of 5' Methylenesulfonate Subunit This example describes the preparation of a subunit suitable for use in preparing polymers with 6-atom unit- length backbones having sulfonamide linkages.
  • Example 8 Preparation of 5'-aminomethanesulfonate Subunit This example describes the preparation of a subunit suitable for use in preparing polymers with 7-atom unit- length backbones having sulfonamide linkages.
  • Example 9 Synthesis of N-methanesulfonate Subunit This example describes the preparation of a subunit containing a sulfonate moiety linked to the morpholino ring nitrogen suitable for preparing polymers with 7-atom unit-length backbones. The steps are described with respect to structures shown in Figure 7.
  • the subunit is prepared by oxidizing a ribonucleoside (Structure 1) with periodate in the 47 presence of aminomethanesulfonic acid (AMSA) and N-ethyl morpholine. The oxidation is followed by reduction with sodium cyanoborohydride in the presence of benzotriazole (used to buffer the reaction mix) to give a morpholino subunit having a methane sulfonic acid group on the morpholino nitrogen (Structure 2) .
  • the 5'hydroxyl (numbered as in the parent ribose) is then oxidized to an aldehyde (Structure 3) and converted to a primary or secondary amine (Structure 4) by reductive alkylation as in Example 5, and tritylated to give the desired subunit of Structure 5.
  • N-methanecarboxylate Subunit This example describes the preparation of a subunit containing a carboxylate moiety linked via a methylene to the morpholino ring nitrogen suitable for preparing polymers with 7-atom unit-length backbones.
  • the subnit can be prepared essentially as in Example 9, but substituting glycine for aminomethanesulfonic acid. Alternatively, it is generally more convenient to prepare it starting with Structure 3 of Figure 5. This is readily alkylated on the morpholino ring nitrogen using chloroacetic acid or bromoacetic acid. The 5' hydroxyl is then converted to a primary amine and tritylated as in Example 9.
  • N4-Benzoylcytidine-2',3'-acetonide (1 mmole) was converted to the 5'-Iodo derivative by reaction with methyltriphenoxyphosphonium iodide in DMF (20ml) under argon at room temperature for 20 hours. Methanol (5 ml) was added and after 30 minutes the mixture was evaporated in vacuo. The residue was dissolved in ethyl acetate and 48 the solution washed with aqueous sodium thiosulfate, then brine. After drying with sodium sulfate and evaporation of the solvent the product was purified by chromatography on silica using isopropanol/chloroform mixtures.
  • the iodo compound (1 mmole) is reacted with potassium cyanide (5 mmol) in Dimethylsulfoxide for 12 hours under argon atmosphere.
  • the nitrile is isolated by pouring the reaction mixture into saturated aqueous sodium dihydrogen phosphate. The mixture is extracted with ethyl acetate, the organic layer washed well with water, dried over sodium sulfate and evaporated in vacuo.
  • the nitrile is purified by chromatography on silica using chloroform/ethylacetate mixtures.
  • the nitrile from the previous paragraph is treated with a mixture of equal parts of DMF and aqueous ammonia at 25°C for 24 hours.
  • the mixture is treated with rhodium on alumina and hydrogenated in a hydrogen atmosphere to provide the amine.
  • the residue is dissolved in 0.2 N HC1 to cleave the acetonide.
  • the amine diol may be purified by ion exchange on a cation exchange column.
  • the amine moiety may be tritylated as in Example 2 to give the amine-protected 2',3'-diol.
  • the sulfide from the previous paragraph was oxidized with excess perbenzoic acid in chloroform to the sulfone. This was immediately, treated with 0.2 N HCl/dioxane to cleave the acetonide group.
  • the diol was purified by chromatography on silica using methanol/chloroform mixtures.
  • the diol sulfone from above is reduced with hydrogen/ palladium on carbon in DMF/methanol in the presence of acetic acid to remove the carbobenzoxy group.
  • One equivalent of tosic acid is added to the mix, the solution is filtered and the filtrate evaporated.
  • the pendant amine is tritylated as in Example 2 to give the amine-protected 2',3'-diol.
  • the benzoyl group on the base is removed by treatment with equal amounts of DMF and CMC aqueous ammonia at room temperature for 24 hours.
  • Example 13 Activation and Coupling To Give Carbamate Linkage
  • This example describes the activation of morpholino- subunits, such as prepared in Example 2, and their sub- sequent coupling via a carbamate linkage to yield a 6- atom unit-length backbone.
  • the example is described with reference to the Structures in Figure 9. Activation Step
  • the activated subunit (Structure 2) is added to the DMF solution of unprotected subunit and incubated at room temperature for 1 hour to give coupled product (Structure 4) .
  • the activated subunit (Structure 2) is added to the DMF solution of deprotected subunit (Structure 3) and incubated at room temperature for 1 hour to give coupled product (Structure 4) .
  • the activated subunit (Structure 2) is added to the DMF solution of deprotected subunit (Structure 3) and incubated at room temperature for 1 hour to give coupled product (Structure 4) .
  • This example describes the oxidation of a ribonu ⁇ cleoside containing a protected amine linked through the 5' methylene, such as prepared in Example 11 or 12, and 54 coupling to the unprotected amine of another subunit to simultaneously form a morpholino ring structure and join the subunits.
  • the example is described with reference to the structures in Figure 14.
  • Tritylated subunit (Structure 2) in methanol, is reacted with 11 mmole of NaI0 4 to give the dialdehyde (Structure 3) .
  • a weak acid is used to neutralize the amine component (Structure 1) and buffer the reaction in this coupling step.
  • Weak acids which have been found suitable for this purpose are: carbonic, ortho and para nitrophenol, and benzotriazole. Accordingly, the dialdehyde (Structure 3) is combined with a suitable salt of Structure 1 in a water/methanol mixture to give the coupled product (Structure 4) .
  • Oligomer of the Sequence 5'-CUGU This example describes the assembly of a short oligomer containing a sulfamide-linked backbone (Structure C-C of Figure 4, wherein X 2 is a nitrogen) coupled as in Example 14.
  • This solution assembly method is particularly useful for large-scale synthesis of short oligomers suitable for subsequent assembly into longer oligomers using the solid-phase method (Example 19) .
  • 5'Sulfamic acid subunits of C, U, and G tritylated on the morpholino ring nitrogen are prepared as in Example 5.
  • the U subunit is then activated by conversion to the sulfamoyl chloride form as in Example 14.
  • the C subunit and the G subunit are deprotected as in Example 14.
  • the deprotected C component (1.1 m mole) is dissolved in 5 ml DMF and 0.3 ml TEA, followed by addition of 1.0 m mole of the activated U component.
  • the deprotected G component is reacted with the activated U component.
  • the CU dimer is deprotected as in Example 14. Better yields of tetramer are obtained when the dimer, after the initial ether precipitation, is thoroughly resuspended in about 2 ml of trifluoroethanol, reprecipitated with 30 ml of ether, and then resuspended in DMF and TEA for subsequent coupling.
  • Coupling to form the desired tetramer entails simply adding 1 m mole of activated GU dimer to the DMF/TEA 56 solution containing 1.1 m mole of deprotected CU dimer.
  • Example 19 Solid-Phase Assembly of Sulfamide-Linked Morpholino Polymer This example describes the use of tetramer blocks, prepared as per Example 18, for solid-phase assembly of a morpholino polymer containing sulfamide intersubunit linkages. Solid-phase assembly provides a rapid method for assembly of longer binding polymers. The use of short oligomer blocks instead of monomers greatly simplifies separation of the final product from failure sequences.
  • This product is purified by silica gel chromatography and then added to a suitable solid support containing primary amine functions (e.g., Long Chain Alkyl Amine Controlled Pore Glass, from Pierce of Rockford, Illinois) .
  • This procedure links the first tritylated subunit to the synthesis support via a linker which is stable to the acidic conditions used for detritylations, but which can be readily cleaved via a beta elimination mechanism using a strong non- nucleophilic base, such as a 1, 8- Diazabicyclo[5.4.0]undec-7-ene (DBU) .
  • a linker which is stable to the acidic conditions used for detritylations, but which can be readily cleaved via a beta elimination mechanism using a strong non- nucleophilic base, such as a 1, 8- Diazabicyclo[5.4.0]undec-7-ene (DBU) .
  • DBU 1, 8- Diazabicyclo[5.4.0]undec-7-ene
  • the coupling cycle for addition of each subunit or oligomer block generally includes deprotection of the terminal backbone moiety, a thorough wash, addition of the next activated subunit or oligomer block, and a thorough wash.
  • the coupling efficiency for each addition can be determined by collecting each detritylation solu ⁇ tion and subsequent wash and quantitating the trityl therein.
  • Detritylation in the present sufamide-linked polymer is achieved by slowly passing through the column a solu- tion of 2% formic acid in trifluoroethanol (or 2% dichlo- roacetic acid in dicloromethane) until the eluant no longer tests positive for trityl (readily determined by adding a drop of eluant to 100 ⁇ l methanesulfonic acid and inspecting for the visible yellow color characteris- tic of the trityl carbonium ion) . Thereafter the support is thoroughly washed to remove excess acid and then washed with DMF containing 1% by volume of N-ethylmor- pholine (NEM) .
  • NEM N-ethylmor- pholine
  • Coupling of the next subunit or oligomer block in the desired polymer sequence entails addition of 58 a concentrated DMF solution containing the activated monomer or oligomer and a molar equivalent of NEM. Since the rate of coupling is a function of concentration it is desirable to add a substantial molar excess of monomer or oligomer relative to the concentration of support-bound growing chains. A 5-fold molar excess of activated monomer or oligomer over that of the growing chains often gives acceptable coupling efficiencies.
  • Required cou ⁇ pling times can be determined by removing at specified time intervals small defined quantities of the support material, thoroughly washing, treating the support with methanesulfonic acid, and then spectrophotometrically quantitating the released trityl carbonium ion (molar absorbance at 409 nm is 45,000 in methane sulfonic acid).
  • the unreacted subunit or oligomer is washed from the support with DMF.
  • the un ⁇ reacted subunit is generally recovered, purified by chromatography, and reused for later synthesis.
  • the sup ⁇ port is thoroughly washed with the solvent trifluoro- ethanol, without added acid. Washing is complete when addition of a drop of the wash eluant to 100 ⁇ l methane ⁇ sulfonic acid shows no yellow color.
  • the synthesis support is treated with 20% DBU in DMF for two hours at room temperature in the presence of 2% diethylmalonate, to tie up the vinylsulfone generated during cleavage of the linker.
  • the released morpholino polymer is washed from the support with DMF and precipi ⁇ tated by adding ethylacetate.
  • the precipitate contains full-length polymer having a 5' methylamine, the bases 59 still protected and a trityl moiety on the terminal morpholino nitrogen.
  • the precipitate con ⁇ tains small amounts of failure sequences.
  • the polymer size can be confirmed by positive ion fast atom mass spectrometry.
  • solubilizing groups to the morpholino polymer this can be done conveniently by detritylting the N-terminal morpholino nitrogen using 2% formic acid in trifluoroethanol.
  • solubilizing moiet is to be added, then the 5'me- thylamine is acetylated with acetic anhydride before the detritylation step.
  • Polyethylene glycol 1000 (from Polysciences Inc., Warrington, Pennsylvania, USA) is thoroughly dried by dissolving in dry DMF and then evaporating the solvent under vacuum. The solid is resuspended in a minimal volume of pure dry DMF and 0.5 mole equivalent (relative to PEG 1000) of bis (p-nitrophenyl)carbonate and 1 mole equivalent of TEA is added and the preparation sealed and incubated overnight at 30°C to give p-nitrophenyl car ⁇ bonate-activated PEG 1000.
  • the full-length morpholino polymer which has been detritylated is added to a substantial molar excess (generally 10- to 20-fold) of activated PEG 1000 and incubated two hours at room temperature. Unreacted PEG 1000 is removed by precipitation of the tailed polymer with ether.
  • Purification at pH 2.5 is general for binding poly- ers wherein about half or more of the base-pairing moieties are of types 1, 2, 3, and 7 of Figure 2.
  • the eluant containing the polymer is desalted by loading on an equivalent-sized column packed with 35 micron chromatographic polypropylene (Cat. No. 4342 from Polysciences, Inc.) and washing thoroughly with solvent A. If baseline separation was achieved in the foregoing cation exchange chromatography, then pure product is obtained simply by eluting with solvent C; otherwise, the product is eluted with a linear gradient ranging from 100% solvent A to 100% solvent C. When the product is somewhat acid sensitive the solution is neutralized with dilute NaOH before drying under reduced pressure. Purification at high pH Purification at pH 11 is generally used for binding polymers wherein above half or more of the base-pairing moieties are at type 4, 5, 6 and 9 of Figure 2.
  • N,N-diethylethanolamine (Aldrich) is added to degas ⁇ sed water to adjust the pH to 11.0 (solvent D) .
  • a cor- responding pH 11 solution 2 N in KC1 (solvent E) is prepared.
  • a third pH 11 solution is prepared by mixing Solvent D 1:1 by volume with chromatographic grade aceto ⁇ nitrile (solvent F) .
  • the fully-deprotected binding polymer, prepared as above, is suspended in solvent D at a concentration of about 1 mg/ml.
  • the pH is adjusted, if necessary, to pH 11 with N,N-diethylethanol-amine.
  • About 10 ml of this polymer solution is placed on a chromatography column 1 cm in diameter and 10 to 20 cm in length which is packed with anion-exchange support Q-Sepharose Fast Flow (Phar ⁇ macia) . After washing the column thoroughly with solvent D, the column is eluted with a linear gradient ranging from 100% solvent D to 100% solvent E and the eluant is monitored at 254 nm. 62
  • the eluant containing the polymer is desalted by loading on an equivalent-sized column of polypropylene and washing thoroughly with solvent D. If baseline separation is achieved in the foregoing anion exchange chromatography then pure product is obtained simply by eluting with solvent F; otherwise, the product is eluted with a linear gradient ranging from 100% solvent D to 100% solvent F. Fractions containing the product are dried under reduced pressure. H. Sequence confirmation
  • One method for confirming the sequence of the poly- mer is to take a small portion of the growing polymer after coupling each oligomer block and use mass spectral analysis to follow the elongation of the polymer. This method is applicable except for those rare cases where two blocks used in the synthesis happen to have exactly the same mass.
  • An indirect method to help verify the correctness of the polymer subunit sequence is to pair the morpholino polymer with its complementary DNA (whose sequence can be confirmed by established methods) and with DNA sequences 63 which might have resulted if the blocks were assembled in the wrong order. Pairing between the polymer and DNA can be evaluated by the occurrence of a hypochromic shift in the 240 to 290 nm wavelength region; such a shift occurs only between the polymer and its complementary sequence.
  • the polymer/DNA duplex can also be distinguished from any partially-mismatched duplex by slowly raising the temper ⁇ ature while monitoring the absorbance in the 240 to 290 nm wavelength region.
  • the perfect duplex will have a melting temperature (corresponding to a 50% reduction in the hypochromicity) generally 10 degrees or more above that of any mismatched duplex.
  • This example describes the preparation, structural confirmation, and assessment of target binding affinity of a simple carbamate-linked morpholino polymer.
  • a carbamate-linked morpholino hexamer wherein all P x moieties are cytosines is assembled from dimer prepared as in Example 13.
  • One third of that dimer preparation is detritylated (as in Example 13) and the remaining two thirds is activated (again as in Example 13) .
  • Half of the activated dimer is reacted with the detritylated dimer to give tetramer, which is purified by silica gel chromatography developed with 6% methanol/94%chloroform.
  • the tetramer is detritylated and reacted with the remain ⁇ ing activated dimer to give hexamer, which is purified by silica gel chromatography developed with 10% methanol/90% 64 chloroform.
  • a 260 units of DNA target p(dG) 6 purchased from Pharmacia LKB, is dis ⁇ solved in 50 microliters of deionized water and 200 65 microliters of DMSO (spectrophotometric grade from Aid- rich Chem. Co.) is added (stock solution A).
  • DMSO spectrophotometric grade from Aid- rich Chem. Co.
  • stock solution B 1.8 mg of the tailed morpholino hexamer, c(mC) 6 -PEG1000, is dis ⁇ solved in 0.36 ml of spectrophotometric grade DMSO (stock solution B) .
  • Phosphate buffer is prepared by adjusting the pH of 0.05 N NaOH to 7.4 using phosphoric acid, followed by addition of EDTA to a final concentration of 0.001 N (Buffer C) .
  • Stock solutions A and B are assayed for the actual concentration of polymer by UV; the absorbance of stock solution A is measured in 0.1 N NaOH and stock solution B is measured in 0.1 N HC1. Measurements at these pH extremes minimize base stacking and other polymer inter ⁇ actions which can give absorbance values not proportional to the component monomers.
  • Stock solutions A and B are diluted with Buffer C to give solutions of a final con ⁇ centration of 10 micromolar in polymer. The required dilutions are calculated using molar absorbencies of 65,000 for solution A, p(dG)6, and 77,100 for solution B, c(mC * ) 6 -PEG1000.
  • Using four matched quartz cuvettes one is filled with 0.5 ml of 10 micromolar p(dG) 6 and 0.5 ml of Buffer C and a second is filled with 0.5 ml of 10 micromolar c(mC * ) 6 -PEG1000 and 0.5 ml of Buffer C. These two cu- vettes are placed in the reference beam of the tempera ⁇ ture-controlled cell housing. Next, a third cuvette is filled with 1 ml of Buffer C and a fourth is filled with 0.5 ml of 10 micromolar p(dG) 6 and 0.5 ml of 10 micromo ⁇ lar c(mC * ) 6 -PEG1000.
  • Example 21 Solution-phase Assembly of Simple Prototype Sulfamide-linked Morpholino Polymer and Assessment of Binding to RNA and DNA Target Sequences This example describes the preparation and target binding of a simple sulfamide-linked morpholino polymer.
  • a sulfamide-linked morpholino hexamer, wherein all P ⁇ moieties are cytosines, is assembled from 5'sulfated 67 methylamine subunit (R is methyl) prepared as in Example 5 and activated as in Example 14. Activated monomer is reacted in DMF with an excess of 5'OH subunit lacking a protective group on the morpholino nitrogen, prepared as in Example 2. The resultant dimer is purified by silica gel chromatography developed with methanol/chloroform mixtures and then deprotected as in Example 14. This product is then reacted with more activated monomer, the chain-extended product purified, and deprotected as above. This cycle is repeated until hexamer is obtained. Before the last .detritylation, the mass of the hexamer was confirmed by negative ion FAB mass spectro ⁇ scopy, which showed M-l 2598.9 (100).
  • the sulfamide-linked hexamer is tailed with PEG-1000, deprotected, purified, and tested for binding to its DNA target, p(dG) 6 , and its RNA tar ⁇ get, poly(G) .
  • the target-binding affinities, expressed in T m values, for this sulfamide-linked hexamer, referred to as s (mC) 6 are tabulated below, along with the cor- responding target-binding affinities for the analogous DNA oligomer, p(dC) ⁇ .
  • T_ value (° C.) s (mC) 6 /p(dG) 6 25 p(dC) 6 /p(dG) ⁇ 29

Abstract

A polymer composition is disclosed composed of morpholino subunit structures which are linked together by uncharged, achiral linkages. These linkages are one to three atoms in length, joining the morpholino nitrogen of one subunit to the 5' exocyclic carbon of an adjacent subunit. Each subunit contains a purine or pyrimidine base-paring moiety effective to bind by hydrogen bonding to a specific base or base-pair in a target polynucleotide.

