WO2001096584A2 - Materials and methods for the control of nematodes - Google Patents

Materials and methods for the control of nematodes Download PDF

Info

Publication number
WO2001096584A2
WO2001096584A2 PCT/US2001/018911 US0118911W WO0196584A2 WO 2001096584 A2 WO2001096584 A2 WO 2001096584A2 US 0118911 W US0118911 W US 0118911W WO 0196584 A2 WO0196584 A2 WO 0196584A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
dna sequence
molecule according
rnai molecule
rnai
Prior art date
Application number
PCT/US2001/018911
Other languages
French (fr)
Other versions
WO2001096584A3 (en
Inventor
Arcady R. Mushegian
Christopher G. Taylor
Jerald S. Feitelson
Alexy M. Eroshkin
Original Assignee
Akkadix Corporation
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 Akkadix Corporation filed Critical Akkadix Corporation
Priority to AU2001275474A priority Critical patent/AU2001275474A1/en
Publication of WO2001096584A2 publication Critical patent/WO2001096584A2/en
Publication of WO2001096584A3 publication Critical patent/WO2001096584A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • C12N15/8241Phenotypically and genetically modified plants via recombinant DNA technology
    • C12N15/8261Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield
    • C12N15/8271Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for stress resistance, e.g. heavy metal resistance
    • C12N15/8279Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for stress resistance, e.g. heavy metal resistance for biotic stress resistance, pathogen resistance, disease resistance
    • C12N15/8285Phenotypically and genetically modified plants via recombinant DNA technology with agronomic (input) traits, e.g. crop yield for stress resistance, e.g. heavy metal resistance for biotic stress resistance, pathogen resistance, disease resistance for nematode resistance
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43536Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from worms
    • C07K14/4354Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from worms from nematodes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A40/00Adaptation technologies in agriculture, forestry, livestock or agroalimentary production
    • Y02A40/10Adaptation technologies in agriculture, forestry, livestock or agroalimentary production in agriculture
    • Y02A40/146Genetically Modified [GMO] plants, e.g. transgenic plants

Definitions

  • Plant parasitic nematodes such as root-knot nematodes (Meloidogyne species) and cyst nematodes (Globodera and Heterodera), attack nearly every food crop, and are among the world's most damaging agricultural pests.
  • root-knot nematodes parasitize more than 2,000 plant species from diverse plant families and represent a tremendous threat to crop production world-wide.
  • biotrophic pathogens have evolved highly specialized and complex feeding relationships with their hosts.
  • Nematodes cause millions of dollars of damage each year to turf grasses, ornamental plants, and food crops. Efforts to eliminate or minimize damage caused by nematodes in agricultural settings have typically involved the use of soil fumigation with materials such as chloropicrin, methyl bromide, and dazomet, which volatilize to spread the active ingredient throughout the soil. Such fumigation materials can be highly toxic and may create an environmental hazard. Various non-fumigant chemicals have also been used, but these too create serious environmental problems and can be highly toxic to humans.
  • Avermectin B 2a is active against the root- knot nematode, Meloidogyne incognita. It is reported to be 10-30 times as potent as commercial contact nematicides when incorporated into soil at 0.16-0.25 kg/ha (Boyce Thompson Institute for Plant Research 58th Annual Report [1981]; Putter, I. et al.
  • Avermectins Novel Insecticides, Acaracides, and Nematicides from a Soil Microorganism
  • Avermectin B 2a is not toxic to tomatoes or cucumbers at rates of up to 10 kg/ha.
  • Fatty acids are a class of natural compounds which occur abundantly in nature and which have interesting and valuable biological activities.
  • Tarjan and Cheo (Tarjan, A.C., P.C. Cheo [1956] "Nematocidal Value of Some Fatty Acids," Bulletin 332, Contribution 884, Agricultural Experiment Station, University of Rhode Island, guitarist, 41 pp.) report the activity of certain fatty acids against nematodes.
  • Sitaramaiah and Singh Sanaramaiah, K., R.S. Singh [1977] Indian J. Nematol 7:58-65
  • Sitaramaiah and Singh (Sitaramaiah, K., R.S. Singh [1977] Indian J. Nematol 7:58-65) also examined the response of nematodes to fatty acids.
  • RNAi double- stranded interfering RNA
  • RNAi-mediated interference RNA-mediated interference
  • RNAi Agrobacterium tumefaciens-mediated transformation of Arabidopsis thaliana
  • Agrobacterium tumefaciens-mediated transformation of Arabidopsis thaliana Choang, C.-F. and E. M. Meyerowitz [2000] "Specific and heritable genetic interference by double-stranded RNA in Arabidopsis thaliana” Proc. Natl. Acad. Sci. USA 97:4985- 4990).
  • Chuang et al. describe the construction of vectors delivering variable levels of RNAi targeted to each of four genes involved in floral development. Severity of abnormal flower development varied between transgenic lines. For one of the genes, AGAMOUS (AG), a strong correlation existed between declining accumulation of mRNA and increasingly severe phenotypes, suggesting that AG-specific endogenous mRNA is the target of RNAi.
  • the subject invention provides novel methods and compositions for controlling nematodes. More specifically, the subject invention provides polynucleotide sequences that encode nematode genes, RNAi that selectively targets mRNA transcripts of these essential nematode genes, and methods of using these sequences in nematode control strategies. Such sequences for use according to the subject invention are summarized in Appendix 1. RNAi molecules disclosed herein can be used to inhibit the expression of one or more of these genes in nematodes. Brief Description of the Drawings
  • Figure 1 Modular Binary Construct System (MBCS): A series of six, 8-base cutter restriction enzyme sites has been placed between the left and right Ti borders of a previously created kan R /tet R binary plasmid.
  • MBCS Modular Binary Construct System
  • Figure 2 An exemplary shuttle vector created for cloning of useful DNA fragments by containing the multi-cloning site (MCS) of a modified Bluescript plasmid flanked by 8-base restriction sites.
  • MCS multi-cloning site
  • Figure 3 An exemplary shuttle vector with exemplary inserts.
  • Figure 4 A suggested RNAi binary vector with exemplary inserts.
  • Figure 5 Exemplary selectable markers for MBCS.
  • Figure 6 Exemplary scorable markers for MCBS.
  • Figure 7 Exemplary RNAi binary vector.
  • Figure 8 Exemplary RNAi shuttle vector.
  • the subject invention provides novel methods and compositions for controlling nematodes. More specifically, the subject invention provides polynucleotide sequences and methods of using these sequences in nematode control strategies.
  • a preferred method for controlling nematodes according to the subject invention provides materials and methods for controlling nematodes by using double-stranded interfering RNA (RNAi), or RNA-mediated interference (RNAi).
  • RNAi double-stranded interfering RNA
  • RNAi RNA-mediated interference
  • RNAi molecules are provided which are useful in methods of killing nematodes and/or inhibiting their growth, development, parasitism or reproduction. RNAi molecules of the invention are also useful for the regulation of levels of specific mRNA in nematodes.
  • dsRNA typically comprises a polynucleotide sequence identical to a target gene (or fragment thereof) linked directly, or indirectly, to a polynucleotide sequence complementary to the sequence of the target gene (or fragment thereof).
  • the dsRNA may comprise a polynucleotide linker (stuffer) sequence of sufficient length to allow for the two polynucleotide sequences to fold over and hybridize to each other; however, a linker sequence is not necessary.
  • the linker (stuffer) sequence is designed to separate the antisense and sense strands of RNAi significantly enough to limit the effects of steric hindrances and allow for the formation of dsRNA molecules.
  • RNA containing a nucleotide sequence identical to a fragment of the target gene is preferred for inhibition; however, RNA sequences with insertions, deletions, and point mutations relative to the target sequence can also be used for inhibition.
  • Sequence identity may optimized by sequence comparison and alignment algorithms known in the art (see Gribskov and Devereux, Sequence Analysis Primer, Stockton Press, 1991, and references cited therein) and calculating the percent difference between the nucleotide sequences by, for example, the Smith- Waterman algorithm as implemented in the BESTFIT software program using default parameters (e.g. , University of Wisconsin Genetic Computing Group).
  • the duplex region of the RNA may be defined functionally as a nucleotide sequence that is capable of hybridizing with a fragment of the target gene transcript.
  • RNA and the target gene are not required to practice the present invention.
  • the invention has the advantage of being able to tolerate sequence variations that might be expected due to genetic mutation, strain polymorphism, or evolutionary divergence.
  • RNA may be synthesized either in vivo or in vitro. Endogenous RNA polymerase of the cell may mediate transcription in vivo, or cloned RNA polymerase can be used for transcription in vivo or in vitro.
  • a regulatory region e.g., promoter, enhancer, silencer, splice donor and acceptor, polyadenylation
  • Inhibition may be targeted by specific transcription in an organ, tissue, or cell type; stimulation of an environmental condition (e.g., infection, stress, temperature, chemical inducers); and/or engineering transcription at a developmental stage or age.
  • RNA strands may or may not be polyadenylated; the RNA strands may or may not be capable of being translated into a polypeptide by a cell's translational apparatus.
  • RNA may be chemically or enzymatically synthesized by manual or automated reactions.
  • the RNA may be synthesized by a cellular RNA polymerase or a bacteriophage RNA polymerase (e.g., T3, T7, SP6).
  • a cellular RNA polymerase or a bacteriophage RNA polymerase e.g., T3, T7, SP6.
  • T3, T7, SP6 bacteriophage RNA polymerase
  • the RNA may be purified prior to introduction into the cell.
  • RNA can be purified from a mixture by extraction with a solvent or resin, precipitation, electrophoresis, chromatography, or a combination thereof.
  • the RNA may be used with no or a minimum of purification to avoid losses due to sample processing.
  • the RNA may be dried for storage or dissolved in an aqueous solution.
  • the solution may contain buffers or salts to promote annealing, and/or stabilization of the duplex strands.
  • RNAi can be targeted to an entire polynucleotide sequence of a gene set forth herein.
  • Preferred RNAi molecules of the instant invention are highly homologous or identical to the polynucleotides summarized in Appendix 1. The homology is preferably greater than 90% and is most preferably greater than 95%.
  • Fragments of genes can also be targeted. These fragments are typically in the approximate size range of about 20 nucleotides. Thus, targeted fragments are preferably at least about 15 nucleotides. In certain embodiments, the gene fragment targeted by the RNAi molecule is about 20-25 nucleotides in length. However, other size ranges can also be used. For example, using a C. elegans microinjection assay, RNAi "fragments" of about 60 nucleotides with between 95 and 100% identity (to a nematode gene) were determined to cause excellent inhibition.
  • RNAi molecules of the subject invention are not limited to those that are targeted to the full-length polynucleotide or gene.
  • the nematode gene product can be inhibited with a RNAi molecule that is targeted to a portion or fragment of the exemplified polynucleotides; high homology (90-95%) or identity is also preferred, but not necessarily essential, for such applications.
  • the polynucleotide sequences identified in Appendix A and shown in the Sequence ID listing are from genes encoding nematode proteins having the functions shown in Appendix 1.
  • the genes exemplified herein are representative of particular classes of proteins which are preferred targets for disruption according to the subject invention. These classes of proteins include, for example, proteins involved in ribosome assembly; neurol transmitter receptors and ligands; electron transport proteins; metabolic pathway proteins; and protein and polynucleotide production, folding, and processing proteins.
  • Genetic regulatory sequences such as promoters, enhancers, and terminators, can be used in genetic constructs to practice the subject invention. Such constructs themselves can also be used for nematode control. Various constructs can be used to achieve expression in specific plant tissues (by using root specific promoters, for example) and/or to target specific nematode tissues (by using targeting elements or adjacent targeting sequences, for example).
  • plant cells are genetically modified to produce at least one RNAi that is designed to be taken up by nematodes during feeding to block expression (or the function of) of a target gene.
  • RNAi can target and reduce (and, in some cases, prevent) the translation of a specific gene product.
  • RNAi can be used to reduce or prevent message translation in any tissue of the nematode because of its ability to cross tissue and cellular boundaries.
  • RNAi that is contacted with a nematode by soaking, injection, or consumption of a food source will cross tissue and cellular boundaries.
  • RNAi can also be used as an epigenetic factor to prevent the proliferation of subsequent generations of nematodes.
  • Nematode polynucleotide sequences disclosed herein demonstrate conserved nucleotide motifs among different nematode genera. conserveed nucleotide motifs strongly suggest that these sequences are associated with viability and/or parasitism and are functionally conserved and expressed in both Meloidogyne incognita (root-knot nematode) and Globodera rostochiensis and Globdera pallids (potato cyst nematodes).
  • the use of these polynucleotides, and RNAi inhibitors thereof, is advantageous because such RNAi can be designed to have broad RNAi specificity and are thus useful for controlling a large number of plant parasitic nematodes in planta.
  • RNAi inhibition of these genes prevents and/or reduces parasitic nematode growth, development, and or parasitism.
  • Methods of the subject invention include the transformation of plant cells with genes or polynucleotides of the present invention, which can be used to produce nematode inhibitors or RNAi in the plants.
  • the transformed plant or plant tissue can express RNAi molecules encoded by the gene or polynucleotide sequence introduced into the plant.
  • Other nematode inhibitors contemplated by the invention include antisense molecules specific to the polynucleotide sequences disclosed herein.
  • the transformation of plants with genetic constructs disclosed herein can be accomplished using techniques well known to those skilled in the art and can involve modification of the gene(s) to optimize expression in the plant to be made resistant to nematode infection and infestation.
  • many tissues of the transgenic plants (such as the roots) can be targeted for transformation.
  • RNA-mediated interference of gene expression.