Description

UNCHARGED MORPHOLINO-BASED POLYMERS HAVING ACHIRAL INTERSUBUNIT LINKAGES
Field of the Invention
The present invention relates to morpholino-based polymers.
References Agarwal, Proc Nat Acad Sci USA, _85_:7079 (1988).
Balgobin, N., et al., Tetrahedron Lett, 22:3667 (1981) .
Beli ova, Tetrahedron Lett, 3_7:3557 (1967) . Blake et al., Biochem, 4:6132 (1985a). Blake et al., Biochem 24:6139 (1985b).
Dikshit et al., Canadian J Chem, _66:2989 (1988). Fild et al., Chem Ber, 1^:142 (1980) . Froehler, et al., Nucleic Acids Res. 1^6:4831 (1988). Fox, J.J., et al., J Am Chem Soc, _8_0:1669 (1958). Gait, "Oligonucleotide Synthesis, A practical Ap¬ proach," pages 31-33, IRL Press, Oxford, England (1984). Goldberg, M. L. et al; Methods in Enzymology 68:206 (1979) .
Greenlee, J Org Chem, 4_9 2632 (1984) . Grunstein, M. et al; Methods in Enzymology 68:379 (1979) .
Himmelsbach, F., and W. Pfleiderer, Tetrahedron Lett, _2_4:3583 (1983) .
Jayaraman, et al., Proc Natl Acad Sci USA 78:1537 (1981) .
Kamimura et al., Chemistry Letters (The Chem. Soc. of Japan) pg. 1051 (1983) .
Lerman, L.S., "DNA Probes: Applications in Genetic and Infectious Disease and Cancer, " Current Comm in Molec Biol, Cold Spring Harbor Laboratory (1986) .
Letsinger and Miller, J A er Chem Soc, 91:3356 (1969) .
McBride et al., J Amer Chem Soc l_fJ8^:2040 (1986) . Miller, et al., Biochemistry 18:5134 (1979). Miller, et al., J Biol Chem 255:6959 (1980). Miller, et al., Biochimie 67:769 (1985). Murakami, et al., Biochemistry 24:4041 (1985).
Niedballa, U., and H. Vorbruggen, J Org Chem, 32:3668 (1974).
Pitha, Biochem Biophys Acta 204:39 (1970a) . Pitha, Biopoly ers _9"965 (1970b). Reese, C.B., and R.S. Saffhill, J Chem Soc Perkin Trans, JL:2937 (1972) .
Smith, et al., J Amer Chem Soc _8 :6204 (1958). Smith, et al., Proc Natl Acad Sci USA 83:2787 (1986). Southern, E.; Methods in Enzymology 68:152 (1979)
Stirchak E.P. et al., Organic Chem. 52^:4202 (1987). Summerton, et al., J Mfolec Biol, 122:145 (1978). Summerton, et al., J Theor Biol, 18_: 61 (1979a). Summerton, J Theor Biol, Jl-' 11 (1979b) . Szostak, J. W. et al; Methods in Enzymology 68:419 (1979) .
Thomas, P.; Methods in Enzymology 100:255 (1983). Toulme et al., Proc Nat Acad Sci, USA 83:1227 (1986) . Trichtinger et al., Tetrahedron Letters 2 :711 (1983) .
Background of the Invention Polymers which are designed for base-specific bind¬ ing to polynucleotides have significant potential both for in vitro detection of specific genetic sequences characteristic of pathogens (Ler an) and for in vivo inactivation of genetic sequences causing many diseases— particularly viral diseases (Belikova, Summerton) .
Standard ribo- and deoxyribonucleotide polymers have been widely used both for detection of complementary genetic sequences, and more recently, for inactivating targeted genetic sequences. However, standard polynucle- otides suffer from a number of limitations when used for base-specific binding to target oligonucleotides. These limitations include (i) restricted passage across biolo¬ gical membranes, (ii) nuclease sensitivity, (iii) target binding which is sensitive to ionic concentration, and (iv) susceptibility to cellular strand-separating mecha¬ nisms.
In principle, the above limitations can be overcome or minimized by designing polynucleic acid analogs in which the bases are linked along an uncharged backbone. Examples of uncharged nucleic acid analogs have been reported. Pitha et al (1970a, b) have disclosed a vari¬ ety of homopolymeric polynucleotide analogs in which the normal sugar-phosphate backbone of nucleic acids is replaced by a polyvinyl backbone. These nucleic acid analogs were reported to have the expected Watson/Crick pairing specificities with complementary polynucleotides, but with substantially reduced T values (Pitha, 1970a) . One serious limitation of this approach is the inability to construct polymers by sequential subunit addition, for producing polymers with a desired base sequence. Thus the polymers cannot be used for base-specific binding to selected target sequenceδolynucleotide analogs containing uncharged, but stereoisomeric, methylphospho- nate linkages between the deoxyribonucleoside subunits have been reported (Miller, 1979, 1980; Jayaraman; Murakami; Blake, 1985a, 1985b; Smith) . More recently a variety of analogous uncharged phosphoramidate-linked oligonucleotide analogs have also been reported (Froehler, 1988) . These polymers comprise deoxynucleo- sides linked by the 3'OH group of one subunit and the 5'OH group of another subunit via an uncharged chiral phosphorous-containing group. These compounds have been shown to bind to and selectively block single-strand polynucleotide target sequences. However, uncharged phosphorous-linked polynucleotide analogs of the type just described have limitations, particularly the cost and difficulty of preparing the polymers.
More recently, deoxyribonucleotide analogs having un- charged and achiral intersubunit linkages have been constructed (Stirchak 1987) . Since these polymers are stereoregular, all polymers having a given subunit se¬ quence will have the same Tm value for a given target nucleotide sequence, thus avoiding some of the limita- tions inherent in chirally-linked polymers. These un¬ charged, achiral deoxyribonucleoside-derived analogs, however, are limited by relatively high cost of starting materials.
Summary of the Invention
It is therefore one general object of the invention to provide a polymer capable of sequence-specific binding to polynucleotides and which overcomes or minimizes many of the problems and limitations associated with poly- nucleotide analog polymers noted above.
The invention includes a polymer composition con¬ taining morpholino ring structures of the form:
Figure imgf000007_0001
The ring structures are linked together by uncharged, achiral linkages, one to three atoms long, joining the morpholino nitrogen of one ring structure to the 5' exocyclic carbon of an adjacent ring structure.
Each ring structure includes a purine or pyrimidine base-pairing moiety Pi which is effective to bind by base-specific hydrogen bonding to a base in a target sequence in a polynucleotide.
These and other objects and features of the invention will become more fully apparent when the following detailed description of the invention is read in conjunction with the accompanying examples and figures.
Brief Description of the Figures Figure 1 shows a basic ^-morpholino ring structure which is linked through uncharged, achiral linkages to form the polymer of the present invention. P, is a purine or pyrimidine base pairing moiety.
Figure 2 shows several exemplary purine- and pyrimidine base-pairing moieties (represented as P{ of the ring structures shown in Figure l) , where X = H, CH3 F, Cl, Br, or I.
Figure 3 shows several preferred subunits having 5- atom (A) , six-atom (B and C) and seven-atom (D-G) linkin groups suitable for forming polymers. Y = 0 or S. X1 = O or S X2 = O, S, CH2, or NR,. X3 = O, S, CH2, or NR2. XA = 0, S, or NR,. X5 = 0, S, CH2, NR2, or S02. n = 0, 1, or 2; when n = 0, X5 is not S02. When n = 1, X5 is CH2 or S02. , = H, CH3, or other group which does not interfere with sequence-specific hydrogen-bonding of the polymer to its target polynucleotide. R2 is an electron withdrawing group, such as methane-sulfonyl, which reduces the pKa of the nitrogen to which it is attached to less than pKa=6. Figure 4 shows a repeating subunit segment of exemplary morpholino-based polymers, designated A-A through G-G, constructed using subunits A-G, respectively, of Figure 3. Y, X,, X2, X3, X4, X5, n, R and R2 are as in Figure 3. Figure 5 shows the steps in the synthesis of several types of morpholino subunits from a ribonucleoside.
Figure 6 shows an alternative synthesis of the basic morpholino subunit.
Figure 7 shows the steps in the synthesis of a mor- pholino subunit designed for construction of polymers with seven-atom repeating-unit backbones.
Figure 8 shows the binding mode for 2-amine- containing purines to polar major-groove sites of respective target base-pairs (Figure 8a) and a representative base sequence of a duplex-binding polymer (Figure 8b) . In Figure 8b, a = Adenine; c = cytosine; g = guanine; t = thymine; u = uracil; D = 2,6-Diaminopurine or 2-aminopurine; G = Guanine or thioguanine; | = high specificity hydrogen bonding; and : = low specificity hydrogen bonding.
Figure 9 shows the steps in linking two morpholino subunits through a carbamate linkage.
Figure 10 shows the activation of sulfamic acid and coupling to form a sulfamide linkage.
SUBSTITUTE SHEET, 6-1
Figure 11 shows the activation of sulfonic acid and coupling to form a sulfonamide linkage.
Figure 12 shows the steps in linking two morpholino subunits through a sulfamate linkage. Figure 13 shows the steps in linking two morpholino subunits through an amide linkage.
Figure 14 shows a subunit coupling procedure which simultaneously generates the morpholino ring structure.
Figure 15 shows thermal denaturation plots for poly(dC)/poly(dG) and. poly(C morpholino)/poly(dG) duplexes where the poly(C morpholino) was constructed according to the present invention.
Figure 16 illustrates a diagnostic solid-support particle employing polymers of the present invention for use in a probe-diagnostic assay.
Detailed Description of the Invention
SUBSTITUTE SHEET The present invention includes a morpholino- based polymer which is designed for base-specific binding to a target sequence of a polynucleotide. The polymer is composed of morpholino-based ring structures which are linked together by uncharged, achiral linkages, one to three atoms long, joining the morpholino nitrogen of one structure to the 5' exocyclic carbon of an adjacent structure.
A. Morpholino-Based Subunits
Figure 1 shows the β-morpholino ring structures on which the polymer subunits are based, where the morpho¬ lino carbon atoms are numbered as in the parent ribose. As seen in Figure 1, the ring structure contains a 5' methylene attached to the A ' carbon in the β-orientation.
Each ring structure includes a purine or pyrimidine or related hydrogen-bonding moiety, PA, attached to the backbone morpholine moiety through a linkage in the β- orientation.
The purine hydrogen-bonding moieties or bases in¬ clude purines as well as purine-like planar ring struc¬ tures having a 5-6 fused ring in which one or more of the atoms, such as N3, N7, or N9 is replaced by a suitable atom, such as carbon. The pyrimidine moieties likewise include pyrimidines as well as pyrimidine-like planar 6- membered rings in which one or more of the atoms, such as NI, is replaced by a suitable atom, such as carbon. Preferred hydrogen-bonding moieties in the invention include the set of purines and pyrimidines shown in Figure 2. Each base includes at least two hydrogen- bonding sites specific for a polynucleotide base or base- pair. Where the polymers are used for sequence-specific binding to single-stranded polynucleotides, the purine 8 structures 1-3 are designed to bind to thymine or uracil bases; structures 7-8, to guanine bases; structures 4-6, to cytosine bases; and structure 9, to adenine bases.
The polymers of the invention are also effective to bind to hydrogen-bonding sites accessible through the major-groove in duplex polynucleotides having mostly purine bases in one strand and mostly pyrimidine bases in the complementary strand, as discussed below.
Because of the similar type and positioning of the two central polar major-groove sites among the different base-pairs of duplex nucleic acids (i.e., the NH4 and 06 of a CG base-pair present the same H-bonding array as the NH6 and 04 of an AT base-pair) , the H-bonding moiety of a duplex-binding polymer must hydrogen-bond to the N7 of its target base-pair in order to uniquely recognize a given base-pair in a target genetic duplex. Thus, where the polymers of the present invention are targeted against duplex genetic sequences (containing predominant¬ ly purines in one strand and predominantly pyrimidines in the other strand) , the hydrogen-bonding moieties of the polymer preferably contain purines having an amine at the 2 position since that amine is suitably positioned for H- bonding to the N7 of the target base-pair. Structures 2 and 3 of Figure 2 provide for specific binding to a TA or UA base-pair, and Structures 4 and 6 provide for specific binding to a CG base-pair. Two bases which are parti¬ cularly useful in a duplex-binding polymer are 2,6-diami- nopurine (Structure 3) and guanine (Structure 4) . Figure 8 illustrates the binding of these two bases to the polar major-groove sites of their respective target base-pairs in duplex nucleic acids.
The morpholino subunits of the instant invention are combined to form polymers by linking the subunits through stable, achiral, uncharged linkages. The linking group of a subunit usually includes a carbonyl or sulfonyl electrophile for reaction with a nucleophile of the subunit to which it is to be linked. As used herein "carbonyl" means a -C=0 or -C=S group, and "sulfonyl" means an 0=S→0 group.
The selection of subunit linking groups for use in polymer synthesis is guided by several considerations.
Initial screening of promising intersubunit linkages
(i.e., those linkages which are predicted to not be unstable and which allow either free rotation about the linkage or which exist in a single conformation) typical¬ ly involves the use of space-filling CPK or computer molecular models of duplex DNA or RNA. The DNA and RNA duplexes are constructed according to parameters deter- mined by x-ray diffraction of oligodeoxyribonucleotides in the B-form and oligoribonucleotide-containing duplexes in the A-form.
In each of these constructed duplexes, one of the two sugar phosphate backbones is removed, and the pro- spective backbone, including the morpholino ring and intersubunit linkage, is replaced, if possible, on the sites of the bases from which the original sugar-phos¬ phate backbone has been removed. Each resulting poly- nucleotide/polymer duplex is then examined for coplana- rity of the Watson/Crick base pairs, torsional and angle strain in the prospective binding polymer backbone, degree of distortion imposed on the nucleic acid strand, and interstrand and intrastrand nonbonded interactions.
In the case of amide-containing linkages, special attention is paid to whether or not amide-containing backbones can readily adopt a conformation in which the amide moieties are planar. This is important because of the substantial energy cost required to force an amide into a nonplanar conformation. 10
Initial studies of this type carried out in support of the present invention showed that for morpholino-based polymers the preferred unit backbone length (i.e., the number of atoms in a repeating backbone chain in the polymer) is 6 atoms. However, the modeling studies also show that certain 5-atom and 7-atom repeating-unit mor¬ pholino-based backbones meet the requirements for binding to targeted genetic sequences.
Since the morpholino structure itself contributes 4 atoms to each repeating backbone unit, the linkages in the five-atom, six-atom, and seven-atom repeating-unit backbone contribute one, two, and three atoms to the backbone length, respectively. In all cases, the linkage between the ring structures is (a) uncharged, (b) achi- ral, (c) stable, and (d) must permit adoption of a con¬ formation suitable for binding to the target polynucleo¬ tide.
Subunit backbone structures judged acceptable in the above modeling studies were then assessed for feasibility of synthesis. The actual chemical stability of the intersubunit linkage was assessed with model compounds or dimers.
Figure 3 shows several preferred β-morpholino sub- unit types, including linkage groups, which meet the constraints and requirements outlined above. It will be appreciated that a polymer may contain more than one linkage type.
Subunit A in Figure 3 contains a 1-atom sulfonyl linkage which forms the five atom repeating-unit backbone shown at A-A in Figure 4, where the morpholino rings are linked by a 1-atom sulfonamide linkage. It is noted here that the corresponding amide linkage (substituting a carbonyl for sulfonyl linkage) is not acceptable due to lack of rotational freedom about the carbon-nitrogen 11 tertiary amide bond.
Subunits B and C in Figure 3 are designed for 6-atom repeating-unit backbones, as shown at B-B and C-C, re¬ spectively, in Figure 4. In Structure B, the atom X linking the 5' morpholino carbon to the carbonyl group may be oxygen or sulfur, but not nitrogen or carbon, due to lack of free rotation about the resultant intersubunit linkage. The C=Y carbonyl group may be either C=0 or C=S, as noted above. In Structure C, the moiety X linking the 5'morpho¬ lino carbon to the sulfonyl (0=S→0) group may be a methy- lene, oxygen, sulfur, or a nitrogen. The nitrogen may be secondary (NH) , or tertiary (NR) , where R is a methyl or other group which does not interfere with polymer binding to the target polynucleotide (as can be easily determined from molecular modeling studies such as those outlined above) .
Subunits D-G in Figure 3 are designed for 7-atom repeating-unit backbones, as shown at D-D through G-G, respectively, in Figure 4. In Structure E, the X can be a secondary nitrogen (NH) , or a tertiary nitrogen (NR) where R is a is a methyl or other group which does not interfere with polymer binding to the target polynucleo¬ tide, as can be determined from molecular modeling stu- dies. In addition, X in Structure E can be an oxygen since the 5'' methylene in such morpholino structures is surprisingly resistant to nucleophilic attack.
Based on the molecular modeling studies of the type described above, both the sulfamate (Structure C-C of Figure 4 wherein X is oxygen) and sulfonate (structure E- E of Figure 4 wherein X is oxygen) linkages were good candidates. Experiments conducted in support of the present invention indicated that the 5' tosylate of the basic morpholino cytosine subunit (Structure 8 of Figure 12
5, where Pi is N4-benzoylated cytosine) are surprisingly resistant to both intermolecular and intramolecular nucleophilic attack on the 5' methylene. This suggested that the corresponding sulfamate, and possibly the sul- fonate also, may be sufficiently stable for intersubunit linkages. Accordingly, a sulfamate-linked dimer (Struc¬ ture C-C of Figure 4, where X is oxygen) was prepared, and assessed for linkage stability under conditions commonly used for polymer synthesis (i.e., detritylation conditions, base-deprotection conditions, and purifica¬ tion conditions, such, as detailed in Example 19) . These studies confirmed that such linkages are adequately stable under conditions typically required for synthesis, deprotection, purification and various applications. In Structure G-G of Figure 4, when n is zero, X must not be S02, and when n is one, X is CH2 or S02.
B. Subunit Synthesis
The most economical starting materials for the synthesis of morpholino-subunits are generally ribon- ucleosides. Typically, ribonucleosides containing hydro¬ gen-bonding moieties or bases (e.g., A, U, G, C) are transformed to their morpholino derivatives to provide a complete set of subunits for polymer synthesis. Where a suitable ribonucleoside is not available, a 1-haloribose or, preferably, a lα-bromoglucose derivative, can be linked to a suitable base and this nucleoside analog then converted to the desired β-morpholino structure via peri- odate cleavage, and closing the resultant dialdehyde on a suitable amine.
Because of the reactivity of the compounds used for subunit synthesis, activation, and/or coupling, it is generally desirable, and often necessary, to protect the exocyclic ring nitrogens of the bases and sometimes the 13 oxygens of U and G. Selection of these protective groups is determined by (i) the relative reactivity of the moiety to be protected, (ii) the type of reactions in¬ volved in subunit synthesis and coupling, and (iii) the stability of the completed polymer prior to base depro- tection.
Methods for base protecting a number of the more common ribonucleosides are given in Example 1. The methods detailed in the example are generally applicable for forming nucleosides with amine-protective groups. Standard base- protective groups used for nucleic acid chemistry are often suitable including the following groups: benzoyl for the N4 of cytosine (C) ; benzoyl or p-nitrobenzoyl for the N6 of adenine (A) ; acetyl, phenyl- acetyl or isobutyryl for the N2 of guanine (G) ; and N2,N6-bisisobutyryl for 2, 6-diaminopurine residues. These protective groups can be removed after polymer assembly by treatment with ammonium hydroxide.
It is sometimes desirable to protect the base por- tion of the morpholino subunit with a group which can be readily removed by other than a nucleophilic base. Suitable base protective groups removable by a strong non-nucleophilic base via a β-elimination mechanism in¬ clude: 2-(4-nitrophenyl)ethoxy carbonyl or 2- (phenyl sulfonyl)ethoxycarbonyl for both the N4 of C and the N6 of A; and the 9-fluorenyl methoxycarbonyl for the N2 of G and the N2 and N6 of 2, 6-diaminopurine. These groups can be removed after polymer assembly by treatment with the strong nonnucleophilic base 1,8-diazabicyclo[5.4.0]- undec-7-ene (DBU) , under stringently anhydrous condi¬ tions.
The syntheses of representative morpholino subunits follow here and are described in detail in Examples 2-10. With reference to the synthesis scheme depicted in Figure 14
5, a base-protected ribonucleoside is reacted with sodium periodate to form a transient 2', 3'-dialdehyde which then closes upon ammonia to form a morpholino-ring having 2' and 3' hydroxyl groups (numbered as in the parent ribose, see Figure 1) . The compound is then treated with sodium cyanoborohydride to reduce the ring hydroxyl groups. The ring nitrogen is preferably protected by trityl derivatization or by a benzhydraloxycarbonyl group for subsequent subunit coupling. The protective group can be added by reacting the morpholino subunit with trityl chloride or with nitrophenyl benzhydryl carbonate or by reacting the dialdehyde with a primary amine, as illustrated in Figure 6 and described in Example 3. The stereochemistry of the nucleoside starting material is retained as long as the pH of the reaction mixture at the iminium stage is not allowed to go above about 10.
The above synthesis results in a morpholino-ring with an available 5'-hydroxyl. The 5'-hydroxl can be converted to other active groups including 5' amine (Example 5) and 5'-sulfonate (Example 6).
In the above morpholino synthesis a variety of nitrogen sources can be used — including ammonia, ammo¬ nium hydroxide, ammonium carbonate, and ammonium bicar¬ bonate. Best results are obtained when the reaction solution is maintained near neutrality during the oxida¬ tion and morpholino ring closure reactions. This can be accomplished by continually titrating the reaction mix or, more conveniently, by using ammonium biborate as the ammonia source. When the solution is too acidic the yield of product is low and when it is too basic, side products (possibly due to epimerization of the 1' and/or 4' carbons) are produced which are difficult to separate from the desired product. It is also noted that the reducing agent can be added before, during, or after the 15 oxidation step with little noticeable effect on product yield.
Ribonucleosides lacking base protection groups can also be successfully oxidized, ring closed, and reduced in aqueous solution to generate the morpholino ring. However, without base protection the number and quantity of undesired side products frequently increases, par¬ ticularly in the case of cytidine.
The subunits formed by the above methods contain a 5'-OH, SH, or amine which is modified, reacted with, and/or activated, to be suitable for coupling to a second morpholino subunit (see below) . For example, Figure 5 shows the conversion of a 5'-OH of a morpholino subunit to a sulfonyl linking moiety to form a subunit (Structure 10) which is linked to form a 5-atom unit-length backbone polymer. Details of the subunit synthesis are given in Example 6.
Alternatively, the subunits are designed to include a sulfonyl or carbonyl group attached directly or in- directly to the morpholino ring nitrogen, which is cou¬ pled to a 5' moiety of a second morpholino subunit (Fi¬ gures 12 and 13) . Subunits of this type are suitable for constructing morpholino polymers with 6-atom (Figure 12) or 7-atom (Figure 13) repeating-unit backbones. An example of the synthesis of a subunit suitable for 7-atom unit-length backbones having an amine at the 5'-carbon atom, and a sulfonyl group linked to the ring nitrogen through a methylene group is detailed in Example 9 (with reference to Figure 7) . A similar synthesis, described in Example 10, is used to prepare morpholino subunits having a 5'-linked primary amine and an acetyl group linked to the ring nitrogen. This subunit is formed by coupling glycine, rather than AMSA, to the 5' ribonucleoside aldehyde 16 group. Examples 11 and 12 describe, with reference to Structure G of Figure 3, the preparation of non-morpho- lino subunits which are converted into morpholino struc¬ tures during polymer assembly.
C. Activation and Coupling Reactions
The subunits prepared as above are coupled, in a controlled, sequential manner, generally by activating the carbonyl or sulfonyl group on one subunit (having protected nitrogen groups) and contacting this activated subunit with another subunit having an unprotected nitro¬ gen. Different types of linkages, such as those illust¬ rated below, may be employed in the construction of a single polymer. A number of closely related variations are possible for the carbonyl-containing linkages giving six-atom backbones, corresponding to Structure B-B in Figure 4. A typical activation and coupling reaction for forming a carbamate linkage (where X is 0 in Structure B-B) is illustrated in Figure 9. Here a base-protected morpho¬ lino subunit with a 5'-OH is reacted with bis-(p-nitro- phenyl) carbonate and triethylamine to yield an activated carbonyl subunit (Structure 2, Figure 9) . This activated subunit is then combined with a second base-protected morpholino subunit which may be blocked at the 5'-OH. Bond formation between the subunits occurs between the annular nitrogen on the morpholino ring of subunit 2 and electrophilic carbonyl group of the first subunit, to form a carbamate linkage, where the carbonyl group is C=0. Details of the coupling reaction are given in Example 13.
Activation of the 5'-OH morpholino subunit with p- nitrophenylchlorothioformate and coupling to a second subunit with an unprotected ring nitrogen yields a thio- 17 carbamate linkage (where Y is S in structure B-B of Figure 4) .
The simplest and most obvious morpholino-type bind¬ ing polymers are the carbamate-linked polymers (type B-B of Figure 4) where X is oxygen. The polymer has been found to effectively bind to its single-stranded DNA target sequence. However, in binding studies with an RNA target, the polymer exhibited unusual binding, as evi¬ denced by a highly atypical hypochromicity profile in the 320 to 230 nm spectral range and lack of a normal thermal denaturation.
Modeling studies conducted in support of the ap¬ plication indicate that in a carbamate-linked polymer bound to DNA existing in a B conformation the backbone of the polymer provides adequate length for binding and the carbamate moieties of the polymer backbone can assume a nearly planar conformation. This modeling result was in good accord with the effective binding of the carbamate- linked polymers to DNA. In contrast, similar modeling studies suggested that binding of the carbamate-linked polymer to an RNA target requires one of the following: (i) the carbamate linkage of the polymer adopt a substan¬ tially nonplanar conformation, or (ii) the RNA target sequence adopt a strained conformation in which base- stacking interactions are quite different from that in a normal A conformation. This observation may explain the atypical binding of a carbamate-linked polymer to an RNA target sequence.
The modeling work further indicated that replacing the carbonyl intersubunit linking moiety with either an achiral sulfonyl-containing intersubunit linkage or with a chiral phosphorous-containing linkage would provide added length of about 0.32 angstrom per intersubunit linkage. This sulfonyl linkage also provides greater 18 rotational freedom about key bonds, and bond angles of the intersubunit linkage compatible with an oligomer backbone conformation suitable for pairing to both RNA and DNA target sequences in their standard conformations. Based on these findings, a number of syntheses of oligo¬ mer structures in which morpholino subunits are joined by sulfonyl moieties were subsequently developed and are described below (Structures A-A, C-C, D-D, and E-E of Figure 4) . The linkage in structure A-A in Figure 4 (five-atom backbone) can be formed according to the reaction scheme shown in Figure 11, and detailed in Example 14. Briefly, a 5'-OH morpholino subunit is protected at its ring nitrogen, converted to a 5'SH subunit, then oxidized to convert the 5'-linked sulhydral group to a sulfonyl group. The sulfonyl group is activated with phosgene, and coupled to a second subunit having an unprotected ring nitrogen, as shown. The polymer assembly is con¬ tinued by deprotecting the morpholino ring nitrogen of the dimer, and reacting the dimer with a third activated subunit.
The sulfamide linkage (corresponding to the linkage in structure C-C in Figure 4, where X is an amine) , is formed by sulfating the 5'-linked amine in a subunit having a protected morpholino ring nitrogen, and then activating with phosgene and reacting this subunit with a second subunit having an unprotected ring nitrogen, as illustrated in Figure 10. Details of the coupling reac¬ tion are given in Example 14. The sulfamate linkage (corresponding to the linkage in Structure C-C in Figure 4, wherein X is 0) is produced by sulfating the morpholino ring nitrogen of a 5' protec¬ ted subunit, then using phosgene to generate the sulf- amoyl chloride. This activated subunit is then mixed 19 with another subunit or oligomer having a free 5'OH. Coupling of the subunits is achieved either with a cata¬ lyst such as silver trifluoromethanesulfonate or use of a strong base to convert the 5' hydroxyl to the anionic form. Conversion of the 5' hydroxyl to the alkoxy can be achieved by KOH and a suitable phase transfer catalyst. This sulfamate coupling is illustrated in Figure 12 and details are given in Example 15.
A number of 7-atom unit length backbones prepared from the morpholino-subunits (corresponding to structures D-D through F-F in Figure 4) allow even more flexibility in the construction of polymers which have specified distances between the base-pairing moieties. Using the 7-atom unit length linkages, distances between the mor- pholino-subunits, and consequently between the base pairing moieties, can be lengthened. Such lengthening of the intersubunit linkage is particularly useful when targeting duplex genetic sequences in a B conformation.
The 7-atom backbone polymers can be readily syn- thesized from the subunits D-F constructed as above, employing the general coupling reactions described above.
For example, Structure D-D in Figure 4 can be produced by
(a) reacting the sulfonyl group of subunit D (Figure 3) with phosgene, and (b) coupling the activated subunit with a second subunit having an unprotected morpholino ring nitrogen.
Similarly, Structure E-E in Figure 4 can be produced by activating the sulfonyl group with phosgene, and coup¬ ling the activated subunit with a second subunit having an unprotected 5'-linked amine.
Structure F-F in Figure 4 can be produced by a similar synthetic method in which the carboxyl group is activated with carbonyldiimidazole or a carbodiimide, and the activated compound is reacted with a second subunit 20 having an unprotected 5'-linked primary amine.
A novel class of linkages corresponding to Structure G-G of Figure 4 can be produced by oxidizing vicinyl hydroxyls of one ribonucleoside subunit and closing the resultant dialdehyde on a primary amine of another sub- unit followed by reduction with cyanoborohydride. In principle this same scheme could also be used to couple a secondary amine of one subunit and a mono-aldehyde of a second subunit; however, the coupling of a ribose-derived dialdehyde to a primary amine proceeds substantially faster and provides a. better yield. Examples 11 and 12 describe the synthesis of ribonucleosides containing a primary amine at the 5' . Their use in formation of mor¬ pholino polymers is illustrated in Figure 14.
D. Assembly of Polymers
After selecting a desired polymer length and recog¬ nition moiety sequence (guidelines for this are presented below) , the polymer is assembled using the general proce- dures described above. One method of polymer assembly involves initial preparation of an appropriate set of di¬ mers, linking selected dimers to form tetramers, linking these to form octamers, and so on. This method is car¬ ried out in solution, substantially according to the coupling methods described with reference to Examples 13- 17. Example 18 outlines such a block assembly synthesis using monomers to form dimers, and dimers to form tetra¬ mers. The synthesis need not involve oligomers of equal size. A particular merit of this block assembly method is that each coupling product is roughly twice the length of its precursors, so purification of the product of each coupling is simplified. Example 18 details the assembly of a 4-subunit polymer formed by this method. 21
The polymers may also be synthesized by stepwise subunit addition on a solid support. However, the op¬ timal synthetic approach often uses a combination of the solution and solid support assembly methods where dimers, trimers, or tetramers are synthesized by solution phase and subsequently assembled into the full-length polymer on a solid support, as described in Example 19.
Typically, a solid support, such as glass beads derivatized with acid-stable, long-chain cleavable lin- kers, are employed as the support material, and prepared for attachment of the first subunit, or block of sub- units, as described in Example 19. The glass beads are reacted with a subunit which generally has a readily cleavable protective group on a nitrogen. Whether the morpholino subunit is linked to the support via its morpholino nitrogen or a group at the 5' position depends on the direction of polymer synthesis, i.e., to which group the next subunit will be attached.
After coupling the second subunit (or oligomer which may be assembled in solution) to the support, any un- reacted nucleophilic sites can be capped by addition of a suitable capping reagent, such as p-nitrophenyl acetate or acetic anhydride, and thereafter the support is washed. The protecting group on the nitrogen of the terminal subunit is removed, typically by acid treatment, and after neutralization, the support is reacted with an excess of the next-in- sequence subunit (or polymer unit) which is activated by one of the methods outlined above. One feature of the solid support assembly method is the need for high coupling efficiencies at each subunit addition step. This high coupling efficiency is general¬ ly achieved by addition of an excess of the activated subunit which maximizes the number of support-bound chains which are chain-elongated. 22
Chain elongation is continued in this manner, with optional capping of failure sequences after each subunit addition, until the polymer of the desired length and sequence is achieved. After addition of the final subunit, the terminal backbone moiety may be reacted with a suitable charged or uncharged group, as described in Example 19. The polymer is then cleaved from the support, e.g., by treatment with either ammonium hydroxide or a non-nucleophilic base suitable for effecting β-elimination in the linker join¬ ing the polymer to the. support. The bases are deprotec¬ ted and the polymer is purified as described below and in Example 19.
E. Polymer Processing and Purification
Binding polymers assembled in solution (Examples 18 and 20) are typically base-deprotected by suspending in DMSO or DMF and layering on the suspension an equal volume of concentrated ammonium hydroxide. The prepara- tion is mixed with shaking and incubated at 30°C for 16 hrs. Workup includes removing the ammonia under reduced pressure. If a protective group (generally trityl or a related acid-labile moiety) is present, this group is cleaved and the crude polymer preparation is suspended in the appropriate buffer for purification (Example 19) .
Binding polymers assembled by a solid-phase method (Example 19) wherein they are linked to the support via an ester linkage can be cleaved from the support by suspending the dried support in DMSO, layering on an equal volume of concentrated NH4OH, capping tightly, and slowly agitating for 16 hrs at 30°C. The solid support material is removed by filtration and the filtrate is treated as described above.
Alternatively, binding polymers linked to the sup- 23 port via a β-elimination-sensitive linker can be cleaved from the support using a strong non-nucleophilic base 1,8 diazabicyclo(5.4.0.)undec-7-ene (DBU) in DMF. Using this approach one can release the polymer with its bases still protected and thus the polymer is suitable for further modification and/or structural confirmation via fast atom bombardment mass spectroscopy.
Purification of the base-deprotected polymer is preferably carried out at pH 2.5 or pH 11, depending on the pKs of the base moieties in the polymer. At pH 2.5 cytosine, adenine, and 2-6-diaminopurine moieties carry a positive charge and guanine carries a partial positive charge. At pH 11 guanine, uracil and hypoxanthine carry a negative charge. For polymers in which about 50% or more of the base- pairing moieties are ionized at pH 2.5, the purification can be carried out by cation ex¬ change on a column of S-Sepharose fast-flow (Pharmacia) developed with a shallow NaCl gradient buffered at pH 2.5. The effluent is monitored at 254 nm and collected in a fraction collector. The full length polymer, which elutes after the shorter failure sequences, can be fur¬ ther purified and desalted on a column of chromatogra¬ phic grade polypropylene (Polysciences Inc.), eluted with an aqueous gradient of acetonitrile adjusted to pH 2.5 with formic acid, with the eluant being monitored at 254 nm. The fractions containing the pure product are neutralized and dried under reduced pressure. Salts may be discarded by dissolving the polymer in trifluoroe- thanol, filtering, and evaporating the trifluoroethanol. For polymers in which about 50% or more of the base- pairing moieties are ionized at pH 11, the purification may be performed on an anion exchange column of Q Sepha- rose fast-flow (Pharmacia) developed with an aqueous pH 11 gradient of NaCl. The full-length polymer, which 24 elutes after shorter failure sequences, is further puri¬ fied and desalted on a polypropylene column eluted with an aqueous pH 11 gradient of acetonitrile. Fractions containing the pure product are processed as above. The purification methods described above should be carried out so that polymers containing adenine base-pairing moieties are not exposed to pH 11 for more than a few hours at room temperature, to avoid potential base lability problems. The details of the purification methods are outlined in Example 19.
In neutral, aqueous solution, longer morpholino polymers may have solubilities only in the sub-micromolar range. Therefore, it may be advantageous to enhance polymer solubility by addition of one or more hydrophilic moieties, e.g., polyethylene glycol. For most of the polymer types disclosed herein, this can be accomplished by cleaving the terminal backbone protective group from the completed polymer, and reacting the polymer, with the bases still in the protected state, with excess of car- bonyldiimidazole-activated polyethylene glycol (PEG) . Thereafter the binding polymer is treated with ammonium hydroxide to remove the base-protected groups, and the polymer is purified as above. The level of solubiliza- tion is easily adjusted through proper selection of the PEG material. Suitable PEG fractions having average molecular weights of 200, 400, 600, 1,000, 1,540, 3,400, 4,000, 6,000, 7,500, and 18,500 daltons are commercially available (e.g., Polysciences, Inc.) with PEG1000 often providing the best solubilization. The solubilizing moiety may be linked to the polymer through a cleavable linkage, if desired, to allow the polymer to be released from the solubilizing agent, e.g., by esterase or pep- tidase enzymes. 25
It will be appreciated that the polymer may be further derivatized or labeled according to known proce¬ dures. For example, the polymer may be radiolabeled by preparing the polymer subunits from radiolabeled ribonu- cleosides or by attaching a radiolabeled amino acid at one terminus. The polymer may be readily derivatized, e.g., employing modifications of the above subunit cou¬ pling reactions, with enzymes, chromophoric groups, or the like, where the polymer is to be used as a diagnostic probe. Further, the polymer may be derivatized with biomolecules which serve to target the polymers to speci¬ fic tissues or cell types.
F. Structural Characterization Fully-protected binding polymers of moderate size (10 to 20 subunits) often give a strong molecular ion in FAB (Fast Atom Bombardment) mass spectroscopy, providing a key confirmation of the polymer length.
Further, COSY-NMR (two-dimensional correlated spec- troscopy) of the deprotected and purified polymer pro¬ vides information on the ratio of the different base-pairing moieties in the polymer as well as quantita¬ tive information on the ratio of binding polymer to any solubilizing or other type moiety which may have been linked thereto.
Mobilities on ion exchange columns also provide information on the number of C + A base-pairing moieties in a polymer when purification is carried out at pH 2.5 and information on the number of G + U residues when the purification is run at pH 11. Structural verification is easiest when the polymers have been assembled from oligo- mer blocks, such as in Examples 18, 19 and 20, since any failure sequences then differ more substantially from the full-length sequences. 26
The UV profiles of the polymers at pH 1, 7, and 13 can provide information about the relative nucleotide composition of the polymer.
Assessment of a morpholino-based polymer's affinity for its target sequence is carried out by examining the melting curve of the polymer/target duplex, as illustra¬ ted in Examples 20 and 21.
Further, comparisons can be made between the melting curve of a regular nucleic acid duplex (such as p(dC) 6/p(dG)6) and the melting curve of a hybrid duplex containing a corresponding morpholino-based polymer (such as (morpholino-based C)6/p(dG)6). Characterization of the synthetic intermediates and the full-length oligomer was achieved by proton NMR and negative ion FAB mass spec- troscopy. With these carbamates of the morpholino oligo¬ mers, the fragmentation of the oligomers is greatly suppressed so that little sequence information is avail¬ able. However, the parent ion signal is quite strong and allows confirmation of the composition of the morpholino oligomer (see Example 20) . High resolution mass spectro- metry of the morpholino-based poly C hexamer provided a satisfactory elemental analysis.
Several features of the proton spectrum of the oligomers were of interest. For example, the length of the oligomers could be ascertained by comparing the integration of the various signals. For example, in a dimer the two 2' protons were separated by 0.5 ppm and gave a 1.02/1 ratio of integrations and for the hexamer, this ratio was 4.65/1 against the expected value of 5/1. The bases of the above hexamer were deprotected by treatment with concentrated ammonia for 24 hours. The 4'- terminal morpholino ring nitrogen was liberated by treat¬ ment of the crude oligomer with 1% formic acid in tri- fluoroethanol. In order to assess the stability of these 27 molecules under these conditions, a precursor dimer was treated with concentrated ammonia for 60 hours; no clea¬ vage of the intersubunit linkage was observed. Under all conditions used to date, no cleavage of the carbamate linkage under acidic conditions has occurred.
The hexamer was taken up in pH 2.5 buffer and puri¬ fied by cation exchange chromatography on S-Sepharose Fast Flow™, eluting with potassium chloride gradients. The chromatograms showed one major peak comprising over 95% of the cytosine- containing materials in the mixture, and confirming that little or no cleavage of the oligomer occurs in the deprotection of the bases and the morpho- lino-amine. After neutralization the hexamer was desalted on a polypropylene column eluted with a water-acetonit- rile gradient.
The purified hexamer was analyzed by **H NMR. The assignment for the protons was made on the basis of a COSY plot. One cytosine base has signals that were found downfield relative to the other bases (8.04 to 7.65 and 6.78 to 5.85 ppm). The relative integrations of these peaks confirmed that the hexamer was deprotected and has been purified intact. The 5' protons were assigned to the signal at 4.35-4.15 downfield of the 1' proton signal at 4.14-3.90 ppm. These chemical shifts run against the trend identified in the protected oligomers where the 1' proton of the same base(s) was always downfield of the 5' protons of the same base(s). Apparently the benzoyl groups in the protected oligomers play a role in shaping the environment of the 1' and 5' protons. The solubility of the hexamer was found to be 4 μM in pH 7.5 buffer. In order to increase water solubility of the hexamer a polyethylene glycol (PEG) tail was attached to the oligomers. 5 equivalents of PEG 1000 was treated with one equivalent of bis (p-nitrophenyl) carbo- 28 nate to give monoactivated PEG. Detritylation of the hexamer with 1% formic acid in trifluoroethanol afforded a free amine. Treatment of the hexamer containing the free amine with activated PEG1000 under standard coupling conditions resulted in attachment of the PEG tail to the hexamer. The bases were deprotected by treatment of the tailed hexamer with concentrated ammonia for 24 hours. The tailed hexamer was taken up in pH 2.5 buffer and purified by cation exchange chromatography on S-Sepharose Fast Flow™ eluted with a potassium chloride gradient. After neutralization the eluant was desalted on a poly¬ propylene column eluted with a water/acetonitrile gradi¬ ent. The tailed hexamer was found to be freely soluble in pH 7.5 buffer in concentrations up to 2 mM. The characterization of the tailed hexamer by the 1H NMR methods employed above was not possible. In the spectrum of the tailed hexamer there was no differentia¬ tion between the signals of the base protons, thus pre¬ cluding the assessment of the oligomer length. Addition- ally, the envelope containing the PEG tail signals ob¬ scured the majority of the signals of the morpholino rings. However, the ion exchange chromatography of the tailed hexamer gave one major peak indicating little or no cleavage of the oligomer during deprotection. The pattern of the chromatogram of the tailed hexamer was the same as found for the free hexamer, except that the tailed hexamer elutes faster than the free hexamer.
The stability of complexes of the tailed hexamer with complementary nucleic acids was investigated by thermal denaturation experiments. Difference spectra between mixed and unmixed samples of the tailed hexamer and the selected phosphodiester complement were obtained from 14°C to 85°C and over the 320 to 260 nm range (see Example 20). As a control, the duplex of p)dC)6 with 29 p(dG)6 was thermally denatured. The difference UV spectrum of the tailed hexamer (morphC)6 with p(dG)6 was similar to that of the control DNA duplex, p(dC)6 with p(dG)6, except that the amount of hypochromicity before denaturation of the (morphC)6/p(dG)6 duplex was much greater than that of the control. The thermal denatura¬ tion of the p(morphC)6/p(dG) 6 duplex gave a Tm value of 62.5°C (see Example 20 and Figure 15). The corresponding DNA/DNA duplex gave a Tm value of 26.5°C.
G. Diagnostic Applications
The target-specific polymers of the invention can be used in a variety of diagnostic assays for detection of RNA or DNA having a given target sequence. In one gene- ral application, the polymers are labeled with a suitable radiolabel or other detectable reporter group. Target polynucleotide, typically a single stranded polynucleo¬ tide which is bound to a solid support, is reacted with the polymer under hybridization conditions, allowed to anneal, and then the sample is examined for the presence of polymer reporter group.
The diagnostic assay can be carried out according to standard procedures, with suitable adjustment of the hybridization conditions to allow polymer hybridization with the target region. In this regard, it is noted that the polymer can be designed for hybridization with the target at a higher melting temperature than the comple¬ mentary polynucleotide strand, since polymer binding does not entail backbone charge repulsion effects. Therefore, the polymer can bind to the target at a temperature above the normal polynucleotide melting temperature, an impor¬ tant advantage of the polymer over conventional oligo¬ nucleotide probes. This binding at elevated temperature minimizes the problem of competition for binding to the 30 target between the probe and any corresponding single- strand oligonucleotide which may be present in the diag¬ nostic mixture.
In a second general type of diagnostic application, the polymers are linked to a solid support, for capture of target RNA or DNA to the support. The solid support, e.g., polymeric microparticles, can be prepared by link¬ ing the polymers to the support according to the methods described above or by conventional derivatization proce- dures. Alternatively, where the polymers are synthesized on a solid support this support may also serve as the assay support.
According to an important feature of this assay system, the target polynucleotide molecules which are captured on the support by base-specific binding to the polymers can be detected on the basis of their backbone charge, since the support-bound polymers are themselves substantially uncharged. To this end, the assay system may also include polycationic reporter molecules which are designed to bind to the fully charged analyte back¬ bone, but not the uncharged (or substantially uncharged) polymer backbone, under selected binding conditions.
In one embodiment the reporter molecules are com¬ posed of a polycationic moiety or tail designed to bind electrostatically to a fully charged polynucleotide, under conditions where the reporter does not bind to the less charged or uncharged binding polymer carried on the diagnostic reagent; one or more reporter groups may be attached to the tail, adapted to produce a signal by which the presence of the reporter can be detected. Methods for forming polycationic molecules and for at¬ taching reporter molecules to cationic compounds are known in the art. 31
Each reporter molecule carries one or more reporter groups, and each polynucleotide can accommodate binding of typically several thousand or more reporter molecules. Thus the system has an amplification factor, in terms of reporter signal per bound analyte molecule, of several orders of magnitude. In addition, the method has the advantage, noted above, that the polynucleotide binding reaction can be carried out under conditions in which binding competition with complementary nucleotide strands does not occur.
The design considerations applied in preparing a polynucleotide binding polymer for use as a diagnostic reagent are governed by the nature of the target analyte and the reaction conditions under which the analyte is to be assayed. As a first consideration, there is selected a non-homopolymeric target base sequence against which the polymer is directed. This target sequence is gener¬ ally single-stranded and preferably unique to the ana¬ lyte being assayed. The probability of occurrence of a given n-base target sequence is approximately (1/4)n. Accordingly, a given n-base target sequence would be expected to occur approximately once in a polymer containing 4n bases. Therefore, the probability P that a given n-base sequence will occur in polynucleotides containing a total of N unique-sequence bases is approximately P=N/4n. To illu¬ strate, the probability P that a 9-base target sequence will be found in a 20 kilobase polynucleotide is about 20xl03/2xl05 or 0.08, the probability that a 16-base target sequence will be present is about 20xl03/4.3xl09 or 0.0000047. From these calculations, it can be seen that a polymer having 9-16 recognition moieties specific for a defined 9-16 base target sequence should have high speci¬ ficity for the target sequence in an assay mixture con- 32 taining only viral genomes, whose greatest complexities correspond to about 400K of unique-sequence bases.
Similar calculations show that a 12 to 16 subunit polymer can provide adequate specificity for a viral or bacterial target sequence in an assay mixture containing viral and bacterial genomic material only; largest geno- mic sizes about 5,000 kilobases. A 16 to 22 subunit polymer can provide adequate specificity for a target sequence in a polynucleotide mixture containing mammalian genomic DNA material; genomic sizes of about 5 billion base pairs of unique-sequence DNA.
The polymer/analyte binding affinity, and particu¬ larly the temperature at which the polymer just binds with the target sequence (the melting temperature, or Tm) can be selectively varied according to the following criteria: (a) number of subunits in the polymer; (b) the number of hydrogen bonds that can be formed between the base-pairing moieties and the corresponding, complemen¬ tary bases of the analyte target sequence; (c) unit length of the polymer backbone; (d) the particular inter¬ subunit linkages; and (e) concentration of denaturants, such as formamide, which reduces the temperature of melting.
From a number of studies on model nucleic acid duplexes it is known that the melting temperature of oligonucleotide duplexes in the 10 to 20 bp range in¬ creases roughly 3°C per additional base pair formed by two hydrogen bonds, and about 6°C per additional base pair formed by three hydrogen bonds. Therefore, the target sequence length originally selected to insure high binding specificity with the polymer may be extended to achieve a desired melting temperature under selected assay conditions.
Also, where the recognition moieties used in con- 33 structing the polymer are the standard nucleic acid bases the target sequence may be selected to have a high per¬ centage of guanine plus cytosine bases to achieve a relatively high polymer/analyte melting temperature. On the other hand to achieve a lower melting temperature a target sequence is selected which contains a relatively high percentage of adenine plus thymine bases.
The binding components in the diagnostic system, as they function in the solid-support diagnostic method just described, are illustrated in Figure 16. Here "S", the assay reagent, is the. solid support having a number of binding polymers attached to its surface through spacer arms indicated by sawtooth lines. In the assay proce¬ dure, the target DNA in single strand form is reacted with the support-bound polymers under hybridization conditions, and the solid support is then washed to remove non-hybridized nucleic acid material.
The washed support is then reacted with the repor¬ ter, under conditions which favor electrostatic binding of the reporter cationic moiety to the target DNA back¬ bone. The reporter shown in Figure 16 is a dicationic molecule having a reporter group R.
After reaction with the reporter solution, typically at room temperature for 1-2 minutes, the reagent is washed to remove unbound reporter, and then the assay reagent is assessed for bound reporter. One approach in determining the amount of reporter associated with the reagent, particularly in the case of fluorescent or chromophoric reporter groups, is to elute the reporter from the reagent with a high salt solution and then assess the eluate for reporterhe polymer of the invention can undergo sequence-specific binding to duplex nucleic acids via base-pair-specific hydrogen bonding sites which are accessible through the major groove of 34 the double helix. This bonding can occur in a duplex region in which at least 70% of the bases on one strand are purines and a corresponding percent of the bases on the other strand are pyrimidines. The duplex binding polymer preferably includes 2-aminopurine or 2, 6-diamino¬ purine hydrogen bonding moieties for binding to T-A or U- A base pairs, and guanine or thioguanine hydrogen-bonding moieties for binding to C-G base pairs as illustrated in Figure 8A. Thus, for these special target sequences (an example of which is shown in Figure 8B) , the polymer of the invention can be .used for diagnostic assays of the types just described, but where the target nucleic acid is in nondenatured, duplex form.
H. Other Applications
The polymers of the instant invention can be used in place of standard RNA or DNA oligomers for a number of standard laboratory procedures. As mentioned above, mor¬ pholino-based polymers can be fixed to a solid support and used to isolate complementary nucleic acid sequences, for example, purification of a specific mRNA from a poly- A fraction (Goldberg et al) . The instant polymers are advantageous for such applications since they are inex¬ pensive and straightforward to prepare from activated subunits.
A large number of applications in molecular biology can be found for labeled morpholino-based polymers. Morpholino-based polymers can be easily and efficiently end-labelled by the inclusion in the last step of the polymer synthesis an activated and labelled morpholino- based subunit or, preferably, an 35S-labelled methionine. The type of label to be used is dependent on the final application of the polymer, and includes radioactive (3H, 1C, 32P, or 35S) nucleosides and biotin. Labelled mor- 35 pholino-based oligonucleotide analogs can act as efficient probes in, for example, colony hybridization
(Grunstein et al) , RNA hybridizations (Thomas) , DNA hybridizations (Southern) , and gene bank screening (Szostak et al) .
The polymers of the invention also have important potential use as therapeutic agents. Recently, uncharged anti-sense nucleic acid analogs, which are nearly iso- structural with DNA, have been used as anti-viral and anti-tumor agents. The polymers of the present invention provide several advantages over the more conventional anti-sense agents.
First, the morpholino polymers are substantially less expensive to synthesize than oligonucleotides. This is due in part to the fact that the morpholino subunits used in polymer synthesis are derived from ribonucleo- sides, rather than the much more expensive deoxyribonu- cleosides. Also, as noted above, the coupling reaction between a phosphorous and an amine of a second subunit occurs under relatively mild conditions, so that protec¬ tion steps and other precautions needed to avoid unwanted reactions are simplified. This is in contrast to stan¬ dard ribo- and deoxyribonucleotide polymer synthesis where coupling through a phosphate ester linkage requires that the coupling reagents be highly reactive and that the reaction be carried out under stringent reaction/pro¬ tection conditions. This advantage in polymer synthesis also applies, of course, to diagnostic uses of the poly¬ mer. Second, polymer binding to its target may give sub¬ stantially better target inactivation, since the polymer- /target duplex is not susceptible to duplex unwinding mechanisms in the cell. 36
Third, the morpholino-based polymer is also more stable within the cell; the polymer backbone linkage is not susceptible to degradation by cellular nucleases.
Fourth, in therapeutic applications involving cel- lular uptake of the compound, the uncharged morpholino polymer is more likely to efficiently enter cells than a charged oligonucleotide.
In the context of therapeutic applications, the morpholino polymers of the present invention may be targeted against double-stranded genetic sequences in which one strand contains predominantly purines and the other strand contains predominantly pyrimidines (e.g., Figure 8B) .
Further, when a messenger RNA is coded by the mostly purine strand of the duplex target sequence, morpholino binding polymers targeted to the duplex have potential for also inactivating the mRNA. Thus such a polymer has the potential for inactivating key genetic sequences of a pathogen in both single-stranded and double-stranded forms.
In 1981 it was reported that short (3 to 7 subunits) methylphosphonate-linked DNA analogs complementary to portions of the Shine-Dalgarano consensus sequence of procaryotic mRNAs were effective in disrupting bacterial protein synthesis in bacterial lysates and in a special permeable strain of bacteria. However, such agents failed to inhibit protein synthesis in normal bacteria (Jayaramon, 1981) .
Experiments performed in support of the instant invention show that polymers of 3 to 5 subunits in length can be effective to block protein synthesis in normal bacteria by using a combination of bases which result in a high target-binding affinity. More specifically, the following oligomers and oligomer combinations can perturb 37 protein synthesis in normal intact bacteria (where D is 2,6-Diaminopurine or adenine; G is Guanine; B is 5-Bromo- uracil, other 5-Halouracil or uracil; sequences are shown with their 5' end to the left) : DGG, BDDG, DDGG; DGGD; GGDG; GDGG; DGGB; GGBG; GGAGG; GGDGG; and the combinations BDD + GGDG; DDG + GDGG; DGG + DGGB; GGD + GGBG; BDDG + GDG; DDGG + DGG; DGGD + GGB GGDG + GBG; BDD + GGDG + GBG.
While other backbone types may be suitable for such binding-enhanced short oligomers (e.g., carbamate-linked deoxyribonucleosides; . Stirchak, 1987) , the morpholino type oligomers of the present invention are preferred on the basis of starting material costs and ease of assem¬ bly. The use of short binding-enhanced oligomers to disrupt the biological activity of an RNA sequence which plays a key role in the metabolism of a target class of organisms but not a correspondingly important role in higher organisms should be broadly adaptable to a variety of pathogenic organisms (e.g., bacteria and fungi) having a cell wall which excludes the entrance of longer poly¬ mers.
The following examples illustrate methods of subunit and polymer synthesis, and uses of the polymer composi- tion of the invention. The examples are in no way in¬ tended to limit the scope of the invention.
Example 1
Base Protection of Ribonucleosides The following ribonucleosides are obtained from
Sigma Chemical Co. (St. Louis, MO) : uridine, guanosine,
5-methyluridine, adenosine, cytidine, 5-bromouridine, and inosine. 38
2, 6-diamino-9- (B-D-ribofuranosyl) -9H-purine (2, 6-di¬ aminopurine riboside) is obtained from Pfaltz and Bauer, Inc., Division of Aceto Chemical Co., Inc. (Waterbury, CT) . The following nucleosides are prepared by the literature methods indicated:
1-β-D-ribofuranosyl)-2-pyrimidinone (2-hydroxy-pyri- midine riboside) is prepared by ..the procedure of Niedballa.
2-amino-9-β-D-ribofuranosyl) -1, 6-dihydro-6hpurine-6 -thione (thioguanosine) is prepared by the procedure of Fox. Dimethoxytrityl chloride, N-6-benzoyladeno- sine, N-4-benzoylcytidine, and N-2-benzoylguanosine are obtained from Sigma Chemicals. 9-fluorenylmethoxycar- bonyl chloride (FMOC chloride) , trimethylchlorosilane, isobutyric anhydride, 4-nitrobenzoyl chloride, naphthalic anhydride, and all organic solvents for reactions and chromatography were obtained from Aldrich Chemical Co. (Milwaukee, Wl) . Silica Gel is obtained from EM Science (Cherry Hill, NJ) . When activation of the subunits is achieved using dihalogenated electrophiles (eg. C0C12 or S02C1F) , better yields of activated subunits are often obtained by using protective groups which leave no acidic protons on the purine and pyrimidine exocyclic amines. Examples of such exocyclic amine moieties are as follows: the N6 of adenine, the N4 of cytosine, the N2 of guanine, and the N2 and N6 of diaminopurine. Suitable protective groups for this purpose include the naphthaloyl group (Dickshit) and the aminidine groups developed by McBride et al (1986) . In addition, use of dihalogenated electrophiles for subunit activation generally requires that the 06 of guanine moieties is protected; this protection is achieved using the diphenylcarboamoyl group
(Trichtinger) . Chem Soc 80:6204 (1958). 39
Guanosine
In order to minimize side reactions during subunit activations it is often desirable to protect the guanine moiety on both the N2 and 06 using the procedure of Trichtinger et al (1983) . The N-2 9-fluorenylmethoxy- carbonyl derivative of guanosine is prepared by the procedure below which is general for the protection of nucleoside amino groups: guanosine (1 mmol) is suspended in pyridine (5 ml) and treated with tri'methyl- chlorosilane (5 mmol) .. After the solution is stirred for 15 minutes, 9-fluorenylmethoxycarbonyl chloride (5 mmol) is added and the solution is maintained at room temperature for 3 hours. The reaction is cooled in an ice bath and water (1 ml) is added. After stirring for 5 minutes cone, ammonia (1 ml) is added, and the reaction is stirred for 15 minutes. The solution is evaporated to near dryness and the residue is dissolved in chloroform (10 ml) . This solution is washed with sodium bicarbonate solution (5 ml, 10%) , dried over sodium sulfate and evaporated. The residue is coevaporated several times with toluene and the product chromatographed on silica gel using a gradient of methanol in methylene chloride (0-50%) . N-2-Isobutyrylguanosine is prepared by the method of Letsinger.
N-2-acetylguanosine is obtained by the method of Reese. N-2-naphthaylguanosine is prepared by the method of Dikshit; this reference provides a general method for the protection of nucleoside amine groups.
Adenosine
The N-6 2-(4-nitrophenyl) -ethoxycarbonyl derivative is prepared by the method of Himmelsbach. 40
N-6 (4-nitrobenzoyl)adenosine is prepared using the procedure above for FMOC-guanosine except that 4-nitro¬ benzoyl chloride is substituted for FMOC chloride.
The N-β 2-(phenylsulfonyl) -ethoxycarbonyl derivative is prepared by the procedure for FMOC guanosine except the 2- (phenylsulfonyl) -ethyl chloroformate (Balgobin) is used as the acylating agent and N-methylimidazole or pyridine is used as the solvent.
N-6 naphthoyladenosine is prepared by the method of Dikshit; this reference provides a general method for the protection of nucleoside amine groups.
2, -diaminopurineriboside
The N-2,N-6-bis (9-fluorenylmethoxycarbonyl) derivative of 2,6-diaminopurine riboside is prepared by the general procedure described for guanosine.
The N-2,N-6-bis (isobutyryl) derivative is prepared by the general procedure described for guanosine.
Thioguanosine
The N-2 (9-fluorenylmethoxycarbonyl) derivative of thioguanosine is prepared by the general procedure described for guanosine.
Uridine
To minimize undesired side products during the subunit activation step it is sometimes desirable to protect the N3 of the uracil moiety. 5'0-tritylated uridine-2' ,3'-acetonide is converted to the N3 anisoyl derivative by the procedure of Kamimura et al (1983) . The product is then treated with hot 80% acetic acid or 0.1 N HC1 in THF to cleave the protective groups on the ribose moiety. 41
Example 2 Synthesis of 5'-OH Morpholino Subunits The steps in the method are illustrated in Figure 5, with reference to structures shown in Figure 5. The base-protected ribonucleoside is oxidized with periodate to a 2'-3' dialdehyde (Structure 1). The dialdehyde is closed on ammonia or primary amine (Structure 2) and the 2' and 3' hydroxyls (numbered as in the parent ribose) are removed by reduction with cyanoborohydride (Structure 3) .
An example of this general synthetic scheme is de¬ scribed below with reference to the synthesis of a base- protected cytosine (Pi*) morpholino subunit. To 1.6 1 of methanol is added, with stirring, 0.1 mole of N-4- benzoylcytidine and 0.105 mole sodium periodate dissolved in 100 ml of water. After 5 minutes, 0.12 mole of ammonium biborate is added, and the mixture is stirred 1 hour at room temperature, chilled and filtered. To the filtrate is added 0.12 mole sodium cyanoborohydride. After 10 minutes, 0.20 mole of toluenesulfonic acid is added. After another 30 minutes, another 0.20 mole of toluenesulfonic acid is added and the mixture is chilled and filtered. The solid precipitate is washed with two 500 ml portions of water and dried under vacuum to give the tosylate salt of the free amine shown in Structure 3. The use of a moderately strong (pKa < 3) aromatic acid, such as toluenesulfonic acid or 2- naphthalenesulfonic acid, provides ease of handling, significantly improved yields, and a high level of product purity.
The base-protected morpholino subunit is then pro¬ tected at the annular nitrogen of the morpholino ring using trityl chloride or benzyhydral nitrophenyl carbonate (Structure 4) . Alternatively, the 5' hydroxyl 42 can be protected with a trialkylsilyl group.
As an example of a protection step, to 2 liters of acetonitrile is added, with stirring, 0.1 mole of the tosylate salt from above followed by 0.26 mole of tri- ethylamine and 0.15 mole of trityl chloride. The mixture is covered and stirred for 1 hour at room temperature after which 100 ml methanol is added, followed by stir¬ ring for 15 minutes. After drying by rotovaping, 400 ml of methanol is added. After the solid is thoroughly sus- pended as a slurry, 5 liters of water is added, the mixture is stirred for 30 minutes and filtered. The solid is washed with 1 liter of water, filtered, and dried under vacuum. The solid is resuspended in 500 ml of dichloromethane, filtered, and rotovaped until precipitation just begins, after which 1 liter of hexane is added and stirred for 15 minutes. The solid is removed by filtering, and dried under vacuum.
The above procedure yields the base-protected morpholino subunit tritylated on the morpholino nitrogen and having a free 5' hydroxyl.
Example 3 Alternative Synthesis of Morpholino Subunits This example describes an alternative preparation of a morpholino subunit containing an acid-labile moiety linked to the morpholino ring nitrogen. The steps are described with respect to Figure 6.
The subunit is prepared by oxidizing a ribonucleoside with periodate, as in Example 2, and closing the resultant dialdehyde (Structure 1) on the primary amine 4,4'-dimethoxybenzhydrylamine (which can be prepared by the method of Greenlee, 1984) buffered with benzotriazole, or p-nitrophenol. Reduction with sodium cyanoborohydride, carried out as in Example 2, gives a 43 morpholino subunit (Structure 2) having a 4,4'- dimethoxybenzhydryl group on the morpholino nitrogen.
This procedure is particularly useful for preparing morpholino subunits from ribonucleosides which do not have a protective group on the base (e.g., uridine) .
Example 4
N-Sulfation of Morpholino Subunit This example describes the preparation of a morpholino subunit protected on its 5' oxygen and sulfated on its morpholino ring nitrogen. The steps are described with reference to Figure 12.
Structure 3 of Figure 5 is silylated with t-butyldi- methlsilyl chloride to give Structure 1 of Figure 12.
This product is then treated with S03/pyridine complex
(with excess pyridine) in dimethylformamide (DMF) to give
Structure 2 of Figure 12.
It should be mentioned that the salts of sulfamic acids (e.g., Structure 7 of Figure 5, and Structure 2 of Figure 12) and the salts of sulfonic acids (e.g., Structure 10 of Figure 5, and Structure 5 of Figure 7) can be easily chromatographed on silica gel using triethylamine/methanol/chloroform mixtures if the silica is first pre-eluted with 2% triethylamine in chloroform.
Example 5 Synthesis of 5'-Sulfamic Acid Morpholino Subunits The steps in the synthesis of 5'-sulfamic acid mor- pholino subunits are described with reference to structures shown in Figure 5.
The 5'hydroxyl of the doubly-protected morpholino subunit (Structure 4, Figure 5) can be converted to the amine as follows. To 500 ml of DMSO is added 1.0 mole of 44 pyridine (Pyr) , 0.5 mole of triflouroacetic acid (TFA) , and 0.1 mole of the morpholino subunit. The mixture is stirred until dissolved, and then 0.5 mole of diisopro- pylcarbodiimide (DIC) or dicyclohexylcarbodiimide (DCC) is added. After 2 hours the reaction mixture is added to 8 liters of rapidly stirred brine, which is stirred for 30 minutes and filtered. The solid is dried briefly, washed with 1 liter of ice cold hexanes, filtered, and the solid is added to 0.2 mole of sodium cyanoborohydride in 1 liter of methanol, stirred for 10 minutes, 0.4 mole of benzotriazole or p-nitrophenol is added, followed by 0.2 mole of methylamine (40% in H20) and the preparation is stirred four hours at room temperature [Note: the benzotriazole or p-nitrophenol buffers the reaction mixture to prevent racemization at the 4' carbon of the subunit at the iminium stage of the reductive alkyla- tion] . Finally, the reaction mixture is poured into 5 liters of water, stirred until a good precipitate forms, and the solid (Structure 6, Figure 5) is collected and dried. This dried product is next suspended in DMF and 4 equivalents of S03/pyridine complex is added. Over a period of several hours, 8 equivalents of triethylamine is added dropwise with stirring. After an additional two hours the preparation is dumped into a large volume of brine and the solid collected by filtration and dried.
This sulfamic acid preparation is then purified by silica gel chromatography.
Example 6 Synthesis of 5'-Sulfonate Morpholino Subunits
The steps in the synthesis of 5'-sulfonate morpholino subunits are described with reference to structures shown in Figure 5. 45
For the following synthesis the morpholino nitrogen should be protected as a carbamate (e.g., Structure 4 of Figure 5) instead of with a trityl group.
The 5' hydroxyl of the doubly-protected morpholino subunit is converted to a sulfhydral as follows. 0.1 mole of the 5'-hydroxyl subunit (Structure 4, Figure 5) is added to 1 liter of pyridine followed by 0.12 mole of toluenesulfonylchloride, and stirred for 3 hours at room temperature to give Structure 8 of Figure 5. After removing the pyridine by rotovapping, 0.5 mole of fresh sodium hydrosulfide in 1 liter of methanol/DMF containing Nal is added and the mixture is stirred at room temperature overnight. The reaction mix is added to 5 liters of water, stirred 20 minutes, and the solid material is collected by filtration and dried to give Structure 9 of Figure 5. This sulfhydral product is next oxidized to the sulfonate (Structure 10 of Figure 5) by dissolving in acetone or t-butanol/water mixture. Magnesium sulfate (0.2 mole) and potassium permanganate (0.5 mole) are added. The mixture is stirred at room temperature until reaction is complete, then filtered, and treated with excess aqueous NaHS03 to decompose KMn04 and Mn02. The filtrate is partitioned between water containing triethylamine hydrochloride and chloroform. The chloroform layer is dried down and purified by silica gel chromatography to give Structure 10 of Figure 5.
Example 7 Synthesis of 5' Methylenesulfonate Subunit This example describes the preparation of a subunit suitable for use in preparing polymers with 6-atom unit- length backbones having sulfonamide linkages.
For the preparation of subnits having Structure C of Figure 3 wherein X2 is CH2 the starting material is the 46
5'aldehyde (Structure 5 of Figure 5). This material is treated with phenyl diphenylphosphinylmethane sulfonate (Fild) , then reduced with H2/Pd on charcoal in a polar solvent, and lastly treated with alcoholic KOH in DMF. The product is reprotected on the morpholino nitrogen with trityl chloride and then purified by silica gel chromatography.
Example 8 Preparation of 5'-aminomethanesulfonate Subunit This example describes the preparation of a subunit suitable for use in preparing polymers with 7-atom unit- length backbones having sulfonamide linkages.
For the preparation of subnits having Structure D of
Figure 3 wherein X3 is a methanesulfonated amine, the starting material is the 5'aldehyde (Structure 5 of
Figure 5) . The 5' aldehyde is converted by reductive alkylation to a secondary amine by the method illustrated in Example 5, except that aminomethanesulfonic acid comprises the amine and ethylmorpholine is used to assure availability of the amine moiety for reaction with the aldehyde. This product is then reacted with methanesulfonyl chloride in the presence of triehtylamine to give the desired product, which is purified by silica gel chromatography.
Example 9 Synthesis of N-methanesulfonate Subunit This example describes the preparation of a subunit containing a sulfonate moiety linked to the morpholino ring nitrogen suitable for preparing polymers with 7-atom unit-length backbones. The steps are described with respect to structures shown in Figure 7.
The subunit is prepared by oxidizing a ribonucleoside (Structure 1) with periodate in the 47 presence of aminomethanesulfonic acid (AMSA) and N-ethyl morpholine. The oxidation is followed by reduction with sodium cyanoborohydride in the presence of benzotriazole (used to buffer the reaction mix) to give a morpholino subunit having a methane sulfonic acid group on the morpholino nitrogen (Structure 2) .
The 5'hydroxyl (numbered as in the parent ribose) is then oxidized to an aldehyde (Structure 3) and converted to a primary or secondary amine (Structure 4) by reductive alkylation as in Example 5, and tritylated to give the desired subunit of Structure 5.
Example 10
Preparation of N-methanecarboxylate Subunit This example describes the preparation of a subunit containing a carboxylate moiety linked via a methylene to the morpholino ring nitrogen suitable for preparing polymers with 7-atom unit-length backbones.
The subnit can be prepared essentially as in Example 9, but substituting glycine for aminomethanesulfonic acid. Alternatively, it is generally more convenient to prepare it starting with Structure 3 of Figure 5. This is readily alkylated on the morpholino ring nitrogen using chloroacetic acid or bromoacetic acid. The 5' hydroxyl is then converted to a primary amine and tritylated as in Example 9.
Example 11
Synthesis of 5'-aminomethyl Riboside Subunit
N4-Benzoylcytidine-2',3'-acetonide (1 mmole) was converted to the 5'-Iodo derivative by reaction with methyltriphenoxyphosphonium iodide in DMF (20ml) under argon at room temperature for 20 hours. Methanol (5 ml) was added and after 30 minutes the mixture was evaporated in vacuo. The residue was dissolved in ethyl acetate and 48 the solution washed with aqueous sodium thiosulfate, then brine. After drying with sodium sulfate and evaporation of the solvent the product was purified by chromatography on silica using isopropanol/chloroform mixtures. The iodo compound (1 mmole) is reacted with potassium cyanide (5 mmol) in Dimethylsulfoxide for 12 hours under argon atmosphere. The nitrile is isolated by pouring the reaction mixture into saturated aqueous sodium dihydrogen phosphate. The mixture is extracted with ethyl acetate, the organic layer washed well with water, dried over sodium sulfate and evaporated in vacuo. The nitrile is purified by chromatography on silica using chloroform/ethylacetate mixtures.
The nitrile from the previous paragraph is treated with a mixture of equal parts of DMF and aqueous ammonia at 25°C for 24 hours. The mixture is treated with rhodium on alumina and hydrogenated in a hydrogen atmosphere to provide the amine. After filtration and evaporation, the residue is dissolved in 0.2 N HC1 to cleave the acetonide. After evaporation the amine diol may be purified by ion exchange on a cation exchange column. When appropriate, the amine moiety may be tritylated as in Example 2 to give the amine-protected 2',3'-diol.
Example 12 Synthesis of 5'-aminoethylsulfonyl Riboside Subunit 2-Aminoethanethiol (2 mmol) is reacted with carbobenzoxychloride (1 mmol) in pyridine. The protected thiol carbamate is purified by Si02 using ethylacetate/hexane mixtures. Under an argon atmosphere the thiol (1 mmol) is dissolved in oxygen-free DMF containing 1.1 mmol oil-free sodium hydride. After evolution of gases the mixture is treated with the N4- 49 benzoylcytidine-2' , 3'-acetonide iodo-compound from Example 11 (at 0°C.) and the mixture stirred at room temperature for 12 hours. The solvent was evaporated in vacuo. After redissolution in chloroform the solution was washed with sodium bicarbonate, then brine, then dried over Na2S04, filtered and evaporated in vacuo. The residue was purified by chromatography on silica gel using chloroform/methanol mixtures.
The sulfide from the previous paragraph was oxidized with excess perbenzoic acid in chloroform to the sulfone. This was immediately, treated with 0.2 N HCl/dioxane to cleave the acetonide group. The diol was purified by chromatography on silica using methanol/chloroform mixtures. The diol sulfone from above is reduced with hydrogen/ palladium on carbon in DMF/methanol in the presence of acetic acid to remove the carbobenzoxy group. One equivalent of tosic acid is added to the mix, the solution is filtered and the filtrate evaporated. When appropriate the pendant amine is tritylated as in Example 2 to give the amine-protected 2',3'-diol. If required, the benzoyl group on the base is removed by treatment with equal amounts of DMF and CMC aqueous ammonia at room temperature for 24 hours.
Example 13 Activation and Coupling To Give Carbamate Linkage This example describes the activation of morpholino- subunits, such as prepared in Example 2, and their sub- sequent coupling via a carbamate linkage to yield a 6- atom unit-length backbone. The example is described with reference to the Structures in Figure 9. Activation Step
Dry, N-protected, 5'hydroxyl morpholino nucleoside 50
(Structure 1) (1 mmol) , prepared as in Example 2, is treated with bis-(p-nitrophenyl) carbonate (BNPC) and tri- ethylamine (TEA) in DMF under anhydrous conditions. The solution is stirred for three hours, then evaporated to dryness. The residue is dissolved in chloroform and chromatographed on silica gel eluting with an appropriate chloroform/ ethanol/ 0.1% TEA mixture to give activated subunit (Structure 2) . Deprotection Step 1.1 mmole morpholino nucleoside (Structure 1) is dissolved in 10 ml trifluoroethanol and 0.1 ml formic acid (or 0.2 ml acetic acid) added - giving a strong yellow color from the trityl carbonium ion - which fades on standing a few minutes. After five minutes the ' trifluoroethanol and acid are removed under reduced pressure and the deprotected subunit (Structure 3) resuspended in 5 ml DMF containing 0.5 ml triethylamine. Coupling
The activated subunit (Structure 2) is added to the DMF solution of unprotected subunit and incubated at room temperature for 1 hour to give coupled product (Structure 4) .
Example 14
Activation of Sulfamic and Sulfonic Acids and Coupling to Give Sulfamide and Sulfonamide Linkages This example describes the activation of sulfamic acid salts (such as prepared in Examples 4 and 5) and the activation of sulfonic acid salts (such as prepared in Examples 6, 7, 8 and 9) and their coupling to form sulfamide and sulfonamide linkages, respectively. The example is described with reference to the structures in Figures 10 and 11. 51
Activation
Ten mmole of the triethylamine salt of sulfated subunit protected on the base and on the nitrogen of the morpholino ring (e.g., Structure 1 of Figures 10 and 11) is dissolved in 10 ml of dichloromethane and then 40 mmole of pyridine is added. This solution is chilled for 15 minutes on a bed of dry ice and then 1.1 mmole of phosgene (20% in Toluene) is slowly added while the solution is rapidly stirred. After addition the solution is allowed to come to room temperature and then washed with aqueous NaHC03, dried, and chromatographed on silica gel eluted with a mixture of chloroform and acetone to give the desired sulfamoyl chloride (e.g., Structure 2 of Figure 10) or sulfonyl chloride (e.g., Structure 2 of Figure 11) . Deprotection
Eleven mmole of the triethylamine salt of sulfated subunit (e.g., Structure 1 of Figure 10 or 11) is dissolved in 200 ml of trifluoroethanol and 0.2 ml of formic acid (or 0.4 ml acetic acid) added. After 5 minutes the solution is concentrated under reduced pressure and the deprotected subunit (e.g., Structure 3 of Figure 10 or 11) precipitated with ether. The preci¬ pitate is then washed thoroughly with ether and then resuspended in 5 ml of DMF containing 0.6 ml of triethyl¬ amine. If an appreciable amount of residual formic or acetic acid remains in the deprotected subunit preparation the subsequent coupling efficiency can be seriously reduced. This reduction in efficiency is probably the result of the sulfamoyl chloride or sulfonyl chloride component reacting with these carboxylate salts to form mixed anhydrides, which in turn fail to react in the desired manner with the morpholino nitrogen of the deprotected component. 52
Coupling
The activated subunit (Structure 2) is added to the DMF solution of deprotected subunit (Structure 3) and incubated at room temperature for 1 hour to give coupled product (Structure 4) .
Example 15 Coupling of sulfamoyl Chloride with Alcohol to Give
Sulfamate Linkage This example describes the coupling of a sulfamoyl chloride (prepared as in Example 4 and activated as in Example 14) with a 5' hydroxyl subunit. This example is described with reference to the structures in Figure 12.
One mmole of the sulfamoyl chloride, prepared as in Example 4 and activated as in Example 14 (Structure 3) , 1 mmole 2,6-di-t-butyl-4-methylpyridine, 0.5 mmole of the alcohol component (Structure 4), and 20 ml of dry toluene are placed in an oven-dried round-bottom flask. After dissolution the reaction mixture is evaporated under reduced pressure and residual toluene removed under high vacuum. The residue is redissolved in methylene chloride
(10 ml) and treated with silver trifluoromethanesulfonate
(2 mmole) . The reaction mixture is stirred at room temperature for several hours to complete cooling. Chloroform (20 ml) is added and the resulting milky suspension added to an acetonitrile solution (20 ml) of tetraethylammonium chloride (5 mmole) . After stirring at room temperature for 30 minutes the excess solvent is removed by rotary evaporator, the residue dissolved in chloroform (150 ml) and filtered into a separatory funnel containing 0.05 N HC1 (20 ml). Following washing, the organic layer is washed with 20 ml water, dried over sodium sulfate, and then dried under vacuum. The residue is chromatographed on silica gel developed with a 53 chloroform/methanol mixture to give the desired product (Structure 5) .
Example 16 Activation and Coupling To Give Amide Linkage
This example describes the activation of the carboxylate subunit prepared in Example 10 and coupling to form an amide linkage. The example is described with reference to the Structures in Figure 13. Activation
10 mmole of the. subunit prepared in Example 10
(Structure 1) is dissolved in DMF containing 20 mmole of p-nitrophenol and 15 mmole of dicyclohexylcarbodiimide.
After 1 hour the product is rotovaped and then purified by silica gel chromatography to give Structure 2.
Deprotection
Eleven mmole of the subunit prepared in Example 10 (Structure 1) is dissolved in 100 ml of dichloromethane, 1 ml of methanol and 1 ml of dichloroacetic acid. After 5 minutes the CH2C12 is removed under reduced pressure and the product washed with ether, dried and dissolved in 20 ml DMF containing 1 ml triethylamine to give Structure 3. Coupling
The activated subunit (Structure 2) is added to the DMF solution of deprotected subunit (Structure 3) and incubated at room temperature for 1 hour to give coupled product (Structure 4) .
Example 17 Simultaneous Morpholino Ring Formation and Subunit Coupling
This example describes the oxidation of a ribonu¬ cleoside containing a protected amine linked through the 5' methylene, such as prepared in Example 11 or 12, and 54 coupling to the unprotected amine of another subunit to simultaneously form a morpholino ring structure and join the subunits. The example is described with reference to the structures in Figure 14. Amine Protection
Ten mmole of ribonucleoside containing a 1° amine linked through the 5' methylene (Structure 1) is reacted with 11 mmole of trityl chloride to protect the amine (Structure 2) . Oxidation
Tritylated subunit (Structure 2) , in methanol, is reacted with 11 mmole of NaI04 to give the dialdehyde (Structure 3) . Coupling If the coupling solution is too acidic the reaction is very slow and if the solution is too basic epimerization of the Structure 3 component appears to occur. A weak acid is used to neutralize the amine component (Structure 1) and buffer the reaction in this coupling step. Weak acids which have been found suitable for this purpose are: carbonic, ortho and para nitrophenol, and benzotriazole. Accordingly, the dialdehyde (Structure 3) is combined with a suitable salt of Structure 1 in a water/methanol mixture to give the coupled product (Structure 4) .
Reduc ion
Either during or after the morpholino ring closure step sodium cyanoborohydride is added to reduce the 2',3' dihydroxymorpholino ring (Structure 4) to the desired morpholino product (Structure 5) .
Example 18 Solution-Phase Block Assembly of Sulfamide-Linked 55
Oligomer of the Sequence 5'-CUGU This example describes the assembly of a short oligomer containing a sulfamide-linked backbone (Structure C-C of Figure 4, wherein X2 is a nitrogen) coupled as in Example 14. This solution assembly method is particularly useful for large-scale synthesis of short oligomers suitable for subsequent assembly into longer oligomers using the solid-phase method (Example 19) .
5'Sulfamic acid subunits of C, U, and G tritylated on the morpholino ring nitrogen are prepared as in Example 5. The U subunit is then activated by conversion to the sulfamoyl chloride form as in Example 14. The C subunit and the G subunit are deprotected as in Example 14. The deprotected C component (1.1 m mole) is dissolved in 5 ml DMF and 0.3 ml TEA, followed by addition of 1.0 m mole of the activated U component. Likewise, the deprotected G component is reacted with the activated U component.
After one hour each of these preparations is added to 100 ml of rapidly stirred brine and the solid collected and washed with water. The GU dimer is dried thoroughly under high vacuum and then activated as in Example 14. The best tetramer coupling results are obtained when purification of the dimer, via silica gel chromatography, is carried out after, rather than before, this activation step.
The CU dimer is deprotected as in Example 14. Better yields of tetramer are obtained when the dimer, after the initial ether precipitation, is thoroughly resuspended in about 2 ml of trifluoroethanol, reprecipitated with 30 ml of ether, and then resuspended in DMF and TEA for subsequent coupling.
Coupling to form the desired tetramer entails simply adding 1 m mole of activated GU dimer to the DMF/TEA 56 solution containing 1.1 m mole of deprotected CU dimer.
Workup of the tetramer entails adding the reaction mixture to brine, washing the solid with water, and drying under vacuum to give the desired tetramer: 5'- CUGU having a sulfamic acid salt at the 5'" end and a trityl on the morpholino nitrogen of the terminal U subunit. The structure of this tetramer is most easily confirmed by negative ion Fast Atom Bombardment mass spectroscopy. As a rule the dominant specie in the spectrum is the molecular ion.
Example 19 Solid-Phase Assembly of Sulfamide-Linked Morpholino Polymer This example describes the use of tetramer blocks, prepared as per Example 18, for solid-phase assembly of a morpholino polymer containing sulfamide intersubunit linkages. Solid-phase assembly provides a rapid method for assembly of longer binding polymers. The use of short oligomer blocks instead of monomers greatly simplifies separation of the final product from failure sequences.
A. Synthesis of short oligomers
The following tetramers are synthesized in solution: 5'-CUGU (Example 18); 5'-UCGG; 5'-GCGC; 5'-CACU.
These tetramers are converted to their activated sulfomoyl chloride form by the general method described in Example 14.
B. Preparation of the first monomer with a cleavable linker and attachment to the solid support
Morpholino C subunit containing a trityl moiety on the morpholino ring nitrogen and having a methylamine on the 5'methylene, prepared as in Example 5, is reacted with a 3-fold molar excess of Bis [2-(succinimidooxycar- 57 bonyloxy)ethyl]sulfone from Pierce of Rockford, Illinois, USA. This product is purified by silica gel chromatography and then added to a suitable solid support containing primary amine functions (e.g., Long Chain Alkyl Amine Controlled Pore Glass, from Pierce of Rockford, Illinois) . This procedure links the first tritylated subunit to the synthesis support via a linker which is stable to the acidic conditions used for detritylations, but which can be readily cleaved via a beta elimination mechanism using a strong non- nucleophilic base, such as a 1, 8- Diazabicyclo[5.4.0]undec-7-ene (DBU) .
C. Stepwise assembly of the polymer bound to the solid support The coupling cycle for addition of each subunit or oligomer block generally includes deprotection of the terminal backbone moiety, a thorough wash, addition of the next activated subunit or oligomer block, and a thorough wash. The coupling efficiency for each addition can be determined by collecting each detritylation solu¬ tion and subsequent wash and quantitating the trityl therein.
Detritylation in the present sufamide-linked polymer is achieved by slowly passing through the column a solu- tion of 2% formic acid in trifluoroethanol (or 2% dichlo- roacetic acid in dicloromethane) until the eluant no longer tests positive for trityl (readily determined by adding a drop of eluant to 100 μl methanesulfonic acid and inspecting for the visible yellow color characteris- tic of the trityl carbonium ion) . Thereafter the support is thoroughly washed to remove excess acid and then washed with DMF containing 1% by volume of N-ethylmor- pholine (NEM) . Coupling of the next subunit or oligomer block in the desired polymer sequence entails addition of 58 a concentrated DMF solution containing the activated monomer or oligomer and a molar equivalent of NEM. Since the rate of coupling is a function of concentration it is desirable to add a substantial molar excess of monomer or oligomer relative to the concentration of support-bound growing chains. A 5-fold molar excess of activated monomer or oligomer over that of the growing chains often gives acceptable coupling efficiencies. Required cou¬ pling times can be determined by removing at specified time intervals small defined quantities of the support material, thoroughly washing, treating the support with methanesulfonic acid, and then spectrophotometrically quantitating the released trityl carbonium ion (molar absorbance at 409 nm is 45,000 in methane sulfonic acid). After coupling is complete the unreacted subunit or oligomer is washed from the support with DMF. The un¬ reacted subunit is generally recovered, purified by chromatography, and reused for later synthesis. The sup¬ port is thoroughly washed with the solvent trifluoro- ethanol, without added acid. Washing is complete when addition of a drop of the wash eluant to 100 μl methane¬ sulfonic acid shows no yellow color.
The above coupling cycle is used to add, in order, the four activated tetramers 5'-CUGU; 5'-UCGG; 5'- GCGC; and 5'-CACU. This results in the following poly¬ mer: support-linker-CCUGUUCGGGCGCCACU-trityl. D. Cleavage from the support
The synthesis support is treated with 20% DBU in DMF for two hours at room temperature in the presence of 2% diethylmalonate, to tie up the vinylsulfone generated during cleavage of the linker. The released morpholino polymer is washed from the support with DMF and precipi¬ tated by adding ethylacetate. The precipitate contains full-length polymer having a 5' methylamine, the bases 59 still protected and a trityl moiety on the terminal morpholino nitrogen. In addition, the precipitate con¬ tains small amounts of failure sequences. At this stage the polymer size can be confirmed by positive ion fast atom mass spectrometry.
E. Addition of solubilizing moieties
If it is desired to add two solubilizing groups to the morpholino polymer this can be done conveniently by detritylting the N-terminal morpholino nitrogen using 2% formic acid in trifluoroethanol. Alternatively, if only one solubilizing moiet is to be added, then the 5'me- thylamine is acetylated with acetic anhydride before the detritylation step.
Polyethylene glycol 1000 (from Polysciences Inc., Warrington, Pennsylvania, USA) is thoroughly dried by dissolving in dry DMF and then evaporating the solvent under vacuum. The solid is resuspended in a minimal volume of pure dry DMF and 0.5 mole equivalent (relative to PEG 1000) of bis (p-nitrophenyl)carbonate and 1 mole equivalent of TEA is added and the preparation sealed and incubated overnight at 30°C to give p-nitrophenyl car¬ bonate-activated PEG 1000.
The full-length morpholino polymer which has been detritylated is added to a substantial molar excess (generally 10- to 20-fold) of activated PEG 1000 and incubated two hours at room temperature. Unreacted PEG 1000 is removed by precipitation of the tailed polymer with ether.
F. Base deprotection The dried polymer is suspended in DMSO, the DMSO solution chilled, and an equal volume of concentrated NH4OH is carefully layered on top of the chilled DMSO, and the container tightly capped. The preparation is incubated at 30°C for eighteen hours. Thereafter, the 60 solution is briefly exposed to aspirator vacuum to remove ammonia.
G. Purification of morpholino polymer
Purification at pH 2.5 is general for binding poly- ers wherein about half or more of the base-pairing moieties are of types 1, 2, 3, and 7 of Figure 2.
Water to be used for chromatography is degassed under aspirator vacuum and phosphoric acid added to give pH 2.5 (solvent A). A corresponding pH 2.5 solution is made 2 N in KC1 (solvent B) . Solvent A is mixed 1:1 by volume with chromatographic-grade acetonitrile to give solvent C.
Load up to about 10 mg of this polymer in 10 ml of solvent A on a chromatography column 1 cm in diameter and 10 to 20 cm in length which is packed with the cation- exchange support S-Sepharose Fast Flow (Pharmacia) . Proportionately larger quantities can be loaded on larger columns of the same length, e.g., up to 60 mg can be loaded on a 2.5 cm diameter column and 250 mg on a 5 cm diameter column. After washing the column thorough with solvent A elute with a linear gradient ranging from 100% solvent A to 100% solvent B and monitor the eluant to 254 nm. The desired binding polymer is generally the last and the largest peak to elute from the column. When the polymer is prepared by block assembly, base-line separations are often achieved. When peak shapes are unsymmetrical the problem generally has been due to insolubility of the binding polymer rather than a lack of capacity of the chromatographic packing. Such a problem, which is most common when the binding polymers do not contain a solubi¬ lizing moiety, can often be solved by reducing the quan¬ tity of binding polymer loaded in a given run. When peaks are symmetrical but base-line separation is not 61 achieved, substantial improvements are usually attained simply by eluting with a shallower gradient.
The eluant containing the polymer is desalted by loading on an equivalent-sized column packed with 35 micron chromatographic polypropylene (Cat. No. 4342 from Polysciences, Inc.) and washing thoroughly with solvent A. If baseline separation was achieved in the foregoing cation exchange chromatography, then pure product is obtained simply by eluting with solvent C; otherwise, the product is eluted with a linear gradient ranging from 100% solvent A to 100% solvent C. When the product is somewhat acid sensitive the solution is neutralized with dilute NaOH before drying under reduced pressure. Purification at high pH Purification at pH 11 is generally used for binding polymers wherein above half or more of the base-pairing moieties are at type 4, 5, 6 and 9 of Figure 2.
N,N-diethylethanolamine (Aldrich) is added to degas¬ sed water to adjust the pH to 11.0 (solvent D) . A cor- responding pH 11 solution 2 N in KC1 (solvent E) is prepared. A third pH 11 solution is prepared by mixing Solvent D 1:1 by volume with chromatographic grade aceto¬ nitrile (solvent F) .
The fully-deprotected binding polymer, prepared as above, is suspended in solvent D at a concentration of about 1 mg/ml. The pH is adjusted, if necessary, to pH 11 with N,N-diethylethanol-amine. About 10 ml of this polymer solution is placed on a chromatography column 1 cm in diameter and 10 to 20 cm in length which is packed with anion-exchange support Q-Sepharose Fast Flow (Phar¬ macia) . After washing the column thoroughly with solvent D, the column is eluted with a linear gradient ranging from 100% solvent D to 100% solvent E and the eluant is monitored at 254 nm. 62
The eluant containing the polymer is desalted by loading on an equivalent-sized column of polypropylene and washing thoroughly with solvent D. If baseline separation is achieved in the foregoing anion exchange chromatography then pure product is obtained simply by eluting with solvent F; otherwise, the product is eluted with a linear gradient ranging from 100% solvent D to 100% solvent F. Fractions containing the product are dried under reduced pressure. H. Sequence confirmation
While mass spectral analysis of the full-length polymer in the fully-protected state, as described ear¬ lier, does serve to confirm both the polymer length and the base composition, it does not provide information on the subunit sequence. Significant sequence information can be obtained from fragmentation patterns of deoxyribo- nucleic acids and carbamate-linked deoxyribonucleoside- derived polymers
(Griffin et al. (1987), Biomed. & Environ. Mass Spectro- metry 11_: 105) ; however, many of the morpholino polymers of the instant invention are quite resistant to fragmen¬ tation and give predominantly the molecular ion with only minimal fragments.
One method for confirming the sequence of the poly- mer is to take a small portion of the growing polymer after coupling each oligomer block and use mass spectral analysis to follow the elongation of the polymer. This method is applicable except for those rare cases where two blocks used in the synthesis happen to have exactly the same mass.
An indirect method to help verify the correctness of the polymer subunit sequence is to pair the morpholino polymer with its complementary DNA (whose sequence can be confirmed by established methods) and with DNA sequences 63 which might have resulted if the blocks were assembled in the wrong order. Pairing between the polymer and DNA can be evaluated by the occurrence of a hypochromic shift in the 240 to 290 nm wavelength region; such a shift occurs only between the polymer and its complementary sequence. The polymer/DNA duplex can also be distinguished from any partially-mismatched duplex by slowly raising the temper¬ ature while monitoring the absorbance in the 240 to 290 nm wavelength region. The perfect duplex will have a melting temperature (corresponding to a 50% reduction in the hypochromicity) generally 10 degrees or more above that of any mismatched duplex.
Example 20 Solution-Phase Assembly of Simple Prototype Morpholino Polymer, Structural Confirmation, Deprotection, Purification, and Assessment of Binding to Target DNA Sequence
This example describes the preparation, structural confirmation, and assessment of target binding affinity of a simple carbamate-linked morpholino polymer.
A carbamate-linked morpholino hexamer wherein all Px moieties are cytosines is assembled from dimer prepared as in Example 13. One third of that dimer preparation is detritylated (as in Example 13) and the remaining two thirds is activated (again as in Example 13) . Half of the activated dimer is reacted with the detritylated dimer to give tetramer, which is purified by silica gel chromatography developed with 6% methanol/94%chloroform. The tetramer is detritylated and reacted with the remain¬ ing activated dimer to give hexamer, which is purified by silica gel chromatography developed with 10% methanol/90% 64 chloroform.
This carbamate-linked 5'OH, base-protected hexamer having a trityl moiety on the morpholino nitrogen is designated c(mC*)6-trityl. Photon NMR gives: δ = 8.25-7.90 (18H, m) , 7.65-7.05 (39H, m) ,
6.16 (1H, bd) , 5.77 (4H, m) , 5.69 (1H, bd) , 4.46 (1H, m) , 4.35-3.80 (25H, m) , 3.56 (2H, m) , 3.25-2.75 (12H, m) , 1.47 )1H, m), 1.24 (1H, m) .
The mass spectrum (3-nitrobenzyl alcohol matrix) shows:
M-l = 2352.6 (2), 459.2 (30), 306.2 (100).
The high-resolution mass spectrum shows an M-l of 2352.8197, which is in good agreement for C120H112N24O29 calculated as 2352.8026. This c(mC*)6-trityl polymer is next detritylated as in Example 13, and then a polyethylene glycol 1000 tail is added followed by base deprotection, as in Example 19. Purification is by cation exchange chromatography fol¬ lowed by desalting on a column of polypropylene, as described in Example 19.
This purified tailed hexamer, c(mC*)6-PEG1000, shows an absorption maximum at 267.1 nm in neutral aqueous solution, with a calculated molar absorbance of 42,800. In aqueous solution at pH 1, the same material shows an absorption maximum at 275.7 nm, with a calculated molar absorbance of 77,100. Proton NMR data for this final product is as follows:
5 = 7.74 (6H, broad d) , 5.97 (6H, broad D) , 5.65 (6H, broad D) , 4.30-4.05 (12H, m) , 4.04-3.80 (18H, m) , a large envelope containing the PEG protons, and several signals of the oligomer, 2.99-2.80 (120H, m) .
To assess target binding affinity 20 A260 units of DNA target p(dG)6, purchased from Pharmacia LKB, is dis¬ solved in 50 microliters of deionized water and 200 65 microliters of DMSO (spectrophotometric grade from Aid- rich Chem. Co.) is added (stock solution A). 1.8 mg of the tailed morpholino hexamer, c(mC)6-PEG1000, is dis¬ solved in 0.36 ml of spectrophotometric grade DMSO (stock solution B) . Phosphate buffer is prepared by adjusting the pH of 0.05 N NaOH to 7.4 using phosphoric acid, followed by addition of EDTA to a final concentration of 0.001 N (Buffer C) .
Stock solutions A and B are assayed for the actual concentration of polymer by UV; the absorbance of stock solution A is measured in 0.1 N NaOH and stock solution B is measured in 0.1 N HC1. Measurements at these pH extremes minimize base stacking and other polymer inter¬ actions which can give absorbance values not proportional to the component monomers. Stock solutions A and B are diluted with Buffer C to give solutions of a final con¬ centration of 10 micromolar in polymer. The required dilutions are calculated using molar absorbencies of 65,000 for solution A, p(dG)6, and 77,100 for solution B, c(mC*)6-PEG1000.
Assessment of target binding affinity is carried out in a double-beam scanning spectrophotometer having a temperature-controlled cell housing which will accom¬ modate two cells in the reference beam and two in the sample beam.
Using four matched quartz cuvettes, one is filled with 0.5 ml of 10 micromolar p(dG)6 and 0.5 ml of Buffer C and a second is filled with 0.5 ml of 10 micromolar c(mC*)6-PEG1000 and 0.5 ml of Buffer C. These two cu- vettes are placed in the reference beam of the tempera¬ ture-controlled cell housing. Next, a third cuvette is filled with 1 ml of Buffer C and a fourth is filled with 0.5 ml of 10 micromolar p(dG)6 and 0.5 ml of 10 micromo¬ lar c(mC*)6-PEG1000. These two cuvettes are placed in the 66 sample beam of the cell housing. The cell housing is then heated to 60 °C and allowed to cool slowly to 14 °C to assure complete pairing between the polymer and its DNA target in the fourth cuvette. A scan is then taken from 320 nm to 240 nm - which shows a substantial absorb¬ ance difference due to a hypochromic shift in polymer- target mixture, centered around 273 nm. The temperature of the cell holder is then raised in 2-degree increments to 80°C, with scans taken after each 2-degree rise. For comparison, the same procedure is used for assessing the binding • affinity of p(dC)6 DNA for its target p(dG)6, which gives a similar but less intense hypochromic shift in the paired state.
Plots of the absorbance difference as a function of temperature for both the morpholino polymer/DNA and the analogous DNA/DNA complexes are shown in Figure 15. The melting temperature, T„, wherein the complex is half melted, is seen to be 62°C for the morpholino polymer/DNA and 30 °C for the DNA/DNA. At low salt concentrations such as used here, the charge repulsion between the anionic DNA backbones substantially destabilizes the DNA/DNA duplex, while there is no corresponding electro¬ static repulsion between the morpholino polymer and its DNA target.
Example 21 Solution-phase Assembly of Simple Prototype Sulfamide-linked Morpholino Polymer and Assessment of Binding to RNA and DNA Target Sequences This example describes the preparation and target binding of a simple sulfamide-linked morpholino polymer.
A sulfamide-linked morpholino hexamer, wherein all Pλ moieties are cytosines, is assembled from 5'sulfated 67 methylamine subunit (R is methyl) prepared as in Example 5 and activated as in Example 14. Activated monomer is reacted in DMF with an excess of 5'OH subunit lacking a protective group on the morpholino nitrogen, prepared as in Example 2. The resultant dimer is purified by silica gel chromatography developed with methanol/chloroform mixtures and then deprotected as in Example 14. This product is then reacted with more activated monomer, the chain-extended product purified, and deprotected as above. This cycle is repeated until hexamer is obtained. Before the last .detritylation, the mass of the hexamer was confirmed by negative ion FAB mass spectro¬ scopy, which showed M-l = 2598.9 (100).
As in Example 20, the sulfamide-linked hexamer is tailed with PEG-1000, deprotected, purified, and tested for binding to its DNA target, p(dG)6, and its RNA tar¬ get, poly(G) . The target-binding affinities, expressed in Tm values, for this sulfamide-linked hexamer, referred to as s (mC)6, are tabulated below, along with the cor- responding target-binding affinities for the analogous DNA oligomer, p(dC)β.
T_ value (° C.) s (mC)6/p(dG)6 25 p(dC)6/p(dG)β 29
s(mC)6/poly(G) 33 s(dC)6/poly(G) 38
While specific embodiments, methods, and uses of the invention have been described, it will be appreciated that various changes and modifications of the invention may be made without departing from the invention. In 68 particular, although preferred polymer backbone struc¬ tures have been described and illustrated, it will be appreciated that other morpholino-based polymers may be constructed according to the backbone constraints and requirements discussed above.

Claims

69IT IS CLAIMED:
1. A polymer composition comprised of morpholino subunit structures of the form:
Figure imgf000072_0001
where (i) the structures are linked together by un¬ charged, achiral linkages, one to three atoms long, joining the morpholino nitrogen of one subunit to the 5', exocyclic carbon of an adjacent subunit, and (ii) Pi is a purine or pyrimidine base-pairing moiety effective to bind by base-specific hydrogen bonding to a base in a polynucleotide.
2. The composition of claim 1, wherein Pi is selec¬ ted from the group consisting of:
Figure imgf000072_0002
X= H, CH3, F, Cl, Br, I 70
3. The composition of claim 1, wherein the linked structures have a form selected from the group consisting of:
(A) (B) (C)
Figure imgf000073_0001
(D) (E) (F)
Figure imgf000073_0002
4. The composition of claim 1, wherein the linkage is of the form:
" o - pi
Figure imgf000073_0003
O =S -**
Figure imgf000073_0004
where X is NH, NCH3, 0, S, or CH2; and, 71
Pj is a purine or pyrimidine base-pairing moiety effective to bind by base-specific hydrogen bonding to a base in a polynucleotide.
5. The composition of claim 1, wherein the linkage is of the form:
Figure imgf000074_0001
where ϊ is 0 or S; and, Pj is a purine or pyrimidine base-pairing moiety effective to bind by base-specific hydrogen bonding to a base in a polynucleotide.
6. The composition of claim 1, wherein the linkage is of the form:
Figure imgf000074_0002
where Pj is a purine or pyrimidine base-pairing moiety effective to bind by base-specific hydrogen bonding to a base in a polynucleotide. 72
7. The composition of claim 1, wherein the linkage is of the form
Figure imgf000075_0001
where R is H or CH3; and,
Pj is a purine or pyrimidine base-pairing moiety effective to bind by base-specific hydrogen bonding to a base in a polynucleotide.
8. The composition of claim 1, which further includes a moiety at one or both termini which is effec¬ tive to enhance the solubility of the polymer in aqueous medium.
9. The composition of claim 7, wherein the ter¬ minal moiety is polyethylene glycol.
10. The composition of claim 1, composed of at least 3 morpholino subunits.
11. The composition of claim 1, wherein at least one of the P± is a 2, 6-diaminopurine.
12. The composition of claim 1, wherein at least one of the Pi is a 5-halouracil. 73
13. The composition of claim 1, wherein at least % of the Pi are 2-amine containing purines.
PCT/US1990/007565 1989-12-20 1990-12-20 Uncharged morpholino-based polymers having achiral intersubunit linkages WO1991009073A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
DE69032167T DE69032167T2 (en) 1989-12-20 1990-12-20 UNCHARGED POLYMERS WITH MORPHOLINO UNITS AND WITH ACHIRAL BINDINGS BETWEEN THESE UNITS
EP91902401A EP0506845B1 (en) 1989-12-20 1990-12-20 Uncharged morpholino-based polymers having achiral intersubunit linkages
KR1019920701480A KR0154317B1 (en) 1989-12-20 1990-12-20 Uncharged morpholino-based polymers having achiral intersubunit linkages
JP50289391A JP3172174B2 (en) 1989-12-20 1990-12-20 Uncharged morpholino-based polymerase with achiral internal subunit linkage
AU71587/91A AU654473B2 (en) 1989-12-20 1990-12-20 Uncharged morpholino-based polymers having achiral intersubunit linkages

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US07/454,055 US5034506A (en) 1985-03-15 1989-12-20 Uncharged morpholino-based polymers having achiral intersubunit linkages
US454,055 1989-12-20

Publications (1)

Publication Number Publication Date
WO1991009073A1 true WO1991009073A1 (en) 1991-06-27

Family

ID=23803109

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/007565 WO1991009073A1 (en) 1989-12-20 1990-12-20 Uncharged morpholino-based polymers having achiral intersubunit linkages

Country Status (9)

Country Link
US (1) US5034506A (en)
EP (1) EP0506845B1 (en)
JP (1) JP3172174B2 (en)
KR (1) KR0154317B1 (en)
AT (1) ATE164081T1 (en)
AU (1) AU654473B2 (en)
CA (1) CA2069906C (en)
DE (1) DE69032167T2 (en)
WO (1) WO1991009073A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993020236A1 (en) * 1992-04-03 1993-10-14 Applied Biosystems, Inc. Probe composition and method
WO1996003417A1 (en) * 1994-07-25 1996-02-08 Hybridon, Inc. Improved methods of detritylation for oligonucleotide synthesis
EP0773950A1 (en) * 1994-07-15 1997-05-21 Isis Pharmaceuticals, Inc. Linked peptide nucleic acids
WO1998032467A2 (en) * 1997-01-24 1998-07-30 Antivirals, Inc. Method and conjugate for treating h. pylori infection
WO2000044897A1 (en) * 1999-01-29 2000-08-03 Avi Biopharma, Inc. Method of treating restenosis by antisense targeting of c-myc
EP2002018A2 (en) * 2006-03-07 2008-12-17 AVI BioPharm, Inc. Antisense antiviral compound and method for treating arenavirus infection