  • RNAi RNA-mediated interference
  • C. elegans Several aspects of root-knot nematode biology make classical genetic studies difficult with this organism. Since root-knot nematodes reproduce by obligatory mitotic parthenogenesis, the opportunity to perform genetic crosses is not available.
  • Microinjection of RNAi can be used to manipulate gene expression in C. elegans (Fire, A., S. Xu, M. K. Montgomery, S. A. Kostas, S. E. Driver, and C. C. Mello. [1998] "Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans" Nature 391 :806- 811).
  • RNAi can turn off specific genes in progeny worms complementary to the coding region of the genes. Moreover, gene inhibition occurs in progeny when RNAi is injected into the body cavity of the adult, indicating the ability of the RNAi to cross cellular boundaries. This RNAi injection method provides a molecular genetic tool that allows for analysis of gene function in root-knot nematodes.
  • RNAi can be taken up by C. elegans by simply soaking the nematodes in a solution RNAi. This results in targeted inhibition of gene expression in the nematode (Maeda, I., Y. Kohara, M. Yamamoto and A. Sugimoto [1999] "RNAi screening with a non-redundant cDNA set" International Worm Meeting, Madison, WI, abstract 565). Nematodes fed E. coli expressing RNAi also demonstrate targeted and heritable inhibition of gene expression (Sarkissian, M., H. Tabara and C. C.
  • one aspect of the instant invention is directed to the control of nematodes comprising contacting nematodes with compositions comprising RNAi molecules specific to the nematode genes disclosed herein.
  • the contacting step may include soaking the nematodes in a solution containing RNAi molecules, feeding nematodes RNAi molecules contained in microbes or plant cells upon which the nematode feeds, or injecting nematodes with RNAi.
  • Nematodes can also be "contacted” and controlled by RNAi expressed in plant tissues that would be consumed, ingested, or frequented by nematodes.
  • RNAi molecules provided to the nematodes may be specific to a single gene.
  • a "cocktail” of RNAi molecules specific to various segments of a single gene can also be used.
  • a "multigene cocktail” of RNAi molecules specific to two or more genes (or segments thereof) may be applied to the nematodes according to the subject invention.
  • nematodes can be directly transformed with RNAi constructs of cDNAs encoding secretory or other essential proteins to reduce expression of the corresponding gene.
  • the transgenic animals can be assayed for inhibition of gene product using immunoassays or for reduced virulence on a host. Progeny of affected worms can also be assayed by similar methods.
  • RNAi Procedures that can be used for the preparation and injection of RNAi include those detailed by Fire et al, (1998; ftp://ciwl.ciwemb.edu).
  • Root-knot nematodes can be routinely monoxenically cultured on Arabidopsis thaliana roots growing on Gamborg's B-5/Gelrite® media.
  • This nematode-host pathosystem is ideally suited for these microinjection experiments since limited root galling results in the parasitic stages (late J2 through adult females) developing outside of the root for easy accessibility for injecting.
  • Another advantage is the parthenogenic reproduction of root- knot nematodes, which makes fertilization by males unnecessary for egg production.
  • RNAi can be injected into the body cavity of parasitic stages of root-knot nematodes feeding on A. thaliana roots using microinjection.
  • Control nematodes can be injected in parallel with only buffer or an unrelated RNAi.
  • Injected nematodes can be monitored for egg production, and the eggs can be collected for the assays described below.
  • Female root-knot nematodes will typically survive and lay more than 250 eggs following 1 ⁇ l injection of buffer.
  • RNAi molecules for efficacy by directly inhibiting growth and development of the nematode feeding upon the microinjected plant cell, or by reducing fecundity and the ability of said nematode to generate pathogenic or viable progeny.
  • RNAi gene interference There are a number of strategies that can be followed to assay for RNAi gene interference. Inhibition of gene expression by RNAi inhibits the accumulation of the corresponding secretory protein in the esophageal gland cells of transgenic J2 hatched from the eggs produced by the injected nematodes.
  • polyclonal antibodies to the target gene product can be used in immunolocalization studies (Hussey, R. S. [1989] "Monoclonal antibodies to secretory granules in esophageal glands of Meloidogyne species" J Nematol. 21:392-398; Borgonie, G, E. van Driessche, C. D. Link, D.
  • Control nematodes injected only with the injection buffer can be processed similar to the RNAi treated nematodes.
  • Another assay is designed to determine the effect of the RNAi on reducing the virulence of J2 progeny of the injected females.
  • Egg masses from injected females can be transferred singly to A. thaliana plates to assess the ability of the transgenic J2 to infect roots.
  • the J2 hatching from the eggs transferred to the plates can be monitored; after 25 days the number of galls with egg laying females can be recorded.
  • the A. thaliana roots can also be stained with acid fuschin to enumerate the number of nematodes in the roots.
  • Egg masses from nematodes injected only with the injection buffer can be handled similarly and used as controls. The treatments can be replicated, and the root infection data can be analyzed statistically. These experiments can be used to assess the importance of the target genes in root-knot nematode 's virulence or viability. By staining the J2 progeny of the injected females with the antibodies, it can be determined whether RNAi blocks expression of the targeted gene.
  • polynucleotides can be used in a variety of ways. These polynucleotides can be used in assays for additional polynucleotides and additional homologous genes, and can be used in tracking the quantitative and temporal expression of parasitism genes in nematodes. These polynucleotides can be cloned into microbes for production and isolation of their gene products. Among the many uses of the isolated gene product is the development of additional inhibitors and modifiers. The protein products of the subject polynucleotides can also be used as diagnostic tools.
  • proteins encoded by the parasitism genes, as identified herein can be used in large scale screenings for additional peptide inhibitors.
  • the use of peptide phage display screening is one method that can be used in this regard.
  • the subject invention also provides new biotechnological strategies for managing nematodes under sustainable agricultural conditions.
  • Antisense technologies can also be used for phytopathogenic nematode control. Antisense technology can be used to interfere with expression of the disclosed endogenous nematode genes. Antisense technology can also be used to alter the components of plants used as targets by the nematodes. For example, the transformation of a plant with the reverse complement of an endogenous gene encoded by a polynucleotide exemplified herein can result in strand co-suppression and gene silencing or inhibition of a target involved in the nematode infection process.
  • the subject invention includes transgenic plants (which are preferably made nematode-resistant in this manner, and other organisms including microbes and phages) comprising RNAi or antisense molecules specific to any of the polynucleotides identified herein.
  • DNA possesses a fundamental property called base complementarity.
  • base complementarity In nature, DNA ordinarily exists in the form of pairs of anti- parallel strands, the bases on each strand projecting from that strand toward the opposite strand.
  • the base adenine (A) on one strand will always be opposed to the base thymine (T) on the other strand, and the base guanine (G) will be opposed to the base cytosine (C).
  • the bases are held in apposition by their ability to hydrogen bond in this specific way. Though each individual bond is relatively weak, the net effect of many adjacent hydrogen bonded bases, together with base stacking effects, is a stable joining of the two complementary strands.
  • hybridization can be highly specific. That is, only strands with a high degree of base complementarity will be able to form stable double-stranded structures.
  • the relationship of the specificity of hybridization to reaction conditions is well known. Thus, hybridization may be used to test whether two pieces of DNA are complementary in their base sequences. It is this hybridization mechanism which facilitates the use of probes of the subject invention to readily detect and characterize DNA sequences of interest.
  • the specifically exemplified polynucleotides of the subject invention can themselves be used as probes. Additional polynucleotide sequences can be added to the ends of (or internally in) the exemplified polynucleotide sequences so that polynucleotides that are longer than the exemplified polynucleotides can also be used as probes. Thus, isolated polynucleotides comprising one or more of the exemplified sequences are within the scope of the subject invention. Polynucleotides that have less nucleotides than the exemplified polynucleotides can also be used and are contemplated within the scope of the present invention.
  • polynucleotides of the subject invention can be used to find additional, homologous (wholly or partially) genes.
  • Probes of the subject invention may be composed of DNA, RNA, or PNA (peptide nucleic acid).
  • the probe will normally have at least about 10 bases, more usually at least about 17 bases, and may have about 100 bases or more. Longer probes can readily be utilized, and such probes can be, for example, several kilobases in length.
  • the probe sequence is designed to be at least substantially complementary to a portion of a gene encoding a protein of interest. The probe need not have perfect complementarity to the sequence to which it hybridizes.
  • the probes may be labeled utilizing techniques that are well known to those skilled in this art.
  • One approach for the use of the subject invention as probes entails first identifying DNA segments that are homologous with the disclosed nucleotide sequences using, for example, Southern blot analysis of a gene bank.
  • Southern blot analysis of a gene bank.
  • One hybridization procedure useful according to the subject invention typically includes the initial steps of isolating the DNA sample of interest and purifying it chemically. Either lysed nematodes or total fractionated nucleic acid isolated from nematodes can be used. Cells can be treated using known techniques to liberate their DNA (and/or RNA). The DNA sample can be cut into pieces with an appropriate restriction enzyme. The pieces can be separated by size through electrophoresis in a gel, usually agarose or acrylamide. The pieces of interest can be transferred to an immobilizing membrane.
  • the particular hybridization technique is not essential to the subject invention. As improvements are made in hybridization techniques, they can be readily applied.
  • the probe and sample can then be combined in a hybridization buffer solution and held at an appropriate temperature until annealing occurs. Thereafter, the membrane is washed free of extraneous materials, leaving the sample and bound probe molecules typically detected and quantified by autoradiography and/or liquid scintillation counting.
  • the probe molecule and nucleic acid sample hybridize by forming a strong non-covalent bond between the two molecules, it can be reasonably assumed that the probe and sample are essentially identical or very similar.
  • the probe's detectable label provides a means for determining in a known manner whether hybridization has occurred.
  • the particular probe is labeled with any suitable label known to those skilled in the art, including radioactive and non-radioactive labels.
  • Typical radioactive labels include 32 P, 35 S, or the like.
  • Non- radioactive labels include, for example, ligands such as biotin or thyroxine, as well as enzymes such as hydrolases or peroxidases, or the various chemiluminescers such as luciferin, or fluorescent compounds like fluorescein and its derivatives.
  • the probes can be made inherently fluorescent as described in International Application No. WO 93/16094.
  • hybridization is conducted under moderate to high stringency conditions by techniques well known in the art, as described, for example, in Keller, G.H., M.M. Manak (1987) DNA Probes, Stockton Press, New York, NY., pp. 169-170.
  • moderate to high stringency conditions for hybridization refers to conditions that achieve the same, or about the same, degree of specificity of hybridization as the conditions "as described herein.” Examples of moderate to high stringency conditions are provided herein. Specifically, hybridization of immobilized DNA on Southern blots with 32 P-labeled gene-specific probes was performed using standard methods (Maniatis et al). In general, hybridization and subsequent washes were carried out under moderate to high stringency conditions that allowed for detection of target sequences with homology to sequences exemplified herein.
  • Tm 81.5°C+16.6 Log[Na+]+0.41(%G+C)-0.61(%formamide)- 600/length of duplex in base pairs.
  • Washes are typically carried out as follows:
  • Tm melting temperature
  • Tm (°C) * 2(number T/A base pairs) +4(number G/C base pairs)
  • salt and/or temperature can be altered to change stringency.
  • salt and/or temperature can be altered to change stringency.
  • Moderate 0.2X or IX SSPE, 65 °C High: 0.1X SSPE, 65°C.
  • polynucleotide sequences of the subject invention include mutations (both single and multiple), deletions, and insertions in the described sequences, and combinations thereof, wherein said mutations, insertions, and deletions permit formation of stable hybrids with a target polynucleotide of interest. Mutations, insertions, and deletions can be produced in a given polynucleotide sequence using standard methods known in the art. Other methods may become known in the future.
  • the mutational, insertional, and deletional variants of the polynucleotide sequences of the invention can be used in the same manner as the exemplified polynucleotide sequences so long as the variants have substantial sequence similarity with the original sequence.
  • substantial sequence similarity refers to the extent of nucleotide similarity that is sufficient to enable the variant polynucleotide to function in the same capacity as the original sequence.
  • this similarity is greater than 50%; more preferably, this similarity is greater than 75%; and most preferably, this similarity is greater than 90%.
  • the degree of similarity needed for the variant to function in its intended capacity will depend upon the intended use of the sequence. It is well within the skill of a person trained in this art to make mutational, insertional, and deletional mutations that are designed to improve the function of the sequence or otherwise provide a methodological advantage.
  • PCR Polymerase Chain Reaction
  • PCR is a repetitive, enzymatic, primed synthesis of a nucleic acid sequence. This procedure is well known and commonly used by those skilled in this art (see U.S. Patent Nos. 4,683,195; 4,683,202; and 4,800,159; Saiki et al, 1985).
  • PCR is based on the enzymatic amplification of a DNA fragment of interest that is flanked by two oligonucleotide primers that hybridize to opposite strands of the target sequence. The primers are oriented with the 3' ends pointing towards each other.
  • thermostable DNA polymerase such as Taq polymerase, which is isolated from the thermophilic bacterium Thermus aquaticus, the amplification process can be completely automated.
  • Other enzymes that can be used are known to those skilled in the art.
  • polynucleotide sequences of the subject invention can be used as, and/or used in the design of, primers for PCR amplification.
  • a certain degree of mismatch can be tolerated between primer and template. Therefore, mutations, deletions, and insertions (especially additions of nucleotides to the 5' end) of the exemplified ppolynucleotides can be used in this manner. Mutations, insertions and deletions can be produced in a given primer by methods known to an ordinarily skilled artisan.
  • the polynucleotide sequences of the instant invention may be "operably linked” to regulatory sequences such as promoters and enhancers.
  • Nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is “operably linked” to DNA encoding a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is “operably linked” to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is “operably linked” to a coding sequence if it is positioned so as to facilitate translation.
  • operably linked means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • Polynucleotides and proteins Polynucleotides and proteins.
  • Polynucleotides of the subject invention can be defined according to several parameters. One characteristic is the biological activity of the protein products as identified herein.
  • the proteins and genes of the subject invention can be further defined by their amino acid and nucleotide sequences.
  • the sequences of the molecules can be defined in terms of homology to certain exemplified sequences as well as in terms of the ability to hybridize with, or be amplified by, certain exemplified probes and primers. Additional primers and probes can readily be constructed by those skilled in the art such that alternate polynucleotide sequences encoding the same amino acid sequences can be used to identify and/or characterize additional genes.
  • the proteins of the subject invention can also be identified based on their immunoreactivity with certain antibodies.
  • polynucleotides and proteins of the subject invention include portions, fragments, variants, and mutants of the full-length sequences as well as fusions and chimerics, so long as the encoded protein retains the characteristic biological activity of the proteins identified herein.
  • variants or variantations refer to nucleotide sequences that encode the same proteins or which encode equivalent proteins having equivalent biological activity.
  • equivalent proteins refers to proteins having the same or essentially the same biological activity as the exemplified proteins.
  • genes within the scope of the subject invention can be identified and obtained through several means.
  • the specific genes exemplified herein may be obtained from root-knot nematodes. Genes, or portions or variants thereof, may also be artificially synthesized by, for example, a gene synthesizer.
  • genes may be readily constructed using standard techniques such as site-directed mutagenesis and other methods of making point mutations and by DNA shuffling, for example.
  • gene and protein fragments can be made using commercially available exonucleases, endonucleases, and proteases according to standard procedures.
  • enzymes such as E /31 can be used to systematically cut off nucleotides from the ends of genes.
  • genes that encode fragments may be obtained using a variety of restriction enzymes. Proteases may be used to directly obtain active fragments of these proteins.
  • molecular techniques for cloning polynucleotides and producing gene constructs of interest are also well known in the art. In vitro evaluation techniques, such as MAXYGEN's "Molecular Breeding" can also be applied to practice the subject invention.
  • antibodies raised against proteins encoded by polynucleotides disclosed herein can be used to identify and isolate proteins from a mixture of proteins. Specifically, antibodies may be raised to the portions of the proteins that are conserved and most distinct from other proteins. These antibodies can then be used to specifically identify equivalent proteins by immunoprecipitation, enzyme linked immunosorbent assay (ELISA), or Western blotting.
  • ELISA enzyme linked immunosorbent assay
  • Antibodies to proteins encoded by polynucleotides disclosed herein, or to equivalent proteins can readily be prepared using standard procedures known in the art. The genes that encode these proteins can be obtained from various organisms.
  • DNA sequences can encode the amino acid sequences encoded by the polynucleotide sequences disclosed herein. It is well within the skill of a person trained in the art to create these alternative DNA sequences encoding proteins having the same, or essentially the same, amino acid sequence. These variant DNA sequences are within the scope of the subject invention. As used herein, reference to "essentially the same" sequence refers to sequences that have amino acid substitutions, deletions, additions, or insertions that do not materially affect biological activity. Fragments retaining the characteristic biological activity are also included in this definition.
  • a further method for identifying genes and polynucleotides (and the proteins encoded thereby) of the subject invention is through the use of oligonucleotide probes.
  • Probes provide a rapid method for identifying genes of the subject invention.
  • the nucleotide segments that are used as probes according to the invention can be synthesized using a DNA synthesizer and standard procedures.
  • the subject invention comprises variant or equivalent proteins (and nucleotide sequences coding for equivalent proteins or for inhibitors of the genes encoding such proteins) having the same or similar biological activity of inhibitors or proteins encoded by the exemplified polynucleotides.
  • Equivalent proteins will have amino acid similarity with an exemplified protein (or peptide).
  • the amino acid and/or nucleotide identity will typically be greater than 60%.
  • the identity will be greater than 75%. More preferably, the identity will be greater than 80%, and even more preferably greater than 90%. Most preferably, the identity will be greater than 95%.
  • RNAi molecules will also have corresponding identities in these preferred ranges.
  • amino acids may be placed in the following classes: non-polar, uncharged polar, basic, and acidic. Conservative substitutions whereby an amino acid of one class is replaced with another amino acid of the same type fall within the scope of the subject invention so long as the substitution does not materially alter the biological activity of the compound.
  • non-conservative substitutions can also be made.
  • the critical factor is that these substitutions must not detract from the ability to manage nematode-caused diseases.
  • An "isolated” or “substantially pure” nucleic acid molecule or polynucleotide is a polynucleotide that is substantially separated from other polynucleotide sequences which naturally accompany a nucleic acid molecule.
  • the term embraces a polynucleotide sequence which was removed from its naturally occurring environment by the hand of man. This includes recombinant or cloned DNA isolates, chemically synthesized analogues and analogues biologically synthesized by heterologous systems.
  • An "isolated” or “purified” protein likewise, is a protein removed from its naturally occurring environment.
  • RNAi polynucleotides within the scope of the present invention can be introduced into a wide variety of microbial or plant hosts. Plant cells can be transformed (made recombinant) in this manner. Microbes, for example, can also be used in the application of RNAi molecules of the subject invention in view of the fact that microbes are a food source for nematodes
  • heterologous gene or polynucleotide there are many methods for introducing a heterologous gene or polynucleotide into a host cell or cells under conditions that allow for stable maintenance and expression of the gene or polynucleotide. These methods are well known to those skilled in the art.
  • Synthetic genes such as, for example, those genes modified to enhance expression in a heterologous host (such as by preferred codon usage or by the use of adjoining, downstream, or upstream enhancers) that are functionally equivalent to the genes (and which encode equivalent proteins) can also be used to transform hosts. Methods for the production of synthetic genes are known in the art.
  • the gene or polynucleotide of interest is introduced via a suitable vector into a microbial host, and said host is applied to the environment in a living state, certain host microbes are preferred.
  • Certain microorganism hosts are known to occupy the phytosphere, phylloplane, phyllosphere, rhizosphere, and/or rhizoplane of one or more crops of interest. These microorganisms can be selected so as to be capable of successfully competing in the particular environment (crop and other habitats) with the wild-type microorganisms, provide for stable maintenance and expression of the gene expressing a polypeptide of interest, and, desirably, provide for improved protection of the protein/peptide from environmental degradation and inactivation.
  • microorganisms A large number of microorganisms is known to inhabit the phylloplane (the surface of the plant leaves) and/or the rhizosphere (the soil surrounding plant roots) of a wide variety of important crops. These microorganisms include bacteria, algae, and fungi.
  • microorganisms such as bacteria, e.g., genera Pseudomonas, Erwinia, Serratia, Klebsiella, Xanthomonas, Streptomyces, Rhizobium, Rhodopseudomonas, Methylophilius, Agrobacterium, Acetobacter, Lactobacillus, Arthrobacter, Azotobacter, Leuconostoc, and Alcaligenes; fungi, particularly yeast, e.g., genera Saccharomyces, Cryptococcus, Kluyveromyces, Sporobolomyces, Rhodotorula, and Aureobasidium.
  • the pigmented microorganisms are particularly yeast.
  • Methods of the subject invention also include the transformation of plants or plant tissue with genes which encode the RNAi molecules of the present invention.
  • the transformed plant or plant tissue expresses antisense RNA and or RNAi. Transformation of cells can be made by those skilled in the art using standard techniques. Materials necessary for these transformations are disclosed herein or are otherwise readily available to the skilled artisan.
  • RNAi molecules of the instant invention can be accomplished by a variety of additional methods that would be apparent to those skilled in the art having the benefit of the subject disclosure.
  • a "cocktail" of two or more RNAi molecules can be used to disrupt one or more of the genes identified herein.
  • the "cocktail” of RNAi molecules may be specific to segments of a single gene or the entire gene.
  • a "multigene cocktail” of RNAi molecules specific to two or more genes (or segments thereof) is also encompassed by the instant invention.
  • the disclosed RNAi molecules, cocktails, and/or multigene cocktails thereof may be used in conjunction with other known nematode control agents and methodologies. Such cocktails can be used to combat the development of resistance by nematodes to a certain inhibitor or inhibitors.
  • compositions of the subject invention which comprise RNAi molecules and carriers can be applied, themselves, directly or indirectly, to locations frequented by, or expected to be frequented by, nematodes.
  • Microbial hosts which were transformed with polynucleotides that encode RNAi molecules, express said RNAi molecules, and which colonize roots (e.g., Pseudomonas, Bacillus, and other genera) can be applied to the sites of the pest, where they will proliferate and be ingested. The result is control of the pest.
  • methods of the subject invention include, for example, the application of recombinant microbes to the pests (or their locations).
  • the recombinant microbes may also be transformed with more than one RNAi molecule thereby delivering a "cocktail" of RNAi molecules to the nematode pests.
  • a carrier may be any substance suitable for delivering the RNAi molecules to the nematode. Acceptable carriers are well known in the art and also are commercially available. For example, such acceptable carriers are described in E.W. Martin's Remington's Pharmaceutical Science, Mack Publishing Company, Easton, PA.
  • a hairy root assay system was developed for testing the anti-nematode activity of RNAi molecules.
  • Agrobacterium rhizogenes Several Agrobacterium rhizogenes strains produce hairy roots on a variety of plant species. A. rhizogenes strains, A4, 15834, 8196 and LBA4404 demonstrate hairy root development on tomato and sugar beet, with A4 being the most efficient. The A. rhizogenes strain K599 demonstrated very efficient formation on transgenic soybean hairy roots and was also effective on sugar beet and Arabidopsis. However, stain K599 failed to produce hairy roots on tomato tissues possibly due to hyper-virulence.
  • Hairy root production Transgenic hairy roots were identified by stable GUS expression in tomato, sugar beet, soybean and Arabidopsis.
  • the construct pAKKHOl (pNOS / NPT-II / tNOS // pSU / GUS / tNOS) was used to produce hairy roots when transformed into A. rhizogenes strains A4 or K599. Transgenic roots were identified by GUS expression.
  • Electro-competent Agrobacterium Protocol [00089] 1. Grow Agrobacterium overnight in 5 mis LB + antibiotics at 30°C on shaker (for Agrobacterium rhizogenes strain K599 no antibiotics are needed). [00090] 2. Use the 5 mis of overnight culture to inoculate 500 mis LB + antibiotics at 30°C on shaker. Grow overnight. [00091 ] 3. Add liquid culture in eight 50 ml polypropylene orange cap tubes. [00092] 4. Centrifuge 10 min., 4000 rpm, 4°C.
  • Example 3 Protocol for Production of Transgenic Hairy Roots on Soybean
  • Seed Sterilization Rinse the soybean seed with 70% ETOH for 2-5 min. Remove and add 20% Clorox and shake for 20-25 min. Rinse 3X with sterile water. Plate the seed, 5 seed per plate, onto l A MSB5 + 2% sucrose + 0.2% gel (referred to as Vi MSB5). Place seed into chamber at 25C, 16/8 photoperiod for 5-7 day (depending on genotype) germination period. After 1 week seedlings can be placed into cold room for longer storage if necessary (not to exceed 2 weeks).
  • Agrobacterium Preparation For Agrobacterium rhizogenes strain K599, take a small sample from frozen glycerol into 25-50 ml of NZYM media with 50 mg/L kanamycin in a 125-250 ml Erlenmyer flask. Place onto shaker at 28-30 °C for 16 - 20 hours. Pour sample into centrifuge tube and centrifuge the bacterium at 4000 rpm for 10 min. Pour off supernatant and re-suspend the pellet with an equal volume of liquid V_ MSB5 + 200 ⁇ M acetosyringone. Use pipette to re-suspend the pellet and homogenize the sample (remove all clumps).
  • O.D. To determine O.D., prepare a 1:10 dilution by putting 900 ⁇ l MSB5 into cuvette and add 100 ⁇ l of bacterial sample. Determine the O.D. 660 and calculate the volume needed to adjust (dilute) OD to approximately 0.2 for inoculation. Check final O.D.
  • Roots should develop in approximately 2-3 weeks. The roots will form primarily from the cut vascular bundles with other roots developing from the small cuts on cotyledon surface. Remove roots (>lcm in length) and place onto replica media with transfers to fresh media every 2 weeks to prevent Agrobacterium overgrowth. After 6-8 weeks on selection the roots can be moved to media without kanamycin, however carbenicillin must remain in media for several months for continued suppression of Agrobacterium. At this stage roots can be used for testing RNAi for nematode control. Sterilized nematodes can be added and observed for RNAi affects.
  • nematodes can be used in appropriate bioassays.
  • Caenorhabditis elegans a bacterial feeding nematode
  • plant parasitic nematodes can be used for bioassay purposes.
  • plant parasitic nematodes include a migratory endo-parasite, Pratylenchus scribneri (lesion), and two sedentary endo-parasites, Meloidogyne javanica (root-knot) and Heterodera schachtii (cyst).
  • RNAi vectors can be tested through expression of the RNAi in E. coli.
  • C. elegans are fed E. coli and assayed for their growth by measuring growth of nematodes, production of eggs and viability of offspring.
  • Another approach is to inject dsRNA directly into living nematodes.
  • soaking nematodes in a solution of in vz ' tr ⁇ -prepared RNAi can quickly establish efficacy of treatment.