Families Citing this family (1825)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521063A (en) * 1985-03-15 1996-05-28 Antivirals Inc. Polynucleotide reagent containing chiral subunits and methods of use
US5217866A (en) * 1985-03-15 1993-06-08 Anti-Gene Development Group Polynucleotide assay reagent and method
US5506337A (en) * 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
US5235033A (en) * 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5470974A (en) * 1985-03-15 1995-11-28 Neu-Gene Development Group Uncharged polynucleotide-binding polymers
US5185444A (en) * 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5378841A (en) * 1989-12-20 1995-01-03 Antivirals Inc. Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5955589A (en) * 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
US5623065A (en) * 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US6753423B1 (en) 1990-01-11 2004-06-22 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced biostability and altered biodistribution of oligonucleotides in mammals
US5514788A (en) * 1993-05-17 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US6339066B1 (en) 1990-01-11 2002-01-15 Isis Pharmaceuticals, Inc. Antisense oligonucleotides which have phosphorothioate linkages of high chiral purity and which modulate βI, βII, γ, δ, Ε, ζ and η isoforms of human protein kinase C
US5591623A (en) * 1990-08-14 1997-01-07 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of cell adhesion
US5914396A (en) * 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US6399754B1 (en) * 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
US20040142899A1 (en) * 1990-01-11 2004-07-22 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced biostability and altered biodistribution of oligonucleotides in mammals
US5959096A (en) * 1992-03-16 1999-09-28 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against human protein kinase C
US5514577A (en) * 1990-02-26 1996-05-07 Isis Pharmaceuticals, Inc. Oligonucleotide therapies for modulating the effects of herpes viruses
US5834607A (en) * 1990-07-27 1998-11-10 Isis Pharmaceuticals, Inc. Amines and methods of making and using the same
US5789573A (en) * 1990-08-14 1998-08-04 Isis Pharmaceuticals, Inc. Antisense inhibition of ICAM-1, E-selectin, and CMV IE1/IE2
US5442049A (en) * 1992-11-19 1995-08-15 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating the effects of cytomegalovirus infections
US6153595A (en) * 1990-08-16 2000-11-28 Isis Pharmaceuticals Inc. Composition and method for treatment of CMV infections
US5595978A (en) * 1990-08-16 1997-01-21 Isis Pharmaceuticals, Inc. Composition and method for treatment of CMV retinites
US5965722A (en) * 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
WO1994008003A1 (en) * 1991-06-14 1994-04-14 Isis Pharmaceuticals, Inc. ANTISENSE OLIGONUCLEOTIDE INHIBITION OF THE ras GENE
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
US6713602B1 (en) * 1991-05-24 2004-03-30 Ole Buchardt Synthetic procedures for peptide nucleic acids
US5719262A (en) * 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5766855A (en) * 1991-05-24 1998-06-16 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity and sequence specificity
US6441130B1 (en) 1991-05-24 2002-08-27 Isis Pharmaceuticals, Inc. Linked peptide nucleic acids
US5714331A (en) * 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
DK51092D0 (en) * 1991-05-24 1992-04-15 Ole Buchardt OLIGONUCLEOTIDE ANALOGUE DESCRIBED BY PEN, MONOMERIC SYNTHONES AND PROCEDURES FOR PREPARING THEREOF, AND APPLICATIONS THEREOF
US6414112B1 (en) 1991-05-24 2002-07-02 Ole Buchardt Peptide nucleic acids having 2,6-diaminopurine nucleobases
BR9206156A (en) * 1991-06-14 1995-10-17 Isis Pharmaceuticals Inc Oligonucleotide or aligonucleotide analogue process of modulating the expression of the human H-RAS gene process of detecting the presence of the H-RAS gene in cells or tissues process of detecting activated H-RAS based on the differential affinity of particular oligonucleotides by activated H-RAS VS, like, process of treating conditions that arise from the activation of the H-RAS oncogene
US5747253A (en) * 1991-08-23 1998-05-05 Isis Pharmaceuticals, Inc. Combinatorial oligomer immunoabsorbant screening assay for transcription factors and other biomolecule binding
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US8153602B1 (en) 1991-11-19 2012-04-10 Isis Pharmaceuticals, Inc. Composition and methods for the pulmonary delivery of nucleic acids
US6235887B1 (en) * 1991-11-26 2001-05-22 Isis Pharmaceuticals, Inc. Enhanced triple-helix and double-helix formation directed by oligonucleotides containing modified pyrimidines
TW393513B (en) * 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
AU3250093A (en) 1991-12-12 1993-07-19 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5359044A (en) * 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US20070032446A1 (en) * 1991-12-24 2007-02-08 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5856455A (en) * 1991-12-24 1999-01-05 Isis Pharmaceuticals, Inc. Gapped 2'-modified oligonucleotides
JP3131222B2 (en) 1991-12-24 2001-01-31 アイシス・ファーマシューティカルス・インコーポレーテッド 2 'modified oligonucleotide having a gap
CA2126451A1 (en) * 1991-12-24 1993-07-08 Brett P. Monia Compositions and methods for modulating .beta.-amyloid
EP1126025A3 (en) * 1992-03-16 2002-02-13 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of protein kinase C
US5885970A (en) * 1992-03-16 1999-03-23 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against human protein kinase C
US6153599A (en) * 1992-03-16 2000-11-28 Isis Pharmaceuticals, Inc. Methoxyethoxy oligonucleotides for modulation of protein kinase C expression
US5681747A (en) * 1992-03-16 1997-10-28 Isis Pharmaceuticals, Inc. Nucleic acid sequences encoding protein kinase C and antisense inhibition of expression thereof
US5922686A (en) * 1992-03-16 1999-07-13 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of protein kinase C
US6537973B1 (en) 1992-03-16 2003-03-25 Isis Pharmaceuticals, Inc. Oligonucleotide inhibition of protein kinase C
US5882927A (en) * 1992-03-16 1999-03-16 Isis Pharmaceuticals, Inc. Oligonucleotide inhibition of protein kinase C
US5916807A (en) * 1992-03-16 1999-06-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against human protein kinase C
US5948898A (en) * 1992-03-16 1999-09-07 Isis Pharmaceuticals, Inc. Methoxyethoxy oligonucleotides for modulation of protein kinase C expression
US6117847A (en) * 1992-03-16 2000-09-12 Isis Pharmaceuticals, Inc. Oligonucleotides for enhanced modulation of protein kinase C expression
US5643780A (en) * 1992-04-03 1997-07-01 Isis Pharmaceuticals, Inc. Compositions and methods for modulating RNA activity through modification of the 5' cap structure of RNA
US5817781A (en) * 1992-06-01 1998-10-06 Gilead Sciences, Inc. Modified internucleoside linkages (II)
US5434257A (en) * 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
ATE282695T1 (en) * 1992-07-20 2004-12-15 Isis Pharmaceuticals Inc PSEUDO HALF-KNOT FORMING RNA THROUGH HYBRIDIZATION OF ANTISESEOLIGON NUCLEOTIDES TO TARGETED RNA SECONDARY STRUCTURES
US6284458B1 (en) * 1992-09-10 2001-09-04 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of hepatitis C virus-associated diseases
US6995146B2 (en) 1992-09-10 2006-02-07 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of hepatitis C virus-associated diseases
US6423489B1 (en) 1992-09-10 2002-07-23 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus-associated diseases
US6433159B1 (en) 1992-09-10 2002-08-13 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus associated diseases
US6174868B1 (en) 1992-09-10 2001-01-16 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of hepatitis C virus-associated diseases
US20040049021A1 (en) * 1992-09-10 2004-03-11 Anderson Kevin P. Compositions and mehtods for treatment of Hepatitis C virus-associated diseases
US6391542B1 (en) 1992-09-10 2002-05-21 Isis Pharmaceuticals, Inc. Compositions and methods for treatment of Hepatitis C virus-associated diseases
US6235886B1 (en) 1993-09-03 2001-05-22 Isis Pharmaceuticals, Inc. Methods of synthesis and use
WO1994006815A1 (en) 1992-09-11 1994-03-31 Isis Pharmaceuticals, Inc. Oligonucleotide and nucleotide amine analogs, methods of synthesis and use
IL107150A0 (en) * 1992-09-29 1993-12-28 Isis Pharmaceuticals Inc Oligonucleotides having a conserved g4 core sequence
US6784290B1 (en) 1992-10-05 2004-08-31 Isis Pharmaceuticals, Inc. Antisense oligonucleotide inhibition of ras
KR100199247B1 (en) * 1992-10-05 1999-06-15 파샬 비. 린네 Antisense oligonucleotide inhibition of the ras gene
US5872242A (en) * 1992-10-05 1999-02-16 Isis Pharmaceuticals, Inc. Antisense oligonucleotide inhibition of ras
US20030013670A1 (en) * 1992-10-05 2003-01-16 Monia Brett P. Antisense oligonucleotide inhibition of ras
CA2170869C (en) 1993-09-03 1999-09-14 Phillip Dan Cook Amine-derivatized nucleosides and oligonucleosides
US5510239A (en) * 1993-10-18 1996-04-23 Isis Pharmaceuticals, Inc. Oligonucleotide modulation of multidrug resistance-associated protein
US5801154A (en) * 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US6399376B1 (en) 1993-11-05 2002-06-04 Isis Pharmaceuticals, Inc. Modulation of vascular cell adhesive molecule expression through oligonucleotide interactions
US6710164B1 (en) 1993-11-22 2004-03-23 Peter E. Nielsen Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5656612A (en) * 1994-05-31 1997-08-12 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US5563255A (en) * 1994-05-31 1996-10-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US6410518B1 (en) 1994-05-31 2002-06-25 Isis Pharmaceuticals, Inc. Antisense oligonucleotide inhibition of raf gene expression
US6090626A (en) * 1994-05-31 2000-07-18 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of raf gene expression
US20060142236A1 (en) * 1994-05-31 2006-06-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US20030119769A1 (en) * 1994-05-31 2003-06-26 Monia Brett P Antisense oligonucleotide modulation of raf gene expression
US5981731A (en) * 1994-05-31 1999-11-09 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of B-raf gene expression
KR100211178B1 (en) * 1994-05-31 1999-07-15 파샬 비. 린네 Antisense oligonucleotide modulation of raf gene expression
US6391636B1 (en) 1994-05-31 2002-05-21 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of raf gene expression
US5665873A (en) * 1995-02-09 1997-09-09 Dana Farber Cancer Institute Glucocorticoid response elements
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
JPH11511128A (en) * 1995-08-01 1999-09-28 ノバルティス・アクチエンゲゼルシャフト Liposome oligonucleotide composition
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US7244622B2 (en) * 1996-04-03 2007-07-17 Applera Corporation Device and method for multiple analyte detection
US20050118605A9 (en) * 1996-06-06 2005-06-02 Baker Brenda F. Oligomeric compounds having modified bases for binding to adenine and guanine and their use in gene modulation
US20040171030A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Oligomeric compounds having modified bases for binding to cytosine and uracil or thymine and their use in gene modulation
US20070275921A1 (en) * 1996-06-06 2007-11-29 Isis Pharmaceuticals, Inc. Oligomeric Compounds That Facilitate Risc Loading
US20050032068A1 (en) * 2002-11-05 2005-02-10 Prakash Thazha P. Sugar and backbone-surrogate-containing oligomeric compounds and compositions for use in gene modulation
US20030044941A1 (en) 1996-06-06 2003-03-06 Crooke Stanley T. Human RNase III and compositions and uses thereof
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20040266706A1 (en) * 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US9096636B2 (en) * 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US6593305B1 (en) * 1996-08-02 2003-07-15 Genesense Technologies Inc. Antitumor antisense sequences directed against R1 and R2 components of ribonucleotide reductase
US5948681A (en) * 1996-08-14 1999-09-07 Children's Hospital Of Philadelphia Non-viral vehicles for use in gene transfer
US6111085A (en) 1996-09-13 2000-08-29 Isis Pharmaceuticals, Inc. Carbamate-derivatized nucleosides and oligonucleosides
WO1998014592A2 (en) 1996-10-01 1998-04-09 Geron Corporation Telomerase reverse transcriptase
US6001991A (en) * 1996-10-04 1999-12-14 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of MDR P-glycoprotein gene expression
US5919638A (en) * 1996-10-08 1999-07-06 Abbott Laboratories Reagents and methods useful for detecting prostate tumors
US20030165971A1 (en) * 1996-10-31 2003-09-04 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
US20050202499A1 (en) 1996-10-31 2005-09-15 Billing-Medel Patricia A. Reagents and methods useful for detecting diseases of the breast
US7235653B2 (en) * 1996-12-31 2007-06-26 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US6077833A (en) * 1996-12-31 2000-06-20 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US6319906B1 (en) 1996-12-31 2001-11-20 Isis Pharmaceuticals Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US20040023917A1 (en) * 1996-12-31 2004-02-05 Bennett C. Frank Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6127533A (en) 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
US20020137904A1 (en) * 1998-03-27 2002-09-26 Patricia A. Billing-Medel Reagents and methods useful for detecting diseases of the gastrointestinal tract
US6716625B1 (en) 1997-04-16 2004-04-06 Claude Selitrennikoff Histidine kinases of Aspergillus and other fungal species, related compositions, and methods of use
US20050208567A1 (en) * 1997-04-25 2005-09-22 Billing-Medel Patricia A Reagents and methods useful for detecting diseases of the prostate
US7541151B2 (en) 1997-06-05 2009-06-02 Duke University Single-cell biosensor for the measurement of GPCR ligands in a test sample
US5891646A (en) * 1997-06-05 1999-04-06 Duke University Methods of assaying receptor activity and constructs useful in such methods
US6528271B1 (en) 1997-06-05 2003-03-04 Duke University Inhibition of βarrestin mediated effects prolongs and potentiates opioid receptor-mediated analgesia
US5962665A (en) 1997-06-16 1999-10-05 Abbott Laboratories Nucleic acid primers and probes for detecting HIV-1 and HIV-2
EP1012331B1 (en) 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
AU8280798A (en) 1997-07-03 1999-01-25 Thomas Jefferson University An improved method for design and selection of efficacious antisense oligonucleotides
US7105496B2 (en) 1998-07-23 2006-09-12 Northwestern University Methods and compositions for inhibiting angiogenesis
US6797691B1 (en) 1997-07-23 2004-09-28 Northwestern University Methods and compositions for inhibiting angiogenesis
US6809193B2 (en) 1997-08-13 2004-10-26 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US6133246A (en) * 1997-08-13 2000-10-17 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US6383808B1 (en) 2000-09-11 2002-05-07 Isis Pharmaceuticals, Inc. Antisense inhibition of clusterin expression
US5877309A (en) 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
US20070149472A1 (en) * 1997-08-13 2007-06-28 Mckay Robert Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
US7252950B1 (en) 1997-09-04 2007-08-07 The Regents Of The University Of California Assays for detecting modulators of cytoskeletal function
US7115884B1 (en) 1997-10-06 2006-10-03 Trustees Of Tufts College Self-encoding fiber optic sensor
US5948902A (en) * 1997-11-20 1999-09-07 South Alabama Medical Science Foundation Antisense oligonucleotides to human serine/threonine protein phosphatase genes
US20040171564A1 (en) * 1997-11-20 2004-09-02 Honkanen Richard E. Antisense oligonucleotide modulation of human serine/threonine protein phosphatase gene expression
EP1049475A4 (en) 1998-01-08 2003-06-04 Univ California KINESIN MOTOR MODULATORS DERIVED FROM THE MARINE SPONGE $i(ADOCIA)
US7189703B2 (en) * 1998-01-09 2007-03-13 Intracell, Llc Treatment and diagnosis of alzheimer's disease
US6764830B1 (en) 1998-01-23 2004-07-20 The Regents Of The University Of California Thermomyces lanuginosus kinesin motor protein and methods of screening for modulators of kinesin proteins
US6326479B1 (en) 1998-01-27 2001-12-04 Boston Probes, Inc. Synthetic polymers and methods, kits or compositions for modulating the solubility of same
CA2319710A1 (en) 1998-01-30 1999-08-05 Jim A. Wright Oligonucleotide sequences complementary to thioredoxin or thioredoxin reductase genes and methods of using same to modulate cell growth
US5968748A (en) * 1998-03-26 1999-10-19 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of human HER-2 expression
US20040186071A1 (en) 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
US7321828B2 (en) 1998-04-13 2008-01-22 Isis Pharmaceuticals, Inc. System of components for preparing oligonucleotides
US6417169B1 (en) 1998-04-23 2002-07-09 Genesense Technologies Inc. Insulin-like growth factor II antisense oligonucleotide sequences and methods of using same to inhibit cell growth
CA2329252A1 (en) 1998-05-21 1999-11-25 Isis Pharmaceuticals Inc. Compositions and methods for topical delivery of oligonucleotides
JP2003524586A (en) 1998-05-21 2003-08-19 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for parenteral administration of oligonucleotides
EP1084227A4 (en) 1998-05-29 2002-06-12 Univ Jefferson Compositions and methods for use in affecting hematopoietic stem cell populations in mammals
EP2045334A1 (en) 1998-06-24 2009-04-08 Illumina, Inc. Decoding of array sensors with microspheres
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6867294B1 (en) * 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
AU5329599A (en) * 1998-07-30 2000-02-21 University Of South Florida Method for the modulation of function of transcription factors
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
US20040009938A1 (en) * 1998-08-07 2004-01-15 Muthiah Manoharan Methods of enhancing renal uptake of oligonucleotides
US6673912B1 (en) 1998-08-07 2004-01-06 Isis Pharmaceuticals, Inc. 2′-O-aminoethyloxyethyl-modified oligonucleotides
US20020172678A1 (en) 2000-06-23 2002-11-21 Napoleone Ferrara EG-VEGF nucleic acids and polypeptides and methods of use
US6175004B1 (en) 1998-09-01 2001-01-16 Isis Pharmaceuticals, Inc. Process for the synthesis of oligonucleotides incorporating 2-aminoadenosine
US6225293B1 (en) 1998-09-02 2001-05-01 Isis Pharmaceuticals, Inc. Methods and compounds for tracking the biodistribution of macromolecule-carrier combinations
US6335194B1 (en) 1998-09-29 2002-01-01 Isis Pharmaceuticals, Inc. Antisense modulation of survivin expression
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
IL142094A0 (en) 1998-09-29 2002-03-10 Gamida Cell Ltd Methods of controlling proliferation and differentiation of stem and progenitor cells
US6069243A (en) * 1998-10-06 2000-05-30 Isis Pharmaceuticals, Inc. Process for oligonucleotide synthesis
US6214986B1 (en) * 1998-10-07 2001-04-10 Isis Pharmaceuticals, Inc. Antisense modulation of bcl-x expression
US6172216B1 (en) * 1998-10-07 2001-01-09 Isis Pharmaceuticals Inc. Antisense modulation of BCL-X expression
US20020138856A1 (en) * 1998-10-23 2002-09-26 Northwestern University Compositions and methods useful for treatment of depressive disorder based on an animal model
US6429027B1 (en) 1998-12-28 2002-08-06 Illumina, Inc. Composite arrays utilizing microspheres
US6300320B1 (en) 1999-01-05 2001-10-09 Isis Pharmaceuticals, Inc. Modulation of c-jun using inhibitors of protein kinase C
WO2000041731A1 (en) 1999-01-19 2000-07-20 The Children's Hospital Of Philadelphia Reverse gene therapy
US6395029B1 (en) 1999-01-19 2002-05-28 The Children's Hospital Of Philadelphia Sustained delivery of polyionic bioactive agents
AU3347000A (en) 1999-01-19 2000-08-01 Children's Hospital Of Philadelphia, The Hydrogel compositions for controlled delivery of virus vectors and methods of use thereof
US7282489B2 (en) * 2000-01-19 2007-10-16 The Children's Hospital Of Philadelphia Compositions and methods for performing reverse gene therapy
US6127124A (en) * 1999-01-20 2000-10-03 Isis Pharmaceuticals, Inc. Fluorescence based nuclease assay
US6121000A (en) * 1999-02-11 2000-09-19 Genesense Technologies, Inc. Antitumor antisense sequences directed against R1 and R2 components of ribonucleotide reductase
ES2296613T3 (en) * 1999-03-18 2008-05-01 Queen Mary And Westfield College INHIBITORS OF ENDOTHELINE SYNTHESIS-1.
US6235891B1 (en) 1999-03-31 2001-05-22 South Alabama Medical Science Foundation Glucocorticoid receptor agonist and decreased PP5
EP1165787A2 (en) 1999-03-31 2002-01-02 The University of North Carolina at Chapel Hill Isolated dna encoding cullin regulators roc1 and roc2, isolated proteins encoded by the same, and methods utilizing the same
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US20030207295A1 (en) * 1999-04-20 2003-11-06 Kevin Gunderson Detection of nucleic acid reactions on bead arrays
ATE413467T1 (en) 1999-04-20 2008-11-15 Illumina Inc DETECTION OF NUCLEIC ACID REACTIONS ON BEAD ARRAYS
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US6331618B1 (en) 1999-05-13 2001-12-18 Pe Corporation (Ny) Compositions of solvents and high concentrations of nucleic acid analogs
EP2360270B1 (en) 1999-05-20 2016-11-09 Illumina, Inc. Combinatorial decoding of random nucleic acid 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
US7534605B2 (en) * 1999-06-08 2009-05-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem CD44 polypeptides, polynucleotides encoding same, antibodies directed thereagainst and method of using same for diagnosing and treating inflammatory diseases
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US6147200A (en) * 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US7668658B2 (en) 1999-10-13 2010-02-23 Sequenom, Inc. Methods for generating databases and databases for identifying polymorphic genetic markers
US20020165188A1 (en) * 1999-10-14 2002-11-07 Meenhard Herlyn Methods for inhibition of tumorigenic properties of melanoma cells
ATE322493T1 (en) * 1999-12-30 2006-04-15 Leuven K U Res & Dev CYCLOHEXENE NUCLEIC ACIDS
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US6559128B1 (en) * 2000-01-21 2003-05-06 Northwestern University Inhibitors of G protein-mediated signaling, methods of making them, and uses thereof
WO2001061043A2 (en) 2000-02-16 2001-08-23 Illumina, Inc. Parallel genotyping of multiple patient samples
US6846486B1 (en) 2000-02-24 2005-01-25 Advanced Biotherapy Concepts, Inc. Method of treating allergy by administering an anti-histamine antibody
AU2001242190A1 (en) * 2000-03-31 2001-10-15 Aventis Pasteur Limited Immunogenic peptides derived from prostate-specific membrane antigen (psma) and uses thereof
EP2202241A3 (en) * 2000-04-13 2010-07-28 Thomas N. Wight Therapeutic compounds and methods for modulating V3, a versican isoform
JP2003531149A (en) 2000-04-13 2003-10-21 ザ・ロツクフエラー・ユニバーシテイ Enhancement of antibody-derived immune response
AU5752601A (en) 2000-05-04 2001-11-12 Avi Biopharma Inc Splice-region antisense composition and method
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
US6686188B2 (en) * 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US6656700B2 (en) * 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
CA2410950A1 (en) * 2000-05-30 2001-12-06 Hans-Michael Wenz Methods for detecting target nucleic acids using coupled ligation and amplification
US20060166227A1 (en) * 2000-06-20 2006-07-27 Stephen Kingsmore Protein expression profiling
AU2001271873A1 (en) 2000-07-06 2002-01-21 Avi Biopharma, Inc. Transforming growth factor beta (TGF-beta) blocking agent-treated stem cell composition and method
FR2811323B1 (en) * 2000-07-07 2006-10-06 Fuma Tech Gmbh HYBRID MATERIAL, USE OF SAID HYBRID MATERIAL, AND METHOD OF MANUFACTURING THE SAME
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
US20080194022A1 (en) * 2000-08-03 2008-08-14 Clarke Michael F Isolation and use of solid tumor stem cells
US6984522B2 (en) * 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
CN1274072C (en) * 2000-08-03 2006-09-06 松下电器产业株式会社 Brushless motor and method of Manufacturing the brushless motor
US8044259B2 (en) 2000-08-03 2011-10-25 The Regents Of The University Of Michigan Determining the capability of a test compound to affect solid tumor stem cells
US8568766B2 (en) 2000-08-24 2013-10-29 Gattadahalli M. Anantharamaiah Peptides and peptide mimetics to treat pathologies associated with eye disease
KR100916949B1 (en) * 2000-08-30 2009-09-14 에이브이아이 바이오파마 인코포레이티드 Method for analysis of oligonucleotide analogs
AU2001290629A1 (en) * 2000-09-07 2002-03-22 Boehringer Ingelheim International G.M.B.H Heat shock response and virus replication
DE60129719T2 (en) * 2000-09-14 2008-06-05 The Mount Sinai School Of Medicine SCREENING METHOD FOR IDENTIFYING G-PROTEINS AND OTHER COMPOUNDS WHICH INFLUENCE THE PHOSPHODIESTERASE (PDE) ACTIVITY
EP1191097A1 (en) * 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
US20030044803A1 (en) * 2000-09-22 2003-03-06 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of JAK1
US20020164576A1 (en) * 2000-09-22 2002-11-07 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of Nrf2
US20020115058A1 (en) * 2000-09-22 2002-08-22 Pedersen Finn Skou Methods for diagnosis and treatment of diseases associated with altered expression of Pik3r1
US20020123474A1 (en) * 2000-10-04 2002-09-05 Shannon Mark E. Human GTP-Rho binding protein2
EP2336166A1 (en) 2000-10-12 2011-06-22 University Of Rochester Compositions that inhibit proliferation of cancer cells
US7045283B2 (en) 2000-10-18 2006-05-16 The Regents Of The University Of California Methods of high-throughput screening for internalizing antibodies
EP1736760A3 (en) 2000-12-11 2008-06-18 President And Fellows Of Harvard College Nanosensors
US7645441B2 (en) 2000-12-22 2010-01-12 Sagres Discovery Inc. Compositions and methods in cancer associated with altered expression of PRLR
US7820447B2 (en) * 2000-12-22 2010-10-26 Sagres Discovery Inc. Compositions and methods for cancer
US20030087252A1 (en) * 2000-12-22 2003-05-08 Morris David W. Novel compositions and methods in cancer associated with altered expression of PRDM11
US20030165878A1 (en) * 2000-12-22 2003-09-04 Morris David W. Novel compositions and methods in cancer associated with altered expression of MCM3AP
US20030232334A1 (en) * 2000-12-22 2003-12-18 Morris David W. Novel compositions and methods for cancer
US7700274B2 (en) * 2000-12-22 2010-04-20 Sagres Discovery, Inc. Compositions and methods in cancer associated with altered expression of KCNJ9
US7892730B2 (en) * 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US20030099963A1 (en) * 2000-12-22 2003-05-29 Morris David W. Novel compositions and methods in cancer associated with altered expression of TBX21
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7279324B2 (en) * 2001-01-23 2007-10-09 Duke University Nucleic acid encoding G-protein coupled receptor with modified DRY motif
US20030036854A1 (en) * 2001-02-06 2003-02-20 The Penn State Research Foundation Apparatus and method for designing proteins and protein libraries
US6573051B2 (en) * 2001-03-09 2003-06-03 Molecular Staging, Inc. Open circle probes with intramolecular stem structures
US7294472B2 (en) * 2001-03-14 2007-11-13 Caden Biosciences Method for identifying modulators of G protein coupled receptor signaling
ES2328796T3 (en) 2001-03-14 2009-11-18 Myriad Genetics, Inc. TSG101-GAG INTERACTION AND USE OF THE SAME.
US7208279B2 (en) * 2001-03-14 2007-04-24 Caden Biosciences, Inc. Method for identifying inhibitors of G protein coupled receptor signaling
US7803982B2 (en) 2001-04-20 2010-09-28 The Mount Sinai School Of Medicine Of New York University T1R3 transgenic animals, cells and related methods
EP1572871A4 (en) * 2001-04-20 2007-11-14 Sinai School Medicine T1r3 a novel taste receptor
WO2002086147A2 (en) * 2001-04-24 2002-10-31 Pharmacia & Upjohn Company Single nucleotide polymorphisms diagnostic for schizophrenia
CA2446619C (en) 2001-05-11 2011-04-26 Elan Corporation, Plc Permeation enhancers
US7803915B2 (en) 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
DK2000545T3 (en) 2001-06-20 2011-11-28 Genentech Inc Compositions and methods for diagnosis and treatment of lung tumor
CA2451643C (en) 2001-06-21 2012-11-13 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
EP1925672A1 (en) 2001-06-22 2008-05-28 Syngeta Participations AG Abiotic stress responsive polynucleotides and polypeptides
US20040241683A1 (en) * 2001-07-06 2004-12-02 Ekker Stephen C Hsst and angiogenesis
US7393656B2 (en) * 2001-07-10 2008-07-01 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for risk stratification
US7381535B2 (en) 2002-07-10 2008-06-03 The Board Of Trustees Of The Leland Stanford Junior Methods and compositions for detecting receptor-ligand interactions in single cells
WO2003067210A2 (en) 2001-07-10 2003-08-14 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for detecting the activation state of the multiple proteins in single cells
EP2246438B1 (en) 2001-07-12 2019-11-27 Illumina, Inc. Multiplex nucleic acid reactions
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US7229774B2 (en) 2001-08-02 2007-06-12 Regents Of The University Of Michigan Expression profile of prostate cancer
US20040096880A1 (en) * 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
WO2003013437A2 (en) * 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
DE60220804D1 (en) * 2001-08-20 2007-08-02 Regenesis Bioremediation Produ BIOSENSOR FOR SMALL MOLECULAR ANALYTES
WO2003020980A2 (en) * 2001-08-28 2003-03-13 Pharmacia & Upjohn Company Single nucleotide polymorphisms diagnostic for schizophrenia
US20040115699A1 (en) * 2001-08-28 2004-06-17 Kaytes Paul S. Single nucleotide polymorphisms diagnostic for schizophrenia
US6852491B2 (en) 2001-09-04 2005-02-08 Abbott Laboratories Amplification and detection reagents for HIV-1
US7205048B2 (en) * 2001-09-17 2007-04-17 Invitrogen Corporation Functionalized fluorescent nanocrystal compositions and methods of making
EP2159044B1 (en) 2001-09-17 2012-05-16 Life Technologies Corporation Nanocrystals
US7214428B2 (en) * 2001-09-17 2007-05-08 Invitrogen Corporation Highly luminescent functionalized semiconductor nanocrystals for biological and physical applications
EP1721977A3 (en) 2001-09-17 2008-10-15 PDL BioPharma, Inc. Methods of diagnosis of cancer, compositions and methods of screening for modulators of cancer
CA2460120A1 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US20070098728A1 (en) * 2001-09-24 2007-05-03 Pedersen Finn S Novel compositions and methods in cancer
ATE441727T1 (en) 2001-10-01 2009-09-15 Sinai School Medicine NOONANIC SYNDROME GENE
ATE516364T1 (en) 2001-10-09 2011-07-15 Isis Pharmaceuticals Inc ANTISENSE MODULATION OF EXPRESSION OF THE INSULIN-LIKE GROWTH FACTOR BINDING PROTEY S 5
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
AU2004236174B2 (en) 2001-10-10 2011-06-02 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
ES2538342T3 (en) 2001-10-10 2015-06-19 Ratiopharm Gmbh Remodeling and glycoconjugation of follicle stimulating hormone (FSH)
EP2305311A3 (en) 2001-10-10 2011-07-20 BioGeneriX AG Glycoconjugation of peptides
US8323903B2 (en) * 2001-10-12 2012-12-04 Life Technologies Corporation Antibody complexes and methods for immunolabeling
US20050069962A1 (en) 2001-10-12 2005-03-31 Archer Robert M Antibody complexes and methods for immunolabeling
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
EP1436426A4 (en) * 2001-10-24 2005-11-02 Singulex Inc Methods for detecting genetic haplotypes by interaction with probes
US20030224380A1 (en) * 2001-10-25 2003-12-04 The General Hospital Corporation Genes and polymorphisms on chromosome 10 associated with Alzheimer's disease and other neurodegenerative diseases
AU2002364945A1 (en) * 2001-10-25 2003-07-09 Neurogenetics, Inc. Genes and polymorphisms on chromosome 10 associated with alzheimer's disease and other neurodegenerative diseases
US20030170678A1 (en) * 2001-10-25 2003-09-11 Neurogenetics, Inc. Genetic markers for Alzheimer's disease and methods using the same
EP1438417B1 (en) * 2001-10-26 2014-05-14 Danisco US Inc. Phytase enzymes, nucleic acid sequences encoding phytase enzymes and vectors and host cells incorporating same
US7510831B2 (en) * 2001-10-26 2009-03-31 Genencor International, Inc. Trichoderma reesei phytase enzymes, nucleic acids encoding such phytase enzymes, vectors and host cells incorporating same and methods of making and using same
WO2003042226A2 (en) * 2001-11-09 2003-05-22 Pharmacia & Upjohn Company Single nucleotide polymorphisms in gh-1
US20060040262A1 (en) * 2002-12-27 2006-02-23 Morris David W Novel compositions and methods in cancer
US20040180344A1 (en) * 2003-03-14 2004-09-16 Morris David W. Novel therapeutic targets in cancer
US20040197778A1 (en) * 2002-12-26 2004-10-07 Sagres Discovery, Inc. Novel compositions and methods in cancer
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
JP2005525095A (en) 2002-01-02 2005-08-25 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
EP1953244A1 (en) 2002-01-09 2008-08-06 Nakamura, Yusuke Cancer profiles
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) * 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US7553619B2 (en) 2002-02-08 2009-06-30 Qiagen Gmbh Detection method using dissociated rolling circle amplification
US7499806B2 (en) * 2002-02-14 2009-03-03 Illumina, Inc. Image processing in microsphere arrays
US20030191075A1 (en) * 2002-02-22 2003-10-09 Cook Phillip Dan Method of using modified oligonucleotides for hepatic delivery
JP2005518212A (en) 2002-02-25 2005-06-23 ジェネンテック・インコーポレーテッド Novel type 1 cytokine receptor GLM-R
US20030182669A1 (en) * 2002-03-19 2003-09-25 Rockman Howard A. Phosphoinositide 3-kinase mediated inhibition of GPCRs
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
AU2003218350A1 (en) 2002-03-21 2003-10-08 Sagres Discovery, Inc. Novel compositions and methods in cancer
US7332585B2 (en) 2002-04-05 2008-02-19 The Regents Of The California University Bispecific single chain Fv antibody molecules and methods of use thereof
US7026120B2 (en) 2002-04-15 2006-04-11 Abbott Laboratories Probes for detecting tumor cells
AU2003230874A1 (en) 2002-04-16 2003-11-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7622117B2 (en) * 2002-04-17 2009-11-24 Dynamis Therapeutics, Inc. 3-deoxyglucosone and skin
AU2003231121B2 (en) 2002-04-26 2008-08-07 University Of Utah Research Foundation Characterization of single stranded nucleic acids by melting analysis using a double strand specific nucleic acid dye
US7015317B2 (en) * 2002-05-02 2006-03-21 Abbott Laboratories Polynucleotides for the detection and quantification of hepatitis B virus nucleic acids
AU2003221575B2 (en) 2002-05-02 2010-06-10 Dalia Barsyte Teneurin C-terminal associated peptides (TCAP) and uses thereof
CA2484676A1 (en) 2002-05-03 2003-11-13 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof, and related methods
US7351542B2 (en) 2002-05-20 2008-04-01 The Regents Of The University Of California Methods of modulating tubulin deacetylase activity
US7176181B2 (en) * 2002-05-21 2007-02-13 Yeda Research And Development Co. Ltd. Compositions and methods of using galectin-8 as an inhibitor of tumor cell growth
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
CA2488048A1 (en) 2002-05-30 2003-12-11 Memorial Sloan-Kettering Cancer Center Kinase suppressor of ras inactivation for therapy of ras mediated tumorigenesis
US7290215B2 (en) * 2002-06-03 2007-10-30 Microsoft Corporation Dynamic wizard interface system and method
WO2003106617A2 (en) * 2002-06-12 2003-12-24 Tel Aviv Medical Center Research Development Fund Oligonucleotides antibodies and kits including same for treating prostate cancer and determining predisposition thereto
AU2003276131A1 (en) * 2002-06-18 2003-12-31 Epigenesis Pharmaceuticals, Inc. A dry powder oligonucleotide formulation, preparation and its uses
WO2004013160A2 (en) 2002-08-05 2004-02-12 University Of Rochester Protein transducing domain/deaminase chimeric proteins, related compounds, and uses thereof
ATE490309T1 (en) * 2002-08-12 2010-12-15 Danisco Us Inc MUTANT E. COLI-APPA PHYTASE ZYMES AND NATURAL VARIANTS THEREOF, NUCLEIC ACIDS ENCODING SUCH PHYTASE ZYMES, VECTORS AND HOST CELLS CONTAINING THE SAME, AND METHODS FOR THE PRODUCTION AND USE THEREOF
AU2003270582A1 (en) * 2002-09-12 2004-04-30 Massachusetts Institute Of Technology Rb pahtway and chromatin remodeling genes that antagonize let-60 ras signaling
BR0314236A (en) 2002-09-13 2005-08-09 Replicor Inc Oligonucleotide formulation, pharmaceutical composition, kit, antiviral compound, preparation of oligonucleotide and methods for selection of an antiviral oligonucleotide for use as an antiviral agent, for prophylaxis or treatment of a viral infection in a patient, for prophylactic treatment of cancer caused by oncoviruses. for identifying a compound that alters the binding of an oligonucleotide to at least one viral component, for purifying oligonucleotide binding to at least one viral component and for enriching oligonucleotides from an oligonucleotide cluster
EP1546170A4 (en) * 2002-09-20 2007-08-29 Univ Yale Riboswitches, methods for their use, and compositions for use with riboswitches
EP2272958A1 (en) 2002-09-26 2011-01-12 ISIS Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
US20040235005A1 (en) * 2002-10-23 2004-11-25 Ernest Friedlander Methods and composition for detecting targets
CA2504929C (en) 2002-11-05 2014-07-22 Charles Allerson Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
WO2004044138A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US9150605B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US9150606B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US7696345B2 (en) * 2002-11-05 2010-04-13 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
US7807802B2 (en) 2002-11-12 2010-10-05 Abbott Lab Polynucleotides for the amplification and detection of Chlamydia trachomatis and Neisseria gonorrhoeae
DK1569695T3 (en) 2002-11-13 2013-08-05 Genzyme Corp ANTISENSE MODULATION OF APOLIPOPROTEIN-B EXPRESSION
JP4986109B2 (en) 2002-11-13 2012-07-25 ジェンザイム・コーポレーション Antisense regulation of apolipoprotein B expression
US20060009378A1 (en) * 2002-11-14 2006-01-12 Itshak Golan Novel galectin sequences and compositions and methods utilizing same for treating or diagnosing arthritis and other chronic inflammatory diseases
CA2506127C (en) 2002-11-15 2013-07-09 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
AU2003298650B2 (en) 2002-11-15 2010-03-11 Musc Foundation For Research Development Complement receptor 2 targeted complement modulators
AU2003298672A1 (en) * 2002-11-19 2005-01-28 Singulex, Inc. Detection of target molecules through interaction with probes
EP1624753B1 (en) 2002-11-21 2012-01-25 The University of Utah Research Foundation Purinergic modulation of smell
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
AU2003299441A1 (en) * 2002-12-19 2004-07-14 Centre National De La Recherche Scientifique - Cnrs Nf-hev compositions and methods of use
US9487823B2 (en) 2002-12-20 2016-11-08 Qiagen Gmbh Nucleic acid amplification
AU2003299694A1 (en) 2002-12-20 2004-07-22 Qiagen Gmbh Nucleic acid amplification
US6977153B2 (en) * 2002-12-31 2005-12-20 Qiagen Gmbh Rolling circle amplification of RNA
JP4177123B2 (en) * 2003-01-10 2008-11-05 富士通株式会社 Wiring pattern verification method, program and apparatus
US9169516B2 (en) 2003-01-24 2015-10-27 University Of Utah Research Foundation Methods of predicting mortality risk by determining telomere length
DK1597366T3 (en) 2003-02-11 2013-02-25 Antisense Therapeutics Ltd Modulation of expression of insulin-like growth factor receptor I
US7002006B2 (en) * 2003-02-12 2006-02-21 Isis Pharmaceuticals, Inc. Protection of nucleosides
US7767387B2 (en) * 2003-06-13 2010-08-03 Sagres Discovery, Inc. Therapeutic targets in cancer
EP2058408A3 (en) 2003-02-14 2009-09-09 Sagres Discovery, Inc. Therapeutic GPCR targets in cancer
US20040170982A1 (en) 2003-02-14 2004-09-02 Morris David W. Novel therapeutic targets in cancer
US6943768B2 (en) 2003-02-21 2005-09-13 Xtellus Inc. Thermal control system for liquid crystal cell
CA2517074A1 (en) 2003-02-27 2004-09-10 Yeda Research And Development Co., Ltd. Nucleic acid molecules, polypeptides, antibodies and compositions containing same useful for treating and detecting influenza virus infection
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
CA2524255C (en) 2003-03-21 2014-02-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
US8043834B2 (en) 2003-03-31 2011-10-25 Qiagen Gmbh Universal reagents for rolling circle amplification and methods of use
JP2007521791A (en) 2003-04-01 2007-08-09 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
US20050043510A1 (en) * 2003-04-10 2005-02-24 Moeckli Randolph A. Affinity purification system using troponin molecules as affinity ligands
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US20040219533A1 (en) * 2003-04-29 2004-11-04 Jim Davis Biological bar code
EP1629105B1 (en) 2003-04-29 2010-09-01 AVI BioPharma, Inc. Compositions for enhancing transport and antisense efficacy of nucleic acid analog into cells
US7709610B2 (en) 2003-05-08 2010-05-04 Facet Biotech Corporation Therapeutic use of anti-CS1 antibodies
US20050025763A1 (en) 2003-05-08 2005-02-03 Protein Design Laboratories, Inc. Therapeutic use of anti-CS1 antibodies
ATE446791T1 (en) * 2003-05-16 2009-11-15 Univ Laval CNS CHLORIDE MODULATION AND USE THEREOF
US7960355B2 (en) * 2003-05-23 2011-06-14 Isis Pharmaceuticals, Inc. Compositions and methods for the modulation of the expression of B7 protein
WO2004106360A2 (en) * 2003-05-23 2004-12-09 Mount Sinai School Of Medicine Of New York University Viral vectors with improved properties
US7897582B2 (en) * 2003-05-23 2011-03-01 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
US7276599B2 (en) * 2003-06-02 2007-10-02 Isis Pharmaceuticals, Inc. Oligonucleotide synthesis with alternative solvents
AU2004253455B2 (en) 2003-06-03 2011-03-03 Eli Lilly And Company Modulation of survivin expression
US20050239089A1 (en) * 2003-06-06 2005-10-27 Johnson Martin D Mobility cassettes
US20060019256A1 (en) * 2003-06-09 2006-01-26 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
GB2417727B (en) 2003-06-13 2008-01-16 Alnylam Europe Ag Double-stranded ribonucleic acid with increased effectiveness in an organism
US20040259100A1 (en) 2003-06-20 2004-12-23 Illumina, Inc. Methods and compositions for whole genome amplification and genotyping
US7572581B2 (en) * 2003-06-30 2009-08-11 Roche Molecular Systems, Inc. 2′-terminator nucleotide-related methods and systems
US7947817B2 (en) * 2003-06-30 2011-05-24 Roche Molecular Systems, Inc. Synthesis and compositions of 2'-terminator nucleotides
WO2005017109A2 (en) * 2003-06-30 2005-02-24 Massachusetts Institute Of Technology Nucleic acids and polypeptides required for cell survival in the absence of rb
US20090088982A1 (en) * 2003-07-31 2009-04-02 Fukushima Noelle H Co-detection of single polypeptide and polynucleotide molecules
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
CA2534382C (en) 2003-08-04 2018-12-11 The Hospital For Sick Children Epm2b gene mutations associated with lafora's disease
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
CA2903196A1 (en) 2003-08-27 2005-03-10 Ophthotech Corporation Combination therapy for the treatment of ocular neovascular disorders
US20050053981A1 (en) * 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US20070123480A1 (en) * 2003-09-11 2007-05-31 Replicor Inc. Oligonucleotides targeting prion diseases
SG146682A1 (en) 2003-09-18 2008-10-30 Isis Pharmaceuticals Inc Modulation of eif4e expression
EP1677822B1 (en) * 2003-09-18 2014-04-23 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
US7125945B2 (en) * 2003-09-19 2006-10-24 Varian, Inc. Functionalized polymer for oligonucleotide purification
US20050222068A1 (en) * 2003-10-23 2005-10-06 Mourich Dan V Method and antisense composition for selective inhibition of HIV infection in hematopoietic cells
US20070281896A1 (en) * 2003-09-30 2007-12-06 Morris David W Novel compositions and methods in cancer
US7662929B2 (en) 2003-10-10 2010-02-16 Alchemia Oncology Pty Limited Antibody that specifically binds hyaluronan synthase
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
CA2747871C (en) 2003-11-17 2018-04-10 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
NZ548204A (en) 2003-12-03 2009-08-28 Coda Therapeutics Nz Ltd Antisense compounds targeted to connexins and methods of use thereof
US7259258B2 (en) * 2003-12-17 2007-08-21 Illumina, Inc. Methods of attaching biological compounds to solid supports using triazine
WO2005060675A2 (en) * 2003-12-19 2005-07-07 Philadelphia Health And Education Corporation(D/B/A Drexel University College Of Medicine (Ducom)) Novel spliced 5-ht1a receptors and methods, kits, and uses relating thereto
US20050136414A1 (en) * 2003-12-23 2005-06-23 Kevin Gunderson Methods and compositions for making locus-specific arrays
WO2005067646A2 (en) 2004-01-07 2005-07-28 Hitachi Chemical Research Center, Inc. Primers and probes for the detection of hiv
JP2007520474A (en) * 2004-01-12 2007-07-26 ジーンセンス テクノロジーズ インコーポレーテッド Antisense oligonucleotides directed to ribonucleotide reductase R2 and its use in combination therapy for the treatment of cancer
WO2006054296A2 (en) 2004-11-17 2006-05-26 Spectrum Dynamics Llc Methods of detecting prostate cancer
WO2005071080A2 (en) 2004-01-20 2005-08-04 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US8778900B2 (en) * 2004-01-22 2014-07-15 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP1 expression
US7468431B2 (en) * 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
WO2005072527A2 (en) * 2004-01-23 2005-08-11 Avi Biopharma, Inc. Antisense oligomers and methods for inducing immune tolerance and immunosuppression
EP2816351A3 (en) * 2004-01-27 2015-03-25 Compugen Ltd. Methods and systems for annotating biomolecular sequences
US7842459B2 (en) * 2004-01-27 2010-11-30 Compugen Ltd. Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis
WO2005074633A2 (en) * 2004-02-03 2005-08-18 The Regents Of The University Of Michigan Compositions and methods for characterizing, regulating, diagnosing, and treating cancer
US7481997B1 (en) 2004-02-12 2009-01-27 Montana State University Snow mountain virus genome sequence, virus-like particles and methods of use
WO2005082110A2 (en) * 2004-02-26 2005-09-09 Illumina Inc. Haplotype markers for diagnosing susceptibility to immunological conditions
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
EP2700720A3 (en) 2004-03-15 2015-01-28 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNASE H
AU2005230684B2 (en) 2004-04-05 2011-10-06 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
US20050260755A1 (en) * 2004-04-06 2005-11-24 Isis Pharmaceuticals, Inc. Sequential delivery of oligomeric compounds
US20050260652A1 (en) * 2004-04-15 2005-11-24 The General Hospital Corporation Compositions and methods that modulate RNA interference
CA2562151C (en) 2004-04-30 2016-09-06 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a c5-modified pyrimidine
EP2322650A1 (en) 2004-05-14 2011-05-18 Rosetta Genomics Ltd MicroRNAs and uses thereof
EP2325331A1 (en) 2004-05-14 2011-05-25 Rosetta Genomics Ltd MicroRNAs and Uses Thereof
AU2005244036A1 (en) * 2004-05-14 2005-11-24 Universite De Laval Phospholipase C gamma modulation and uses thereof for management of pain and nociception
AU2005245996A1 (en) 2004-05-21 2005-12-01 Atonomics A/S Surface acoustic wave sensor comprising a hydrogel
EP3225633B1 (en) 2004-05-21 2020-03-25 The UAB Research Foundation Variable lymphocyte receptors, related polypeptides and nucleic acids, and uses thereof
WO2005116081A2 (en) 2004-05-24 2005-12-08 Genvault Corporation Stable protein storage and stable nucleic acid storage in recoverable form
US7524827B2 (en) 2004-06-01 2009-04-28 Pronai Therapeutics, Inc. Methods and compositions for the inhibition of gene expression
US20090048192A1 (en) * 2004-06-03 2009-02-19 Isis Pharmaceuticals, Inc. Double Strand Compositions Comprising Differentially Modified Strands for Use in Gene Modulation
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
JP2008509681A (en) * 2004-06-09 2008-04-03 マクギル・ユニバーシテイ Polynucleotides encoding the acetylcholine-dependent chloride channel subunit of CENORHABDITISELEGANS
US7858323B2 (en) 2004-06-09 2010-12-28 The Regents Of The University Of Michigan Phage microarray profiling of the humoral response to disease
US7745125B2 (en) * 2004-06-28 2010-06-29 Roche Molecular Systems, Inc. 2′-terminator related pyrophosphorolysis activated polymerization
USRE48960E1 (en) 2004-06-28 2022-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
EP2500430B1 (en) 2004-06-28 2017-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
US8314072B2 (en) 2004-07-02 2012-11-20 Avi Biopharma, Inc. Antisense antibacterial method and compound
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
WO2006033732A1 (en) * 2004-08-17 2006-03-30 Invitrogen Corporation Synthesis of highly luminescent colloidal particles
WO2006023880A2 (en) * 2004-08-23 2006-03-02 Isis Pharmaceuticals, Inc. Compounds and methods for the characterization of oligonucleotides
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US8129352B2 (en) * 2004-09-16 2012-03-06 Avi Biopharma, Inc. Antisense antiviral compound and method for treating ssRNA viral infection
WO2006030442A2 (en) 2004-09-16 2006-03-23 Gamida-Cell Ltd. Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
DK1809303T3 (en) 2004-09-23 2019-06-11 Arc Medical Devices Inc PHARMACEUTICAL COMPOSITION AND METHODS IN RELATION TO FIBROSE ADHESION OR INFLAMMATORIC DISEASE USING LOW SULPHATE FUCAN
ATE435025T1 (en) * 2004-10-18 2009-07-15 Sinai School Medicine INHIBITION OF TUMOR GROWTH AND METASTASIS FORMATION WITH PEPTIDES DERIVED FROM ATF2
US7368086B2 (en) * 2004-10-29 2008-05-06 Invitrogen Corporation Functionalized fluorescent nanocrystals, and methods for their preparation and use
US7524829B2 (en) * 2004-11-01 2009-04-28 Avi Biopharma, Inc. Antisense antiviral compounds and methods for treating a filovirus infection
US20090118132A1 (en) * 2004-11-04 2009-05-07 Roche Molecular Systems, Inc. Classification of Acute Myeloid Leukemia
US8440610B2 (en) * 2004-11-12 2013-05-14 Massachusetts Institute Of Technology Mapkap kinase-2 as a specific target for blocking proliferation of P53-defective cells
WO2006050999A2 (en) * 2004-11-15 2006-05-18 Obe Therapy Biotechnology S.A.S Methods of reducing body fat
US7842794B2 (en) 2004-12-17 2010-11-30 Roche Molecular Systems, Inc. Reagents and methods for detecting Neisseria gonorrhoeae
DE102004063599B4 (en) 2004-12-30 2007-07-12 Bayer Innovation Gmbh Shortened wound healing processes by means of novel fiber fleeces
KR20070104574A (en) * 2004-12-30 2007-10-26 신벤션 아게 Combination comprising an agent providing a signal, an implant material and a drug
WO2006076650A2 (en) * 2005-01-12 2006-07-20 Applera Corporation Compositions, methods, and kits for selective amplification of nucleic acids
EP2014708A1 (en) * 2005-01-13 2009-01-14 Cinvention Ag Shaped body made of a composite material
KR20070102717A (en) * 2005-01-24 2007-10-19 신벤션 아게 Metal containing composite materials
AU2006257283A1 (en) 2005-02-03 2006-12-21 Coda Therapeutics Limited Anti-connexin compounds and uses thereof
CA2596506C (en) 2005-02-09 2021-04-06 Avi Biopharma, Inc. Antisense composition and method for treating muscle atrophy
WO2006123246A2 (en) * 2005-02-11 2006-11-23 Aurelium Biopharma Inc. Methods for identifying chemotherapeutic resistance in non-hematopoietic tumors
RU2007137489A (en) 2005-03-10 2009-04-20 Дженентек, Инк. (Us) METHODS AND COMPOSITIONS FOR MODULATION OF VESSEL INTEGRITY
JP2008532913A (en) * 2005-03-18 2008-08-21 シンベンション アーゲー Process for the preparation of porous sintered metal materials
US20060256599A1 (en) * 2005-03-22 2006-11-16 Malin Patricia J Database of electronically profiled cells and methods for generating and using same
US7476733B2 (en) 2005-03-25 2009-01-13 The United States Of America As Represented By The Department Of Health And Human Services Development of a real-time PCR assay for detection of pneumococcal DNA and diagnosis of pneumococccal disease
US20060240032A1 (en) * 2005-03-31 2006-10-26 Hinrichs David J Immunomodulating compositions and methods for use in the treatment of human autoimmune diseases
US8309303B2 (en) 2005-04-01 2012-11-13 Qiagen Gmbh Reverse transcription and amplification of RNA with simultaneous degradation of DNA
CA2605512A1 (en) * 2005-04-22 2006-10-26 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the binding of sr proteins and by interfering with secondary rna structure.
US7838502B2 (en) * 2005-05-06 2010-11-23 University Of Massachusetts Medical School Compositions and methods to modulate H. influenzae pathogenesis