  • P. scribneri The P. scribneri in vitro feeding assay uses a corn root exudate (CRE) as a feeding stimulus and both the red dye Amaranth or potassium, arsenate as feeding indicators. Feeding is confirmed after seven days by the presence of red stained intestinal cells in live worms exposed to the Amaranth or death of worms exposed to arsenate. This bioassay is used to test soluble toxins or RNAi.
  • P. scribneri has also been cultured on wild type roots of corn, rice and Arabidopsis, and on A. rhizogenes-induced hairy roots of sugar beet and tomato. P. scribneri is very valuable in evaluating transgenic hairy roots because of the non-specific feeding of these worms.
  • M javanica Nematode eggs are sterilized using bleach and are used to inoculate hairy roots expressing RNAi. Nematodes are assessed for their growth by measuring knots, egg masses or production of viable eggs. An alternative approach is to microinject dsRNA directly into root feeding sites or into living female nematodes.
  • H schachtii Cultures of this nematode were maintained on sugar beets. Nematodes eggs are sterilized using bleach and used to inoculate hairy roots expressing RNAi. Nematodes can be assessed for their growth by measuring knots, egg masses or production of viable eggs.
  • Modular Binary Construct System An important aspect of the subject disclosure is the Modular Binary Construct System.
  • the MBCS eases the burden of construct development by creating modular pieces of DNA that can be easily added, removed, or replaced with the use of low frequency cutting restriction enzymes (8-base cutters). These constructs are useful for delivery of a variety of genes to plant cells and is not limited to the delivery of RNAi genes.
  • 8-base cutters 8-base cutters
  • kan R and tet R MCBS aids the testing of constructs using different strains of Agrobacterium rhizogenes in different plant species.
  • a series of shuttle vectors were created that aid in the cloning of useful DNA fragments by containing the multi-cloning site (MCS) of a modified Bluescript plasmid flanked by 8- base restriction sites ( Figure 2). With six 8-base cutter sites, each site is, preferably, reserved for a particular function ( Figures 3 and 4). Because of the close proximity of the Eme I and Sgfl sites to the left and right border of the binary vector, these sites are, preferably, reserved for gene tagging and enhancer trap experiments.
  • the Not I site is, preferably, reserved for plant selectable markers ( Figure 5).
  • the Pac I site is reserved, preferably, for Plant Scorable Markers ( Figure 6).
  • the Asc I site is, preferably, reserved for R ⁇ Ai experiments ( Figures 7 and 8), while the Sbfl site is, preferably, reserved for anti-nematode proteins.
  • the restriction sites that are denoted in the Figures are, preferably, reserved for the denoted insertions; however, the MCBS binary and shuttle vectors do not require the restriction sites to contain these suggested inserts.
  • Plant Selectable Markers for MBCS To further develop the MBCS, a series of plant selectable markers were added to the MBCS ( Figure 5). Plant selectable markers that were added to the MBCS include: p ⁇ OS/ ⁇ PT-II/t ⁇ OS (kan R ), p ⁇ OS/Bar/t ⁇ OS (basta R for dicots), pUBI/Intron-Bar/t ⁇ OS (basta 11 for monocots), and pUBI/Intron-PMI/t ⁇ OS (mannitol isomerase 11 ).
  • Reporter Genes for MBCS Four exemplary reporter genes are used in the MBCS are provided in Figure 6 and Appendix 2. GUS, a nuclear localized GUS, GEP, and the anthocyanin transcriptional activator papIC genes into the MBCS.
  • Promoters for MBCS We cloned several useful constitutive and nematode-inducible promoters ( Figures 6, 7 and Appendix 2). Constitutive promoters include the SuperUbiquitin promoter from pine (pSU) and two promoter regions from the Strawberry Banding Vein virus (pSBVj and pSBV 2 ). Seven nematode-inducible promoters from Arabidopsis were also been cloned.
  • RNAi shuttle vector to be used is adapted from the Modular Binary Construct System (MBCS - See Example 5).
  • RNAi shuttle vectors preferably comprise a promoter, intron, antisense RNAi, stuffer fragment, sense RNAi, and terminator (See Figures 7 and 8 and Appendix 2 for more details).
  • the plant promoter can be constitutive, tissue-specific or nematode-inducible.
  • the intron is necessary to eliminate expression in Agrobacterium.
  • RNAi molecules comprise nematode-specific sequences and are disclosed herein. These genes are associated with pathogenesis, growth, or other cellular function in nematodes.
  • An exemplary group of RNAi sequences for use in plant/nematode control may be based upon:
  • RNAi constructs allow for the formation of dsRNA molecules (the sense and antisense strands join to form the dsRNA).
  • the terminator sequence adds a poly-A tail for transcriptional termination.
  • the RNAi shuttle vector can then be subcloned into the MBCS and transformed into Agrobacterium rhizogenes.
  • RNAi vector once introduced into the MBCS can subsequently (as a binary vector) be transformed in A. rhizogenes using, for example, the electroporation protocol of Example 2. Once the A. rhizogenes is confirmed to contain the plasmid, it is then used in generating hairy roots (See Example 3). Using this protocol transgenic hairy roots expressing RNAi are isolated, cultured and tested.
  • RNAi expressing hairy roots can be inoculated with sterilized nematodes. Infested hairy roots can be observed and the effect on nematodes determined.
  • An alternative approach involves the microinjection of RNAi directly into root feeding sites (giant-cells for root- knot nematode, and syncytia for cyst nematodes) or into living female nematodes.
  • One aspect of the subject invention is the transformation of plants with genes encoding proteins of the present invention. Transformation of plants as described herein can be used to improve the resistance of these plants to attack by the target pest.
  • RNAi molecules as disclosed or suggested herein can be inserted into plant cells using a variety of techniques which are well known in the art.
  • cloning vectors for example, pBR322, pUC series, M13mp series, pACYC184, pMON, etc.
  • biolistics microparticle bombardment
  • Agrobacterium tumefaciens ox Agrobacterium rhizogenes- mediated transformation
  • electroporation as well as other possible methods.
  • the genetically modified-cell(s) can be screened via a vector carried-selectable marker that confers on the transformed plant cells resistance to a biocide or an antibiotic, such as kanamycin, G418, bleomycin, hygromycin, chloramphenicol, or bialophos, inter alia.
  • the transformed cell will be regenerated into a morphologically normal plant.
  • the transgene(s) in the transgenic plant is relatively stable and can be inherited by progeny plants.
  • a transformation event involves a germ line cell
  • the inserted DNA an corresponding phenotypic trait(s) will be transmitted to progeny plants.
  • Such plants can be grown in the normal manner and crossed with plants that have the same transformed hereditary factors or other hereditary factors.
  • the resulting hybrid individuals have the corresponding phenotypic properties.

Abstract

The subject invention provides novel methods and compositions for controlling nematodes. More specifically, the subject invention provides RNAi molecules, polynucleotide sequences, and methods of using these sequences in nematode control.

Description

DESCRIPTION
MATERIALS AND METHODS FOR THE CONTROL OF NEMATODES
Background of the Invention
[0001] Plant parasitic nematodes, such as root-knot nematodes (Meloidogyne species) and cyst nematodes (Globodera and Heterodera), attack nearly every food crop, and are among the world's most damaging agricultural pests. For example, root-knot nematodes parasitize more than 2,000 plant species from diverse plant families and represent a tremendous threat to crop production world-wide. These biotrophic pathogens have evolved highly specialized and complex feeding relationships with their hosts.
[0002] Nematodes cause millions of dollars of damage each year to turf grasses, ornamental plants, and food crops. Efforts to eliminate or minimize damage caused by nematodes in agricultural settings have typically involved the use of soil fumigation with materials such as chloropicrin, methyl bromide, and dazomet, which volatilize to spread the active ingredient throughout the soil. Such fumigation materials can be highly toxic and may create an environmental hazard. Various non-fumigant chemicals have also been used, but these too create serious environmental problems and can be highly toxic to humans.
[0003] Some research articles have been published concerning the effects of δ- endotoxins from B. thuringiensis species on the viability of nematodes. See, for example, Bottjer, Bone and Gill ([1985] Experimental Parasitology 60:239-244); Ignoffo and Dropkin (Ignoffo, CM., Dropkin, V.H. [1977] J Kans. Entomol Soc. 50:394-398); and Ciordia, H. and W.E. Bizzell ([1961] Jour, of Parasitology 47:41 [abstract]). Several patents have issued describing the control of nematodes with B.t. See, for example, U.S. Patent Nos. 4,948,734; 5,093,120; 5,281,530; 5,426,049; 5,439,881; 5,236,843; 5,322,932; 5,151,363; 5,270,448; 5,350,577; 5,667,993; and 5,670,365. The development of resistance by insects to B.t. toxins is one obstacle to the successful use of such toxins. [0004] The pesticidal activity of avermectins is well known. The avermectins are disaccharide derivatives of pentacyclic, 16-membered lactones. They can be divided into four major compounds: Ala, A2a, Bla, and B2a; and four minor compounds: Alb, A2b, Blb, and B2b. The isolation and purification of these compounds is also described in U.S. Patent No. 4,310,519, issued January 12, 1982. Avermectin B2a is active against the root- knot nematode, Meloidogyne incognita. It is reported to be 10-30 times as potent as commercial contact nematicides when incorporated into soil at 0.16-0.25 kg/ha (Boyce Thompson Institute for Plant Research 58th Annual Report [1981]; Putter, I. et al. [1981] "Avermectins: Novel Insecticides, Acaracides, and Nematicides from a Soil Microorganism," Experientia 37:963-964). Avermectin B2a is not toxic to tomatoes or cucumbers at rates of up to 10 kg/ha.
[0005] Fatty acids are a class of natural compounds which occur abundantly in nature and which have interesting and valuable biological activities. Tarjan and Cheo (Tarjan, A.C., P.C. Cheo [1956] "Nematocidal Value of Some Fatty Acids," Bulletin 332, Contribution 884, Agricultural Experiment Station, University of Rhode Island, Kingston, 41 pp.) report the activity of certain fatty acids against nematodes. In 1977 Sitaramaiah and Singh (Sitaramaiah, K., R.S. Singh [1977] Indian J. Nematol 7:58-65) also examined the response of nematodes to fatty acids. The results of these tests with short chain acids were equivocal, showing nematode-inhibitory action in some instances and stimulatory activity in other instances. Phytotoxicity of these acids was observed at higher concentrations. The short chain fatty acids were also examined by Malik and Jairajpuri (Malik, Z., M.S. Jairajpuri [1977] Nematol medit. 12:73-79), who observed nematode toxicity at high concentrations of the fatty acids.
[0006] Notwithstanding the foregoing (some of the limitations of and problems associated with these approaches are discussed above), there is a need for safe and effective alternatives for controlling nematodes.
[0007] One method for disrupting normal cellular processes is by the use double- stranded interfering RNA (RNAi), or RNA-mediated interference (RNAi). When RNAi corresponding to a sense and antisense sequence of a target mRNA is introduced into a cell, the targeted mRNA is degraded and protein translation of that message is stopped. Although not yet fully understood, the mechanism of this post-transcriptional gene silencing appears to be at least partially due to the generation of small RNA molecules, about 21 - 25 nucleotides in length, that correspond to the sense and antisense pieces of the RNAi introduced into the cell (Bass, B. L. [2000] "Double-stranded RNA as a template for gene silencing" Cell 101:235-238).
[0008] The specificity of this gene silencing mechanism appears to be extremely high, blocking expression only of targeted genes, while leaving other genes unaffected. A recent example of the use of RNAi; to inhibit genetic function in plants used Agrobacterium tumefaciens-mediated transformation of Arabidopsis thaliana (Chuang, C.-F. and E. M. Meyerowitz [2000] "Specific and heritable genetic interference by double-stranded RNA in Arabidopsis thaliana" Proc. Natl. Acad. Sci. USA 97:4985- 4990). Chuang et al. describe the construction of vectors delivering variable levels of RNAi targeted to each of four genes involved in floral development. Severity of abnormal flower development varied between transgenic lines. For one of the genes, AGAMOUS (AG), a strong correlation existed between declining accumulation of mRNA and increasingly severe phenotypes, suggesting that AG-specific endogenous mRNA is the target of RNAi.
Brief Summary of the Invention [0009] The subject invention provides novel methods and compositions for controlling nematodes. More specifically, the subject invention provides polynucleotide sequences that encode nematode genes, RNAi that selectively targets mRNA transcripts of these essential nematode genes, and methods of using these sequences in nematode control strategies. Such sequences for use according to the subject invention are summarized in Appendix 1. RNAi molecules disclosed herein can be used to inhibit the expression of one or more of these genes in nematodes. Brief Description of the Drawings
[00010] Figure 1 : Modular Binary Construct System (MBCS): A series of six, 8-base cutter restriction enzyme sites has been placed between the left and right Ti borders of a previously created kanR/tetR binary plasmid.
[00011] Figure 2: An exemplary shuttle vector created for cloning of useful DNA fragments by containing the multi-cloning site (MCS) of a modified Bluescript plasmid flanked by 8-base restriction sites.
[00012] Figure 3: An exemplary shuttle vector with exemplary inserts.
[00013] Figure 4: A suggested RNAi binary vector with exemplary inserts.
[00014] Figure 5: Exemplary selectable markers for MBCS.
[00015] Figure 6: Exemplary scorable markers for MCBS.
[00016] Figure 7: Exemplary RNAi binary vector.
[00017] Figure 8: Exemplary RNAi shuttle vector.
Brief Description of the Sequences [00018] Brief Description of the Sequences can be found in Appendix I.
Detailed Disclosure of the Invention
[00019] The subject invention provides novel methods and compositions for controlling nematodes. More specifically, the subject invention provides polynucleotide sequences and methods of using these sequences in nematode control strategies. A preferred method for controlling nematodes according to the subject invention provides materials and methods for controlling nematodes by using double-stranded interfering RNA (RNAi), or RNA-mediated interference (RNAi). The terms RNAi and RNAi are used interchangeably herein unless otherwise noted.
[00020] In one embodiment of the invention, RNAi molecules are provided which are useful in methods of killing nematodes and/or inhibiting their growth, development, parasitism or reproduction. RNAi molecules of the invention are also useful for the regulation of levels of specific mRNA in nematodes.
[00021] dsRNA (RNAi) typically comprises a polynucleotide sequence identical to a target gene (or fragment thereof) linked directly, or indirectly, to a polynucleotide sequence complementary to the sequence of the target gene (or fragment thereof). The dsRNA may comprise a polynucleotide linker (stuffer) sequence of sufficient length to allow for the two polynucleotide sequences to fold over and hybridize to each other; however, a linker sequence is not necessary. The linker (stuffer) sequence is designed to separate the antisense and sense strands of RNAi significantly enough to limit the effects of steric hindrances and allow for the formation of dsRNA molecules.
[00022] RNA containing a nucleotide sequence identical to a fragment of the target gene is preferred for inhibition; however, RNA sequences with insertions, deletions, and point mutations relative to the target sequence can also be used for inhibition. Sequence identity may optimized by sequence comparison and alignment algorithms known in the art (see Gribskov and Devereux, Sequence Analysis Primer, Stockton Press, 1991, and references cited therein) and calculating the percent difference between the nucleotide sequences by, for example, the Smith- Waterman algorithm as implemented in the BESTFIT software program using default parameters (e.g. , University of Wisconsin Genetic Computing Group). Alternatively, the duplex region of the RNA may be defined functionally as a nucleotide sequence that is capable of hybridizing with a fragment of the target gene transcript.
[00023] As disclosed herein, 100% sequence identity between the RNA and the target gene is not required to practice the present invention. Thus the invention has the advantage of being able to tolerate sequence variations that might be expected due to genetic mutation, strain polymorphism, or evolutionary divergence.
[00024] RNA may be synthesized either in vivo or in vitro. Endogenous RNA polymerase of the cell may mediate transcription in vivo, or cloned RNA polymerase can be used for transcription in vivo or in vitro. For transcription from a transgene in vivo or an expression construct, a regulatory region (e.g., promoter, enhancer, silencer, splice donor and acceptor, polyadenylation) may be used to transcribe the RNA strand (or strands). Inhibition may be targeted by specific transcription in an organ, tissue, or cell type; stimulation of an environmental condition (e.g., infection, stress, temperature, chemical inducers); and/or engineering transcription at a developmental stage or age. The RNA strands may or may not be polyadenylated; the RNA strands may or may not be capable of being translated into a polypeptide by a cell's translational apparatus. RNA may be chemically or enzymatically synthesized by manual or automated reactions. The RNA may be synthesized by a cellular RNA polymerase or a bacteriophage RNA polymerase (e.g., T3, T7, SP6). The use and production of an expression construct are known in the art (see, for example, WO 97/32016; U.S. Pat. Nos. 5,593,874; 5,698,425; 5,712,135; 5,789,214; and 5,804,693; and the references cited therein). If synthesized chemically or by in vitro enzymatic synthesis, the RNA may be purified prior to introduction into the cell. For example, RNA can be purified from a mixture by extraction with a solvent or resin, precipitation, electrophoresis, chromatography, or a combination thereof. Alternatively, the RNA may be used with no or a minimum of purification to avoid losses due to sample processing. The RNA may be dried for storage or dissolved in an aqueous solution. The solution may contain buffers or salts to promote annealing, and/or stabilization of the duplex strands.
[00025] Preferably and most conveniently, RNAi can be targeted to an entire polynucleotide sequence of a gene set forth herein. Preferred RNAi molecules of the instant invention are highly homologous or identical to the polynucleotides summarized in Appendix 1. The homology is preferably greater than 90% and is most preferably greater than 95%.
[00026] Fragments of genes can also be targeted. These fragments are typically in the approximate size range of about 20 nucleotides. Thus, targeted fragments are preferably at least about 15 nucleotides. In certain embodiments, the gene fragment targeted by the RNAi molecule is about 20-25 nucleotides in length. However, other size ranges can also be used. For example, using a C. elegans microinjection assay, RNAi "fragments" of about 60 nucleotides with between 95 and 100% identity (to a nematode gene) were determined to cause excellent inhibition.
[00027] Thus, RNAi molecules of the subject invention are not limited to those that are targeted to the full-length polynucleotide or gene. The nematode gene product can be inhibited with a RNAi molecule that is targeted to a portion or fragment of the exemplified polynucleotides; high homology (90-95%) or identity is also preferred, but not necessarily essential, for such applications.
[00028] The polynucleotide sequences identified in Appendix A and shown in the Sequence ID listing are from genes encoding nematode proteins having the functions shown in Appendix 1. The genes exemplified herein are representative of particular classes of proteins which are preferred targets for disruption according to the subject invention. These classes of proteins include, for example, proteins involved in ribosome assembly; neurol transmitter receptors and ligands; electron transport proteins; metabolic pathway proteins; and protein and polynucleotide production, folding, and processing proteins.
[00029] Genetic regulatory sequences, such as promoters, enhancers, and terminators, can be used in genetic constructs to practice the subject invention. Such constructs themselves can also be used for nematode control. Various constructs can be used to achieve expression in specific plant tissues (by using root specific promoters, for example) and/or to target specific nematode tissues (by using targeting elements or adjacent targeting sequences, for example).
[00030] In a specific embodiment of the subject invention, plant cells, preferably root cells, are genetically modified to produce at least one RNAi that is designed to be taken up by nematodes during feeding to block expression (or the function of) of a target gene. As is known in the art, RNAi can target and reduce (and, in some cases, prevent) the translation of a specific gene product. RNAi can be used to reduce or prevent message translation in any tissue of the nematode because of its ability to cross tissue and cellular boundaries. Thus, RNAi that is contacted with a nematode by soaking, injection, or consumption of a food source will cross tissue and cellular boundaries. RNAi can also be used as an epigenetic factor to prevent the proliferation of subsequent generations of nematodes.
[00031] Nematode polynucleotide sequences disclosed herein demonstrate conserved nucleotide motifs among different nematode genera. Conserved nucleotide motifs strongly suggest that these sequences are associated with viability and/or parasitism and are functionally conserved and expressed in both Meloidogyne incognita (root-knot nematode) and Globodera rostochiensis and Globdera pallids (potato cyst nematodes). The use of these polynucleotides, and RNAi inhibitors thereof, is advantageous because such RNAi can be designed to have broad RNAi specificity and are thus useful for controlling a large number of plant parasitic nematodes in planta. Because the genes identified in this disclosure are associated with nematode survival and/or parasitism, RNAi inhibition of these genes (arising from contacting nematodes with compositions comprising RNAi molecules) prevents and/or reduces parasitic nematode growth, development, and or parasitism.
[00032] Methods of the subject invention include the transformation of plant cells with genes or polynucleotides of the present invention, which can be used to produce nematode inhibitors or RNAi in the plants. In one embodiment, the transformed plant or plant tissue can express RNAi molecules encoded by the gene or polynucleotide sequence introduced into the plant. Other nematode inhibitors contemplated by the invention include antisense molecules specific to the polynucleotide sequences disclosed herein. The transformation of plants with genetic constructs disclosed herein can be accomplished using techniques well known to those skilled in the art and can involve modification of the gene(s) to optimize expression in the plant to be made resistant to nematode infection and infestation. Furthermore, it is known in the art that many tissues of the transgenic plants (such as the roots) can be targeted for transformation.
[00033] RNA-mediated interference (RNAi) of gene expression. Several aspects of root-knot nematode biology make classical genetic studies difficult with this organism. Since root-knot nematodes reproduce by obligatory mitotic parthenogenesis, the opportunity to perform genetic crosses is not available. Microinjection of RNAi can be used to manipulate gene expression in C. elegans (Fire, A., S. Xu, M. K. Montgomery, S. A. Kostas, S. E. Driver, and C. C. Mello. [1998] "Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans" Nature 391 :806- 811). Microinjecting (into adult nematodes) RNAi can turn off specific genes in progeny worms complementary to the coding region of the genes. Moreover, gene inhibition occurs in progeny when RNAi is injected into the body cavity of the adult, indicating the ability of the RNAi to cross cellular boundaries. This RNAi injection method provides a molecular genetic tool that allows for analysis of gene function in root-knot nematodes.
[00034] RNAi can be taken up by C. elegans by simply soaking the nematodes in a solution RNAi. This results in targeted inhibition of gene expression in the nematode (Maeda, I., Y. Kohara, M. Yamamoto and A. Sugimoto [1999] "RNAi screening with a non-redundant cDNA set" International Worm Meeting, Madison, WI, abstract 565). Nematodes fed E. coli expressing RNAi also demonstrate targeted and heritable inhibition of gene expression (Sarkissian, M., H. Tabara and C. C. Mello [1999] "A mut-6 screen for RNAi deficient mutants" International Worm Meeting, Madison, WI, abstract 741; Timmons, 1. and A. Fire [1998] "Specific interference by ingested dsRNA" Nature 395:854; WO 99/32619, hereby incorporated by reference in its entirety).
[00035] Accordingly, one aspect of the instant invention is directed to the control of nematodes comprising contacting nematodes with compositions comprising RNAi molecules specific to the nematode genes disclosed herein. The contacting step may include soaking the nematodes in a solution containing RNAi molecules, feeding nematodes RNAi molecules contained in microbes or plant cells upon which the nematode feeds, or injecting nematodes with RNAi. Nematodes can also be "contacted" and controlled by RNAi expressed in plant tissues that would be consumed, ingested, or frequented by nematodes.
[00036] The RNAi molecules provided to the nematodes may be specific to a single gene. A "cocktail" of RNAi molecules specific to various segments of a single gene can also be used. In addition, a "multigene cocktail" of RNAi molecules specific to two or more genes (or segments thereof) may be applied to the nematodes according to the subject invention.
[00037] In addition to RNAi uptake mediated by transgenic plants, nematodes can be directly transformed with RNAi constructs of cDNAs encoding secretory or other essential proteins to reduce expression of the corresponding gene. The transgenic animals can be assayed for inhibition of gene product using immunoassays or for reduced virulence on a host. Progeny of affected worms can also be assayed by similar methods.
[00038] Procedures that can be used for the preparation and injection of RNAi include those detailed by Fire et al, (1998; ftp://ciwl.ciwemb.edu). Root-knot nematodes can be routinely monoxenically cultured on Arabidopsis thaliana roots growing on Gamborg's B-5/Gelrite® media. This nematode-host pathosystem is ideally suited for these microinjection experiments since limited root galling results in the parasitic stages (late J2 through adult females) developing outside of the root for easy accessibility for injecting. Another advantage is the parthenogenic reproduction of root- knot nematodes, which makes fertilization by males unnecessary for egg production. The RNAi can be injected into the body cavity of parasitic stages of root-knot nematodes feeding on A. thaliana roots using microinjection. Control nematodes can be injected in parallel with only buffer or an unrelated RNAi. Injected nematodes can be monitored for egg production, and the eggs can be collected for the assays described below. Female root-knot nematodes will typically survive and lay more than 250 eggs following 1 μl injection of buffer.
[00039] Alternatively, methods are available for microinjecting materials directly into the plant root cells upon which nematodes feed: giant cells or syncytial cells (Bockenlioff, A. and F.M.W. Grundler [1994] "Studies on the nutrient uptake by the beet cyst nematode Heterodera schachtii by in situ microinjection of fluorescent probes into the feeding structures in Arabidopsis thaliana" Parasitology 109:249-254). This provides an excellent test system to screen RNAi molecules for efficacy by directly inhibiting growth and development of the nematode feeding upon the microinjected plant cell, or by reducing fecundity and the ability of said nematode to generate pathogenic or viable progeny.
[00040] There are a number of strategies that can be followed to assay for RNAi gene interference. Inhibition of gene expression by RNAi inhibits the accumulation of the corresponding secretory protein in the esophageal gland cells of transgenic J2 hatched from the eggs produced by the injected nematodes. In the first assay, polyclonal antibodies to the target gene product can be used in immunolocalization studies (Hussey, R. S. [1989] "Monoclonal antibodies to secretory granules in esophageal glands of Meloidogyne species" J Nematol. 21:392-398; Borgonie, G, E. van Driessche, C. D. Link, D. de Waele, and A. Coomans [1994] "Tissue treatment for whole mount internal lectin staining in the nematodes Caenorhabditis elegans, Panagrolaimus superbus and Acrobeloides maximus" Histochemistry 101:379-384) to monitor the synthesis of the target protein in the gland cells of progeny of the injected nematodes, or in any other nematode tissue that fails to express the essential targeted gene. Interference of endogenous gene activity by the RNAi eliminates binding of the antibodies to secretory granules in the glands, or any other target tissue, of the transgenic nematodes, and can be monitored by these in situ hybridization experiments. Control nematodes injected only with the injection buffer can be processed similar to the RNAi treated nematodes. [00041] Another assay is designed to determine the effect of the RNAi on reducing the virulence of J2 progeny of the injected females. Egg masses from injected females can be transferred singly to A. thaliana plates to assess the ability of the transgenic J2 to infect roots. The J2 hatching from the eggs transferred to the plates can be monitored; after 25 days the number of galls with egg laying females can be recorded. The A. thaliana roots can also be stained with acid fuschin to enumerate the number of nematodes in the roots. Egg masses from nematodes injected only with the injection buffer can be handled similarly and used as controls. The treatments can be replicated, and the root infection data can be analyzed statistically. These experiments can be used to assess the importance of the target genes in root-knot nematode 's virulence or viability. By staining the J2 progeny of the injected females with the antibodies, it can be determined whether RNAi blocks expression of the targeted gene.