EP2298829B1 (en) 2005-05-31 2017-09-20 École Polytechnique Fédérale de Lausanne (EPFL) Triblock copolymers for cytoplasmic delivery of gene-based drugs
EP1904111A4 (en) 2005-06-03 2009-08-19 Univ Johns Hopkins Compositions and methods for decreasing microrna expression for the treatment of neoplasia
US20070105133A1 (en) * 2005-06-13 2007-05-10 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
WO2006135886A2 (en) * 2005-06-13 2006-12-21 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
US20090264299A1 (en) * 2006-02-24 2009-10-22 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
EP3492602A1 (en) 2005-06-15 2019-06-05 Complete Genomics, Inc. Single molecule arrays for genetic and chemical analysis
US8067208B2 (en) 2005-06-30 2011-11-29 Roche Molecular Systems, Inc. Probes and methods for hepatitis C virus typing using multidimensional probe analysis
MX2008000131A (en) * 2005-07-01 2008-04-04 Cinv Ag Medical devices comprising a reticulated composite material.
EP1902087A1 (en) * 2005-07-01 2008-03-26 Cinvention Ag Process for the production of porous reticulated composite materials
WO2007007317A1 (en) 2005-07-07 2007-01-18 Yissum Research Development Company Of The Hebrew University Of Jerusalem Nucleic acid agents for downregulating h19, and methods of using same
WO2007009094A2 (en) 2005-07-13 2007-01-18 Avi Biopharma, Inc. Antisense antibacterial method and compound
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US7790694B2 (en) * 2005-07-13 2010-09-07 Avi Biopharma Inc. Antisense antibacterial method and compound
US7776532B2 (en) 2005-08-11 2010-08-17 Synthetic Genomics, Inc. Method for in vitro recombination
US20100015604A1 (en) 2005-08-17 2010-01-21 Evriklia Lianidou Composition and method for determination of ck19 expression
EP2338991B1 (en) 2005-08-29 2017-01-18 Regulus Therapeutics Inc. Methods for use in modulating MIR-122a
WO2007030691A2 (en) * 2005-09-08 2007-03-15 Avi Biopharma, Inc. Antisense antiviral compound and method for treating picornavirus infection
US8329668B2 (en) 2005-09-08 2012-12-11 Avi Biopharma, Inc. Antisense antiviral compound and method for treating picornavirus infection
EP1762627A1 (en) 2005-09-09 2007-03-14 Qiagen GmbH Method for the activation of a nucleic acid for performing a polymerase reaction
EP2612870A1 (en) 2005-09-12 2013-07-10 The Regents of the University of Michigan Recurrent gene fusions in prostate cancer
US8906609B1 (en) 2005-09-26 2014-12-09 Arrowhead Center, Inc. Label-free biomolecule sensor based on surface charge modulated ionic conductance
CN101321877B (en) 2005-10-03 2013-04-10 应用生物系统有限责任公司 Compositions, methods, and kits for amplifying nucleic acids
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for diagnosis of preeclamsia
EP2392645A1 (en) 2005-10-14 2011-12-07 MUSC Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
US8080534B2 (en) 2005-10-14 2011-12-20 Phigenix, Inc Targeting PAX2 for the treatment of breast cancer
US8652467B2 (en) * 2005-10-14 2014-02-18 The Regents Of The University Of Michigan Dek protein compositions and methods of using the same
JP2009511727A (en) * 2005-10-18 2009-03-19 シンベンション アーゲー Thermosetting particles and manufacturing method thereof
US7794951B2 (en) * 2005-10-18 2010-09-14 University Of Massachusetts Medical School SREBP2gc transcription factors and uses thereof
JP5671206B2 (en) 2005-10-21 2015-02-18 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Mutation of c-KIT oncogene in melanoma
CA2627025A1 (en) 2005-10-28 2007-05-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of huntingtin gene
US7723112B2 (en) * 2005-10-31 2010-05-25 The Regents Of The University Of Michigan Compositions and methods for treating and diagnosing cancer
EP1961065A4 (en) 2005-10-31 2009-11-11 Oncomed Pharm Inc Compositions and methods for diagnosing and treating cancer
US7723477B2 (en) * 2005-10-31 2010-05-25 Oncomed Pharmaceuticals, Inc. Compositions and methods for inhibiting Wnt-dependent solid tumor cell growth
WO2007056326A2 (en) * 2005-11-04 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of nav1.8 gene
US8501704B2 (en) 2005-11-08 2013-08-06 Sarepta Therapeutics, Inc. Immunosuppression compound and treatment method
WO2007056331A2 (en) 2005-11-09 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor v leiden mutant gene
CA2630602A1 (en) 2005-11-21 2007-05-31 Isis Pharmaceuticals, Inc. Modulation of eif4e-bp2 expression
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
WO2007064945A2 (en) 2005-12-01 2007-06-07 Pronai Therapeutics, Inc. Cancer therapies and pharmaceutical compositions used therein
US20090215055A1 (en) * 2005-12-13 2009-08-27 Erasmus University Medical Center Rotterdam Genetic Brain Tumor Markers
ES2433669T3 (en) 2005-12-13 2013-12-12 The Trustees Of The University Of Pennsylvania Methods for phototransfecting nucleic acid in living cells
US9157066B2 (en) 2005-12-13 2015-10-13 The Trustees Of The University Of Pennsylvania Transcriptome transfer produces cellular phenotype conversion
US10647960B2 (en) * 2005-12-13 2020-05-12 The Trustees Of The University Of Pennsylvania Transcriptome transfer produces cellular phenotype conversion
US8313901B2 (en) * 2005-12-21 2012-11-20 Yale University Methods and compositions related to the modulation of riboswitches
EP1976567B1 (en) * 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
EP2216339A1 (en) 2006-01-16 2010-08-11 Compugen Ltd. Novel nucleotide and amino acid sequences, and methods of use thereof for diagnosis
CN101426536A (en) 2006-01-20 2009-05-06 妇女儿童健康研究院公司 Method of treatment, prophylaxis and diagnosis of pathologies of the bone
SI2161038T1 (en) 2006-01-26 2014-05-30 Isis Pharmaceuticals, Inc. Compositions and their uses directed to Huntingtin
DE602007009487D1 (en) * 2006-01-27 2010-11-11 Isis Pharmaceutical Inc 6-MODIFIED BICYCLIC NUCLEIC ACID ANALOGUE
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
US8129515B2 (en) 2006-01-27 2012-03-06 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for the use in modulation of microRNAs
US8229398B2 (en) * 2006-01-30 2012-07-24 Qualcomm Incorporated GSM authentication in a CDMA network
US20080019961A1 (en) * 2006-02-21 2008-01-24 Regents Of The University Of Michigan Hedgehog signaling pathway antagonist cancer treatment
EP2495337A1 (en) 2006-02-24 2012-09-05 Callida Genomics, Inc. High throughput genome sequencing on DNA arrays
EP2010677A4 (en) * 2006-02-24 2010-04-14 Investigen Inc Methods and compositions for detecting polynucleotides
NZ571791A (en) 2006-03-08 2012-03-30 Archemix Llc Complement binding aptamers and anti-C5 agents useful in the treatment of ocular disorders
US7914988B1 (en) * 2006-03-31 2011-03-29 Illumina, Inc. Gene expression profiles to predict relapse of prostate cancer
AU2007233109B2 (en) 2006-03-31 2010-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 gene
US8007790B2 (en) 2006-04-03 2011-08-30 Stowers Institute For Medical Research Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
US20100055116A1 (en) 2006-04-13 2010-03-04 Liou Hsiou-Chi Methods and Compositions for Targeting c-Rel
WO2007123391A1 (en) * 2006-04-20 2007-11-01 Academisch Ziekenhuis Leiden Therapeutic intervention in a genetic disease in an individual by modifying expression of an aberrantly expressed gene.
CN101437943A (en) 2006-05-03 2009-05-20 波罗的科技发展有限公司 Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease
EP2015758B1 (en) 2006-05-05 2014-04-02 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression apob
HUE036995T2 (en) * 2006-05-10 2018-08-28 Sarepta Therapeutics Inc Oligonucleotide analogs having cationic intersubunit linkages
US8785407B2 (en) * 2006-05-10 2014-07-22 Sarepta Therapeutics, Inc. Antisense antiviral agent and method for treating ssRNA viral infection
CA2651815A1 (en) * 2006-05-10 2007-11-22 Dxterity Diagnostics Detection of nucleic acid targets using chemically reactive oligonucleotide probes
CN101490074B (en) * 2006-05-11 2013-06-26 Isis制药公司 5'-modified bicyclic nucleic acid analogs
US20070265215A1 (en) * 2006-05-11 2007-11-15 Iversen Patrick L Antisense restenosis composition and method
PL2194128T3 (en) 2006-05-11 2012-12-31 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the PCSK9 gene
US7666854B2 (en) * 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
BRPI0712034A2 (en) * 2006-05-19 2012-01-10 Alnylam Pharmaceuticals Inc aha rnai modulation and therapeutic uses thereof
EP1857548A1 (en) * 2006-05-19 2007-11-21 Academisch Ziekenhuis Leiden Means and method for inducing exon-skipping
EP2192200B1 (en) 2006-05-22 2012-10-24 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
WO2007137301A2 (en) * 2006-05-23 2007-11-29 Isis Pharmaceuticals, Inc. Modulation of chrebp expression
US11001881B2 (en) 2006-08-24 2021-05-11 California Institute Of Technology Methods for detecting analytes
US8637436B2 (en) 2006-08-24 2014-01-28 California Institute Of Technology Integrated semiconductor bioarray
EP2489745B1 (en) 2006-06-05 2017-01-11 California Institute Of Technology Real time micro arrays
US9115389B2 (en) * 2006-06-30 2015-08-25 Rosetta Genomics Ltd. Method for detecting a target nucleic acid comprising two portions using probes having a first portion complementary to the first portion of the target nucleic acid and a second portion substantially complementary to the second portion of the target nucleic acid
WO2008011473A2 (en) 2006-07-19 2008-01-24 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hbxip
US8048626B2 (en) 2006-07-28 2011-11-01 California Institute Of Technology Multiplex Q-PCR arrays
EP2412383A1 (en) 2006-07-28 2012-02-01 Children's Memorial Hospital Methods of inhibiting tumor cell aggressiveness using the microenvironment of human embryonic stem cells
US11525156B2 (en) 2006-07-28 2022-12-13 California Institute Of Technology Multiplex Q-PCR arrays
EP2049664B1 (en) 2006-08-11 2011-09-14 Prosensa Technologies B.V. Single stranded oligonucleotides complementary to repetitive elements for treating DNA repeat instability associated genetic disorders
US11560588B2 (en) 2006-08-24 2023-01-24 California Institute Of Technology Multiplex Q-PCR arrays
US20080261216A1 (en) * 2006-09-08 2008-10-23 The Regents Of The University Of Michigan HERV Group II Viruses In Lymphoma And Cancer
JP2010502752A (en) * 2006-09-11 2010-01-28 イェール ユニバーシティー Lysine riboswitch, structure-based compound design using lysine riboswitch, and methods and compositions for use with lysine riboswitch
WO2008073175A2 (en) 2006-09-14 2008-06-19 The Regents Of The University Of California Nanoplasmonic molecular ruler for nuclease activity and dna footprinting
WO2008036933A2 (en) 2006-09-21 2008-03-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the hamp gene
CA2927045A1 (en) 2006-10-03 2008-04-10 Muthiah Manoharan Lipid containing formulations
US20100166743A1 (en) 2006-10-06 2010-07-01 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
EP1914303A1 (en) * 2006-10-09 2008-04-23 Qiagen GmbH Thermus eggertssonii DNA polymerases
US20080096193A1 (en) * 2006-10-24 2008-04-24 Charles Robert Bupp Methods and compositions for detecting polynucleotides
JP2010507595A (en) 2006-10-27 2010-03-11 ザ・ユニヴァーシティ・オヴ・ウェスタン・オンタリオ Inhibition of SOX9 function in the treatment of pathophysiological conditions associated with proteoglycans
US7910354B2 (en) 2006-10-27 2011-03-22 Complete Genomics, Inc. Efficient arrays of amplified polynucleotides
US8999317B2 (en) 2006-11-01 2015-04-07 University Of Rochester Methods and compositions related to the structure and function of APOBEC3G
US20090111705A1 (en) * 2006-11-09 2009-04-30 Complete Genomics, Inc. Selection of dna adaptor orientation by hybrid capture
CA2669832A1 (en) * 2006-11-15 2008-05-22 Coda Therapeutics, Inc. Improved methods and compositions for wound healing
US20080242560A1 (en) * 2006-11-21 2008-10-02 Gunderson Kevin L Methods for generating amplified nucleic acid arrays
AU2006351377A1 (en) * 2006-11-30 2008-06-05 University Of British Columbia Poxviridae treatment comprising TAP-1 and/or TAP-2 as a molecular adjuvant
WO2009024834A2 (en) * 2006-12-05 2009-02-26 Rosetta Genomics Ltd Nucleic acids involved in viral infection
CA2672297A1 (en) 2006-12-11 2008-06-19 University Of Utah Research Foundation Compositions and methods for treating pathologic angiogenesis and vascular permeability
US9938641B2 (en) * 2006-12-18 2018-04-10 Fluidigm Corporation Selection of aptamers based on geometry
US7858772B2 (en) * 2006-12-22 2010-12-28 Roche Molecular Systems, Inc. Compounds and methods for synthesis and purification of oligonucleotides
EP2097448A4 (en) 2006-12-22 2010-07-21 Univ Utah Res Found Method of detecting ocular diseases and pathologic conditions and treatment of same
US7820389B2 (en) * 2007-01-10 2010-10-26 Geneohm Sciences, Inc. Inhibition of mismatch hybridization by a universal competitor DNA
JP2010515464A (en) * 2007-01-11 2010-05-13 イエール・ユニバーシテイ Compositions and methods for targeted inactivation of HIV cell surface receptors
US20100086526A1 (en) * 2007-01-16 2010-04-08 Abraham Hochberg Nucleic acid constructs and methods for specific silencing of h19
BRPI0806916A2 (en) * 2007-01-19 2014-04-29 Cinv Ag NON-DEGRADABLE POROUS IMPLANT MADE BY DUST
EP2106439B1 (en) 2007-01-24 2014-11-12 The Regents of the University of Michigan Compositions and methods for treating and diagnosing pancreatic cancer
WO2008092099A2 (en) * 2007-01-26 2008-07-31 Rosetta Genomics, Ltd. Compositions and methods for treating hematopoietic malignancies
US20100196403A1 (en) * 2007-01-29 2010-08-05 Jacob Hochman Antibody conjugates for circumventing multi-drug resistance
AU2008212820B2 (en) 2007-02-09 2014-01-30 Northwestern University Particles for detecting intracellular targets
US7771947B2 (en) * 2007-02-23 2010-08-10 Investigen, Inc. Methods and compositions for rapid light-activated isolation and detection of analytes
AU2008220438A1 (en) * 2007-02-27 2008-09-04 Rosetta Genomics Ltd. Composition and methods for modulating cell proliferation and cell death
US9006206B2 (en) 2007-02-27 2015-04-14 Rosetta Genomics Ltd. Composition and methods for modulating cell proliferation and cell death
EP2129803A4 (en) * 2007-02-27 2010-11-03 Univ Northwestern Molecule attachment to nanoparticles
US20080206862A1 (en) * 2007-02-28 2008-08-28 Cinvention Ag High surface cultivation system bag
EP2126040A1 (en) * 2007-02-28 2009-12-02 Cinvention Ag High surface cultivation system
EP2126115A2 (en) 2007-03-01 2009-12-02 Rosetta Genomics Ltd Diagnosis and prognosis of various types of cancers
EP2471925A1 (en) 2007-03-22 2012-07-04 Yale University Methods and compositions related to riboswitches that control alternative splicing
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
WO2012070037A2 (en) 2010-11-22 2012-05-31 Rosetta Genomics Ltd. Methods and materials for classification of tissue of origin of tumor samples
US20100273172A1 (en) 2007-03-27 2010-10-28 Rosetta Genomics Ltd. Micrornas expression signature for determination of tumors origin
JP5350360B2 (en) 2007-03-29 2013-11-27 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting the expression of genes from Ebola
CA2690281A1 (en) 2007-05-11 2008-11-20 The Johns Hopkins University Biomarkers for melanoma
WO2008141176A1 (en) * 2007-05-11 2008-11-20 The Trustees Of The University Of Pennsylvania Methods of treatment of skin ulcers
EA200971050A1 (en) 2007-05-11 2010-06-30 Томас Джефферсон Юниверсити METHODS OF TREATMENT AND PREVENTION OF NEURODEGENERATIVE DISEASES AND DISORDERS
EP3351270A1 (en) 2007-05-23 2018-07-25 The Trustees of the University of Pennsylvania Targeted carriers for intracellular drug delivery
KR20100017905A (en) 2007-05-29 2010-02-16 예일 유니버시티 Riboswitches and methods and compositions for use of and with riboswitches
MX2009012647A (en) * 2007-05-29 2009-12-14 Univ Yale Methods and compositions related to riboswitches that control alternative splicing and rna processing.
EP2160464B1 (en) 2007-05-30 2014-05-21 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
US7807372B2 (en) * 2007-06-04 2010-10-05 Northwestern University Screening sequence selectivity of oligonucleotide-binding molecules using nanoparticle based colorimetric assay
ES2798758T3 (en) 2007-06-06 2020-12-14 Sarepta Therapeutics Inc Soluble her2 and her3 splice variant proteins, splice exchange oligonucleotides and their use in the treatment of disease
EP2173760B2 (en) 2007-06-08 2015-11-04 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
US20090324596A1 (en) * 2008-06-30 2009-12-31 The Trustees Of Princeton University Methods of identifying and treating poor-prognosis cancers
US10745701B2 (en) 2007-06-28 2020-08-18 The Trustees Of Princeton University Methods of identifying and treating poor-prognosis cancers
CA2691673A1 (en) 2007-06-29 2009-01-08 Avi Biopharma, Inc. Tissue specific peptide conjugates and methods
CA2692579C (en) * 2007-07-05 2016-05-03 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
EP2319926B1 (en) 2007-07-05 2016-08-31 Arrowhead Research Corporation DSRNA for treating viral infection
AU2008275304B2 (en) 2007-07-06 2012-07-26 The Regents Of The University Of Michigan Recurrent gene fusions in prostate cancer
WO2009008725A2 (en) * 2007-07-12 2009-01-15 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
JP5706157B2 (en) * 2007-07-12 2015-04-22 プロセンサ テクノロジーズ ビー.ブイ.Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs or tissues
CN101821277B (en) * 2007-08-15 2014-05-07 Isis制药公司 Tetrahydropyran nucleic acid analogs
CN101802182A (en) 2007-08-21 2010-08-11 诺达利蒂公司 Be used to diagnose, the method for prognosis and methods of treatment
US9422363B2 (en) 2007-08-28 2016-08-23 Uab Research Foundation Synthetic apolipoprotein E mimicking polypeptides and methods of use
CA2697957A1 (en) 2007-08-28 2009-03-12 Uab Research Foundation Synthetic apolipoprotein e mimicking polypeptides and methods of use
LT2769729T (en) 2007-09-04 2019-05-10 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
US8138318B2 (en) 2007-09-13 2012-03-20 Abbott Laboratories Hepatitis B pre-S2 nucleic acid
WO2009039192A2 (en) 2007-09-17 2009-03-26 The Regents Of The University Of Californina Internalizing human monoclonal antibodies targeting prostate cancer cells in situ
US8445217B2 (en) 2007-09-20 2013-05-21 Vanderbilt University Free solution measurement of molecular interactions by backscattering interferometry
US7951785B2 (en) * 2007-09-21 2011-05-31 California Institute Of Technology NFIA in glial fate determination, glioma therapy and astrocytoma treatment
JP2010539978A (en) 2007-10-02 2010-12-24 アムジェン インコーポレイテッド Increased erythropoietin using nucleic acids that can hybridize to micro-RNA and its precursors
JP5600064B2 (en) 2007-10-26 2014-10-01 アカデミシュ ジーケンハウス ライデン Means and methods for offsetting myopathy
USRE48468E1 (en) 2007-10-26 2021-03-16 Biomarin Technologies B.V. Means and methods for counteracting muscle disorders
US7820391B2 (en) 2007-11-06 2010-10-26 Osmetech Molecular Diagnostics Baseless nucleotide analogues and uses thereof
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
USRE47320E1 (en) 2007-11-20 2019-03-26 Ionis Pharmaceuticals, Inc. Modulation of CD40 expression
US8546556B2 (en) * 2007-11-21 2013-10-01 Isis Pharmaceuticals, Inc Carbocyclic alpha-L-bicyclic nucleic acid analogs
DK2610351T3 (en) 2007-12-05 2015-09-28 Complete Genomics Inc Base efficient provision of sequencing reactions
EP2848688A1 (en) 2007-12-10 2015-03-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor VII gene
PT2231861E (en) * 2007-12-13 2015-02-03 Philip Morris Products Sa Transgenic plants modified for reduced cadmium transport, derivative products, and related methods
AU2008343841A1 (en) 2007-12-21 2009-07-09 Coda Therapeutics, Inc. Use of inhibitors of connexin43 for treatment of fibrotic conditions
JP2011507859A (en) * 2007-12-21 2011-03-10 コーダ セラピューティクス, インコーポレイテッド Use of anti-connexin polynucleotides for the treatment of surgical adhesions
JP2011507861A (en) * 2007-12-21 2011-03-10 コーダ セラピューティクス, インコーポレイテッド Use of anti-connexin polypeptide agents in combination with anti-connexin polynucleotide agents for the treatment of fibrotic conditions
AU2008345033B2 (en) 2007-12-28 2014-04-03 Sarepta Therapeutics, Inc. Immunomodulatory agents and methods of use
EP3100718B1 (en) 2008-01-02 2019-11-27 Arbutus Biopharma Corporation Improved compositions and methods for the delivery of nucleic acids
US20110038920A1 (en) * 2008-01-07 2011-02-17 Ryoichi Mori Wound healing compositions and treatments
AU2009208035B2 (en) 2008-01-22 2014-04-03 Dogenes Inc. Compositions and methods for detecting Juvenile Renal Dysplasia or calcium oxalate stones in dogs
CA2708661A1 (en) 2008-01-27 2009-07-30 Rosetta Genomics Ltd. Methods and compositions for diagnosing complications of pregnancy
CA2713667A1 (en) * 2008-01-31 2009-08-06 Compugen Ltd. Cd55 isoform and uses therof in cancer detection, monitoring and therapy
US8530640B2 (en) * 2008-02-07 2013-09-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
AU2009210872A1 (en) * 2008-02-08 2009-08-13 Prosensa Holding Bv Methods and means for treating DNA repeat instability associated genetic disorders
KR101397407B1 (en) 2008-03-05 2014-06-19 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibiting expression of Eg5 and VEGF genes
US8426378B2 (en) 2008-03-21 2013-04-23 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucelosides and methods for their use
WO2009124238A1 (en) * 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
US8802923B2 (en) 2008-04-10 2014-08-12 A.B. Seeds Ltd. Compositions and methods for enhancing oil content in plants
DK2982753T3 (en) 2008-04-18 2018-09-03 Baxter Int Microsphere-based composition to prevent and / or reverse newly started autoimmune diabetes
US10150990B2 (en) 2008-04-21 2018-12-11 Roche Molecular Systems, Inc. Ribonucleotide tag nucleic acid detection
CA2722668A1 (en) * 2008-04-29 2009-11-05 Wyeth Llc Methods for treating inflammation
EP2119783A1 (en) 2008-05-14 2009-11-18 Prosensa Technologies B.V. Method for efficient exon (44) skipping in Duchenne Muscular Dystrophy and associated means
US8082730B2 (en) * 2008-05-20 2011-12-27 Caterpillar Inc. Engine system having particulate reduction device and method
CA2725138A1 (en) 2008-05-21 2009-11-26 Kinemed, Inc. Compositions and methods of treatment using modulators of motoneuron diseases
CA2635187A1 (en) 2008-06-05 2009-12-05 The Royal Institution For The Advancement Of Learning/Mcgill University Oligonucleotide duplexes and uses thereof
US20100209957A1 (en) * 2008-06-20 2010-08-19 Genvault Corporation Biosample storage devices and methods of use thereof
KR20110017005A (en) * 2008-07-15 2011-02-18 에프. 호프만-라 로슈 아게 Compositions and methods for inhibiting expression of tgf-beta receptor genes
WO2010007526A1 (en) 2008-07-17 2010-01-21 Andreas Pfuetzner Biomarkers for cardiodiabetes
ES2433691T3 (en) 2008-07-17 2013-12-12 Ikfe Institut Für Klinische Forschung Und Entwicklung Gmbh Biomarkers for insulin resistance and beta cell dysfunction
AU2009279682B2 (en) 2008-08-04 2015-01-22 University Of Miami STING (stimulator of interferon genes), a regulator of innate immune responses
JP5420668B2 (en) 2008-08-25 2014-02-19 エクスカリアード・ファーマシューティカルズ,インコーポレイテッド Antisense oligonucleotides for connective tissue growth factor and uses thereof
EP2331690B1 (en) 2008-09-02 2016-01-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of mutant egfr gene
ES2574137T3 (en) 2008-09-03 2016-06-15 Quantumdx Group Limited Strategies and methods for detecting nucleic acids by biosensors
JP5822726B2 (en) 2008-09-03 2015-11-24 クワンタムディーエックス・グループ・リミテッド Design, synthesis and use of synthetic nucleotides containing charge tags
EP2334792B1 (en) 2008-09-12 2020-09-09 GenTegra LLC Matrices and media for storage and stabilization of biomolecules
EP3067359A1 (en) 2008-09-23 2016-09-14 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
EP2356129B1 (en) * 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Substituted alpha-l-bicyclic nucleosides
WO2010036696A1 (en) * 2008-09-24 2010-04-01 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acid analogs
EP3109321B1 (en) 2008-09-25 2019-05-01 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
CA2738485A1 (en) 2008-09-26 2010-04-01 Oncomed Pharmaceuticals, Inc. Frizzled-binding agents and uses thereof
US8551789B2 (en) 2010-04-01 2013-10-08 OncoMed Pharmaceuticals Frizzled-binding agents and their use in screening for WNT inhibitors
EP2568044B1 (en) 2008-09-29 2015-04-08 The Trustees of The University of Pennsylvania Tumor vascular marker-targeted vaccines
MX339820B (en) 2008-10-03 2016-06-13 Curna Inc Treatment of apolipoprotein-a1 related diseases by inhibition of natural antisense transcript to apolipoprotein-a1.
CA2740000C (en) 2008-10-09 2017-12-12 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010042933A2 (en) 2008-10-10 2010-04-15 Northwestern University Inhibition and treatment of prostate cancer metastasis
WO2010045509A2 (en) 2008-10-15 2010-04-22 Isis Pharmaceuticals, Inc. Modulation of factor 11 expression
EP3848461A1 (en) 2008-10-20 2021-07-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
ES2714787T3 (en) 2008-10-24 2019-05-30 Sarepta Therapeutics Inc Exon skipping compositions for DMD
US8987435B2 (en) 2008-10-24 2015-03-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
CN102264374B (en) 2008-10-24 2015-01-07 Isis制药公司 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
US8309306B2 (en) * 2008-11-12 2012-11-13 Nodality, Inc. Detection composition
RU2011124146A (en) * 2008-11-17 2012-12-27 Ф.Хоффманн-Ля Рош Аг COMPOSITIONS AND METHODS OF SUPPRESSING EXPRESSION OF FACTOR VII GENES
MX2011005429A (en) 2008-11-24 2011-06-21 Univ Northwestern Polyvalent rna-nanoparticle compositions.
WO2010061393A1 (en) 2008-11-30 2010-06-03 Compugen Ltd. He4 variant nucleotide and amino acid sequences, and methods of use thereof
RU2746478C2 (en) 2008-12-04 2021-04-14 КьюРНА, Инк. Treatment of tumors of diseases related to the genom-suppressor by therapy of natural transcript inhibition in anti-significant orientation regarding this gene
JP5971948B2 (en) 2008-12-04 2016-08-17 クルナ・インコーポレーテッド Treatment of vascular endothelial growth factor (VEGF) -related diseases by suppression of natural antisense transcripts against VEGF
WO2010065662A2 (en) 2008-12-04 2010-06-10 Curna, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
US20100256196A1 (en) 2008-12-04 2010-10-07 Ikfe Biomarkers for Atherosclerosis
KR101829469B1 (en) 2008-12-04 2018-03-30 큐알엔에이, 인크. Treatment of erythropoietin (epo) related diseases by inhibition of natural antisense transcript to epo
EP2633854B1 (en) 2008-12-05 2015-09-16 Yeda Research And Development Co. Ltd. miRNA-9 or miRNA-9* for use in treating ALS
JP5855462B2 (en) 2008-12-10 2016-02-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. DsRNA compositions targeting GNAQ and methods for inhibiting expression
WO2010067207A1 (en) 2008-12-11 2010-06-17 Ikfe Gmbh Biomarkers for insulin sensitizer drug response
CA2746508A1 (en) 2008-12-17 2010-07-15 Avi Biopharma, Inc. Antisense compositions and methods for modulating contact hypersensitivity or contact dermatitis
CA2748265C (en) 2008-12-22 2018-04-03 University Of Utah Research Foundation Monochrome multiplex quantitative pcr
US8645115B2 (en) 2008-12-22 2014-02-04 Trustees Of Boston University Modular nucleic acid-based circuits for counters, binary operations, memory and logic
US20100209350A1 (en) 2008-12-30 2010-08-19 Andreas Pfuetzner Biomarkers for Adipose Tissue Activity
US8206929B2 (en) 2009-01-07 2012-06-26 Roche Molecular Systems, Inc. Nucleic acid amplification with allele-specific suppression of sequence variants
WO2010079428A2 (en) 2009-01-07 2010-07-15 Ikfe Gmbh Biomarkers for appetite regulation
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
US20100294952A1 (en) * 2009-01-15 2010-11-25 Northwestern University Controlled agent release and sequestration
CA3036963A1 (en) 2009-01-29 2010-08-05 Arbutus Biopharma Corporation Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
CN102307996A (en) * 2009-02-03 2012-01-04 弗·哈夫曼-拉罗切有限公司 Compositions and methods for inhibiting expression of PTP1B genes
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8536320B2 (en) 2009-02-06 2013-09-17 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
US20100233733A1 (en) * 2009-02-10 2010-09-16 Nodality, Inc., A Delaware Corporation Multiple mechanisms for modulation of the pi3 kinase pathway
KR101682735B1 (en) 2009-02-12 2016-12-06 큐알엔에이, 인크. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
ES2658626T3 (en) 2009-02-12 2018-03-12 Curna, Inc. Treatment of diseases related to glial cell-derived neurotrophic factor (GDNF) by inhibition of natural antisense transcript to GDNF
US7964355B2 (en) * 2009-02-17 2011-06-21 Investigen, Inc. Assays based on detection of photobleaching reaction products from dye catalytic complex
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
EP2403863B1 (en) 2009-03-02 2013-08-28 Alnylam Pharmaceuticals Inc. Nucleic acid chemical modifications
JP6250263B2 (en) 2009-03-04 2017-12-20 クルナ・インコーポレーテッド Treatment of SIRT1-related diseases by suppression of natural antisense transcripts against sirtuin 1 (SIRT1)
EP2406376A1 (en) 2009-03-12 2012-01-18 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
ES2656290T3 (en) 2009-03-16 2018-02-26 Curna, Inc. Treatment of diseases related to nuclear factor (derived from erythroid 2) similar to 2 (NRF2) by inhibition of natural antisense transcript to NRF2
EP2408920B1 (en) 2009-03-17 2017-03-08 CuRNA, Inc. Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
DK2414544T3 (en) * 2009-04-01 2014-06-16 Dxterity Diagnostics Inc Probe amplification-dependent chemical ligation
US8309356B2 (en) * 2009-04-01 2012-11-13 Yale University Pseudocomplementary oligonucleotides for targeted gene therapy
KR20120022938A (en) 2009-04-15 2012-03-12 노오쓰웨스턴 유니버시티 Delivery of oligonucleotide-functionalized nanoparticles
EP3524275A1 (en) 2009-04-22 2019-08-14 Massachusetts Institute Of Technology Innate immune supression enables repeated delivery of long rna molecules
US8871494B2 (en) 2009-04-24 2014-10-28 Wisconsin Alumni Research Foundation Over-production of secondary metabolites by over-expression of the VEA gene
CA2759899A1 (en) 2009-04-24 2010-10-28 Prosensa Technologies B.V. Oligonucleotide comprising an inosine for treating dmd
EP2424987B1 (en) 2009-05-01 2017-11-15 CuRNA, Inc. Treatment of hemoglobin (hbf/hbg) related diseases by inhibition of natural antisense transcript to hbf/hbg
EP2427572B1 (en) * 2009-05-01 2013-08-28 Illumina, Inc. Sequencing methods
CA2760706C (en) 2009-05-05 2019-08-06 Alnylam Pharmaceuticals, Inc. Methods of delivering oligonucleotides to immune cells
KR20220150411A (en) 2009-05-05 2022-11-10 알닐람 파마슈티칼스 인코포레이티드 Lipid compositions
JP5883782B2 (en) 2009-05-06 2016-03-15 クルナ・インコーポレーテッド Treatment of lipid transport metabolism gene-related diseases by suppression of natural antisense transcripts on lipid transport metabolism genes
WO2010129746A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
KR101742334B1 (en) 2009-05-08 2017-06-01 큐알엔에이, 인크. Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family
KR20120069610A (en) * 2009-05-15 2012-06-28 에프. 호프만-라 로슈 아게 Compositions and methods for inhibiting expression of glucocorticoid receptor (gcr) genes
WO2010132665A1 (en) 2009-05-15 2010-11-18 Yale University Gemm riboswitches, structure-based compound design with gemm riboswitches, and methods and compositions for use of and with gemm riboswitches
DK2432881T3 (en) 2009-05-18 2018-02-26 Curna Inc TREATMENT OF REPROGRAMMING FACTOR-RELATED DISEASES BY INHIBITING NATURAL ANTISENSE TRANSCRIPTS TO A REPROGRAMMING FACTOR
CA2682429A1 (en) * 2009-05-20 2010-11-20 Gary Levy An assay for measuring plasma fgl-2 and methods and uses thereof
KR101703695B1 (en) 2009-05-22 2017-02-08 큐알엔에이, 인크. Treatment of transcription factor e3 (tfe3) and insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to tfe3
CN103221541B (en) 2009-05-28 2017-03-01 库尔纳公司 Antiviral gene relevant disease is treated by the natural antisense transcript suppressing antiviral gene
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
PL3431076T3 (en) 2009-06-10 2022-01-31 Arbutus Biopharma Corporation Improved lipid formulation
DK2977455T3 (en) 2009-06-15 2020-07-13 Complete Genomics Inc PROGRESS FOR LONG-FRAGMENT READING SEQUENCE
JP6128846B2 (en) 2009-06-16 2017-05-17 クルナ・インコーポレーテッド Treatment of PON1 gene-related diseases by suppression of natural antisense transcripts against paraoxonase (PON1)
WO2010148050A2 (en) 2009-06-16 2010-12-23 Curna, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
US8859515B2 (en) 2009-06-24 2014-10-14 Curna, Inc. Treatment of tumor necrosis factor receptor 2 (TNFR2) related diseases by inhibition of natural antisense transcript to TNFR2
EP2446037B1 (en) 2009-06-26 2016-04-20 CuRNA, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
WO2011005861A1 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
US20120252869A1 (en) 2009-07-24 2012-10-04 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
GB0913258D0 (en) 2009-07-29 2009-09-02 Dynex Technologies Inc Reagent dispenser
US9523701B2 (en) 2009-07-29 2016-12-20 Dynex Technologies, Inc. Sample plate systems and methods
CA2769665A1 (en) 2009-08-05 2011-02-10 Opko Curna, Llc Treatment of insulin gene (ins) related diseases by inhibition of natural antisense transcript to an insulin gene (ins)
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
CA2770071C (en) 2009-08-07 2014-07-15 Ohmx Corporation Enzyme triggered redox altering chemical elimination (e-trace) immunoassay
CN102625841A (en) 2009-08-11 2012-08-01 欧科库尔纳有限责任公司 Treatment of Adiponectin (ADIPOQ) related diseases by inhibition of natural antisense transcript to an Adiponectin (ADIPOQ)
US8409802B2 (en) 2009-08-14 2013-04-02 Roche Molecular Systems, Inc. Format of probes to detect nucleic acid differences
US9029338B2 (en) 2009-08-14 2015-05-12 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
US20120157324A1 (en) 2009-08-17 2012-06-21 Yale University Methylation biomarkers and methods of use
US8791087B2 (en) 2009-08-21 2014-07-29 Curna, Inc. Treatment of ‘C terminus of HSP70-interacting protein’ (CHIP)related diseases by inhibition of natural antisense transcript to CHIP
CN102482671B (en) 2009-08-25 2017-12-01 库尔纳公司 IQGAP relevant diseases are treated by suppressing the natural antisense transcript of ' gtpase activating protein containing IQ die bodys ' (IQGAP)
CA2772715C (en) 2009-09-02 2019-03-26 Genentech, Inc. Mutant smoothened and methods of using the same
US10072287B2 (en) 2009-09-10 2018-09-11 Centrillion Technology Holdings Corporation Methods of targeted sequencing
US10174368B2 (en) 2009-09-10 2019-01-08 Centrillion Technology Holdings Corporation Methods and systems for sequencing long nucleic acids
WO2011038228A1 (en) 2009-09-24 2011-03-31 President And Fellows Of Harvard College Bent nanowires and related probing of species
ES2664591T3 (en) 2009-09-25 2018-04-20 Curna, Inc. Treatment of phylagrin-related diseases (flg) by modulating the expression and activity of the FLG gene
CA2780741C (en) 2009-10-12 2023-04-04 Smith Holdings, Llc Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
CA2777448C (en) 2009-10-14 2023-02-28 Ilan Sela Compositions for controlling varroa mites in bees
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
WO2011050194A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
US20110129832A1 (en) * 2009-10-27 2011-06-02 Swift Biosciences, Inc. Polynucleotide Primers and Probes
MX2012004905A (en) 2009-10-30 2012-07-23 Univ British Columbia Gna11 mutations in melanoma.
CA2779099C (en) 2009-10-30 2021-08-10 Northwestern University Templated nanoconjugates
KR102138780B1 (en) 2009-11-02 2020-07-29 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Foot and mouth disease virus (fmdv) consensus proteins, coding sequences therefor and vaccines made therefrom
US20110268810A1 (en) 2009-11-02 2011-11-03 Yale University Polymeric materials loaded with mutagenic and recombinagenic nucleic acids
WO2011056215A1 (en) 2009-11-03 2011-05-12 Landers James P Versatile, visible method for detecting polymeric analytes
WO2011054939A2 (en) 2009-11-09 2011-05-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of kif10 genes
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
BR122023023194A2 (en) 2009-11-12 2023-12-26 The University Of Western Australia ANTISENSE OLIGONUCLEOTIDE, COMPOSITION AND RESPECTIVE USE
JP5991922B2 (en) 2009-11-13 2016-09-14 サレプタ セラピューティクス, インコーポレイテッド Antisense antiviral compounds and methods for treating influenza virus infection
US8916362B2 (en) 2009-11-23 2014-12-23 Swift Biosciences, Inc. Devices comprising a polynucleotide to extend single stranded target molecules
TWI507524B (en) 2009-11-30 2015-11-11 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
US8614071B2 (en) 2009-12-11 2013-12-24 Roche Molecular Systems, Inc. Preferential amplification of mRNA over DNA using chemically modified primers
WO2011084455A2 (en) 2009-12-16 2011-07-14 Opko Curna, Llc. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
US20110152349A1 (en) * 2009-12-18 2011-06-23 Anke Geick Compositions and methods for inhibiting expression of il-18 genes
RU2619185C2 (en) 2009-12-23 2017-05-12 Курна, Инк. Treatment of diseases associated with uncoupling proteins 2 (ucp2), by inhibiting of natural antisense transcript to ucp2
KR101891352B1 (en) 2009-12-23 2018-08-24 큐알엔에이, 인크. Treatment of hepatocyte growth factor (hgf) related diseases by inhibition of natural antisense transcript to hgf
CA2785451C (en) 2009-12-24 2019-01-22 Prosensa Technologies B.V. Molecule for treating an inflammatory disorder
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
JP5982288B2 (en) 2009-12-29 2016-08-31 カッパーアールエヌエー,インコーポレイテッド Treatment of tumor protein 63-related diseases by inhibition of natural antisense transcripts against tumor protein 63 (p63)
US20120289583A1 (en) 2009-12-31 2012-11-15 Curna, Inc. Treatment of insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to irs2 and transcription factor e3 (tfe3)
NO2521784T3 (en) 2010-01-04 2018-05-05
EP2521785B1 (en) 2010-01-06 2022-03-09 CuRNA, Inc. Inhibition of natural antisense transcript to a pancreatic developmental gene for use in a treatment of pancreatic developmental gene related diseases
ES2664866T3 (en) 2010-01-11 2018-04-23 Curna, Inc. Treatment of diseases related to sex hormone binding globulin (shbg) by inhibition of the natural antisense transcript to shbg
US8779118B2 (en) 2010-01-11 2014-07-15 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
WO2011088076A2 (en) 2010-01-12 2011-07-21 Yale University Structured rna motifs and compounds and methods for their use
US8883739B2 (en) 2010-01-19 2014-11-11 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for male reproductive disorders
WO2011090949A2 (en) 2010-01-19 2011-07-28 Illumina, Inc. Methods and compositions for processing chemical reactions
CA2786568A1 (en) 2010-01-25 2011-07-28 Curna, Inc. Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
WO2011097407A1 (en) 2010-02-04 2011-08-11 Ico Therapeutics Inc. Dosing regimens for treating and preventing ocular disorders using c-raf antisense
TW201129365A (en) * 2010-02-05 2011-09-01 Hoffmann La Roche Compositions and methods for inhibiting expression of IKK2 genes
JP5976548B2 (en) 2010-02-22 2016-08-23 カッパーアールエヌエー,インコーポレイテッド Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcripts against PYCR1
WO2011105902A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-beta (c8-beta) and uses thereof
WO2011105900A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-alpha (c8-alpha) and uses thereof
SG183335A1 (en) 2010-02-23 2012-09-27 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
EP2539357B1 (en) 2010-02-26 2017-06-14 Memorial Sloan-Kettering Cancer Center Methods and compositions for the detection and treatment of cancer involving mirnas and mirna inhibitors and targets
EP2542249A4 (en) 2010-03-05 2013-08-07 Tissue Genesis Inc Methods and compositions to support tissue integration and inosculation of transplanted tissue and transplanted engineered penile tissue with adipose stromal cells
AU2011224570B2 (en) 2010-03-08 2014-08-14 Monsanto Technology Llc Polynucleotide molecules for gene regulation in plants
WO2011112516A1 (en) 2010-03-08 2011-09-15 Ico Therapeutics Inc. Treating and preventing hepatitis c virus infection using c-raf kinase antisense oligonucleotides
US9068185B2 (en) 2010-03-12 2015-06-30 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
WO2011112732A2 (en) 2010-03-12 2011-09-15 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US20130101512A1 (en) 2010-03-12 2013-04-25 Chad A. Mirkin Crosslinked polynucleotide structure
EP3450979A3 (en) 2010-03-17 2019-04-24 The Regents of The University of Michigan Using phage epitopes to profile the immune response
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
US8889350B2 (en) 2010-03-26 2014-11-18 Swift Biosciences, Inc. Methods and compositions for isolating polynucleotides
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
WO2011123745A2 (en) 2010-04-02 2011-10-06 Opko Curna Llc Treatment of colony-stimulating factor 3 (csf3) related diseases by inhibition of natural antisense transcript to csf3
WO2011127337A2 (en) 2010-04-09 2011-10-13 Opko Curna Llc Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
WO2011133695A2 (en) 2010-04-20 2011-10-27 Swift Biosciences, Inc. Materials and methods for nucleic acid fractionation by solid phase entrapment and enzyme-mediated detachment
US20110262406A1 (en) 2010-04-21 2011-10-27 Yale University Compositions and methods for targeted inactivation of hiv cell surface receptors
US20110293585A1 (en) 2010-04-21 2011-12-01 Helix Therapeutics, Inc. Compositions and methods for treatment of lysosomal storage disorders
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
WO2011139714A2 (en) 2010-04-26 2011-11-10 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-trna synthetase
US8961960B2 (en) 2010-04-27 2015-02-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
EP2563911B1 (en) 2010-04-28 2021-07-21 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of alanyl trna synthetases
WO2011139702A2 (en) 2010-04-28 2011-11-10 Isis Pharmaceuticals, Inc. Modified nucleosides and oligomeric compounds prepared therefrom
AU2011248490B2 (en) 2010-04-29 2016-11-10 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Asparaginyl tRNA synthetases
SI2563920T1 (en) 2010-04-29 2017-05-31 Ionis Pharmaceuticals, Inc. Modulation of transthyretin expression
US9034320B2 (en) 2010-04-29 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Valyl-tRNA synthetases
JP5976638B2 (en) 2010-05-03 2016-08-23 エータイアー ファーマ, インコーポレイテッド Innovative discovery of therapeutic, diagnostic and antibody compositions related to protein fragments of arginyl tRNA synthetase
US8981045B2 (en) 2010-05-03 2015-03-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of methionyl-tRNA synthetases
SG185027A1 (en) 2010-05-03 2012-11-29 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
EP2566495B1 (en) 2010-05-03 2017-03-01 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-trna synthetases
WO2011139387A1 (en) 2010-05-03 2011-11-10 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
US9062302B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
US10232374B2 (en) 2010-05-05 2019-03-19 Miroculus Inc. Method of processing dried samples using digital microfluidic device
WO2011143124A2 (en) 2010-05-10 2011-11-17 The Regents Of The University Of California Endoribonuclease compositions and methods of use thereof
JP5866106B2 (en) 2010-05-12 2016-02-17 コロンビア ユニヴァーシティ Method for producing enteroendocrine cells that produce and secrete insulin
US9050373B2 (en) 2010-05-13 2015-06-09 The Charlotte-Mecklenburg Hospital Authority Pharmaceutical compositions comprising antisense oligonucleotides and methods of using same
CA3090304A1 (en) 2010-05-13 2011-11-17 Sarepta Therapeutics, Inc. Antisense modulation of interleukins 17 and 23 signaling
EP2568996B1 (en) 2010-05-14 2017-10-04 aTyr Pharma, Inc. Therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-trna synthetases
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
US9034598B2 (en) 2010-05-17 2015-05-19 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
RU2620978C2 (en) 2010-05-26 2017-05-30 Курна, Инк. Treatment of diseases associated with methionine sulfoxide reductase a (msra), by msra natural antisense transcript inhibition
RU2585229C2 (en) 2010-05-26 2016-05-27 Курна, Инк. Treatment of diseases associated with atonal homolog 1 (aton1) by inhibiting natural antisense transcript of gene aton1
US20130203045A1 (en) 2010-05-26 2013-08-08 University Of Virginia Patent Foundation Method for detecting nucleic acids based on aggregate formation
AU2011258106B2 (en) 2010-05-27 2017-02-23 Pangu Biopharma Limited Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
CA2799501C (en) 2010-05-28 2022-02-15 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
CA3102008A1 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
US9228240B2 (en) 2010-06-03 2016-01-05 California Institute Of Technology Methods for detecting and quantifying viable bacterial endo-spores
WO2011156278A1 (en) 2010-06-07 2011-12-15 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
US8846637B2 (en) 2010-06-08 2014-09-30 Isis Pharmaceuticals, Inc. Substituted 2′-amino and 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
US9638632B2 (en) 2010-06-11 2017-05-02 Vanderbilt University Multiplexed interferometric detection system and method
WO2011163466A1 (en) 2010-06-23 2011-12-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (nrg-1)
ES2863526T3 (en) 2010-06-23 2021-10-11 Curna Inc Treatment of voltage-controlled sodium channel alpha subunit (SCNA) -related diseases by inhibiting natural antisense transcription to SCNA
US20130209463A1 (en) 2010-06-30 2013-08-15 Compugen Ltd. Polypeptides and uses thereof as a drug for treatment of multiple sclerosis, rheumatoid arthritis and other autoimmune disorders
EP2593125B1 (en) 2010-07-12 2017-11-01 aTyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-trna synthetases
RU2611190C2 (en) 2010-07-14 2017-02-21 Курна, Инк. Treatment of diseases related with gene dlg by inhibition of natural antisense transcript of dlg gene
AU2011280958B2 (en) 2010-07-22 2014-10-09 Massachusetts Institute Of Technology Multiple input biologic classifier circuits for cells
WO2012021554A1 (en) 2010-08-09 2012-02-16 Yale University Cyclic di-gmp-ii riboswitches, motifs, and compounds, and methods for their use
US8198429B2 (en) 2010-08-09 2012-06-12 Avi Biopharma, Inc. Antisense antiviral compounds and methods for treating a filovirus infection
EP2603607B1 (en) 2010-08-11 2016-04-06 Celula, Inc. Genotyping dna
US9029506B2 (en) 2010-08-25 2015-05-12 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-tRNA synthetases
WO2012028697A1 (en) 2010-09-01 2012-03-08 Eth Zürich, Institute Of Molecular Biology And Biophysics Affinity purification system based on donor strand complementation
US10017763B2 (en) 2010-09-03 2018-07-10 Sarepta Therapeutics, Inc. dsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US20120070829A1 (en) 2010-09-10 2012-03-22 Bio-Rad Laboratories, Inc. Size selection of dna for chromatin analysis
US8483969B2 (en) 2010-09-17 2013-07-09 Illuminia, Inc. Variation analysis for multiple templates on a solid support
US20130210901A1 (en) 2010-09-20 2013-08-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method of treating neurodegenerative diseases
JP5974012B2 (en) 2010-10-05 2016-08-23 ジェネンテック, インコーポレイテッド Mutant smoothened and method of using the same
ES2640755T3 (en) 2010-10-06 2017-11-06 Curna, Inc. Treatment of diseases related to Sialidase 4 (neu4) by inhibition of the natural antisense transcript to the neu4 gene
EP2625292B1 (en) 2010-10-07 2018-12-05 The General Hospital Corporation Biomarkers of cancer
WO2012052872A2 (en) 2010-10-17 2012-04-26 Yeda Research And Development Co. Ltd. Methods and compositions for the treatment of insulin-associated medical conditions
EP2851426B1 (en) 2010-10-18 2018-08-22 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of RRM2 genes
US8648053B2 (en) 2010-10-20 2014-02-11 Rosalind Franklin University Of Medicine And Science Antisense oligonucleotides that target a cryptic splice site in Ush1c as a therapeutic for Usher syndrome
WO2012054723A2 (en) 2010-10-22 2012-04-26 Opko Curna Llc Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
US8753816B2 (en) 2010-10-26 2014-06-17 Illumina, Inc. Sequencing methods
RU2611195C2 (en) 2010-10-27 2017-02-21 Курна, Инк. Treatment of interferon-related developmental regulator 1 (ifrd1) associated diseases by inhibition of natural antisense transcript to ifrd1
CN103270174B (en) 2010-10-28 2016-08-24 生命技术公司 Chemical reinforcing type Primer composition, method and test kit
CA2821299C (en) 2010-11-05 2019-02-12 Frank J. Steemers Linking sequence reads using paired code tags
CN110123830A (en) 2010-11-09 2019-08-16 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting the lipid of the expression of Eg5 and VEGF gene to prepare
AU2011326364B2 (en) 2010-11-12 2016-08-11 Sarepta Therapeutics, Inc. Antisense antibacterial compounds and methods
AU2011325956B2 (en) 2010-11-12 2016-07-14 The General Hospital Corporation Polycomb-associated non-coding RNAs
CA3077910A1 (en) 2010-11-17 2012-05-24 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
US10000752B2 (en) 2010-11-18 2018-06-19 Curna, Inc. Antagonat compositions and methods of use
CA2818824A1 (en) 2010-11-23 2012-05-31 Joseph Collard Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
EP2649182A4 (en) 2010-12-10 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
EP2663323B1 (en) 2011-01-14 2017-08-16 The General Hospital Corporation Methods targeting mir-128 for regulating cholesterol/lipid metabolism
EP2663639B1 (en) 2011-01-14 2017-09-13 Life Technologies Corporation Methods for isolation, identification, and quantification of mirnas
WO2012100078A1 (en) 2011-01-19 2012-07-26 Ohmx Corporation Enzyme triggered redox altering chemical elimination (e-trace) immmunoassay
DK3037536T3 (en) 2011-01-28 2020-01-13 Illumina Inc OLIGONUCLEOTID REPLACEMENT FOR DI-TAGGED AND DIRECTORY LIBRARIES
EP2670864B1 (en) 2011-01-31 2017-03-08 Illumina, Inc. Methods for reducing nucleic acid damage
KR101697396B1 (en) 2011-02-02 2017-01-17 엑스칼리아드 파마슈티컬즈, 인코포레이티드 Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
EP2670404B1 (en) 2011-02-02 2018-08-29 The Trustees of Princeton University Sirtuin modulators as virus production modulators
CN103501793A (en) 2011-02-08 2014-01-08 夏洛特-梅克伦堡医院(商业用名:卡罗来纳保健系统) Antisense oligonucleotides
US9365897B2 (en) 2011-02-08 2016-06-14 Illumina, Inc. Selective enrichment of nucleic acids
US9327037B2 (en) 2011-02-08 2016-05-03 The Johns Hopkins University Mucus penetrating gene carriers
WO2012109495A1 (en) 2011-02-09 2012-08-16 Metabolic Solutions Development Company, Llc Cellular targets of thiazolidinediones
EP3940084A1 (en) 2011-02-09 2022-01-19 Bio-Rad Laboratories, Inc. Analysis of nucleic acids
WO2012112606A1 (en) 2011-02-15 2012-08-23 Bio-Rad Laboratories, Inc. Detecting methylati0n in a subpopulation of genomic dna
US9562853B2 (en) 2011-02-22 2017-02-07 Vanderbilt University Nonaqueous backscattering interferometric methods
EP2689028B1 (en) 2011-03-23 2017-08-30 Pacific Biosciences Of California, Inc. Isolation of polymerase-nucleic acid complexes and loading onto substrates
EP3406267A1 (en) 2011-03-25 2018-11-28 Children's Medical Center Corporation Lin28-mediated control of let-7 biogenesis
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
AU2012236099A1 (en) 2011-03-31 2013-10-03 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
US20120252682A1 (en) 2011-04-01 2012-10-04 Maples Corporate Services Limited Methods and systems for sequencing nucleic acids
WO2012138453A1 (en) 2011-04-03 2012-10-11 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
EP2697256A1 (en) 2011-04-15 2014-02-19 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
JP2014512826A (en) 2011-04-25 2014-05-29 バイオ−ラド ラボラトリーズ インコーポレイテッド Methods and compositions for nucleic acid analysis
WO2012149154A1 (en) 2011-04-26 2012-11-01 Swift Biosciences, Inc. Polynucleotide primers and probes
US9353371B2 (en) 2011-05-02 2016-05-31 Ionis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with usher syndrome
WO2012151268A1 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system for high throughput optical and label free detection of analytes
WO2012151289A2 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system to detect aggregate formation on a substrate
WO2012150960A1 (en) 2011-05-05 2012-11-08 Avi Biopharma, Inc. Peptide oligonucleotide conjugates
CN105132284B (en) 2011-05-13 2018-04-03 加利福尼亚大学董事会 Photo-thermal substrate for selective transfectional cell
EP2710145B1 (en) 2011-05-17 2015-12-09 Dxterity Diagnostics Incorporated Methods and compositions for detecting target nucleic acids
ES2703133T3 (en) 2011-05-24 2019-03-07 Rosetta Genomics Ltd Methods and compositions for determining heart failure or a risk of heart failure
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
ES2653247T3 (en) 2011-06-09 2018-02-06 Curna, Inc. Treatment of frataxin-related diseases (FXN) by inhibiting the natural antisense transcript to the FXN gene
EP3320922A1 (en) 2011-06-10 2018-05-16 Ionis Pharmaceuticals, Inc. Methods for modulating kallikrein (klkb1) expression
CA3191066A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
CN103890000B (en) 2011-06-21 2017-09-01 阿尔尼拉姆医药品有限公司 (ANGPTL3) the iRNA compositions of angiopoietin-like 3 and its application method
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
EP4134433A1 (en) 2011-06-23 2023-02-15 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
EP2726153B1 (en) 2011-06-29 2018-03-28 Ionis Pharmaceuticals, Inc. Methods for modulating kallikrein (klkb1) expression
CA2836855C (en) 2011-06-30 2020-07-14 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
WO2013001372A2 (en) 2011-06-30 2013-01-03 University Of Oslo Methods and compositions for inhibition of activation of regulatory t cells
TW202244278A (en) 2011-06-30 2022-11-16 美商艾羅海德製藥公司 Compositions and methods for inhibiting gene expression of hepatitis b virus
JP6109165B2 (en) 2011-07-01 2017-04-05 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Herpesvirus vaccines and methods of use
AU2012284259A1 (en) 2011-07-15 2014-03-06 Sarepta Therapeutics, Inc. Methods and compositions for manipulating translation of protein isoforms from alternative initiation start sites
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
WO2013019960A1 (en) 2011-08-03 2013-02-07 Bio-Rad Laboratories, Inc. Filtering small nucleic acids using permeabilized cells
BR112014002737B1 (en) 2011-08-04 2022-08-02 Yeda Research And Development Co. Ltd. MIR-135
US9040678B2 (en) 2011-08-05 2015-05-26 Illumina, Inc. Functionalization and purification of molecules by reversible group exchange
JP6125505B2 (en) 2011-09-06 2017-05-10 カッパーアールエヌエー,インコーポレイテッド Treatment of voltage-gated sodium channel alpha subunit related diseases with small molecules
US9840715B1 (en) 2011-09-13 2017-12-12 Monsanto Technology Llc Methods and compositions for delaying senescence and improving disease tolerance and yield in plants
CA2848695A1 (en) 2011-09-13 2013-03-21 Monsanto Technology Llc Methods and composition for weed control comprising inhibiting ppg oxidase
US10760086B2 (en) 2011-09-13 2020-09-01 Monsanto Technology Llc Methods and compositions for weed control
US10829828B2 (en) 2011-09-13 2020-11-10 Monsanto Technology Llc Methods and compositions for weed control
US10806146B2 (en) 2011-09-13 2020-10-20 Monsanto Technology Llc Methods and compositions for weed control
EP3382027A3 (en) 2011-09-13 2018-10-31 Monsanto Technology LLC Methods and compositions for weed control
EP2755466A4 (en) 2011-09-13 2015-04-15 Monsanto Technology Llc Methods and compositions for weed control
CN103957697B (en) 2011-09-13 2017-10-24 孟山都技术公司 Method and composition for Weeds distribution
AU2012308320C1 (en) 2011-09-14 2018-08-23 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds
EP2755692B1 (en) 2011-09-14 2020-11-25 Northwestern University Nanoconjugates able to cross the blood-brain barrier
US9920326B1 (en) 2011-09-14 2018-03-20 Monsanto Technology Llc Methods and compositions for increasing invertase activity in plants
US20140234373A1 (en) 2011-09-16 2014-08-21 Georfia Regents University Methods of Promoting Immune Tolerance
US9580713B2 (en) 2011-09-17 2017-02-28 Yale University Fluoride-responsive riboswitches, fluoride transporters, and methods of use
US9801948B2 (en) 2011-09-21 2017-10-31 Yale University Antimicrobial compositions and methods of use thereof
CA2847943C (en) 2011-09-23 2017-08-29 F.Hoffmann-La Roche Ag Use of g-clamp for improved allele-specific pcr
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
WO2013055865A1 (en) 2011-10-11 2013-04-18 The Brigham And Women's Hospital, Inc. Micrornas in neurodegenerative disorders
AU2012322618A1 (en) 2011-10-14 2014-05-29 Genentech, Inc. Anti-HtrA1 antibodies and methods of use
CN103998921B (en) 2011-10-14 2016-04-20 贝克顿·迪金森公司 Square wave thermal cycle
EP2768967B1 (en) 2011-10-17 2017-12-06 Ohmx Corporation Single, direct detection of hemoglobin a1c percentage using enzyme triggered redox altering chemical elimination (e-trace) immunoassay
CN104080958A (en) 2011-10-19 2014-10-01 纽亘技术公司 Compositions and methods for directional nucleic acid amplification and sequencing
US9145559B2 (en) 2011-10-27 2015-09-29 Yeda Research And Development Co. Ltd. Methods of treating cancer
WO2013063482A2 (en) 2011-10-28 2013-05-02 The Regents Of The University Of California Flt3 mutations associated with drug resistance in aml patients having activating mutations in flt3
US9073851B2 (en) 2011-10-28 2015-07-07 Board Of Regents, The University Of Texas System Compositions and methods for treating cancer
US10837879B2 (en) 2011-11-02 2020-11-17 Complete Genomics, Inc. Treatment for stabilizing nucleic acid arrays
JP2014532720A (en) 2011-11-04 2014-12-08 オームクス コーポレイション New chemistry used in biosensors
EP2780299A4 (en) 2011-11-18 2015-04-22 Sarepta Therapeutics Inc Functionally-modified oligonucleotides and subunits thereof
ES2832531T3 (en) 2011-11-30 2021-06-10 Sarepta Therapeutics Inc Oligonucleotides for the treatment of repeat expansion diseases
JP6496549B2 (en) 2011-11-30 2019-04-03 サレプタ セラピューティクス, インコーポレイテッド Induced exon inclusion in spinal muscular atrophy
WO2013082529A1 (en) 2011-12-02 2013-06-06 Yale University Enzymatic synthesis of poly(amine-co-esters) and methods of use thereof for gene delivery
US9895451B2 (en) 2011-12-02 2018-02-20 Yale University Formulations for targeted release of agents to low pH tissue environments or cellular compartments and methods of use thereof
US10465042B2 (en) 2011-12-02 2019-11-05 Yale University Poly(amine-co-ester) nanoparticles and methods of use thereof
SI2788487T1 (en) 2011-12-08 2018-09-28 Sarepta Therapeutics, Inc. Oligonucleotide analogues targeting human lmna
CA2895077A1 (en) 2011-12-12 2013-06-20 Beverly Packard In vivo delivery of oligonucleotides
WO2013090804A2 (en) 2011-12-14 2013-06-20 The Johns Hopkins University Nanoparticles with enhanced mucosal penetration or decreased inflammation
AU2012354062B2 (en) 2011-12-16 2017-09-07 Targetgene Biotechnologies Ltd Compositions and methods for modifying a predetermined target nucleic acid sequence
WO2013090648A1 (en) 2011-12-16 2013-06-20 modeRNA Therapeutics Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013096455A1 (en) 2011-12-20 2013-06-27 Dana-Farber Cancer Institute, Inc. Methods for diagnosing and treating oncogenic kras-associated cancer
US9115394B2 (en) 2011-12-22 2015-08-25 Roche Molecular Systems, Inc. Methods and reagents for reducing non-specific amplification
EP2794880B1 (en) 2011-12-22 2018-05-02 Ionis Pharmaceuticals, Inc. Methods for modulating metastasis-associated-in-lung-adenocarcinoma-transcript-1(malat-1) expression
EP2798089B1 (en) 2011-12-30 2018-05-23 Bio-rad Laboratories, Inc. Methods and compositions for performing nucleic acid amplification reactions
GB201122458D0 (en) 2011-12-30 2012-02-08 Univ Wageningen Modified cascade ribonucleoproteins and uses thereof
CA2860739A1 (en) 2012-01-09 2013-07-18 Ohmx Corporation Enzyme cascade methods for e-trace assay signal amplification
EP2802674B1 (en) 2012-01-11 2020-12-16 Ionis Pharmaceuticals, Inc. Compositions and methods for modulation of ikbkap splicing
US9415020B2 (en) 2012-01-19 2016-08-16 The Johns Hopkins University Nanoparticle formulations with enhanced mucosal penetration
GB2513793B (en) 2012-01-26 2016-11-02 Nugen Tech Inc Compositions and methods for targeted nucleic acid sequence enrichment and high efficiency library generation
WO2013112053A1 (en) 2012-01-27 2013-08-01 Prosensa Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
JP2015512616A (en) 2012-02-01 2015-04-30 コンピュゲン エルティーディー. C1ORF32 antibody and its use for the treatment of cancer
WO2013120003A1 (en) 2012-02-08 2013-08-15 Isis Pharmaceuticals, Inc. Modulation of rna by repeat targeting
US9862994B2 (en) 2012-02-09 2018-01-09 Dana-Farber Cancer Institute, Inc. Selective nucleic acid amplification from nucleic acid pools
WO2013121426A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
EP3222627B1 (en) 2012-02-15 2019-08-07 Pacific Biosciences of California, Inc. Polymerase enzyme substrates with protein shield
JP2015509366A (en) 2012-02-22 2015-03-30 ブレインステム バイオテック リミテッド Generation of neural stem cells and motor neurons
US10034902B2 (en) 2012-02-22 2018-07-31 Exostem Biotec Ltd. MicroRNAs for the generation of astrocytes
CA2866625C (en) 2012-03-13 2020-12-08 Swift Biosciences, Inc. Methods and compositions for size-controlled homopolymer tailing of substrate polynucleotides by a nucleic acid polymerase
CA2867262C (en) 2012-03-15 2021-03-16 Curna, Inc. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
EP2639238A1 (en) 2012-03-15 2013-09-18 Universität Bern Tricyclic nucleosides and oligomeric compounds prepared therefrom
US9610362B2 (en) 2012-03-16 2017-04-04 Valerion Therapeutics, Llc Antisense conjugates for decreasing expression of DMPK
US9340784B2 (en) 2012-03-19 2016-05-17 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating alpha-1-antitrypsin expression
EA031110B1 (en) 2012-03-20 2018-11-30 Сарепта Терапьютикс, Инк. Boronic acid conjugates of oligonucleotide analogues
US20150086584A1 (en) 2012-03-22 2015-03-26 University Of Miami Multi-specific binding agents
EP2831232A4 (en) 2012-03-30 2015-11-04 Univ Washington Methods for modulating tau expression for reducing seizure and modifying a neurodegenerative syndrome
AU2013243948A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with human disease
CN108949772A (en) 2012-04-02 2018-12-07 现代泰克斯公司 For generating the modification polynucleotides of biological agent relevant to human diseases and protein
JP6375289B2 (en) 2012-04-05 2018-08-15 マサチューセッツ インスティテュート オブ テクノロジー Immune stimulating composition and method of use thereof
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
WO2013154798A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
US20150087055A1 (en) 2012-04-13 2015-03-26 Massachusetts Institute Of Technology Analog and mixed-signal computation and circuits in living cells
WO2013159091A2 (en) 2012-04-20 2013-10-24 Aptamir Therapeutics, Inc. Mirna modulators of thermogenesis
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
WO2013163628A2 (en) 2012-04-27 2013-10-31 Duke University Genetic correction of mutated genes
US9533068B2 (en) 2012-05-04 2017-01-03 The Johns Hopkins University Drug loaded microfiber sutures for ophthalmic application
JP6392209B2 (en) 2012-05-04 2018-09-19 ザ・ジョンズ・ホプキンス・ユニバーシティー Lipid-based drug carriers for rapid permeation through the mucus lining
US9273949B2 (en) 2012-05-11 2016-03-01 Vanderbilt University Backscattering interferometric methods
EP2850190B1 (en) 2012-05-16 2020-07-08 Translate Bio MA, Inc. Compositions and methods for modulating mecp2 expression
EP2850186B1 (en) 2012-05-16 2018-12-19 Translate Bio MA, Inc. Compositions and methods for modulating smn gene family expression
EP2850189B8 (en) 2012-05-16 2019-01-23 Translate Bio MA, Inc. Compositions and methods for modulating gene expression
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
WO2013173789A2 (en) 2012-05-17 2013-11-21 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
EP2852606B1 (en) 2012-05-22 2019-08-07 Ionis Pharmaceuticals, Inc. Modulation of enhancer rna mediated gene expression
AU2013264742B2 (en) 2012-05-24 2018-10-04 A.B. Seeds Ltd. Compositions and methods for silencing gene expression
CA2874521A1 (en) 2012-05-24 2013-11-28 Dana-Farber Cancer Institute, Inc. Targeting the glutamine to pyruvate pathway for treatment of oncogenic kras-associated cancer
PT3241902T (en) 2012-05-25 2018-05-28 Univ California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US9487780B2 (en) 2012-06-01 2016-11-08 Ionis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with fibronectin
WO2013181665A1 (en) 2012-06-01 2013-12-05 Isis Pharmaceuticals, Inc. Antisense compounds targeting genes associated with fibronectin
WO2013184209A1 (en) 2012-06-04 2013-12-12 Ludwig Institute For Cancer Research Ltd. Mif for use in methods of treating subjects with a neurodegenerative disorder
EP2861787B1 (en) 2012-06-18 2017-09-20 Nugen Technologies, Inc. Compositions and methods for negative selection of non-desired nucleic acid sequences
EP3461895B1 (en) 2012-06-25 2020-07-01 Ionis Pharmaceuticals, Inc. Modulation of ube3a-ats expression
US20150011396A1 (en) 2012-07-09 2015-01-08 Benjamin G. Schroeder Methods for creating directional bisulfite-converted nucleic acid libraries for next generation sequencing
US20140038182A1 (en) 2012-07-17 2014-02-06 Dna Logix, Inc. Cooperative primers, probes, and applications thereof
WO2014014518A1 (en) 2012-07-18 2014-01-23 Dana-Farber Cancer Institute, Inc. Methods for treating, preventing and predicting risk of developing breast cancer
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
US20150175979A1 (en) 2012-07-23 2015-06-25 La Jolla Institute For Allergy And Immunology Ptprs and proteoglycans in autoimmune disease
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US10870099B2 (en) 2012-07-26 2020-12-22 Illumina, Inc. Compositions and methods for the amplification of nucleic acids
ES2773489T3 (en) 2012-07-27 2020-07-13 Ionis Pharmaceuticals Inc Modulation of diseases related to the renin-angiotensin system (RAS) using angiotensinogens
CA2880833A1 (en) 2012-08-03 2014-02-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
WO2014028739A1 (en) 2012-08-15 2014-02-20 Isis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified capping protocols
CN104781271B (en) 2012-08-20 2018-07-06 加利福尼亚大学董事会 The polynucleotides of group with bio-reversible
US10111962B2 (en) 2012-09-25 2018-10-30 Genzyme Corporation Peptide-linked morpholino antisense oligonucleotides for treatment of myotonic dystrophy
AR092982A1 (en) 2012-10-11 2015-05-13 Isis Pharmaceuticals Inc MODULATION OF THE EXPRESSION OF ANDROGEN RECEIVERS
US9523094B2 (en) 2012-10-11 2016-12-20 Ionis Pharmaceuticals, Inc. Methods of treating kennedy's disease
EP2906255B1 (en) 2012-10-12 2023-02-22 Ionis Pharmaceuticals, Inc. Antisense compounds and uses thereof
EP3459549B1 (en) 2012-10-12 2022-04-06 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2014062989A2 (en) 2012-10-18 2014-04-24 Monsanto Technology Llc Methods and compositions for plant pest control
US10548962B2 (en) 2012-10-22 2020-02-04 The Board Of Regents For Oklahoma State University Use of the salmonella SPP type III secretion proteins as a protective vaccination
US9266959B2 (en) 2012-10-23 2016-02-23 Oncomed Pharmaceuticals, Inc. Methods of treating neuroendocrine tumors using frizzled-binding agents
US20140322706A1 (en) 2012-10-24 2014-10-30 Jon Faiz Kayyem Integrated multipelx target analysis
JP1628115S (en) 2012-10-24 2019-04-01
US20150299803A1 (en) 2012-11-05 2015-10-22 Pronai Therapeutics, Inc. Methods of Using Biomarkers for the Treatment of Cancer by Modulation of BCL2 Expression
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
WO2014093305A2 (en) 2012-12-13 2014-06-19 Ymir Genomics, Llc Micrornas and uses thereof
AU2013364158A1 (en) 2012-12-20 2015-07-09 Sarepta Therapeutics, Inc. Improved exon skipping compositions for treating muscular dystrophy
US10683505B2 (en) 2013-01-01 2020-06-16 Monsanto Technology Llc Methods of introducing dsRNA to plant seeds for modulating gene expression
BR112015015976A2 (en) 2013-01-01 2018-04-10 A. B. Seeds Ltd. method for introducing dsrna into seeds to modulate gene expression
WO2014113540A1 (en) 2013-01-16 2014-07-24 Iowa State University Research Foundation, Inc. A deep intronic target for splicing correction on spinal muscular atrophy gene
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
CA2898326C (en) 2013-01-18 2022-05-17 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
US9956294B2 (en) 2013-01-18 2018-05-01 H. Lee Moffitt Cancer Center And Research Institute, Inc. Targeted sensitization of non-del(5q) malignant cells
US10000767B2 (en) 2013-01-28 2018-06-19 Monsanto Technology Llc Methods and compositions for plant pest control
US9701708B2 (en) 2013-01-31 2017-07-11 Ionis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified coupling protocols
US9359444B2 (en) 2013-02-04 2016-06-07 Oncomed Pharmaceuticals Inc. Methods and monitoring of treatment with a Wnt pathway inhibitor
US10568975B2 (en) 2013-02-05 2020-02-25 The Johns Hopkins University Nanoparticles for magnetic resonance imaging tracking and methods of making and using thereof
KR102169899B1 (en) 2013-02-14 2020-10-26 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of apolipoprotein c-iii (apociii) expression in lipoprotein lipase deficient (lpld) populations
US20150366890A1 (en) 2013-02-25 2015-12-24 Trustees Of Boston University Compositions and methods for treating fungal infections
CN105163755A (en) 2013-03-12 2015-12-16 犹他大学研究基金会 Compositions and methods for inducing apoptosis
US20160024181A1 (en) 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
MX2015012334A (en) 2013-03-13 2016-02-05 Monsanto Technology Llc Methods and compositions for weed control.
WO2014142850A1 (en) 2013-03-13 2014-09-18 Illumina, Inc. Methods and compositions for nucleic acid sequencing
WO2014164761A1 (en) 2013-03-13 2014-10-09 Monsanto Technology Llc Methods and compositions for weed control
EP2971010B1 (en) 2013-03-14 2020-06-10 ModernaTX, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
ES2807379T3 (en) 2013-03-14 2021-02-22 Ionis Pharmaceuticals Inc Compositions and methods to regulate the expression of Tau
US9168300B2 (en) 2013-03-14 2015-10-27 Oncomed Pharmaceuticals, Inc. MET-binding agents and uses thereof
PE20160046A1 (en) 2013-03-14 2016-02-14 Alnylam Pharmaceuticals Inc COMPOSITION OF RNAi AGAINST THE C5 COMPONENT OF THE COMPLEMENT AND METHODS FOR ITS USE
US20140329881A1 (en) 2013-03-14 2014-11-06 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
EP2971167B1 (en) 2013-03-14 2019-07-31 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US20140283211A1 (en) 2013-03-14 2014-09-18 Monsanto Technology Llc Methods and Compositions for Plant Pest Control
JP2016521119A (en) 2013-03-14 2016-07-21 サレプタ セラピューティクス, インコーポレイテッド Exon skipping composition for treating muscular dystrophy
EP3520895A1 (en) 2013-03-15 2019-08-07 Genmark Diagnostics Inc. Fluid container with cantilevered lance
EP2968991A2 (en) 2013-03-15 2016-01-20 Sarepta Therapeutics, Inc. Improved compositions for treating muscular dystrophy
EP2971165A4 (en) 2013-03-15 2016-11-23 Moderna Therapeutics Inc Removal of dna fragments in mrna production process
US10324011B2 (en) 2013-03-15 2019-06-18 The Trustees Of Princeton University Methods and devices for high throughput purification
US9347095B2 (en) 2013-03-15 2016-05-24 Bio-Rad Laboratories, Inc. Digital assays for mutation detection
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
CA2907357A1 (en) 2013-03-15 2014-09-18 Kinemed, Inc. Biomarkers
US10568328B2 (en) 2013-03-15 2020-02-25 Monsanto Technology Llc Methods and compositions for weed control
CN114045295A (en) 2013-03-15 2022-02-15 宾夕法尼亚大学理事会 Foot and Mouth Disease Virus (FMDV) consensus proteins, coding sequences thereof, and vaccines made therefrom
US10052364B2 (en) 2013-03-15 2018-08-21 The Trustees Of Columbia University In The City Of New York Osteocalcin as a treatment for cognitive disorders
CN110186835B (en) 2013-03-15 2022-05-31 Gpb科学有限公司 On-chip microfluidic processing of particles
WO2014144092A1 (en) 2013-03-15 2014-09-18 Nugen Technologies, Inc. Sequential sequencing
RU2015144149A (en) 2013-03-15 2017-04-21 Интермьюн, Инк. PROTEOMIC MARKERS ILF
EP4286517A3 (en) 2013-04-04 2024-03-13 President and Fellows of Harvard College Therapeutic uses of genome editing with crispr/cas systems
US10544461B2 (en) 2013-04-16 2020-01-28 The Johns Hopkins University Diagnostic and prognostic test for sturge-weber syndrome, klippel-trenaunay-weber syndrome, and port-wine stains (PWSS)
BR112015027377B1 (en) 2013-05-01 2023-01-10 Ionis Pharmaceuticals, Inc OLIGOMERIC COMPOUNDS WITH CONJUGATE GROUPS, COMPOSITIONS COMPRISING THE SAID COMPOUNDS AND USES THEREOF
EP2994167B1 (en) 2013-05-06 2020-05-06 Alnylam Pharmaceuticals, Inc. Dosages and methods for delivering lipid formulated nucleic acid molecules
CN111187811B (en) 2013-05-06 2024-03-08 加利福尼亚太平洋生物科学股份有限公司 Real-time electronic sequencing
MX2015015891A (en) 2013-05-22 2016-06-02 Telomere Diagnostics Inc Measures of short telomere abundance.
SG10201913872XA (en) 2013-05-22 2020-03-30 Alnylam Pharmaceuticals Inc SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
US20160113911A1 (en) 2013-06-06 2016-04-28 The General Hospital Corporation Methods and compositions for the treatment of cancer
EP3007704B1 (en) 2013-06-13 2021-01-06 Antisense Therapeutics Ltd Combination therapy for acromegaly
WO2014203189A1 (en) 2013-06-18 2014-12-24 Rosetta Genomics Ltd. Nanocarrier system for micrornas and uses thereof
ES2778462T3 (en) 2013-06-21 2020-08-10 Ionis Pharmaceuticals Inc Compounds and methods to modulate apolipoprotein C-III expression to improve a diabetic profile
CN105452461B (en) 2013-07-02 2021-04-13 Ionis制药公司 Modulators of growth hormone receptors
HUE056760T2 (en) 2013-07-11 2022-03-28 Modernatx Inc Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
US10077236B2 (en) 2013-07-15 2018-09-18 The Regents Of The University Of California Azacyclic constrained analogs of FTY720
US9850496B2 (en) 2013-07-19 2017-12-26 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
EP3030663B1 (en) 2013-07-19 2019-09-04 Monsanto Technology LLC Compositions and methods for controlling leptinotarsa
TWI772856B (en) 2013-07-19 2022-08-01 美商百健Ma公司 Compositions for modulating tau expression
US9944998B2 (en) 2013-07-25 2018-04-17 Bio-Rad Laboratories, Inc. Genetic assays
RS59991B1 (en) 2013-08-08 2020-04-30 Scripps Research Inst A method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
TW201536329A (en) 2013-08-09 2015-10-01 Isis Pharmaceuticals Inc Compounds and methods for modulation of dystrophia myotonica-protein kinase (DMPK) expression
EP3033625B1 (en) 2013-08-13 2020-01-22 The Scripps Research Institute Cysteine-reactive ligand discovery in proteomes
EP3038627B1 (en) 2013-08-28 2020-03-04 Ionis Pharmaceuticals, Inc. Modulation of prekallikrein (pkk) expression
WO2015031837A1 (en) 2013-08-29 2015-03-05 City Of Hope Cell penetrating conjugates and methods of use thereof
AU2014315287A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US11236338B2 (en) 2013-09-05 2022-02-01 Sarepta Therapeutics, Inc. Antisense-induced exon2 inclusion in acid alpha-glucosidase
DK3044315T3 (en) 2013-09-11 2019-05-20 Synthena Ag NUCLEAR INSURANCE AND PROCEDURES FOR TREATMENT OF PUMPS DISEASE
SG10201913950XA (en) 2013-09-11 2020-03-30 Eagle Biologics Inc Liquid protein formulations containing viscosity-lowering agents
SG10201806787VA (en) 2013-09-13 2018-09-27 Ionis Pharmaceuticals Inc Modulators of complement factor b
US9708360B2 (en) 2013-09-30 2017-07-18 Geron Corporation Phosphorodiamidate backbone linkage for oligonucleotides
EP3052626A1 (en) 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
US10519446B2 (en) 2013-10-04 2019-12-31 Novartis Ag Organic compounds to treat hepatitis B virus
TW202310853A (en) 2013-10-04 2023-03-16 美國西奈山伊坎醫學院 Compositions and methods for inhibiting expression of the alas1 gene
WO2015051366A2 (en) 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
CA2925129C (en) 2013-10-04 2023-04-04 Novartis Ag 3' end caps for rnai agents for use in rna interference
EP3055426B1 (en) 2013-10-09 2019-06-19 The United States of America as represented by The Secretary Department of Health and Human Services Detection of hepatitis delta virus (hdv) for the diagnosis and treatment of sjögren's syndrome and lymphoma
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
EP3057693A4 (en) 2013-10-16 2017-08-09 The University Of British Columbia Device for formulating particles at small volumes
CA2928779A1 (en) 2013-10-21 2015-04-30 The General Hospital Corporation Methods relating to circulating tumor cell clusters and the treatment of cancer
US9758546B2 (en) 2013-10-21 2017-09-12 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
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
USD881409S1 (en) 2013-10-24 2020-04-14 Genmark Diagnostics, Inc. Biochip cartridge
WO2015066341A2 (en) 2013-10-30 2015-05-07 Green Life Biotech, Llc Pathogenesis quantification systems and treatment methods for citrus greening blight
US10301622B2 (en) 2013-11-04 2019-05-28 Northwestern University Quantification and spatio-temporal tracking of a target using a spherical nucleic acid (SNA)
EP3066200A1 (en) 2013-11-04 2016-09-14 Monsanto Technology LLC Compositions and methods for controlling arthropod parasite and pest infestations
CA2929596C (en) 2013-11-13 2022-07-05 Nugen Technologies, Inc. Compositions and methods for identification of a duplicate sequencing read
JP2016537028A (en) 2013-11-18 2016-12-01 クリスパー セラピューティクス アーゲー CRISPR-CAS System Materials and Methods
EP3556869B1 (en) 2013-11-26 2023-08-02 Illumina Inc. Compositions and methods for polynucleotide sequencing
ES2797679T3 (en) 2013-12-02 2020-12-03 Ionis Pharmaceuticals Inc Antisense compounds and their uses
CN112107693B (en) 2013-12-03 2023-05-26 西北大学 Liposome particles, method for preparing said liposome particles and use thereof
WO2015085274A1 (en) 2013-12-05 2015-06-11 Centrillion Technology Holdings Corporation Methods for sequencing nucleic acids
EP3077430A4 (en) 2013-12-05 2017-08-16 Centrillion Technology Holdings Corporation Modified surfaces
CN111118121A (en) 2013-12-05 2020-05-08 生捷科技控股公司 Preparation of patterned arrays
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
US10385388B2 (en) 2013-12-06 2019-08-20 Swift Biosciences, Inc. Cleavable competitor polynucleotides
UA119253C2 (en) 2013-12-10 2019-05-27 Біолоджикс, Інк. Compositions and methods for virus control in varroa mite and bees
WO2015085350A1 (en) 2013-12-10 2015-06-18 Conexio Genomics Pty Ltd Methods and probes for identifying gene alleles
WO2015089277A1 (en) 2013-12-12 2015-06-18 The Regents Of The University Of California Methods and compositions for modifying a single stranded target nucleic acid
CA2931090A1 (en) 2013-12-12 2015-06-18 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
AU2014364520B2 (en) 2013-12-20 2020-01-02 The General Hospital Corporation Methods and assays relating to circulating tumor cells
JP6678583B2 (en) 2013-12-20 2020-04-22 バイオメッド バレー ディスカバリーズ,インコーポレイティド Cancer treatment using a combination of type 2 MEK inhibitor and ERK inhibitor
US9243289B2 (en) 2013-12-23 2016-01-26 Roche Molecular Systems, Inc. Method for screening reagents used in PCR assays
AU2014369900B2 (en) 2013-12-24 2021-05-20 Ionis Pharmaceuticals, Inc. Modulation of angiopoietin-like 3 expression
UA121462C2 (en) 2014-01-15 2020-06-10 Монсанто Текнолоджі Елелсі Methods and compositions for weed control using epsps polynucleotides
JP6666852B2 (en) 2014-01-16 2020-03-18 イルミナ インコーポレイテッド Gene expression panel for prognosis of prostate cancer recurrence
ES2838804T3 (en) 2014-01-31 2021-07-02 Univ Temple BAG3 as a target for the treatment of heart failure
AU2015214264B2 (en) 2014-02-04 2018-12-20 Curis, Inc. Mutant Smoothened and methods of using the same
CA2936158C (en) 2014-02-05 2023-06-13 Yeda Research And Development Co. Ltd. Use of mir-135 or precursor thereof for the treatment and diagnosis of a bipolar disease
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2015127389A1 (en) 2014-02-23 2015-08-27 The Johns Hopkins University Hypotonic enema formulations and methods of use
WO2015131107A1 (en) 2014-02-28 2015-09-03 Nugen Technologies, Inc. Reduced representation bisulfite sequencing with diversity adaptors
US10036019B2 (en) 2014-03-17 2018-07-31 Ionis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
MX2016012123A (en) 2014-03-19 2017-01-19 Ionis Pharmaceuticals Inc Compositions for modulating ataxin 2 expression.
US10006027B2 (en) 2014-03-19 2018-06-26 Ionis Pharmaceuticals, Inc. Methods for modulating Ataxin 2 expression
US10350264B2 (en) 2014-03-27 2019-07-16 Dana-Farber Cancer Institute, Inc. Compositions and methods for modulating NCOA4-mediated autophagic targeting of ferritin
US11060139B2 (en) 2014-03-28 2021-07-13 Centrillion Technology Holdings Corporation Methods for sequencing nucleic acids
ES2868305T3 (en) 2014-03-28 2021-10-21 Univ Washington Through Its Center For Commercialization Vaccines against breast and ovarian cancer
JP6574239B2 (en) 2014-03-30 2019-09-11 セフィエド Modified thymine polynucleotide oligomers and methods
US11091770B2 (en) 2014-04-01 2021-08-17 Monsanto Technology Llc Compositions and methods for controlling insect pests
ES2926869T3 (en) 2014-04-01 2022-10-31 Biogen Ma Inc Compositions to modulate the expression of SOD-1
TWI638047B (en) 2014-04-09 2018-10-11 史基普研究協會 Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
US10221416B2 (en) 2014-04-24 2019-03-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
CA2943894A1 (en) 2014-05-01 2015-11-05 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating complement factor b expression
EP3137115B1 (en) 2014-05-01 2020-10-14 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
EP3757215A3 (en) 2014-05-01 2021-03-17 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating growth hormone receptor expression
US9382540B2 (en) 2014-05-01 2016-07-05 Isis Pharmaceuticals, Inc Compositions and methods for modulating angiopoietin-like 3 expression
KR102366078B1 (en) 2014-05-01 2022-02-21 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating pkk expression
WO2015171918A2 (en) 2014-05-07 2015-11-12 Louisiana State University And Agricultural And Mechanical College Compositions and uses for treatment thereof
US11918695B2 (en) 2014-05-09 2024-03-05 Yale University Topical formulation of hyperbranched polymer-coated particles
EP3721875B1 (en) 2014-05-09 2023-11-08 Yale University Particles coated with hyperbranched polyglycerol and methods for their preparation
US10335500B2 (en) 2014-05-12 2019-07-02 The Johns Hopkins University Highly stable biodegradable gene vector platforms for overcoming biological barriers
WO2015175539A1 (en) 2014-05-12 2015-11-19 The Johns Hopkins University Engineering synthetic brain penetrating gene vectors
TW201607559A (en) 2014-05-12 2016-03-01 阿尼拉製藥公司 Methods and compositions for treating a SERPINC1-associated disorder
WO2015175977A2 (en) 2014-05-16 2015-11-19 Geller Bruce L Antisense antibacterial compounds and methods
US10391098B2 (en) 2014-05-19 2019-08-27 Board Of Regents, The University Of Texas System Antisense antibacterial compounds and methods
US10570169B2 (en) 2014-05-22 2020-02-25 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
TWI787678B (en) 2014-05-23 2022-12-21 美商健臻公司 Inhibiting or downregulating glycogen synthase by creating premature stop codons using antisense oligonucleotides
US10456445B2 (en) 2014-06-02 2019-10-29 Children's Medical Center Corporation Methods and compositions for immunomodulation
WO2015191596A1 (en) 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies using platinum agents and agents that target tumor-associated stroma or tumor cells
US10758614B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies targeting tumor-associated stroma or tumor cells and topoisomerase
US10799584B2 (en) 2014-06-09 2020-10-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies using agents that target tumor-associated stroma or tumor cells and alkylating agents
WO2015191617A2 (en) 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies using anti-metabolites and agents that target tumor-associated stroma or tumor cells
US10758526B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies using agents that target tumor-associated stroma or tumor cells and other pathways
WO2015191590A2 (en) 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and microtubules
US11034757B2 (en) 2014-06-09 2021-06-15 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and tumor vasculature
WO2015190922A1 (en) 2014-06-10 2015-12-17 Erasmus University Medical Center Rotterdam Antisense oligonucleotides useful in treatment of pompe disease
EP3154696B1 (en) 2014-06-10 2020-04-08 DxTerity Diagnostics Incorporated Devices and methods for collecting and stabilizing biological samples
TW201620526A (en) 2014-06-17 2016-06-16 愛羅海德研究公司 Compositions and methods for inhibiting gene expression of alpha-1 antitrypsin
AU2015280252A1 (en) 2014-06-23 2017-01-12 Monsanto Technology Llc Compositions and methods for regulating gene expression via RNA interference
US11807857B2 (en) 2014-06-25 2023-11-07 Monsanto Technology Llc Methods and compositions for delivering nucleic acids to plant cells and regulating gene expression
WO2015200697A1 (en) 2014-06-25 2015-12-30 The General Hospital Corporation Targeting human satellite ii (hsatii)
AU2015291151B2 (en) 2014-07-15 2019-10-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Isolated polypeptides of CD44 and uses thereof
JP2017524357A (en) * 2014-07-16 2017-08-31 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chimeric polynucleotide
US9951327B1 (en) 2014-07-17 2018-04-24 Integrated Dna Technologies, Inc. Efficient and rapid method for assembling and cloning double-stranded DNA fragments
EP3169699A4 (en) 2014-07-18 2018-06-20 The University of Washington Cancer vaccine compositions and methods of use thereof
CA3176503A1 (en) 2014-07-21 2016-01-28 Illumina, Inc Polynucleotide enrichment using crispr-cas systems
EP3171895A1 (en) 2014-07-23 2017-05-31 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
ES2752761T3 (en) 2014-07-24 2020-04-06 Abbott Molecular Inc Compositions for the detection and analysis of mycobacterium tuberculosis
US11072681B2 (en) 2014-07-28 2021-07-27 The Regents Of The University Of California Compositions and methods of making polymerizing nucleic acids
CA2955842A1 (en) 2014-07-29 2016-02-04 Monsanto Technology Llc Compositions and methods for controlling insect pests
MX2017001432A (en) 2014-07-31 2017-05-09 Uab Res Found Apoe mimetic peptides and higher potency to clear plasma cholesterol.
DK3174976T3 (en) 2014-08-01 2020-11-23 Gpb Scient Inc Particle processing methods and systems
US11046721B2 (en) 2014-08-04 2021-06-29 The Trustees Of The University Of Pennsylvania Ruthenium-based photolinkers and methods of use
US10980744B2 (en) 2014-08-08 2021-04-20 The Regents Of The University Of California High density peptide polymers
CA2958431A1 (en) 2014-08-19 2016-02-25 Northwestern University Protein/oligonucleotide core-shell nanoparticle therapeutics
US10465188B2 (en) 2014-08-22 2019-11-05 Auckland Uniservices Limited Channel modulators
WO2016030899A1 (en) 2014-08-28 2016-03-03 Yeda Research And Development Co. Ltd. Methods of treating amyotrophic lateral scleroses
ES2928500T3 (en) 2014-08-29 2022-11-18 Alnylam Pharmaceuticals Inc Patisiran for use in the treatment of transthyretin-mediated amyloidosis
JP2017526367A (en) 2014-08-29 2017-09-14 チルドレンズ メディカル センター コーポレーション Methods and compositions for the treatment of cancer
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
AU2015311563B2 (en) 2014-09-05 2019-11-28 Rsem, Limited Partnership Compositions and methods for treating and preventing inflammation
WO2016040589A1 (en) 2014-09-12 2016-03-17 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
US10913973B2 (en) 2014-09-17 2021-02-09 Board Of Regents, The University Texas System Methods and devices related to toehold-based strand displacement with loop-mediated isothermal amplification
US10556020B2 (en) 2014-09-26 2020-02-11 University Of Massachusetts RNA-modulating agents
JP2017534295A (en) 2014-09-29 2017-11-24 ザ ジャクソン ラボラトリー High-efficiency, high-throughput generation of genetically modified mammals by electroporation
AU2015325055B2 (en) 2014-10-01 2021-02-25 Eagle Biologics, Inc. Polysaccharide and nucleic acid formulations containing viscosity-lowering agents
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016057061A2 (en) 2014-10-10 2016-04-14 Massachusetts Eye And Ear Infirmary Efficient delivery of therapeutic molecules in vitro and in vivo
EP3206751A4 (en) 2014-10-14 2018-06-13 The J. David Gladstone Institutes Compositions and methods for reactivating latent immunodeficiency virus
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
US11370823B2 (en) 2014-10-29 2022-06-28 Massachusetts Eye And Ear Infirmary Efficient delivery of therapeutic molecules to cells of the inner ear
US11071790B2 (en) 2014-10-29 2021-07-27 Massachusetts Eye And Ear Infirmary Method for efficient delivery of therapeutic molecules in vitro and in vivo
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
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
US9598722B2 (en) 2014-11-11 2017-03-21 Genmark Diagnostics, Inc. Cartridge for performing assays in a closed sample preparation and reaction system
EP3218513B1 (en) 2014-11-11 2018-10-31 Illumina, Inc. Polynucleotide amplification using crispr-cas systems
EP3831481A1 (en) 2014-11-11 2021-06-09 Genmark Diagnostics Inc. Instrument and cartridge for performing assays in a closed sample preparation and reaction system
CN113846101A (en) 2014-11-17 2021-12-28 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
WO2016081621A1 (en) 2014-11-18 2016-05-26 Yale University Formulations for targeted release of agents under low ph conditions and methods of use thereof
US10682422B2 (en) 2014-11-18 2020-06-16 Yale University Formulations for targeted release of agents under low pH conditions and methods of use thereof
JP7175608B2 (en) 2014-11-19 2022-11-21 ザ トラスティーズ オブ コロンビア ユニバーシティ イン ザ シティ オブ ニューヨーク Osteocalcin as a treatment for age-related frailty
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
WO2016086104A1 (en) 2014-11-25 2016-06-02 Ionis Pharmaceuticals, Inc. Modulation of ube3a-ats expression
CA2969464A1 (en) 2014-12-12 2016-06-16 Tod M. Woolf Compositions and methods for editing nucleic acids in cells utilizing oligonucleotides
US9909169B2 (en) 2014-12-17 2018-03-06 Roche Molecular Systems, Inc. Allele-specific amplification of nucleic acids using blocking oligonucleotides for wild type suppression
BR112017014116A2 (en) 2014-12-30 2018-03-06 Telomere Diagnostics Inc quantitative multiplex pcr
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
AU2015372560B2 (en) 2014-12-31 2021-12-02 Board Of Regents, The University Of Texas System Antisense antibacterial compounds and methods
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
US10538763B2 (en) 2015-01-16 2020-01-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of DUX4
AU2016206475B2 (en) 2015-01-16 2021-11-04 City Of Hope Cell penetrating antibodies
US9434947B2 (en) 2015-01-20 2016-09-06 Oregon Health & Science University Modulation of KCNH2 isoform expression by oligonucleotides as a therapeutic approach for long QT syndrome
PL3256589T3 (en) 2015-01-22 2022-02-21 Monsanto Technology Llc Compositions and methods for controlling leptinotarsa
JP2018506715A (en) 2015-01-23 2018-03-08 ヴァンダービルト ユニバーシティー Robust interferometer and method of use
EP3250184A1 (en) 2015-01-27 2017-12-06 The Johns Hopkins University Hypotonic hydrogel formulations for enhanced transport of active agents at mucosal surfaces
US10676726B2 (en) 2015-02-09 2020-06-09 Duke University Compositions and methods for epigenome editing
KR20210135626A (en) 2015-02-10 2021-11-15 일루미나, 인코포레이티드 The method and the composition for analyzing the cellular constituent
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
US10450342B2 (en) 2015-02-23 2019-10-22 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
WO2016135559A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of human genetic diseases including hemoglobinopathies
HUE063406T2 (en) 2015-02-24 2024-01-28 Hope City Chemically encoded spatially addressed library screening platforms
CA2977965C (en) 2015-02-26 2021-12-21 Ionis Pharmaceuticals, Inc. Allele specific modulators of p23h rhodopsin
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
EP3268475B1 (en) 2015-03-11 2020-10-21 Yissum Research and Development Company of the Hebrew University of Jerusalem Ltd. Decoy oligonucleotides for the treatment of diseases
MA41795A (en) 2015-03-18 2018-01-23 Sarepta Therapeutics Inc EXCLUSION OF AN EXON INDUCED BY ANTISENSE COMPOUNDS IN MYOSTATIN
US9708647B2 (en) 2015-03-23 2017-07-18 Insilixa, Inc. Multiplexed analysis of nucleic acid hybridization thermodynamics using integrated arrays
US10300452B2 (en) 2015-03-24 2019-05-28 Pacific Biosciences Of California, Inc. Methods and compositions for single molecule composition loading
US10168342B2 (en) 2015-03-27 2019-01-01 The Scripps Research Institute Lipid probes and uses thereof
BR112017020750A2 (en) 2015-03-27 2018-06-26 Harvard College modified t-cells and methods of producing and using them
EP4218771A1 (en) 2015-03-27 2023-08-02 Yeda Research and Development Co. Ltd Methods of treating motor neuron diseases
WO2016161201A2 (en) 2015-03-31 2016-10-06 Oncosec Medical Incorporated Systems and methods for improved tissue-sensing based electroporation
US10961532B2 (en) 2015-04-07 2021-03-30 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
US20180126014A1 (en) 2015-04-15 2018-05-10 Yale University Compositions for enhancing delivery of agents across the blood brain barrier and methods of use thereof
CN104845967B (en) 2015-04-15 2020-12-11 苏州新海生物科技股份有限公司 Oligonucleotide fragment, method for selectively amplifying target nucleic acid sequence variant by using same and application of oligonucleotide fragment
KR102104163B1 (en) 2015-04-16 2020-04-23 아이오니스 파마수티컬즈, 인코포레이티드 Composition for modulating C9ORF72 expression
US10407678B2 (en) 2015-04-16 2019-09-10 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
WO2016176617A2 (en) 2015-04-29 2016-11-03 New York University Method for treating high-grade gliomas
EP3291677A4 (en) 2015-05-04 2019-02-13 Monsanto Technology LLC Compositions and methods for controlling arthropod parasite and pest infestations
WO2016181393A1 (en) 2015-05-11 2016-11-17 Yeda Research And Development Co. Ltd. Citrin inhibitors for the treatment of cancer
CA2986228A1 (en) 2015-05-19 2016-11-24 Sarepta Therapeutics, Inc. Peptide oligonucleotide conjugates
US10787664B2 (en) 2015-05-26 2020-09-29 City Of Hope Compounds of chemically modified oligonucleotides and methods of use thereof
US10196701B2 (en) 2015-06-01 2019-02-05 The Penn State Research Foundation Hepatitis B virus capsid assembly
EP3302497A4 (en) 2015-06-01 2019-01-16 Sarepta Therapeutics, Inc. Antisense-induced exon exclusion in type vii collagen
ES2886480T3 (en) 2015-06-01 2021-12-20 Univ Temple Methods and compositions for RNA-guided treatment of HIV infection
EP3302053B1 (en) 2015-06-02 2021-03-17 Monsanto Technology LLC Compositions and methods for delivery of a polynucleotide into a plant
EP3302030A4 (en) 2015-06-03 2019-04-24 Monsanto Technology LLC Methods and compositions for introducing nucleic acids into plants
EP3303634B1 (en) 2015-06-03 2023-08-30 The Regents of The University of California Cas9 variants and methods of use thereof
WO2016196897A1 (en) 2015-06-04 2016-12-08 Sarepta Therapeutics, Inc. Methods and compounds for treatment of lymphocyte-related diseases and conditions
CN108026494A (en) 2015-06-05 2018-05-11 米罗库鲁斯公司 Limitation evaporation and the digital microcurrent-controlled apparatus and method of air matrix of surface scale
CN208562324U (en) 2015-06-05 2019-03-01 米罗库鲁斯公司 Digital microcurrent-controlled (DMF) device of air matrix
WO2016201111A1 (en) 2015-06-09 2016-12-15 Centrillion Technology Holdings Corporation Methods for sequencing nucleic acids
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
CA2990852A1 (en) 2015-06-26 2016-12-29 Beth Israel Deaconess Medical Center, Inc. Cancer therapy targeting tetraspanin 33 (tspan33) in myeloid derived suppressor cells
EP3313989A4 (en) 2015-06-29 2018-12-05 Ionis Pharmaceuticals, Inc. Modified crispr rna and modified single crispr rna and uses thereof
US9861621B2 (en) 2015-06-29 2018-01-09 Biomed Valley Discoveries, Inc. LPT-723 and immune checkpoint inhibitor combinations and methods of treatment
EP3314027A4 (en) 2015-06-29 2019-07-03 Caris Science, Inc. Therapeutic oligonucleotides
IL256622B2 (en) 2015-07-10 2023-09-01 Ionis Pharmaceuticals Inc Modulators of diacyglycerol acyltransferase 2 (dgat2)
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
WO2017011565A1 (en) 2015-07-14 2017-01-19 Abbott Molecular Inc. Compositions and methods for identifying drug resistant tuberculosis
CA2993652A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
WO2017021963A1 (en) 2015-08-03 2017-02-09 Biokine Therapeutics Ltd. Cxcr4 binding agents for treatment of diseases
WO2017023861A1 (en) 2015-08-03 2017-02-09 The Regents Of The University Of California Compositions and methods for modulating abhd2 activity
EP3332009A1 (en) 2015-08-04 2018-06-13 Yeda Research and Development Co., Ltd. Methods of screening for riboswitches and attenuators
JP7104462B2 (en) 2015-08-06 2022-07-21 シティ・オブ・ホープ Cell-permeable protein-antibody conjugate and usage
CN108271387B (en) 2015-08-07 2023-06-27 箭头药业股份有限公司 RNAi therapy of hepatitis B virus infection
CN106434873B (en) 2015-08-13 2021-08-27 生捷科技控股公司 Method for synchronizing nucleic acid molecules
WO2017035278A1 (en) 2015-08-24 2017-03-02 Halo-Bio Rnai Therapeutics, Inc. Polynucleotide nanoparticles for the modulation of gene expression and uses thereof
US10889813B2 (en) 2015-09-02 2021-01-12 Alnylam Pharmaceuticals, Inc. Programmed cell death 1 ligand 1 (PD-L1) iRNA compositions and methods of use thereof
US9499861B1 (en) 2015-09-10 2016-11-22 Insilixa, Inc. Methods and systems for multiplex quantitative nucleic acid amplification
EP3347714B1 (en) 2015-09-10 2021-03-10 InSilixa Inc. Methods for multiplex quantitative nucleic acid amplification
WO2017049286A1 (en) 2015-09-17 2017-03-23 Moderna Therapeutics, Inc. Polynucleotides containing a morpholino linker
CA2998287A1 (en) 2015-09-24 2017-04-20 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
MX2018003685A (en) 2015-09-24 2018-08-15 Univ California Synthetic sphingolipid-like molecules, drugs, methods of their synthesis and methods of treatment.
AR106135A1 (en) 2015-09-24 2017-12-13 Ionis Pharmaceuticals Inc KIRSTEN RAT SARCOMA EXPRESSION MODULATORS (KRAS)
EP4285912A2 (en) 2015-09-25 2023-12-06 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
CN108738310A (en) 2015-09-30 2018-11-02 萨勒普塔医疗公司 Method for treating muscular dystrophy
JP2018530325A (en) 2015-10-08 2018-10-18 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Compounds and methods for modulating the expression of angiotensinogen
MA45819A (en) 2015-10-09 2018-08-15 Sarepta Therapeutics Inc COMPOSITIONS AND METHODS FOR TREATING DUCHENNE MUSCLE DYSTROPHY AND RELATED DISORDERS
US11905526B2 (en) 2015-10-13 2024-02-20 Duke University Genome engineering with Type I CRISPR systems in eukaryotic cells
EP3362102A1 (en) 2015-10-14 2018-08-22 Life Technologies Corporation Ribonucleoprotein transfection agents
EP3985116A1 (en) 2015-10-15 2022-04-20 City of Hope Compounds and compositions including phosphorothioated oligodeoxynucleotide, and methods of use thereof
SK500652015A3 (en) 2015-10-15 2017-05-03 Ústav Polymérov Sav A method for altering the functional state of mRNA allowing its selective and specific recognition
WO2017075038A1 (en) 2015-10-26 2017-05-04 Rana Therapeutics, Inc. Nanoparticle formulations for delivery of nucleic acid complexes
US11369692B2 (en) 2015-10-28 2022-06-28 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of Duchenne Muscular Dystrophy
CA3001362C (en) 2015-10-30 2020-10-13 Genentech, Inc. Anti-htra1 antibodies and methods of use thereof
AU2016349954B2 (en) 2015-11-05 2022-08-25 Antisense Therapeutics Ltd Mobilizing leukemia cells
CN109328231A (en) 2015-11-06 2019-02-12 克里斯普治疗股份公司 For treating the material and method of 1A type glycogen storage disease
US20190046555A1 (en) 2015-11-06 2019-02-14 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
CN108348478A (en) 2015-11-06 2018-07-31 Ionis 制药公司 Apolipoprotein (a) is adjusted to express
EP3373939A4 (en) 2015-11-10 2019-06-26 B.G. Negev Technologies and Applications Ltd., at Ben-Gurion University Means and methods for reducing tumorigenicity of cancer stem cells
CA3005878A1 (en) 2015-11-19 2017-05-26 The Brigham And Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
US20170145394A1 (en) 2015-11-23 2017-05-25 The Regents Of The University Of California Tracking and manipulating cellular rna via nuclear delivery of crispr/cas9
US10946106B2 (en) 2015-11-30 2021-03-16 The Regents Of The University Of California Tumor-specific payload delivery and immune activation using a human antibody targeting a highly specific tumor cell surface antigen
EP3967758A1 (en) 2015-12-01 2022-03-16 CRISPR Therapeutics AG Materials and methods for treatment of alpha-1 antitrypsin deficiency
WO2017096395A1 (en) 2015-12-04 2017-06-08 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
EP3387127A1 (en) 2015-12-07 2018-10-17 Erasmus University Medical Center Rotterdam Enzymatic replacement therapy and antisense therapy for pompe disease
WO2017106304A1 (en) 2015-12-15 2017-06-22 Sarepta Therapeutics, Inc. Peptide oligonucleotide conjugates
CA3007108A1 (en) 2015-12-17 2017-06-22 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase
WO2017106767A1 (en) 2015-12-18 2017-06-22 The Scripps Research Institute Production of unnatural nucleotides using a crispr/cas9 system
AU2016379399B2 (en) 2015-12-23 2022-12-08 Board Of Regents, The University Of Texas System Antisense antibacterial compounds and methods
EP3394262A4 (en) 2015-12-23 2019-12-25 Oregon State University Antisense antibacterial compounds and methods
WO2017109757A1 (en) 2015-12-23 2017-06-29 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis and/or frontal temporal lobular degeneration
EP3400300A4 (en) 2016-01-05 2019-08-07 Ionis Pharmaceuticals, Inc. Methods for reducing lrrk2 expression
JP7349788B2 (en) 2016-01-06 2023-09-25 ザ・ユニバーシティ・オブ・ブリティッシュ・コロンビア Branch mixer and its use and manufacturing method
SG11201805993UA (en) 2016-01-15 2018-08-30 Jackson Lab Genetically modified non-human mammals by multi-cycle electroporation of cas9 protein
US10627396B2 (en) 2016-01-29 2020-04-21 Vanderbilt University Free-solution response function interferometry
US20190038771A1 (en) 2016-02-02 2019-02-07 Crispr Therapeutics Ag Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
CN109071625A (en) 2016-02-04 2018-12-21 柯瑞斯公司 Smooth mutant and its application method
MX2018009904A (en) 2016-02-15 2019-07-08 Univ Temple Excision of retroviral nucleic acid sequences.
WO2017143042A2 (en) 2016-02-16 2017-08-24 Yale University Compositions for enhancing targeted gene editing and methods of use thereof
AU2017221424A1 (en) 2016-02-16 2018-09-20 Yale University Compositions and methods for treatment of cystic fibrosis
EP3416689B1 (en) 2016-02-18 2023-01-18 CRISPR Therapeutics AG Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
CN109312347A (en) 2016-02-19 2019-02-05 希望之城 Bispecific aptamer
JP7033072B2 (en) 2016-02-25 2022-03-09 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド Treatment for fibrosis targeting SMOC2
JP7025340B2 (en) 2016-02-26 2022-02-24 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー Multiplexed single molecule RNA visualization using a two-probe proximity ligation system
JP2019512014A (en) 2016-02-26 2019-05-09 イェール ユニバーシティーYale University Compositions and methods of using piRNA in cancer diagnostics and therapeutics
US20210189062A1 (en) 2016-03-01 2021-06-24 Alexion Pharmaceuticals, Inc. Biodegradable activated polymers for therapeutic delivery
WO2017155858A1 (en) 2016-03-07 2017-09-14 Insilixa, Inc. Nucleic acid sequence identification using solid-phase cyclic single base extension
WO2017152270A1 (en) 2016-03-08 2017-09-14 KeMyth Biotech Co., Ltd. Use of pneumolysin peptides as antagonists against toll-like receptor 4 and methods of treating toll-like receptor 4 related diseases
CA3013797A1 (en) 2016-03-09 2017-09-14 Ionis Pharmaceuticals, Inc. Methods and compositions for inhibiting pmp22 expression
US11083799B2 (en) 2016-03-16 2021-08-10 Crispr Therapeutics Ag Materials and methods for treatment of hereditary haemochromatosis
WO2017161172A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Methods of modulating keap1
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