[00042] Additional uses of polynucleotides. The polynucleotide sequences exemplified herein can be used in a variety of ways. These polynucleotides can be used in assays for additional polynucleotides and additional homologous genes, and can be used in tracking the quantitative and temporal expression of parasitism genes in nematodes. These polynucleotides can be cloned into microbes for production and isolation of their gene products. Among the many uses of the isolated gene product is the development of additional inhibitors and modifiers. The protein products of the subject polynucleotides can also be used as diagnostic tools. For example, proteins encoded by the parasitism genes, as identified herein, can be used in large scale screenings for additional peptide inhibitors. The use of peptide phage display screening is one method that can be used in this regard. Thus, the subject invention also provides new biotechnological strategies for managing nematodes under sustainable agricultural conditions.
[00043] Antisense technologies can also be used for phytopathogenic nematode control. Antisense technology can be used to interfere with expression of the disclosed endogenous nematode genes. Antisense technology can also be used to alter the components of plants used as targets by the nematodes. For example, the transformation of a plant with the reverse complement of an endogenous gene encoded by a polynucleotide exemplified herein can result in strand co-suppression and gene silencing or inhibition of a target involved in the nematode infection process. Thus, the subject invention includes transgenic plants (which are preferably made nematode-resistant in this manner, and other organisms including microbes and phages) comprising RNAi or antisense molecules specific to any of the polynucleotides identified herein.
[00044] Polynucleotide probes. DNA possesses a fundamental property called base complementarity. In nature, DNA ordinarily exists in the form of pairs of anti- parallel strands, the bases on each strand projecting from that strand toward the opposite strand. The base adenine (A) on one strand will always be opposed to the base thymine (T) on the other strand, and the base guanine (G) will be opposed to the base cytosine (C). The bases are held in apposition by their ability to hydrogen bond in this specific way. Though each individual bond is relatively weak, the net effect of many adjacent hydrogen bonded bases, together with base stacking effects, is a stable joining of the two complementary strands. These bonds can be broken by treatments such as high pH or high temperature, and these conditions result in the dissociation, or "denaturation," of the two strands. If the DNA is then placed in conditions which make hydrogen bonding of the bases thermodynamically favorable, the DNA strands will anneal, or "hybridize," and reform the original double- stranded DNA. If carried out under appropriate conditions, this hybridization can be highly specific. That is, only strands with a high degree of base complementarity will be able to form stable double-stranded structures. The relationship of the specificity of hybridization to reaction conditions is well known. Thus, hybridization may be used to test whether two pieces of DNA are complementary in their base sequences. It is this hybridization mechanism which facilitates the use of probes of the subject invention to readily detect and characterize DNA sequences of interest.
[00045] The specifically exemplified polynucleotides of the subject invention can themselves be used as probes. Additional polynucleotide sequences can be added to the ends of (or internally in) the exemplified polynucleotide sequences so that polynucleotides that are longer than the exemplified polynucleotides can also be used as probes. Thus, isolated polynucleotides comprising one or more of the exemplified sequences are within the scope of the subject invention. Polynucleotides that have less nucleotides than the exemplified polynucleotides can also be used and are contemplated within the scope of the present invention. For example, for some purposes, it might be useful to use a conserved sequence from an exemplified polynucleotide wherein the conserved sequence comprises a portion of an exemplified sequence. Thus, polynucleotides of the subject invention can be used to find additional, homologous (wholly or partially) genes.
[00046] Probes of the subject invention may be composed of DNA, RNA, or PNA (peptide nucleic acid). The probe will normally have at least about 10 bases, more usually at least about 17 bases, and may have about 100 bases or more. Longer probes can readily be utilized, and such probes can be, for example, several kilobases in length. The probe sequence is designed to be at least substantially complementary to a portion of a gene encoding a protein of interest. The probe need not have perfect complementarity to the sequence to which it hybridizes. The probes may be labeled utilizing techniques that are well known to those skilled in this art.
[00047] One approach for the use of the subject invention as probes entails first identifying DNA segments that are homologous with the disclosed nucleotide sequences using, for example, Southern blot analysis of a gene bank. Thus, it is possible, without the aid of biological analysis, to know in advance the probable activity of many new polynucleotides, and of the individual gene products expressed by a given polynucleotide. Such an analysis provides a rapid method for identifying commercially valuable compositions.
[00048] One hybridization procedure useful according to the subject invention typically includes the initial steps of isolating the DNA sample of interest and purifying it chemically. Either lysed nematodes or total fractionated nucleic acid isolated from nematodes can be used. Cells can be treated using known techniques to liberate their DNA (and/or RNA). The DNA sample can be cut into pieces with an appropriate restriction enzyme. The pieces can be separated by size through electrophoresis in a gel, usually agarose or acrylamide. The pieces of interest can be transferred to an immobilizing membrane.
[00049] The particular hybridization technique is not essential to the subject invention. As improvements are made in hybridization techniques, they can be readily applied. [00050] The probe and sample can then be combined in a hybridization buffer solution and held at an appropriate temperature until annealing occurs. Thereafter, the membrane is washed free of extraneous materials, leaving the sample and bound probe molecules typically detected and quantified by autoradiography and/or liquid scintillation counting. As is well known in the art, if the probe molecule and nucleic acid sample hybridize by forming a strong non-covalent bond between the two molecules, it can be reasonably assumed that the probe and sample are essentially identical or very similar. The probe's detectable label provides a means for determining in a known manner whether hybridization has occurred.
[00051] In the use of the nucleotide segments as probes, the particular probe is labeled with any suitable label known to those skilled in the art, including radioactive and non-radioactive labels. Typical radioactive labels include 32P, 35S, or the like. Non- radioactive labels include, for example, ligands such as biotin or thyroxine, as well as enzymes such as hydrolases or peroxidases, or the various chemiluminescers such as luciferin, or fluorescent compounds like fluorescein and its derivatives. In addition, the probes can be made inherently fluorescent as described in International Application No. WO 93/16094.
[00052] Various degrees of stringency of hybridization can be employed. The more stringent the conditions, the greater the complementarity that is required for duplex formation. Stringency can be controlled by temperature, probe concentration, probe length, ionic strength, time, and the like. Preferably, hybridization is conducted under moderate to high stringency conditions by techniques well known in the art, as described, for example, in Keller, G.H., M.M. Manak (1987) DNA Probes, Stockton Press, New York, NY., pp. 169-170.
[00053] As used herein "moderate to high stringency" conditions for hybridization refers to conditions that achieve the same, or about the same, degree of specificity of hybridization as the conditions "as described herein." Examples of moderate to high stringency conditions are provided herein. Specifically, hybridization of immobilized DNA on Southern blots with 32P-labeled gene-specific probes was performed using standard methods (Maniatis et al). In general, hybridization and subsequent washes were carried out under moderate to high stringency conditions that allowed for detection of target sequences with homology to sequences exemplified herein. For double-stranded DNA gene probes, hybridization was carried out overnight at 20-25 ° C below the melting temperature (Tm) of the DNA hybrid in 6X SSPE, 5X Denhardt's solution, 0.1% SDS, 0.1 mg/ml denatured DNA. The melting temperature is described by the following formula from Beltz et al. (1983):
[00054] Tm=81.5°C+16.6 Log[Na+]+0.41(%G+C)-0.61(%formamide)- 600/length of duplex in base pairs.
Washes are typically carried out as follows:
(1) Twice at room temperature for 15 minutes in IX SSPE, 0.1% SDS (low stringency wash).
(2) Once at Tm-20°C for 15 minutes in 0.2X SSPE, 0.1% SDS (moderate stringency wash).
[00055] For oligonucleotide probes, hybridization was carried out overnight at 10-20°C below the melting temperature (Tm) of the hybrid in 6X SSPE, 5X Denhardt's solution, 0.1% SDS, 0.1 mg/ml denatured DNA. Tm for oligonucleotide probes was determined by the following formula from Suggs et al. (1981):
[00056] Tm (°C)*=2(number T/A base pairs) +4(number G/C base pairs)
[00057] Washes were typically carried out as follows:
[00058] (1) Twice at room temperature for 15 minutes IX SSPE, 0.1% SDS
(low stringency wash). [00059] (2) Once at the hybridization temperature for 15 minutes in IX SSPE,
0.1% SDS (moderate stringency wash). [00060] In general, salt and/or temperature can be altered to change stringency. With a labeled DNA fragment of greater than about 70 or so bases in length, the following conditions can be used:
Low: 1 or 2X SSPE, room temperature
Low: l or 2X SSPE, 42°C
Moderate: 0.2X or IX SSPE, 65 °C High: 0.1X SSPE, 65°C.
[00061] Duplex formation and stability depend on substantial complementarity between the two strands of a hybrid, and, as noted above, a certain degree of mismatch can be tolerated. Therefore, polynucleotide sequences of the subject invention include mutations (both single and multiple), deletions, and insertions in the described sequences, and combinations thereof, wherein said mutations, insertions, and deletions permit formation of stable hybrids with a target polynucleotide of interest. Mutations, insertions, and deletions can be produced in a given polynucleotide sequence using standard methods known in the art. Other methods may become known in the future.
[00062] The mutational, insertional, and deletional variants of the polynucleotide sequences of the invention can be used in the same manner as the exemplified polynucleotide sequences so long as the variants have substantial sequence similarity with the original sequence. As used herein, substantial sequence similarity refers to the extent of nucleotide similarity that is sufficient to enable the variant polynucleotide to function in the same capacity as the original sequence. Preferably, this similarity is greater than 50%; more preferably, this similarity is greater than 75%; and most preferably, this similarity is greater than 90%. The degree of similarity needed for the variant to function in its intended capacity will depend upon the intended use of the sequence. It is well within the skill of a person trained in this art to make mutational, insertional, and deletional mutations that are designed to improve the function of the sequence or otherwise provide a methodological advantage.
[00063] PCR technology. Polymerase Chain Reaction (PCR) is a repetitive, enzymatic, primed synthesis of a nucleic acid sequence. This procedure is well known and commonly used by those skilled in this art (see U.S. Patent Nos. 4,683,195; 4,683,202; and 4,800,159; Saiki et al, 1985). PCR is based on the enzymatic amplification of a DNA fragment of interest that is flanked by two oligonucleotide primers that hybridize to opposite strands of the target sequence. The primers are oriented with the 3' ends pointing towards each other. Repeated cycles of heat denaturation of the template, annealing of the primers to their complementary sequences, and extension of the annealed primers with a DNA polymerase result in the amplification of the segment defined by the 5' ends of the PCR primers. Since the extension product of each primer can serve as a template for the other primer, each cycle essentially doubles the amount of DNA fragment produced in the previous cycle. This results in the exponential accumulation of the specific target fragment, up to several million-fold in a few hours. By using a thermostable DNA polymerase such as Taq polymerase, which is isolated from the thermophilic bacterium Thermus aquaticus, the amplification process can be completely automated. Other enzymes that can be used are known to those skilled in the art.
[00064] The polynucleotide sequences of the subject invention (and portions thereof such as conserved regions and portions that serve to distinguish these sequences from previously-known sequences) can be used as, and/or used in the design of, primers for PCR amplification. In performing PCR amplification, a certain degree of mismatch can be tolerated between primer and template. Therefore, mutations, deletions, and insertions (especially additions of nucleotides to the 5' end) of the exemplified ppolynucleotides can be used in this manner. Mutations, insertions and deletions can be produced in a given primer by methods known to an ordinarily skilled artisan.
[00065] The polynucleotide sequences of the instant invention may be "operably linked" to regulatory sequences such as promoters and enhancers. Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is "operably linked" to DNA encoding a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is "operably linked" to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is "operably linked" to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
[00066] Polynucleotides and proteins. Polynucleotides of the subject invention can be defined according to several parameters. One characteristic is the biological activity of the protein products as identified herein. The proteins and genes of the subject invention can be further defined by their amino acid and nucleotide sequences. The sequences of the molecules can be defined in terms of homology to certain exemplified sequences as well as in terms of the ability to hybridize with, or be amplified by, certain exemplified probes and primers. Additional primers and probes can readily be constructed by those skilled in the art such that alternate polynucleotide sequences encoding the same amino acid sequences can be used to identify and/or characterize additional genes. The proteins of the subject invention can also be identified based on their immunoreactivity with certain antibodies.
[00067] The polynucleotides and proteins of the subject invention include portions, fragments, variants, and mutants of the full-length sequences as well as fusions and chimerics, so long as the encoded protein retains the characteristic biological activity of the proteins identified herein. As used herein, the terms "variants" or "variations" of genes refer to nucleotide sequences that encode the same proteins or which encode equivalent proteins having equivalent biological activity. As used herein, the term "equivalent proteins" refers to proteins having the same or essentially the same biological activity as the exemplified proteins.
[00068] It will be apparent to a person skilled in this art that genes within the scope of the subject invention can be identified and obtained through several means. The specific genes exemplified herein may be obtained from root-knot nematodes. Genes, or portions or variants thereof, may also be artificially synthesized by, for example, a gene synthesizer.
[00069] Variations of genes may be readily constructed using standard techniques such as site-directed mutagenesis and other methods of making point mutations and by DNA shuffling, for example. In addition, gene and protein fragments can be made using commercially available exonucleases, endonucleases, and proteases according to standard procedures. For example, enzymes such as E /31 can be used to systematically cut off nucleotides from the ends of genes. In addition, genes that encode fragments may be obtained using a variety of restriction enzymes. Proteases may be used to directly obtain active fragments of these proteins. Of course, molecular techniques for cloning polynucleotides and producing gene constructs of interest are also well known in the art. In vitro evaluation techniques, such as MAXYGEN's "Molecular Breeding" can also be applied to practice the subject invention.