IL303936A (en) 2016-03-18 2023-08-01 Caris Science Inc Oligonucleotide probes and uses thereof
US11845938B2 (en) 2016-03-31 2023-12-19 City Of Hope Aptamer compositions and the use thereof
WO2017173453A1 (en) 2016-04-01 2017-10-05 The Brigham And Women's Hospital, Inc. Stimuli-responsive nanoparticles for biomedical applications
WO2017180835A1 (en) 2016-04-13 2017-10-19 Ionis Pharmaceuticals, Inc. Methods for reducing c9orf72 expression
WO2017181163A2 (en) 2016-04-16 2017-10-19 Oncocyte Corporation Methods and compositions for detection and diagnosis of breast cancer
EP3445405A4 (en) 2016-04-18 2019-12-18 Sarepta Therapeutics, Inc. Antisense oligomers and methods of using the same for treating diseases associated with the acid alpha-glucosidase gene
EP3445388A2 (en) 2016-04-18 2019-02-27 CRISPR Therapeutics AG Materials and methods for treatment of hemoglobinopathies
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
AU2017258642B2 (en) 2016-04-29 2023-08-31 Sarepta Therapeutics, Inc. Oligonucleotide analogues targeting human LMNA
AU2017259821A1 (en) 2016-05-04 2018-12-13 Abilita Bio, Inc. Methods and platform for preparing multispanning membrane proteins
WO2017191503A1 (en) 2016-05-05 2017-11-09 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
CA3022319A1 (en) 2016-05-06 2017-11-09 Tod M. Woolf Improved methods for genome editing with and without programmable nucleases
CN116397007A (en) 2016-05-11 2023-07-07 伊鲁米那股份有限公司 Polynucleotide enrichment and amplification Using the ARGONAUTE System
WO2017197128A1 (en) 2016-05-11 2017-11-16 Yale University Poly(amine-co-ester) nanoparticles and methods of use thereof
WO2017201315A1 (en) 2016-05-18 2017-11-23 Roche Sequencing Solutions, Inc. Quantitative real time pcr amplification using an electrowetting-based device
US11472824B2 (en) 2016-05-24 2022-10-18 Sarepta Therapeutics, Inc. Processes for preparing phosphorodiamidate morpholino oligomers
WO2017205496A1 (en) 2016-05-24 2017-11-30 Sarepta Therapeutics, Inc. Processes for preparing oligomers
MA45362A (en) 2016-05-24 2019-04-10 Sarepta Therapeutics Inc PROCESSES FOR THE PREPARATION OF MORPHOLINO OLIGOMERS OF PHOSPHORODIAMIDATE
MX2018014129A (en) 2016-05-24 2019-04-29 Sarepta Therapeutics Inc Pharmaceutical composition comprising eteplirsen.
SG11201809499UA (en) 2016-05-24 2018-12-28 Sarepta Therapeutics Inc Processes for preparing phosphorodiamidate morpholino oligomers
CA3024456A1 (en) 2016-05-24 2017-11-30 Sarepta Therapeutics, Inc. Processes for preparing phosphorodiamidate morpholino oligomers
IL298701B2 (en) 2016-05-25 2024-03-01 Caris Science Inc Oligonucleotide probes and uses thereof
US11828755B2 (en) 2016-06-09 2023-11-28 The Regents Of The University Of California Biomarker concentration and signal amplification for use in paper-based immunoassays and a single platform for extracting, concentrating, and amplifying DNA
US11779657B2 (en) 2016-06-10 2023-10-10 City Of Hope Compositions and methods for mitochondrial genome editing
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
US10544457B2 (en) 2016-06-14 2020-01-28 Pacific Biosciences Of California, Inc. Methods and compositions for enriching compositions for polymerase enzyme complexes
JP7069135B2 (en) 2016-06-15 2022-05-17 ユニヴァーシティー オブ ユタ リサーチ ファウンデーション Compositions and Methods for Using Albumin-Based Nanopharmaceuticals
US10337051B2 (en) 2016-06-16 2019-07-02 The Regents Of The University Of California Methods and compositions for detecting a target RNA
EP3471781A4 (en) 2016-06-17 2020-05-06 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
WO2017223528A1 (en) 2016-06-24 2017-12-28 The Scripps Research Institute Novel nucleoside triphosphate transporter and uses thereof
WO2018002812A1 (en) 2016-06-29 2018-01-04 Crispr Therapeutics Ag Materials and methods for treatment of myotonic dystrophy type 1 (dm1) and other related disorders
AU2017290614C1 (en) 2016-06-29 2024-01-18 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
EP3478313B1 (en) 2016-06-29 2022-05-04 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis (als) and other related disorders
KR20190024977A (en) 2016-06-30 2019-03-08 사렙타 쎄러퓨틱스 인코퍼레이티드 Exon skipping oligomer for myopathies
CN109152792B (en) 2016-06-30 2021-09-14 萨勒普塔医疗公司 Process for preparing phosphoric acid diamide morpholino oligomer
AU2017292173B2 (en) 2016-07-06 2022-01-13 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of pain related disorders
EP3481856A1 (en) 2016-07-06 2019-05-15 Crispr Therapeutics AG Materials and methods for treatment of pain related disorders
WO2018007871A1 (en) 2016-07-08 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of transthyretin amyloidosis
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
EP3485032B1 (en) 2016-07-12 2021-02-17 Life Technologies Corporation Compositions and methods for detecting nucleic acid regions
BR112019000356A2 (en) 2016-07-15 2019-04-16 Ionis Pharmaceuticals, Inc. compounds and methods for smn2 modulation
KR20230088522A (en) 2016-07-21 2023-06-19 맥스시티 인코포레이티드 Methods and compositions for modifying genomic dna
US10711300B2 (en) 2016-07-22 2020-07-14 Pacific Biosciences Of California, Inc. Methods and compositions for delivery of molecules and complexes to reaction sites
WO2018020323A2 (en) 2016-07-25 2018-02-01 Crispr Therapeutics Ag Materials and methods for treatment of fatty acid disorders
CN109963564B (en) 2016-07-27 2022-08-12 利兰斯坦福初级大学董事会 Disaggregated cell-penetrating complexes for nucleic acid delivery
JOP20170161A1 (en) 2016-08-04 2019-01-30 Arrowhead Pharmaceuticals Inc RNAi Agents for Hepatitis B Virus Infection
NL2017295B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Antisense oligomeric compound for Pompe disease
NL2017294B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Natural cryptic exon removal by pairs of antisense oligonucleotides.
CN109715781A (en) 2016-08-22 2019-05-03 米罗库鲁斯公司 Feedback system for the parallel drop control in digital microcurrent-controlled equipment
WO2018039629A2 (en) 2016-08-25 2018-03-01 Northwestern University Micellar spherical nucleic acids from thermoresponsive, traceless templates
MX2019002339A (en) 2016-09-02 2019-05-16 Dicerna Pharmaceuticals Inc 4'-phosphate analogs and oligonucleotides comprising the same.
US20190218261A1 (en) 2016-09-13 2019-07-18 The Jackson Laboratory Targeted enhanced dna demethylation
WO2018053434A1 (en) 2016-09-16 2018-03-22 The Johns Hopkins University Protein nanocages with enhanced mucus penetration for targeted tissue and intracellular delivery
CA3036572A1 (en) 2016-09-19 2018-03-22 Genmark Diagnostics, Inc. Instrument for processing cartridge for performing assays in a closed sample preparation and reaction system
EP3515445A4 (en) 2016-09-20 2020-05-27 The Regents of The University of Colorado, A Body Corporate Synthesis of backbone modified morpholino oligonucleotides and chimeras using phosphoramidite chemistry
JOP20190065A1 (en) 2016-09-29 2019-03-28 Ionis Pharmaceuticals Inc Compounds and methods for reducing tau expression
CN110023494A (en) 2016-09-30 2019-07-16 加利福尼亚大学董事会 The nucleic acid modifying enzyme and its application method of RNA guidance
US11400161B2 (en) 2016-10-06 2022-08-02 Ionis Pharmaceuticals, Inc. Method of conjugating oligomeric compounds
WO2018081817A2 (en) 2016-10-31 2018-05-03 University Of Massachusetts Targeting microrna-101-3p in cancer therapy
JOP20190104A1 (en) 2016-11-10 2019-05-07 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
EP3538153A4 (en) 2016-11-11 2020-06-24 The Regents of the University of California Anti-cd46 antibodies and methods of use
WO2018089967A1 (en) 2016-11-14 2018-05-17 Virginia Commonwealth University Inhibitors of cancer invasion, attachment, and/or metastasis
TW202313978A (en) 2016-11-23 2023-04-01 美商阿尼拉製藥公司 Serpina1 irna compositions and methods of use thereof
EP3330276A1 (en) 2016-11-30 2018-06-06 Universität Bern Novel bicyclic nucleosides and oligomers prepared therefrom
US11033570B2 (en) 2016-12-02 2021-06-15 Cold Spring Harbor Laboratory Modulation of Lnc05 expression
AU2017378153B2 (en) 2016-12-13 2024-03-28 Seattle Children's Hospital (dba Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
TWI790217B (en) 2016-12-16 2023-01-21 美商阿尼拉製藥公司 METHODS FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES USING TRANSTHYRETIN (TTR) iRNA COMPOSITIONS
WO2018112470A1 (en) 2016-12-16 2018-06-21 The Brigham And Women's Hospital, Inc. Co-delivery of nucleic acids for simultaneous suppression and expression of target genes
SG10202012839TA (en) 2016-12-19 2021-01-28 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
CN110337308B (en) 2016-12-19 2023-09-01 萨勒普塔医疗公司 Exon-skipping oligomer conjugates for muscular dystrophy
IL267246B2 (en) 2016-12-19 2023-03-01 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
WO2018126082A1 (en) 2016-12-28 2018-07-05 Miroculis Inc. Digital microfluidic devices and methods
CN110268060A (en) 2017-01-10 2019-09-20 箭头药业股份有限公司 α -1 antitrypsin (AAT) RNAi substance, composition and application method comprising AAT RNAi substance
EP3570829A4 (en) 2017-01-18 2021-03-10 The Scripps Research Institute Photoreactive ligands and uses thereof
RU2021133626A (en) 2017-01-23 2022-01-31 Регенерон Фармасьютикалз, Инк. HSD17B13 OPTIONS AND THEIR APPLICATIONS
WO2018140329A1 (en) 2017-01-24 2018-08-02 Tsavachidou Dimitra Methods for constructing copies of nucleic acid molecules
US20200040061A1 (en) 2017-02-22 2020-02-06 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
WO2018154387A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Compositions and methods for gene editing
EP3585899A1 (en) 2017-02-22 2020-01-01 CRISPR Therapeutics AG Materials and methods for treatment of primary hyperoxaluria type 1 (ph1) and other alanine-glyoxylate aminotransferase (agxt) gene related conditions or disorders
US20200216857A1 (en) 2017-02-22 2020-07-09 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
WO2018154439A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 1 (sca1) and other spinocerebellar ataxia type 1 protein (atxn1) gene related conditions or disorders
EP3596099A4 (en) 2017-03-06 2020-12-09 Singular Genomics Systems, Inc. Nucleic acid sequencing-by-synthesis (sbs) methods that combine sbs cycle steps
WO2018165564A1 (en) 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
WO2018169960A1 (en) 2017-03-17 2018-09-20 The Johns Hopkins University Nanoparticle formulations for enhanced drug delivery to the bladder
JOP20190215A1 (en) 2017-03-24 2019-09-19 Ionis Pharmaceuticals Inc Modulators of pcsk9 expression
US20180284123A1 (en) 2017-03-30 2018-10-04 California Institute Of Technology Barcoded rapid assay platform useful for efficient analysis of candidate molecules and methods of making and using the platform
WO2018187493A1 (en) 2017-04-04 2018-10-11 Yale University Compositions and methods for in utero delivery
US11623219B2 (en) 2017-04-04 2023-04-11 Miroculus Inc. Digital microfluidics apparatuses and methods for manipulating and processing encapsulated droplets
TWI801377B (en) 2017-04-18 2023-05-11 美商阿尼拉製藥公司 Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
EP3612546B1 (en) 2017-04-20 2022-07-13 Synthena AG Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
CN110536694A (en) 2017-04-20 2019-12-03 Atyr 医药公司 For treating pulmonary inflammatory composition and method
WO2018193428A1 (en) 2017-04-20 2018-10-25 Synthena Ag Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
AU2018255972A1 (en) 2017-04-21 2019-10-31 Phi Therapeutics, Inc. Compositions comprising propionibacterium acnes bacteriophages for treating acne
US20200384115A1 (en) 2017-04-21 2020-12-10 The Broad Institute , Inc. Targeted delivery to beta cells
CA3061738A1 (en) 2017-04-28 2018-11-01 Auckland Uniservices Limited Methods of treatment and novel constructs
US11453891B2 (en) 2017-05-10 2022-09-27 The Regents Of The University Of California Directed editing of cellular RNA via nuclear delivery of CRISPR/CAS9
US11299776B2 (en) 2017-05-10 2022-04-12 Board Of Regents, The University Of Texas System Methods and devices related to amplifying nucleic acid at a variety of temperatures
BR112019023608A2 (en) 2017-05-12 2020-05-26 Crispr Therapeutics Ag MATERIALS AND METHODS FOR HANDLED CELLS AND THEIR USES IN IMMUNO-ONCOLOGY
EP3626821A4 (en) 2017-05-18 2021-03-03 Kyoto University Composition for prevention or treatment of spinocerebellar ataxia type 36
US11554178B2 (en) 2017-06-30 2023-01-17 City Of Hope Compositions and methods of modulating macrophage activity
TWI821192B (en) 2017-07-11 2023-11-11 美商新索思股份有限公司 Incorporation of unnatural nucleotides and methods thereof
JP2020526558A (en) 2017-07-13 2020-08-31 ノースウェスタン ユニバーシティ Common and direct methods for preparing oligonucleotide functionalized metal-organic framework nanoparticles
EP3652317A1 (en) 2017-07-13 2020-05-20 Alnylam Pharmaceuticals, Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
EP3651807B1 (en) 2017-07-13 2023-11-01 City of Hope Phosphorothioate-conjugated peptides and methods of using the same
CA3069179A1 (en) 2017-07-13 2019-01-17 Massachusetts Institute Of Technology Targeting the hdac2-sp3 complex to enhance synaptic function
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
CN110892258A (en) 2017-07-24 2020-03-17 米罗库鲁斯公司 Digital microfluidic system and method with integrated plasma collection device
US11459306B2 (en) 2017-07-31 2022-10-04 The Trustees Of Columbia University In The City Of New York Compounds, compositions, and methods for treating T-cell acute lymphoblastic leukemia
KR20200035092A (en) 2017-08-03 2020-04-01 신톡스, 인크. Cytokine conjugates for the treatment of autoimmune diseases
NL2019390B1 (en) 2017-08-04 2019-02-21 Univ Leiden Screening Method
EP3665202A1 (en) 2017-08-09 2020-06-17 Massachusetts Institute Of Technology Albumin binding peptide conjugates and methods thereof
CN111542326A (en) 2017-08-11 2020-08-14 希望之城 RNA aptamers to transferrin receptor (TfR)
CA3071033A1 (en) 2017-08-18 2019-02-21 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
LT3675826T (en) 2017-08-31 2023-09-11 Life Technologies Corporation Cationic lipid compositions for tissue-specific delivery
CN115582155A (en) 2017-09-01 2023-01-10 米罗库鲁斯公司 Digital microfluidic device and method of use thereof
CN111315890A (en) 2017-09-08 2020-06-19 生命技术公司 Methods and compositions for enhancing homologous recombination
US20190119701A1 (en) 2017-09-08 2019-04-25 Life Technologies Corporation Methods for improved homologous recombination and compositions thereof
WO2019051173A1 (en) 2017-09-08 2019-03-14 Ionis Pharmaceuticals, Inc. Modulators of smad7 expression
JP2021508333A (en) 2017-09-19 2021-03-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Therapeutic compositions and methods for transthyretin (TTR) -mediated amyloidosis
EA201991450A1 (en) 2017-09-22 2019-12-30 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXONISM SKIP IN MUSCULAR DYSTROPHY
US20200254002A1 (en) 2017-09-28 2020-08-13 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
EP3687547A1 (en) 2017-09-28 2020-08-05 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
JP2020536058A (en) 2017-09-28 2020-12-10 サレプタ セラピューティクス, インコーポレイテッド Combination therapy to treat muscular dystrophy
MX2020004043A (en) 2017-10-17 2021-05-27 Crispr Therapeutics Ag Compositions and methods for gene editing for hemophilia a.
US11555189B2 (en) 2017-10-18 2023-01-17 Sarepta Therapeutics, Inc. Antisense oligomer compounds
US11099202B2 (en) 2017-10-20 2021-08-24 Tecan Genomics, Inc. Reagent delivery system
US20210180091A1 (en) 2017-10-26 2021-06-17 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
EP3704245A1 (en) 2017-11-01 2020-09-09 Novartis AG Synthetic rnas and methods of use
EP3707155A2 (en) 2017-11-09 2020-09-16 Vertex Pharmaceuticals Incorporated Crispr/cas systems for treatment of dmd
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
US11691141B2 (en) 2017-11-13 2023-07-04 Roche Sequencing Solutions, Inc. Devices for sample analysis using epitachophoresis
WO2019099610A1 (en) 2017-11-16 2019-05-23 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
US10953036B2 (en) 2017-11-20 2021-03-23 University Of Georgia Research Foundation, Inc. Compositions and methods of modulating HIF-2A to improve muscle generation and repair
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
US20190153440A1 (en) 2017-11-21 2019-05-23 Casebia Therapeutics Llp Materials and methods for treatment of autosomal dominant retinitis pigmentosa
US11728007B2 (en) 2017-11-30 2023-08-15 Grail, Llc Methods and systems for analyzing nucleic acid sequences using mappability analysis and de novo sequence assembly
WO2019113149A1 (en) 2017-12-05 2019-06-13 Crispr Therapeutics Ag Crispr-cas9 modified cd34+ human hematopoietic stem and progenitor cells and uses thereof
JP2021506239A (en) 2017-12-14 2021-02-22 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Composite antisense compounds and their use
JP2021506251A (en) 2017-12-14 2021-02-22 クリスパー セラピューティクス アーゲー New RNA programmable endonuclease system, as well as its use in genome editing and other applications
MX2020006012A (en) 2017-12-18 2020-09-14 Alnylam Pharmaceuticals Inc High mobility group box-1 (hmgb1) irna compositions and methods of use thereof.
US11939575B2 (en) 2017-12-19 2024-03-26 City Of Hope Modified tracrRNAs gRNAs, and uses thereof
EP3728594A1 (en) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Materials and methods for treatment of usher syndrome type 2a
EP3728595A1 (en) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Materials and methods for treatment of usher syndrome type 2a and/or non-syndromic autosomal recessive retinitis pigmentosa (arrp)
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
US11242568B2 (en) 2017-12-29 2022-02-08 City Of Hope DNA methylation diagnostic test for breast cancer
WO2019140330A1 (en) 2018-01-12 2019-07-18 Casebia Therapeutics Limited Liability Partnership Compositions and methods for gene editing by targeting transferrin
CN111902537A (en) 2018-01-15 2020-11-06 Ionis制药公司 Modulators of DNM2 expression
EP3740472A1 (en) 2018-01-19 2020-11-25 Synthena AG Tricyclo-dna nucleoside precursors and processes for preparing the same
WO2019147743A1 (en) 2018-01-26 2019-08-01 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
EP3749768A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
EP3749767A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2019155465A1 (en) 2018-02-08 2019-08-15 Yeda Research And Development Co. Ltd. Methods of identifying and using agents for treating diseases associated with intestinal barrier dysfunction
JP7317029B2 (en) 2018-02-12 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド Modified compounds and uses thereof
EP3752616A1 (en) 2018-02-16 2020-12-23 CRISPR Therapeutics AG Compositions and methods for gene editing by targeting fibrinogen-alpha
AU2019224159A1 (en) 2018-02-26 2020-10-01 Synthorx, Inc. IL-15 conjugates and uses thereof
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
EP3759127A4 (en) 2018-03-02 2022-03-30 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-beta precursor protein
CN112105625A (en) 2018-03-07 2020-12-18 赛诺菲 Nucleotide precursors, nucleotide analogs, and oligomeric compounds containing the same
AU2019234836A1 (en) 2018-03-13 2020-09-17 The Regents Of The University Of California Inhibitors of integrin alpha 2 beta 1 and methods of use
EP3768834A1 (en) 2018-03-19 2021-01-27 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and uses thereof
EP3768694A4 (en) 2018-03-22 2021-12-29 Ionis Pharmaceuticals, Inc. Methods for modulating fmr1 expression
CA3095545A1 (en) 2018-03-30 2019-10-03 Rheinische Friedrich-Wilhelms-Universitat Bonn Aptamers for targeted activaton of t cell-mediated immunity
JP2021520781A (en) 2018-04-06 2021-08-26 チルドレンズ メディカル センター コーポレーションChildren’S Medical Center Corporation Compositions and Methods for Somatic Cell Reprogramming and Imprinting Modulation
JP7275164B2 (en) 2018-04-11 2023-05-17 アイオーニス ファーマシューティカルズ, インコーポレーテッド Regulators of EZH2 expression
WO2019204668A1 (en) 2018-04-18 2019-10-24 Casebia Therapeutics Limited Liability Partnership Compositions and methods for knockdown of apo(a) by gene editing for treatment of cardiovascular disease
US20210189460A1 (en) 2018-04-25 2021-06-24 Qiagen Sciences Llc Sequential paired-end sequencing
UY38225A (en) 2018-05-09 2019-11-29 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN3
JP7438135B2 (en) 2018-05-09 2024-02-26 アイオーニス ファーマシューティカルズ, インコーポレーテッド Compounds and methods for reducing expression of FXI
TW202016304A (en) 2018-05-14 2020-05-01 美商阿尼拉製藥公司 Angiotensinogen (agt) irna compositions and methods of use thereof
US10765760B2 (en) 2018-05-29 2020-09-08 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
CN113166191A (en) 2018-05-30 2021-07-23 Dtx医药有限公司 Lipid-modified nucleic acid compounds and methods
US10987428B2 (en) 2018-06-01 2021-04-27 City Of Hope Phosphorothioate-conjugated miRNAs and methods of using the same
EP3806868A4 (en) 2018-06-13 2022-06-22 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
EP3807411A4 (en) 2018-06-14 2022-08-03 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing stmn2 expression
SG11202011864XA (en) 2018-06-27 2020-12-30 Ionis Pharmaceuticals Inc Compounds and methods for reducing lrrk2 expression
BR112020025824A2 (en) 2018-06-28 2021-03-23 Crispr Therapeutics Ag compositions and methods for genome editing by inserting donor polynucleotides
SG11202100077PA (en) 2018-07-25 2021-02-25 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn2 expression
TW202020153A (en) 2018-07-27 2020-06-01 美商薩羅塔治療公司 Exon skipping oligomers for muscular dystrophy
US10857174B2 (en) 2018-07-27 2020-12-08 United States Government As Represented By The Department Of Veterans Affairs Morpholino oligonucleotides useful in cancer treatment
EP3830301A1 (en) 2018-08-01 2021-06-09 Mammoth Biosciences, Inc. Programmable nuclease compositions and methods of use thereof
EP3830278A4 (en) 2018-08-01 2022-05-25 University of Georgia Research Foundation, Inc. Compositions and methods for improving embryo development
US20210189431A1 (en) 2018-08-10 2021-06-24 Yale University Compositions and methods for embryonic gene editing in vitro
JP2021533767A (en) 2018-08-13 2021-12-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Hepatitis B virus (HBV) dsRNA substance composition and its usage
EP3837367A1 (en) 2018-08-16 2021-06-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
AU2019325255A1 (en) 2018-08-20 2021-04-15 Rogcon, Inc. Antisense oligonucleotides targeting SCN2A for the treatment of SCN1A encephalopathies
WO2020047229A1 (en) 2018-08-29 2020-03-05 University Of Massachusetts Inhibition of protein kinases to treat friedreich ataxia
US20210338815A1 (en) 2018-08-31 2021-11-04 Yale University Compositions and methods for enhancing triplex and nuclease-based gene editing
EP3844287A1 (en) 2018-08-31 2021-07-07 Yale University Compositions and methods for enhancing donor oligonucleotide-based gene editing
WO2020051507A1 (en) 2018-09-06 2020-03-12 The Broad Institute, Inc. Nucleic acid assemblies for use in targeted delivery
US10724052B2 (en) 2018-09-07 2020-07-28 Crispr Therapeutics Ag Universal donor cells
EP3620520A1 (en) 2018-09-10 2020-03-11 Universidad del Pais Vasco Novel target to treat a metabolic disease in an individual
US20230011240A1 (en) 2018-09-11 2023-01-12 Singular Genomics Systems, Inc. Modified archaeal family b polymerases
AU2019339509A1 (en) 2018-09-14 2021-05-13 Northwestern University Programming protein polymerization with DNA
JP2022500003A (en) 2018-09-18 2022-01-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA composition and its usage
TW202023573A (en) 2018-09-19 2020-07-01 美商Ionis製藥公司 Modulators of pnpla3 expression
US20210393740A1 (en) 2018-09-19 2021-12-23 La Jolla Institute For Immunology Ptprs and proteoglycans in rheumatoid arthritis
WO2020074742A1 (en) 2018-10-12 2020-04-16 F. Hoffmann-La Roche Ag Detection methods for epitachophoresis workflow automation
MX2021004455A (en) 2018-10-17 2021-08-11 Crispr Therapeutics Ag Compositions and methods for delivering transgenes.
WO2020086834A1 (en) 2018-10-25 2020-04-30 Singular Genomics Systems, Inc. Nucleotide analogues
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
CN113646430A (en) 2018-11-15 2021-11-12 Ionis制药公司 Modulators of IRF5 expression
NL2022043B1 (en) 2018-11-21 2020-06-03 Akershus Univ Hf Tagmentation-Associated Multiplex PCR Enrichment Sequencing
US20220025366A1 (en) 2018-11-21 2022-01-27 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing prion expression
IL263184A (en) 2018-11-21 2020-05-31 Yarden Yosef Method of treating cancer and compositions for same
WO2020112195A1 (en) 2018-11-30 2020-06-04 Yale University Compositions, technologies and methods of using plerixafor to enhance gene editing
WO2020118259A1 (en) 2018-12-06 2020-06-11 Northwestern University Protein crystal engineering through dna hybridization interactions
EA202191601A1 (en) 2018-12-13 2022-01-19 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXON SKIP IN MUSCULAR DYSTROPHY
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
JP2022512265A (en) 2018-12-17 2022-02-03 イルミナ ケンブリッジ リミテッド Primer oligonucleotides for sequencing
CA3103744A1 (en) 2018-12-17 2020-06-25 Pietro GATTI-LAFRANCONI Compositions for use in polynucleotide sequencing
HUE064532T2 (en) 2018-12-20 2024-03-28 Vir Biotechnology Inc Combination hbv therapy
AU2019406186A1 (en) 2018-12-20 2021-07-15 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of KCNT1 related disorders
EP3674702A1 (en) 2018-12-27 2020-07-01 Imec VZW Method for sequencing a polynucleotide using a biofet
WO2020142754A2 (en) 2019-01-04 2020-07-09 Mammoth Biosciences, Inc. Programmable nuclease improvements and compositions and methods for nucleic acid amplification and detection
US10822653B1 (en) 2019-01-08 2020-11-03 Singular Genomics Systems, Inc. Nucleotide cleavable linkers and uses thereof
TW202043471A (en) 2019-01-16 2020-12-01 美商健贊公司 Serpinc1 irna compositions and methods of use thereof
WO2020157760A1 (en) 2019-01-31 2020-08-06 Bar Ilan University Neoantigens created by aberrant-induced splicing and uses thereof in enhancing immunotherapy
SG11202107399WA (en) 2019-01-31 2021-08-30 Ionis Pharmaceuticals Inc Modulators of yap1 expression
CN113811334B (en) 2019-02-01 2024-02-20 利兰斯坦福初级大学董事会 Lytic cell penetrating complexes for delivery of nucleic acids to the lung
SG11202107354WA (en) 2019-02-06 2021-08-30 Synthorx Inc Il-2 conjugates and methods of use thereof
EP3921418A4 (en) 2019-02-06 2023-02-08 Singular Genomics Systems, Inc. Compositions and methods for nucleic acid sequencing
CA3129532A1 (en) 2019-02-15 2020-08-20 Crispr Therapeutics Ag Gene editing for hemophilia a with improved factor viii expression
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
WO2020170240A1 (en) 2019-02-21 2020-08-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reduction drug-induced nephrotoxicity
US11504425B2 (en) 2019-02-26 2022-11-22 Wayne State University Amphiphilic oligodeoxynucleotide conjugates as adjuvant enhancers
JP2022523214A (en) 2019-02-27 2022-04-21 アイオーニス ファーマシューティカルズ, インコーポレーテッド Modulator of MALAT1 expression
EP3935184A1 (en) 2019-03-04 2022-01-12 King Abdullah University Of Science And Technology Compositions and methods of targeted nucleic acid enrichment by loop adapter protection and exonuclease digestion
GB2595606B (en) 2019-03-07 2022-09-21 Univ California CRISPR-Cas effector polypeptides and methods of use thereof
CN113825834A (en) 2019-03-11 2021-12-21 索伦托药业有限公司 Improved process for DNA construct integration using RNA-guided endonuclease
SG11202109741VA (en) 2019-03-12 2021-10-28 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
EP3937780A4 (en) 2019-03-14 2022-12-07 InSilixa, Inc. Methods and systems for time-gated fluorescent-based detection
MA55515A (en) 2019-03-28 2022-02-09 Sarepta Therapeutics Inc METHODS OF TREATING MUSCULAR DYSTROPHY WITH CASIMERSEN
CA3198063A1 (en) 2019-03-29 2020-10-08 Mitsubishi Tanabe Pharma Corporation Compound, method and pharmaceutical composition for modulating expression of dux4
TW202043251A (en) 2019-03-29 2020-12-01 美商Ionis製藥公司 Compounds and methods for modulating ube3a-ats
AU2020253823A1 (en) 2019-03-29 2021-10-14 Dicerna Pharmaceuticals, Inc. Compositions and methods for the treatment of KRAS associated diseases or disorders
EP3953041A4 (en) 2019-04-08 2023-01-25 Miroculus Inc. Multi-cartridge digital microfluidics apparatuses and methods of use
WO2020214763A1 (en) 2019-04-18 2020-10-22 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US11814464B2 (en) 2019-04-29 2023-11-14 Yale University Poly(amine-co-ester) polymers and polyplexes with modified end groups and methods of use thereof
BR112021021686A2 (en) 2019-05-03 2022-03-22 Dicerna Pharmaceuticals Inc Double-stranded nucleic acid inhibitor molecules with short sense strands
WO2020225606A1 (en) 2019-05-08 2020-11-12 Crispr Therapeutics Ag Crispr/cas all-in-two vector systems for treatment of dmd
CN114126628A (en) 2019-05-13 2022-03-01 维尔生物科技有限公司 Compositions and methods for treating Hepatitis B Virus (HBV) infection
EP3969583A1 (en) 2019-05-14 2022-03-23 F. Hoffmann-La Roche AG Devices and methods for sample analysis
EP3969591A1 (en) 2019-05-15 2022-03-23 Squadra Lifesciences, Inc Aptamers against glioblastoma
US20220396794A1 (en) 2019-06-04 2022-12-15 Apterna Limited APTAMERS AGAINST TRANSFERRIN RECEPTOR (TfR)
EP3983543A4 (en) 2019-06-14 2023-05-03 The Scripps Research Institute Reagents and methods for replication, transcription, and translation in semi-synthetic organisms
CA3142521A1 (en) 2019-06-17 2020-12-24 Vertex Pharmaceuticals Incorporated Compositions and methods for editing beta-globin for treatment of hemaglobinopathies
US20220296633A1 (en) 2019-06-19 2022-09-22 Sarepta Therapeutics, Inc. Methods for treating muscular dystrophy
US20220372474A1 (en) 2019-06-21 2022-11-24 Yale University Hydroxymethyl-modified gamma-pna compositions and methods of use thereof
WO2020257776A1 (en) 2019-06-21 2020-12-24 Yale University Peptide nucleic acid compositions with modified hoogsteen binding segments and methods of use thereof
US11377655B2 (en) 2019-07-16 2022-07-05 Pacific Biosciences Of California, Inc. Synthetic nucleic acids having non-natural structures
BR112022001315A2 (en) 2019-07-25 2022-04-12 Aspen Neuroscience Inc Methods for identification of dopaminergic neurons and progenitor cells
US11524298B2 (en) 2019-07-25 2022-12-13 Miroculus Inc. Digital microfluidics devices and methods of use thereof
WO2021021673A1 (en) 2019-07-26 2021-02-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
US20220280656A1 (en) 2019-07-31 2022-09-08 Yale University Compositions and methods for treating sickle cell disease
EP4007811A2 (en) 2019-08-01 2022-06-08 Alnylam Pharmaceuticals, Inc. Carboxypeptidase b2 (cpb2) irna compositions and methods of use thereof
WO2021022109A1 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. SERPIN FAMILY F MEMBER 2 (SERPINF2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021025899A1 (en) 2019-08-02 2021-02-11 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer pharmaceutical compositions
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
MX2022001770A (en) 2019-08-14 2022-05-20 Codiak Biosciences Inc Extracellular vesicle linked to molecules and uses thereof.
KR20220062517A (en) 2019-08-15 2022-05-17 아이오니스 파마수티컬즈, 인코포레이티드 Linkage-modified oligomeric compounds and uses thereof
CN114555128A (en) 2019-08-15 2022-05-27 新索思股份有限公司 Combination immunooncology therapy with IL-2 conjugates
CA3148135A1 (en) 2019-08-23 2021-03-04 Carolina E. CAFFARO Il-15 conjugates and uses thereof
JP2022546699A (en) 2019-08-30 2022-11-07 イェール ユニバーシティー Compositions and methods for delivering nucleic acids to cells
JP2022546570A (en) 2019-09-03 2022-11-04 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting expression of the LECT2 gene
MX2022002663A (en) 2019-09-05 2022-04-07 Crispr Therapeutics Ag Universal donor cells.
AU2020341186A1 (en) 2019-09-05 2022-03-03 Crispr Therapeutics Ag Universal donor cells
EP4025582A1 (en) 2019-09-05 2022-07-13 Sanofi Oligonucleotides containing nucleotide analogs
JP2022547078A (en) 2019-09-10 2022-11-10 シンソークス, インコーポレイテッド IL-2 conjugates and methods of use for treating autoimmune diseases
TW202124406A (en) 2019-09-10 2021-07-01 美商歐姆尼歐美公司 Reversible modification of nucleotides
JP2022547678A (en) 2019-09-11 2022-11-15 ボシュ ヘルス アイルランド リミテッド Methods of treating non-alcoholic fatty liver disease (NAFLD) using IL-17RA antibodies
US11512295B2 (en) 2019-09-12 2022-11-29 Singular Genomics Systems, Inc. Modified thermoccocus polymerases
EP4038189A1 (en) 2019-10-04 2022-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021071909A1 (en) 2019-10-08 2021-04-15 Life Technologies Corporation Compositions and methods for homology-directed recombination
JP2022552249A (en) 2019-10-14 2022-12-15 アストラゼネカ・アクチエボラーグ Modulators of PNPLA3 expression
WO2021076828A1 (en) 2019-10-18 2021-04-22 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
BR112022007540A2 (en) 2019-10-22 2022-07-12 Alnylam Pharmaceuticals Inc COMPONENTS COMPLEMENTARY C3 IRNA COMPOSITIONS AND METHODS OF USE THEREOF
SG11202110571YA (en) 2019-10-25 2021-10-28 Illumina Cambridge Ltd Methods for generating, and sequencing from, asymmetric adaptors on the ends of polynucleotide templates comprising hairpin loops
MX2022004388A (en) 2019-11-01 2022-05-06 Alnylam Pharmaceuticals Inc HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF.
US20230040920A1 (en) 2019-11-01 2023-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
IL291786A (en) 2019-11-04 2022-06-01 Synthorx Inc Interleukin 10 conjugates and uses thereof
TW202132568A (en) 2019-11-13 2021-09-01 美商阿尼拉製藥公司 Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
US20230056569A1 (en) 2019-11-22 2023-02-23 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
JP2023503635A (en) 2019-11-27 2023-01-31 クリスパー・セラピューティクス・アクチェンゲゼルシャフト Methods of synthesizing RNA molecules
JP2023504191A (en) 2019-12-04 2023-02-01 ザ・ボード・オブ・トラスティーズ・オブ・ザ・リーランド・スタンフォード・ジュニア・ユニバーシティ Enhancement of blood-brain barrier drug transport by targeting endogenous regulators
EP4073251A1 (en) 2019-12-13 2022-10-19 Alnylam Pharmaceuticals, Inc. Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
EP4077676A1 (en) 2019-12-18 2022-10-26 Novartis AG Compositions and methods for the treatment of hemoglobinopathies
US20230054569A1 (en) 2019-12-18 2023-02-23 Alia Therapeutics Srl Compositions and methods for treating retinitis pigmentosa
CA3165862A1 (en) 2019-12-23 2021-07-01 The Regents Of The University Of California Stabilization of mhc complexes
CA3165571C (en) 2019-12-23 2023-02-07 Singular Genomics Systems, Inc. Methods for long read sequencing
EP4085135A4 (en) 2019-12-31 2023-08-02 Singular Genomics Systems, Inc. Polynucleotide barcodes for long read sequencing
US11859172B2 (en) 2020-01-02 2024-01-02 The Trustees Of Columbia University In The City Of New York Programmable and portable CRISPR-Cas transcriptional activation in bacteria
MX2022008738A (en) 2020-01-15 2022-09-23 Dicerna Pharmaceuticals Inc 4'-o-methylene phosphonate nucleic acids and analogues thereof.
US20230057461A1 (en) 2020-01-27 2023-02-23 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Rab13 and net1 antisense oligonucleotides to treat metastatic cancer
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
WO2021163281A1 (en) 2020-02-12 2021-08-19 Accutar Biotechnology Inc. Antisense oligonucleotides and their use for treating pendred syndrome
MX2022010052A (en) 2020-02-18 2022-09-05 Alnylam Pharmaceuticals Inc Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof.
US11034942B1 (en) 2020-02-27 2021-06-15 Singular Genomics Systems, Inc. Modified pyrococcus polymerases and uses thereof
PE20221913A1 (en) 2020-02-28 2022-12-23 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS FOR MODULATING SMN2
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
WO2021178778A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin (ttr)
WO2021178893A2 (en) 2020-03-06 2021-09-10 Singular Genomics Systems, Inc. Linked paired strand sequencing
EP4114948A1 (en) 2020-03-06 2023-01-11 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
TW202204615A (en) 2020-03-26 2022-02-01 美商阿尼拉製藥公司 Coronavirus irna compositions and methods of use thereof
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
BR112022020145A2 (en) 2020-04-06 2023-01-03 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR SILENCING THE MYOC EXPRESSION
TW202204617A (en) 2020-04-07 2022-02-01 美商艾爾妮蘭製藥公司 Compositions and methods for silencing scn9a expression
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
EP4133076A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Angiotensin-converting enzyme 2 (ace2) irna compositions and methods of use thereof
EP4117682A4 (en) 2020-04-24 2023-09-13 Singular Genomics Systems, Inc. Modified nucleotides and uses thereof
EP4143319A1 (en) 2020-04-27 2023-03-08 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
CA3181198A1 (en) 2020-04-30 2021-11-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
JP2023524065A (en) 2020-05-01 2023-06-08 アイオーニス ファーマシューティカルズ, インコーポレーテッド Compounds and methods for modulating ATXN1
CA3181786A1 (en) 2020-05-01 2021-11-04 The Regents Of The University Of California Inhibitors of alpha 2 beta 1 integrin and methods of use thereof
WO2021230286A1 (en) 2020-05-12 2021-11-18 田辺三菱製薬株式会社 Compound, method and pharmaceutical composition for regulating expression of ataxin 3
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
WO2021231827A2 (en) 2020-05-15 2021-11-18 Life Technologies Corporation Sirna sequences targeting coronavirus-2
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
US20210355463A1 (en) 2020-05-15 2021-11-18 Crispr Therapeutics Ag Messenger rna encoding cas9 for use in genome-editing systems
WO2021231675A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
EP4150086A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
EP4153747A2 (en) 2020-05-22 2023-03-29 Wave Life Sciences Ltd. Double stranded oligonucleotide compositions and methods relating thereto
US11408000B2 (en) 2020-06-03 2022-08-09 Triplet Therapeutics, Inc. Oligonucleotides for the treatment of nucleotide repeat expansion disorders associated with MSH3 activity
EP4161552A1 (en) 2020-06-05 2023-04-12 The Broad Institute, Inc. Compositions and methods for treating neoplasia
WO2021252557A1 (en) 2020-06-09 2021-12-16 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
KR20230026455A (en) 2020-06-18 2023-02-24 알닐람 파마슈티칼스 인코포레이티드 Xanthine dehydrogenase (XDH) iRNA compositions and methods of use thereof
JP2023531444A (en) 2020-06-19 2023-07-24 イェール ユニバーシティー Poly(amine-co-ester) polymers with modified end groups and enhanced pulmonary delivery
BR112022026316A2 (en) 2020-06-24 2023-03-07 Vir Biotechnology Inc ENGINEERED HEPATITIS B VIRUS NEUTRALIZING ANTIBODIES AND THEIR USES
TW202216203A (en) 2020-06-25 2022-05-01 美商欣爍克斯公司 Immuno oncology combination therapy with il-2 conjugates and anti-egfr antibodies
KR20230029837A (en) 2020-06-29 2023-03-03 아이오니스 파마수티컬즈, 인코포레이티드 Compounds and methods for modulating PLP1
EP4179079A1 (en) 2020-07-10 2023-05-17 Horizon Discovery Limited Method for producing genetically modified cells
EP4153606A2 (en) 2020-07-13 2023-03-29 Singular Genomics Systems, Inc. Methods of sequencing complementary polynucleotides
MX2023001541A (en) 2020-08-04 2023-03-08 Dicerna Pharmaceuticals Inc Systemic delivery of oligonucleotides.
KR20230084469A (en) 2020-08-04 2023-06-13 애보트 라피드 다이어그노스틱스 인터내셔널 언리미티드 컴퍼니 Assays to detect SARS-COV-2
IL300528A (en) 2020-08-07 2023-04-01 Fortis Therapeutics Inc Immunoconjugates targeting cd46 and methods of use thereof
EP4200250A1 (en) 2020-08-18 2023-06-28 Regenacellx.SL Compositions and methods for detecting sars-cov-2 spike protein
EP4204002A1 (en) 2020-08-31 2023-07-05 Yale University Compositions and methods for delivery of nucleic acids to cells
TW202227102A (en) 2020-09-22 2022-07-16 瑞典商阿斯特捷利康公司 Method of treating fatty liver disease
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
EP4222264A1 (en) 2020-09-30 2023-08-09 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
EP3978608A1 (en) 2020-10-05 2022-04-06 SQY Therapeutics Oligomeric compound for dystrophin rescue in dmd patients throughout skipping of exon-51
TW202228786A (en) 2020-10-09 2022-08-01 美商欣爍克斯公司 Immuno oncology therapies with il-2 conjugates
AU2021356693A1 (en) 2020-10-09 2023-06-15 Synthorx, Inc. Immuno oncology combination therapy with il-2 conjugates and pembrolizumab
EP4228637A1 (en) 2020-10-15 2023-08-23 Yeda Research and Development Co. Ltd Method of treating myeloid malignancies
WO2022084331A2 (en) 2020-10-20 2022-04-28 Sanofi Novel ligands for asialoglycoprotein receptor
AU2021365822A1 (en) 2020-10-21 2023-06-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
CA3200595A1 (en) 2020-11-13 2022-05-19 Alnylam Pharmaceuticals, Inc. Coagulation factor v (f5) irna compositions and methods of use thereof
CA3201661A1 (en) 2020-11-18 2022-05-27 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
WO2022106695A1 (en) 2020-11-23 2022-05-27 Alpha Anomeric Sas Nucleic acid duplexes
WO2022113056A1 (en) 2020-11-30 2022-06-02 Crispr Therapeutics Ag Gene-edited natural killer cells
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
GB2603454A (en) 2020-12-09 2022-08-10 Ucl Business Ltd Novel therapeutics for the treatment of neurodegenerative disorders
GB202019692D0 (en) 2020-12-14 2021-01-27 Apterna Ltd Aptamer-sirna fusions
WO2022140126A1 (en) 2020-12-21 2022-06-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Dna vector and uses thereof for detecting hiv and siv
WO2022140535A1 (en) 2020-12-23 2022-06-30 Sarepta Therapeutics, Inc. Compositions comprising exon skipping oligonucleotide conjugates for treating muscular dystrophy
JP2024501288A (en) 2020-12-23 2024-01-11 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー Compositions of modified TREM and uses thereof
EP4271798A1 (en) 2020-12-30 2023-11-08 CRISPR Therapeutics AG Compositions and methods for differentiating stem cells into nk cells
KR20230146007A (en) 2020-12-31 2023-10-18 크리스퍼 테라퓨틱스 아게 universal donor cells
WO2022150260A1 (en) 2021-01-05 2022-07-14 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT 9 (C9) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US20240067954A1 (en) 2021-01-05 2024-02-29 Horizon Discovery Limited Method for producing genetically modified cells
US11486001B2 (en) 2021-02-08 2022-11-01 Singular Genomics Systems, Inc. Methods and compositions for sequencing complementary polynucleotides
TW202245843A (en) 2021-02-12 2022-12-01 美商欣爍克斯公司 Skin cancer combination therapy with il-2 conjugates and cemiplimab
BR112023015761A2 (en) 2021-02-12 2023-11-07 Alnylam Pharmaceuticals Inc SUPEROXIDE DISMUTASE 1 (SOD1) IRNA COMPOSITIONS AND METHODS OF USE THEREOF TO TREAT OR PREVENT NEURODEGENERATIVE DISEASES ASSOCIATED WITH SUPEROXIDE DISMUTASE 1 (SOD1)
TW202302148A (en) 2021-02-12 2023-01-16 美商欣爍克斯公司 Lung cancer combination therapy with il-2 conjugates and an anti-pd-1 antibody or antigen-binding fragment thereof
JP2024506711A (en) 2021-02-17 2024-02-14 ロンザ セールス アーゲー Extracellular vesicles linked to biologically active molecules via optimized linkers and anchor moieties
WO2022182864A1 (en) 2021-02-25 2022-09-01 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods and methods of use thereof
EP4298218A1 (en) 2021-02-26 2024-01-03 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
KR20230150843A (en) 2021-03-04 2023-10-31 알닐람 파마슈티칼스 인코포레이티드 Angiopoietin-like 3 (ANGPTL3) iRNA compositions and methods of using the same
WO2022187697A1 (en) 2021-03-05 2022-09-09 The Board Of Trustees Of The Leland Stanford Junior University In vivo dna assembly and analysis
US11884977B2 (en) 2021-03-12 2024-01-30 Singular Genomics Systems, Inc. Nanoarrays and methods of use thereof
EP4263868A1 (en) 2021-03-12 2023-10-25 Singular Genomics Systems, Inc. Nanoarrays and methods of use thereof
EP4305169A1 (en) 2021-03-12 2024-01-17 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
US20220288181A1 (en) 2021-03-12 2022-09-15 Northwestern University Antiviral vaccines using spherical nucleic acids
JP2024512608A (en) 2021-03-24 2024-03-19 ジェネンテック, インコーポレイテッド Efficient TCR gene editing in T lymphocytes
KR20230162024A (en) 2021-03-29 2023-11-28 알닐람 파마슈티칼스 인코포레이티드 Huntingtin (HTT) iRNA preparation composition and method of use thereof
EP4314293A1 (en) 2021-04-01 2024-02-07 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
EP4326873A1 (en) 2021-04-22 2024-02-28 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating cancer
BR112023022284A2 (en) 2021-04-26 2023-12-26 Alnylam Pharmaceuticals Inc COMPOSITIONS OF TRANSMEMBRANE PROTEASE IRNA, SERINE 6 (TMPRSS6) AND METHODS OF USE THEREOF
EP4294920A1 (en) 2021-04-27 2023-12-27 Singular Genomics Systems, Inc. High density sequencing and multiplexed priming
WO2022232343A1 (en) 2021-04-29 2022-11-03 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
IL307937A (en) 2021-04-30 2023-12-01 Sarepta Therapeutics Inc Treatment methods for muscular dystrophy
WO2022235537A1 (en) 2021-05-03 2022-11-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (ttr) mediated amyloidosis
EP4341401A1 (en) 2021-05-18 2024-03-27 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022251644A1 (en) 2021-05-28 2022-12-01 Lyell Immunopharma, Inc. Nr4a3-deficient immune cells and uses thereof
CN115404240A (en) 2021-05-28 2022-11-29 上海环码生物医药有限公司 Constructs, methods for making circular RNA and uses thereof
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
TW202317762A (en) 2021-06-02 2023-05-01 美商艾拉倫製藥股份有限公司 Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
AU2022286947A1 (en) 2021-06-02 2023-12-14 Lyell Immunopharma, Inc. NR4A-deficient cells expressing c-Jun and uses thereof
WO2022256534A1 (en) 2021-06-03 2022-12-08 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
IL308743A (en) 2021-06-04 2024-01-01 Alnylam Pharmaceuticals Inc HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
AR126070A1 (en) 2021-06-08 2023-09-06 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR TREATING OR PREVENTING STARGARDT DISEASE AND/OR DISORDERS ASSOCIATED WITH RETINOL BORDER PROTEIN 4 (RBP4)
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
CA3223192A1 (en) 2021-06-18 2022-12-22 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ifnar1 expression
US20230014010A1 (en) 2021-06-23 2023-01-19 Crispr Therapeutics Ag Engineered cells with improved protection from natural killer cell killing
WO2022269518A2 (en) 2021-06-23 2022-12-29 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
KR20240026203A (en) 2021-06-30 2024-02-27 알닐람 파마슈티칼스 인코포레이티드 Methods and compositions for treating angiotensinogen (AGT)-related disorders
WO2023285431A1 (en) 2021-07-12 2023-01-19 Alia Therapeutics Srl Compositions and methods for allele specific treatment of retinitis pigmentosa
TW202333748A (en) 2021-07-19 2023-09-01 美商艾拉倫製藥股份有限公司 Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
KR20240037293A (en) 2021-07-23 2024-03-21 알닐람 파마슈티칼스 인코포레이티드 Beta-catenin (CTNNB1) iRNA composition and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
IL310244A (en) 2021-08-03 2024-03-01 Alnylam Pharmaceuticals Inc Transthyretin (ttr) irna compositions and methods of use thereof
CA3228255A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing angiotensinogen (agt)
AR126771A1 (en) 2021-08-13 2023-11-15 Alnylam Pharmaceuticals Inc RNAi COMPOSITIONS AGAINST FACTOR XII (F12) AND THEIR METHODS OF USE
US11833221B2 (en) 2021-09-01 2023-12-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds for reducing DMPK expression
WO2023034515A2 (en) 2021-09-03 2023-03-09 Sarepta Therapeutics, Inc. Delivery of anitsense oligomers by mirror image peptides
WO2023034920A2 (en) 2021-09-03 2023-03-09 Singular Genomics Systems, Inc. Amplification oligonucleotides
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
AU2022345881A1 (en) 2021-09-20 2024-03-21 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
US20230096386A1 (en) 2021-09-30 2023-03-30 Illumina Cambridge Limited Polynucleotide sequencing
CA3233242A1 (en) 2021-09-30 2023-04-06 Sarepta Therapeutics, Inc. Antisense oligonucleotides having one or more abasic units
WO2023070086A1 (en) 2021-10-22 2023-04-27 Sarepta Therapeutics, Inc. Morpholino oligomers for treatment of peripheral myelin protein 22 related diseases
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023076451A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023086295A2 (en) 2021-11-10 2023-05-19 University Of Rochester Antisense oligonucleotides for modifying protein expression
WO2023086292A2 (en) 2021-11-10 2023-05-19 University Of Rochester Gata4-targeted therapeutics for treatment of cardiac hypertrophy
GB202117758D0 (en) 2021-12-09 2022-01-26 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023118349A1 (en) 2021-12-21 2023-06-29 Alia Therapeutics Srl Type ii cas proteins and applications thereof
WO2023122750A1 (en) 2021-12-23 2023-06-29 Synthorx, Inc. Cancer combination therapy with il-2 conjugates and cetuximab
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
US20230212667A1 (en) 2021-12-29 2023-07-06 Illumina Cambridge Limited Methods of nucleic acid sequencing using surface-bound primers
US11772093B2 (en) 2022-01-12 2023-10-03 Miroculus Inc. Methods of mechanical microfluidic manipulation
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023159189A1 (en) 2022-02-18 2023-08-24 Yale University Branched poly(amine-co-ester) polymers for more efficient nucleic expression
WO2023168427A1 (en) 2022-03-03 2023-09-07 Yale University Compositions and methods for delivering therapeutic polynucleotides for exon skipping
WO2023172885A1 (en) 2022-03-09 2023-09-14 The Board Of Trustees Of The Leland Stanford Junior University Branched lipid charge-altering releasable transporters for nucleic acid delivery
WO2023172665A2 (en) 2022-03-10 2023-09-14 Singular Genomics Systems, Inc. Nucleic acid delivery scaffolds
WO2023178230A1 (en) 2022-03-17 2023-09-21 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer conjugates
WO2023177866A1 (en) 2022-03-18 2023-09-21 Dicerna Pharmaceuticals, Inc. Decarboxylative acetoxylation using mn(ii) or mn(iii) reagent for synthesis of 4'-acetoxy- nucleoside and use thereof for synthesis of corresponding 4'-(dimethoxyphosphoryl)methoxy- nucleotide
WO2023194359A1 (en) 2022-04-04 2023-10-12 Alia Therapeutics Srl Compositions and methods for treatment of usher syndrome type 2a
WO2023212294A1 (en) 2022-04-29 2023-11-02 Broadwing Bio Llc Angiopoietin-related protein 7-specific antibodies and uses thereof
WO2023212298A1 (en) 2022-04-29 2023-11-02 Broadwing Bio Llc Bispecific antibodies and methods of treating ocular disease
WO2023212293A1 (en) 2022-04-29 2023-11-02 Broadwing Bio Llc Complement factor h related 4-specific antibodies and uses thereof
WO2023225665A1 (en) 2022-05-19 2023-11-23 Lyell Immunopharma, Inc. Polynucleotides targeting nr4a3 and uses thereof
WO2023230201A1 (en) 2022-05-26 2023-11-30 The Board Of Trustees Of The Leland Stanford Junior University Small molecule conjugated charge-altering releasable transporters for nucleic acid delivery
WO2023231959A2 (en) 2022-05-30 2023-12-07 Shanghai Circode Biomed Co., Ltd Synthetic circular rna compositions and methods of use thereof
WO2023233339A1 (en) 2022-06-01 2023-12-07 Crispr Therapeutics Ag Compositions and methods for differentiating stem cells into nk cells
WO2023233342A2 (en) 2022-06-01 2023-12-07 Crispr Therapeutics Ag Gene-edited natural killer cells
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2024006927A1 (en) 2022-06-29 2024-01-04 Life Technologies Corporation Compositions, kits, and methods for detecting nucleic acids using intra-channel multiplexing
WO2024015962A1 (en) 2022-07-15 2024-01-18 Pacific Biosciences Of California, Inc. Blocked asymmetric hairpin adaptors
WO2024015924A2 (en) 2022-07-15 2024-01-18 Entrada Therapeutics, Inc. Hybrid oligonucleotides
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024039776A2 (en) 2022-08-18 2024-02-22 Alnylam Pharmaceuticals, Inc. Universal non-targeting sirna compositions and methods of use thereof
WO2024044352A1 (en) 2022-08-26 2024-02-29 The General Hospital Corporation Methods and compositions for prognosis and treatment of dilated cardiomyopathy and heart failure
WO2024050261A1 (en) 2022-08-29 2024-03-07 University Of Rochester Antisense oligonucleotide-based anti-fibrotic therapeutics
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
WO2024056880A2 (en) 2022-09-16 2024-03-21 Alia Therapeutics Srl Enqp type ii cas proteins and applications thereof
WO2024064237A2 (en) 2022-09-21 2024-03-28 Sarepta Therapeutics, Inc. Dmd antisense oligonucleotide-mediated exon skipping efficiency