[00070] Other molecular techniques can also be applied using the teachings provided herein. For example, antibodies raised against proteins encoded by polynucleotides disclosed herein can be used to identify and isolate proteins from a mixture of proteins. Specifically, antibodies may be raised to the portions of the proteins that are conserved and most distinct from other proteins. These antibodies can then be used to specifically identify equivalent proteins by immunoprecipitation, enzyme linked immunosorbent assay (ELISA), or Western blotting. Antibodies to proteins encoded by polynucleotides disclosed herein, or to equivalent proteins, can readily be prepared using standard procedures known in the art. The genes that encode these proteins can be obtained from various organisms.
[00071] Because of the redundancy of the genetic code, a variety of different DNA sequences can encode the amino acid sequences encoded by the polynucleotide sequences disclosed herein. It is well within the skill of a person trained in the art to create these alternative DNA sequences encoding proteins having the same, or essentially the same, amino acid sequence. These variant DNA sequences are within the scope of the subject invention. As used herein, reference to "essentially the same" sequence refers to sequences that have amino acid substitutions, deletions, additions, or insertions that do not materially affect biological activity. Fragments retaining the characteristic biological activity are also included in this definition.
[00072] A further method for identifying genes and polynucleotides (and the proteins encoded thereby) of the subject invention is through the use of oligonucleotide probes. Probes provide a rapid method for identifying genes of the subject invention. The nucleotide segments that are used as probes according to the invention can be synthesized using a DNA synthesizer and standard procedures.
[00073] The subject invention comprises variant or equivalent proteins (and nucleotide sequences coding for equivalent proteins or for inhibitors of the genes encoding such proteins) having the same or similar biological activity of inhibitors or proteins encoded by the exemplified polynucleotides. Equivalent proteins will have amino acid similarity with an exemplified protein (or peptide). The amino acid and/or nucleotide identity will typically be greater than 60%. Preferably, the identity will be greater than 75%. More preferably, the identity will be greater than 80%, and even more preferably greater than 90%. Most preferably, the identity will be greater than 95%. RNAi molecules will also have corresponding identities in these preferred ranges. These identities are as determined using standard alignment techniques for determining amino acid and/or nucleotide identity. The identity/similarity will be highest in critical regions of the protein or gene including those regions that account for biological activity or that are involved in the determination of three-dimensional configuration that is ultimately responsible for the biological activity. In this regard, certain amino acid substitutions are acceptable and can be expected if these substitutions are in regions which are not critical to activity or are conservative amino acid substitutions which do not affect the three- dimensional configuration of the molecule. For example, amino acids may be placed in the following classes: non-polar, uncharged polar, basic, and acidic. Conservative substitutions whereby an amino acid of one class is replaced with another amino acid of the same type fall within the scope of the subject invention so long as the substitution does not materially alter the biological activity of the compound. Below is a list of examples of amino acids belonging to various classes
Class of Amino Acid Examples of Amino Acids
Nonpolar Ala, Val, Leu, He, Pro, Met, Phe,
Trp
Uncharged Polar Gly, Ser, Thr, Cys, Tyr, Asn, Gin
Acidic Asp, Glu
Basic Lys, Arg, His
[00074] In some instances, non-conservative substitutions can also be made. The critical factor is that these substitutions must not detract from the ability to manage nematode-caused diseases.
[00075] An "isolated" or "substantially pure" nucleic acid molecule or polynucleotide is a polynucleotide that is substantially separated from other polynucleotide sequences which naturally accompany a nucleic acid molecule. The term embraces a polynucleotide sequence which was removed from its naturally occurring environment by the hand of man. This includes recombinant or cloned DNA isolates, chemically synthesized analogues and analogues biologically synthesized by heterologous systems. An "isolated" or "purified" protein, likewise, is a protein removed from its naturally occurring environment.
[00076] Recombinant hosts. The genes, antisense, and RNAi polynucleotides within the scope of the present invention can be introduced into a wide variety of microbial or plant hosts. Plant cells can be transformed (made recombinant) in this manner. Microbes, for example, can also be used in the application of RNAi molecules of the subject invention in view of the fact that microbes are a food source for nematodes
[00077] There are many methods for introducing a heterologous gene or polynucleotide into a host cell or cells under conditions that allow for stable maintenance and expression of the gene or polynucleotide. These methods are well known to those skilled in the art. Synthetic genes, such as, for example, those genes modified to enhance expression in a heterologous host (such as by preferred codon usage or by the use of adjoining, downstream, or upstream enhancers) that are functionally equivalent to the genes (and which encode equivalent proteins) can also be used to transform hosts. Methods for the production of synthetic genes are known in the art.
[00078] Where the gene or polynucleotide of interest is introduced via a suitable vector into a microbial host, and said host is applied to the environment in a living state, certain host microbes are preferred. Certain microorganism hosts are known to occupy the phytosphere, phylloplane, phyllosphere, rhizosphere, and/or rhizoplane of one or more crops of interest. These microorganisms can be selected so as to be capable of successfully competing in the particular environment (crop and other habitats) with the wild-type microorganisms, provide for stable maintenance and expression of the gene expressing a polypeptide of interest, and, desirably, provide for improved protection of the protein/peptide from environmental degradation and inactivation.
[00079] A large number of microorganisms is known to inhabit the phylloplane (the surface of the plant leaves) and/or the rhizosphere (the soil surrounding plant roots) of a wide variety of important crops. These microorganisms include bacteria, algae, and fungi. Of particular interest are microorganisms, such as bacteria, e.g., genera Pseudomonas, Erwinia, Serratia, Klebsiella, Xanthomonas, Streptomyces, Rhizobium, Rhodopseudomonas, Methylophilius, Agrobacterium, Acetobacter, Lactobacillus, Arthrobacter, Azotobacter, Leuconostoc, and Alcaligenes; fungi, particularly yeast, e.g., genera Saccharomyces, Cryptococcus, Kluyveromyces, Sporobolomyces, Rhodotorula, and Aureobasidium. Of particular interest are the pigmented microorganisms.
[00080] Methods of the subject invention also include the transformation of plants or plant tissue with genes which encode the RNAi molecules of the present invention. In one embodiment, the transformed plant or plant tissue expresses antisense RNA and or RNAi. Transformation of cells can be made by those skilled in the art using standard techniques. Materials necessary for these transformations are disclosed herein or are otherwise readily available to the skilled artisan.
[00081] Additional methods and formulations for control of pests. Control of nematode pests using the RNAi molecules of the instant invention can be accomplished by a variety of additional methods that would be apparent to those skilled in the art having the benefit of the subject disclosure. A "cocktail" of two or more RNAi molecules can be used to disrupt one or more of the genes identified herein. The "cocktail" of RNAi molecules may be specific to segments of a single gene or the entire gene. A "multigene cocktail" of RNAi molecules specific to two or more genes (or segments thereof) is also encompassed by the instant invention. In another embodiment of the instant invention, the disclosed RNAi molecules, cocktails, and/or multigene cocktails thereof, may be used in conjunction with other known nematode control agents and methodologies. Such cocktails can be used to combat the development of resistance by nematodes to a certain inhibitor or inhibitors.
[00082] Compositions of the subject invention which comprise RNAi molecules and carriers can be applied, themselves, directly or indirectly, to locations frequented by, or expected to be frequented by, nematodes. Microbial hosts which were transformed with polynucleotides that encode RNAi molecules, express said RNAi molecules, and which colonize roots (e.g., Pseudomonas, Bacillus, and other genera) can be applied to the sites of the pest, where they will proliferate and be ingested. The result is control of the pest. Thus, methods of the subject invention include, for example, the application of recombinant microbes to the pests (or their locations). The recombinant microbes may also be transformed with more than one RNAi molecule thereby delivering a "cocktail" of RNAi molecules to the nematode pests. A carrier may be any substance suitable for delivering the RNAi molecules to the nematode. Acceptable carriers are well known in the art and also are commercially available. For example, such acceptable carriers are described in E.W. Martin's Remington's Pharmaceutical Science, Mack Publishing Company, Easton, PA.
[00083] All patents, patent applications, provisional applications, and publications referred to or cited herein are incorporated by reference in their entirety to the extent they are not inconsistent with the explicit teachings of this specification.
[00084] Following are examples that illustrate procedures for practicing the invention. These examples should not be construed as limiting. All percentages are by weight and all solvent mixture proportions are by volume unless otherwise noted.
Example 1— Production of Hairy Roots for RNAi Testing
[00085] A hairy root assay system was developed for testing the anti-nematode activity of RNAi molecules.
[00086] Agrobacterium rhizogenes: Several Agrobacterium rhizogenes strains produce hairy roots on a variety of plant species. A. rhizogenes strains, A4, 15834, 8196 and LBA4404 demonstrate hairy root development on tomato and sugar beet, with A4 being the most efficient. The A. rhizogenes strain K599 demonstrated very efficient formation on transgenic soybean hairy roots and was also effective on sugar beet and Arabidopsis. However, stain K599 failed to produce hairy roots on tomato tissues possibly due to hyper-virulence.
[00087] Hairy root production: Transgenic hairy roots were identified by stable GUS expression in tomato, sugar beet, soybean and Arabidopsis. The construct pAKKHOl (pNOS / NPT-II / tNOS // pSU / GUS / tNOS) was used to produce hairy roots when transformed into A. rhizogenes strains A4 or K599. Transgenic roots were identified by GUS expression.
Example 2 — Protocol for Electro-competent Agrobacterium and Electroporation [00088] Electro-competent Agrobacterium Protocol: [00089] 1. Grow Agrobacterium overnight in 5 mis LB + antibiotics at 30°C on shaker (for Agrobacterium rhizogenes strain K599 no antibiotics are needed). [00090] 2. Use the 5 mis of overnight culture to inoculate 500 mis LB + antibiotics at 30°C on shaker. Grow overnight. [00091 ] 3. Add liquid culture in eight 50 ml polypropylene orange cap tubes. [00092] 4. Centrifuge 10 min., 4000 rpm, 4°C.
[00093] 5. Resuspend cells in each tube with 20 mis 10% glycerol (on ice) [00094] 6. Centrifuge 10 min., 4000 rpm, 4°C.
[00095] 7. Resuspend cells in each tube with 10 mis 10% glycerol (on ice). [00096] 8. Centrifuge 10 min., 4000 rpm , 4°C.
[00097] 9. Resuspend cells in each tube with 2 mis 10% glycerol (on ice). [00098] 10. Aliquot 50 μl into cold Eppendorf tube and place onto dry ice. [00099] 11. Store electro-competent cells at -80°C. These cells can be used for up to two years.
[000100] Electroporations:
[000101] 1. Add 1 μl to 5 μl of DNA (resuspended in H2O and not TE or other buffer) to 50 μl of Agrobacterium electrocompetent cells and mix. [000102] 2. Transfer 20 μl of ON Al Agrobacterium mix to cuvette. [000103] 3. Electroporate:
25μF, 400 Ω resistance, 2.5 volts (0.2cm cuvette) or 1.8 volts (0.1cm cuvette for BioRad electroporator. 330 μF, 4000 kΩ, low w, fast charge rate for BRL Electroporator. [000104] 4. Add 1ml of LB and transfer to Eppendorf tube. [000105] 5. Shake at 30°C for 2 hours. [000106] 6. Centrifuge down cells (2 min. 14 krpm). [000107] 7. Plate all onto LB + antibiotics (most Agrobacterium strains are naturally streptomycin resistant).
Example 3 — Protocol for Production of Transgenic Hairy Roots on Soybean [000108] Seed Sterilization. Rinse the soybean seed with 70% ETOH for 2-5 min. Remove and add 20% Clorox and shake for 20-25 min. Rinse 3X with sterile water. Plate the seed, 5 seed per plate, onto lA MSB5 + 2% sucrose + 0.2% gel (referred to as Vi MSB5). Place seed into chamber at 25C, 16/8 photoperiod for 5-7 day (depending on genotype) germination period. After 1 week seedlings can be placed into cold room for longer storage if necessary (not to exceed 2 weeks).
[000109] Agrobacterium Preparation. For Agrobacterium rhizogenes strain K599, take a small sample from frozen glycerol into 25-50 ml of NZYM media with 50 mg/L kanamycin in a 125-250 ml Erlenmyer flask. Place onto shaker at 28-30 °C for 16 - 20 hours. Pour sample into centrifuge tube and centrifuge the bacterium at 4000 rpm for 10 min. Pour off supernatant and re-suspend the pellet with an equal volume of liquid V_ MSB5 + 200 μM acetosyringone. Use pipette to re-suspend the pellet and homogenize the sample (remove all clumps). To determine O.D., prepare a 1:10 dilution by putting 900 μl MSB5 into cuvette and add 100 μl of bacterial sample. Determine the O.D.660 and calculate the volume needed to adjust (dilute) OD to approximately 0.2 for inoculation. Check final O.D.
[000110] Explant Preparation and inoculation. Place a sterile filter paper onto plates of 1/2 MSB5. Cut soybean cotyledons just above the shoot apex and place onto plate. Lightly scar the cotyledon's abaxial surface (flat side, upper surface that reaches toward sun) with a scalpel blade. Cut each cotyledon transversely into 2-3 pieces (no smaller than 1 cm). Add approximately 10 ml of prepared bacterial solution to each plate and allow cotyledons to incubate for 1 hr. Remove the bacteria using a vacuum aspirator fitted with sterile pipette tip, ensure that there is no standing liquid. Orient all explants with abaxial surface up and wrap plates for a 3 day co-culture, 25°C in light (16/8 photoperiod).