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS63303341A (en) * 1987-06-03 1988-12-09 Kuraray Co Ltd Photosensitive composition

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE171185T1 (en) * 1985-03-15 1998-10-15 Antivirals Inc POLYNUCLEOTIDE IMMUNOTESTING AGENTS AND METHODS
DE69034209T2 (en) * 1989-09-15 2006-08-10 Southern Research Institute, Birmingham 2'-DEOXY-4'-THIORIBONUCLEOSIDE AS ANTIVIRAL AND ANTIBODY

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS63303341A (en) * 1987-06-03 1988-12-09 Kuraray Co Ltd Photosensitive composition

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Izv. Akad. Nauk SSSR, Ser. Khim Number 4, (1971) (BELIKOV et al.); pages 849-851 "Effect of the polarity of substituents on the decomposition rate of beta-nitroamines"; see Abstract. *
Nucleic Acids Research Vol 17, Number 15 (1989), (STIRCHAK, E.P.), pages 6129-6141, "Uncharged stereoregular nucleic acid analogs: 2. Morpholino nucleoside oligomers with carbamate internucleoside linkages"; see entire document. *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993020236A1 (en) * 1992-04-03 1993-10-14 Applied Biosystems, Inc. Probe composition and method
WO1993020239A1 (en) 1992-04-03 1993-10-14 Applied Biosystems, Inc. Probe method and composition for detecting different dna sequences
EP0773950A1 (en) * 1994-07-15 1997-05-21 Isis Pharmaceuticals, Inc. Linked peptide nucleic acids
EP0773950A4 (en) * 1994-07-15 2000-05-17 Isis Pharmaceuticals Inc Linked peptide nucleic acids
WO1996003417A1 (en) * 1994-07-25 1996-02-08 Hybridon, Inc. Improved methods of detritylation for oligonucleotide synthesis
WO1998032467A2 (en) * 1997-01-24 1998-07-30 Antivirals, Inc. Method and conjugate for treating h. pylori infection
WO1998032467A3 (en) * 1997-01-24 1998-09-17 Antivirals Inc Method and conjugate for treating h. pylori infection
WO2000044897A1 (en) * 1999-01-29 2000-08-03 Avi Biopharma, Inc. Method of treating restenosis by antisense targeting of c-myc
EP2002018A2 (en) * 2006-03-07 2008-12-17 AVI BioPharm, Inc. Antisense antiviral compound and method for treating arenavirus infection
EP2002018A4 (en) * 2006-03-07 2009-03-25 Avi Biopharma Inc Antisense antiviral compound and method for treating arenavirus infection
US7855283B2 (en) 2006-03-07 2010-12-21 Avi Biopharma, Inc. Antisense antiviral compound and method for treating arenavirus infection
AU2007223776B2 (en) * 2006-03-07 2014-05-22 Sarepta Therapeutics, Inc. Antisense antiviral compound and method for treating Arenavirus infection

Also Published As

Publication number Publication date
KR0154317B1 (en) 1998-12-01
ATE164081T1 (en) 1998-04-15
EP0506845A1 (en) 1992-10-07
EP0506845B1 (en) 1998-03-18
DE69032167D1 (en) 1998-04-23
AU7158791A (en) 1991-07-18
CA2069906C (en) 1996-11-26
JP3172174B2 (en) 2001-06-04
EP0506845A4 (en) 1992-10-21
JPH05504564A (en) 1993-07-15
CA2069906A1 (en) 1991-06-21
AU654473B2 (en) 1994-11-10
KR920703692A (en) 1992-12-18
DE69032167T2 (en) 1998-10-29
US5034506A (en) 1991-07-23

Similar Documents

Publication Publication Date Title
EP0506845B1 (en) Uncharged morpholino-based polymers having achiral intersubunit linkages
EP0962463B1 (en) Uncharged morpholino-based polymers having phosphorus-containing chiral intersubunit linkages
US5235033A (en) Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5185444A (en) Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5521063A (en) Polynucleotide reagent containing chiral subunits and methods of use
US5378841A (en) Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5217866A (en) Polynucleotide assay reagent and method
US5142047A (en) Uncharged polynucleotide-binding polymers
AU676704B2 (en) Sequence-specific binding polymers for duplex nucleic acids
AU665560B2 (en) Sequence-specific binding polymers for duplex nucleic acids
US5470974A (en) Uncharged polynucleotide-binding polymers

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP KR

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 2069906

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1019920701480

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1991902401

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1991902401

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1991902401

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2069906

Country of ref document: CA

Kind code of ref document: A