[000111] Hairy root selection and maintenance. After 3 day co-culture, wash explants with liquid Vi MSB5 + 500 mg/L carbenicillin. Transfer the explants abaxial side up to selection media, V2 MSB5 supplemented with 500 mg/L carbenicillin and 200 mg/L kanamycin. Roots should develop in approximately 2-3 weeks. The roots will form primarily from the cut vascular bundles with other roots developing from the small cuts on cotyledon surface. Remove roots (>lcm in length) and place onto replica media with transfers to fresh media every 2 weeks to prevent Agrobacterium overgrowth. After 6-8 weeks on selection the roots can be moved to media without kanamycin, however carbenicillin must remain in media for several months for continued suppression of Agrobacterium. At this stage roots can be used for testing RNAi for nematode control. Sterilized nematodes can be added and observed for RNAi affects.
Example 4 — Testing of RNAi for Plant Parasitic Nematode Control.
[000112] Various types of nematodes can be used in appropriate bioassays. For example, Caenorhabditis elegans, a bacterial feeding nematode, and plant parasitic nematodes can be used for bioassay purposes. Examples of plant parasitic nematodes include a migratory endo-parasite, Pratylenchus scribneri (lesion), and two sedentary endo-parasites, Meloidogyne javanica (root-knot) and Heterodera schachtii (cyst).
[000113] C. elegans: RNAi vectors can be tested through expression of the RNAi in E. coli. C. elegans are fed E. coli and assayed for their growth by measuring growth of nematodes, production of eggs and viability of offspring. Another approach is to inject dsRNA directly into living nematodes. Finally, soaking nematodes in a solution of in vz'trø-prepared RNAi can quickly establish efficacy of treatment.
[000114] P. scribneri: The P. scribneri in vitro feeding assay uses a corn root exudate (CRE) as a feeding stimulus and both the red dye Amaranth or potassium, arsenate as feeding indicators. Feeding is confirmed after seven days by the presence of red stained intestinal cells in live worms exposed to the Amaranth or death of worms exposed to arsenate. This bioassay is used to test soluble toxins or RNAi. P. scribneri has also been cultured on wild type roots of corn, rice and Arabidopsis, and on A. rhizogenes-induced hairy roots of sugar beet and tomato. P. scribneri is very valuable in evaluating transgenic hairy roots because of the non-specific feeding of these worms.
[000115] M javanica: Nematode eggs are sterilized using bleach and are used to inoculate hairy roots expressing RNAi. Nematodes are assessed for their growth by measuring knots, egg masses or production of viable eggs. An alternative approach is to microinject dsRNA directly into root feeding sites or into living female nematodes.
[000116] H schachtii: Cultures of this nematode were maintained on sugar beets. Nematodes eggs are sterilized using bleach and used to inoculate hairy roots expressing RNAi. Nematodes can be assessed for their growth by measuring knots, egg masses or production of viable eggs.
Example 5 — Plant Expression Vectors for RNAi
[000117] Modular Binary Construct System (MBCS): An important aspect of the subject disclosure is the Modular Binary Construct System. The MBCS eases the burden of construct development by creating modular pieces of DNA that can be easily added, removed, or replaced with the use of low frequency cutting restriction enzymes (8-base cutters). These constructs are useful for delivery of a variety of genes to plant cells and is not limited to the delivery of RNAi genes. To develop this system, a series of six, 8-base cutter restriction enzyme sites was placed between the left and right Ti borders of a previously created kanR/tetR binary plasmid (Figure 1). The production of both kanR and tetR MCBS aids the testing of constructs using different strains of Agrobacterium rhizogenes in different plant species. In addition to the MBCS, a series of shuttle vectors were created that aid in the cloning of useful DNA fragments by containing the multi-cloning site (MCS) of a modified Bluescript plasmid flanked by 8- base restriction sites (Figure 2). With six 8-base cutter sites, each site is, preferably, reserved for a particular function (Figures 3 and 4). Because of the close proximity of the Eme I and Sgfl sites to the left and right border of the binary vector, these sites are, preferably, reserved for gene tagging and enhancer trap experiments. The Not I site is, preferably, reserved for plant selectable markers (Figure 5). The Pac I site is reserved, preferably, for Plant Scorable Markers (Figure 6). The Asc I site is, preferably, reserved for RΝAi experiments (Figures 7 and 8), while the Sbfl site is, preferably, reserved for anti-nematode proteins. The restriction sites that are denoted in the Figures are, preferably, reserved for the denoted insertions; however, the MCBS binary and shuttle vectors do not require the restriction sites to contain these suggested inserts.
[000118] Plant Selectable Markers for MBCS: To further develop the MBCS, a series of plant selectable markers were added to the MBCS (Figure 5). Plant selectable markers that were added to the MBCS include: pΝOS/ΝPT-II/tΝOS (kanR), pΝOS/Bar/tΝOS (bastaR for dicots), pUBI/Intron-Bar/tΝOS (basta11 for monocots), and pUBI/Intron-PMI/tΝOS (mannitol isomerase11). [000119] Reporter Genes for MBCS: Four exemplary reporter genes are used in the MBCS are provided in Figure 6 and Appendix 2. GUS, a nuclear localized GUS, GEP, and the anthocyanin transcriptional activator papIC genes into the MBCS.
[000120] Promoters for MBCS: We cloned several useful constitutive and nematode-inducible promoters (Figures 6, 7 and Appendix 2). Constitutive promoters include the SuperUbiquitin promoter from pine (pSU) and two promoter regions from the Strawberry Banding Vein virus (pSBVj and pSBV2). Seven nematode-inducible promoters from Arabidopsis were also been cloned.
[000121] The following Scorable marker clones have been constructed and placed in the MBCS, NPT-II binary vector (pNOS/NPT-II/tNOS):
Intron/GUS/tNos Intron/NLS-GUS/tNOS Intron/GFP/tNOS pSU/Intron/GUS/tNOS pSU/Intron/NLS-GUS/tNOS pSU/Intron GFP/tNOS pSBV,/Intron GUS/tNOS pSBV,/Intron/NLS-GUS/tNOS pSBV ^Intron/GFP/tNOS pSBV2/Intron/GUS/tNOS pSBV2/Intron NLS-GUS/tNOS pSBV2/Intron/GFP/tNOS pKT/Intron GFP/tNOS pKA/Intron/GFP/tNOS
Example 6 — Control of Plant parasitic nematodes using RNAi inplanta
[000122] Production of RNAi Vector. The RNAi shuttle vector to be used is adapted from the Modular Binary Construct System (MBCS - See Example 5). RNAi shuttle vectors preferably comprise a promoter, intron, antisense RNAi, stuffer fragment, sense RNAi, and terminator (See Figures 7 and 8 and Appendix 2 for more details). The plant promoter can be constitutive, tissue-specific or nematode-inducible. The intron is necessary to eliminate expression in Agrobacterium.
[000123] The anti-sense and sense RNAi molecules comprise nematode-specific sequences and are disclosed herein. These genes are associated with pathogenesis, growth, or other cellular function in nematodes. An exemplary group of RNAi sequences for use in plant/nematode control may be based upon:
[000124] 1. Genes specific for nematode esophageal gland cells.
[000125] 2. Genes specific for plant parasitic nematodes but not other free living nematodes. [000126] 3. Genes common to all plant parasitic nematodes.
[000127] 4. Genes common to all nematodes (nematode-specific).
[000128] 5. Genes specific for important tissues or cell types.
[000129] 6. Genes from large gene families.
[000130] 7. Genes involved in nematode signal transduction or other cellular pathways.
[000131] Appropriate RNAi constructs allow for the formation of dsRNA molecules (the sense and antisense strands join to form the dsRNA). The terminator sequence adds a poly-A tail for transcriptional termination. The RNAi shuttle vector can then be subcloned into the MBCS and transformed into Agrobacterium rhizogenes.
[000132] Plant Transformation with RNAi Vectors. An exemplary transformation system for generating hairy roots using Agrobacterium rhizogenes is provided below. The RNAi vector once introduced into the MBCS can subsequently (as a binary vector) be transformed in A. rhizogenes using, for example, the electroporation protocol of Example 2. Once the A. rhizogenes is confirmed to contain the plasmid, it is then used in generating hairy roots (See Example 3). Using this protocol transgenic hairy roots expressing RNAi are isolated, cultured and tested.
[000133] Testing of RNAi Vector for Nematode or Plant Pathogen Resistance. RNAi expressing hairy roots can be inoculated with sterilized nematodes. Infested hairy roots can be observed and the effect on nematodes determined. An alternative approach involves the microinjection of RNAi directly into root feeding sites (giant-cells for root- knot nematode, and syncytia for cyst nematodes) or into living female nematodes.
Example 7 - Insertion of Genes Into Plants
[000134] One aspect of the subject invention is the transformation of plants with genes encoding proteins of the present invention. Transformation of plants as described herein can be used to improve the resistance of these plants to attack by the target pest.
[000135] Genes, polynucleotides, and/or RNAi molecules as disclosed or suggested herein can be inserted into plant cells using a variety of techniques which are well known in the art. For example, a large number of cloning vectors, for example, pBR322, pUC series, M13mp series, pACYC184, pMON, etc., are available for preparation for the insertion of foreign genes into higher plants via injection, biolistics (microparticle bombardment), Agrobacterium tumefaciens, ox Agrobacterium rhizogenes- mediated transformation, or electroporation as well as other possible methods. Once the inserted DNA has been integrated into the genome, the genetically modified-cell(s) can be screened via a vector carried-selectable marker that confers on the transformed plant cells resistance to a biocide or an antibiotic, such as kanamycin, G418, bleomycin, hygromycin, chloramphenicol, or bialophos, inter alia. The transformed cell will be regenerated into a morphologically normal plant. The transgene(s) in the transgenic plant is relatively stable and can be inherited by progeny plants.
[000136] If a transformation event involves a germ line cell, then the inserted DNA an corresponding phenotypic trait(s) will be transmitted to progeny plants. Such plants can be grown in the normal manner and crossed with plants that have the same transformed hereditary factors or other hereditary factors. The resulting hybrid individuals have the corresponding phenotypic properties.
[000137] It should be understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application.

Claims

We claim:
1. An RNAi molecule, optionally comprising a linker, wherein at least one strand of said RNAi is encoded by a DNA sequence selected from the group consisting of SEQ ID NO: 1 through SEQ ID NO: 139.
2. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 1.
3. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
2.
4. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 3.
5. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 4.
6. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
5.
7. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 6.
8. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
7.
9. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 8.
10. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 9.
11. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
10.
12. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 11.
13. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 12.
14. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 13.
15. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 14.
16. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 15.
17. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 16.
18. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 17.
19. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 18.
20. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 19.
21. An RNAi molecule according to claim 1 , wherein said DNA sequence is SEQ ID NO : 20.
22. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
21.
23. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
22.
24. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 23.
25. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 24.
26. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
25.
27. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 26.
28. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
27.
29. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 28.
30. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 29.
31. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 30.
32. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 31.
33. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
32.
34. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 33.
35. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 34.
36. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
35.
37. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 36.
38. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ DD NO : 37.
39. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 38.
40. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 39.
41. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 40.
42. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 41.
43. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 42.
44. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
43.
45. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 44.
46. An RN-Ai molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
45.
47. An RN-Ai molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 46.
48. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
47.
49. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 48.
50. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 49.
51. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 50.
52. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 51.
53. An RN-Ai molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
52.
54. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 53.
55. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
54.
56. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
55.
57. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 56.
58. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
57.
59. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 58.
60. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 59.
61. An RNAi molecule according to claim 1 , wherein said DNA sequence is SEQ ID NO : 60.
62. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 61.
63. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 62.
64. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 63.
65. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 64.
66. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
65.
67. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 66.
68. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 67.
69. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 68.
70. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 69.
71. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 70.
72. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 71.
73. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
72.
74. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 73.
75. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 74.
76. An RNAi molecule according to claim 1, wherein said DNA.sequence is SEQ ID NO: 75.
77. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
76.
78. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 77.
79. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 78.
80. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 79.
81. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 80.
82. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 81.
83. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 82.
84. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 83.
85. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 84.
86. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 85.
87. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 86.
88. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
87.
89. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 88.
90. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 89.
91. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 90.
92. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 91.
93. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 92.
94. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 93.
95. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 94.
96. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 95.
97. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 96.
98. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 97.
99. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO:
98.
100. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 99.
101. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 100.
102. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 101.
103. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 102.
104. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 103.
105. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED NO: 104.
106. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 105.
107. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 106.
108. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 107.
109. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID NO: 108.
110. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 9.
111. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 0.
112. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 1.
113. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 2.
114. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID .
115. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 4.
116. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 5.
117. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED 6.
118. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 7.
119. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED 8.
120. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 9.
121. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 0.
122. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 1.
123. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED .
124. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID .
125. An RN-Ai molecule according to claim 1, wherein said DNA sequence is SEQ ID .
126. An RN-Ai molecule according to claim 1, wherein said DNA sequence is SEQ ID 5.
127. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 6.
128. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED .
129. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED 8.
130. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ED 9.
131. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 0.
132. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 1.
133. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 2.
134. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 3.
135. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 4.
136. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 5.
137. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 6.
138. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 7.
139. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 8.
140. An RNAi molecule according to claim 1, wherein said DNA sequence is SEQ ID 9.
141. A transgenic plant or transgenic plant tissue comprising an RNAi molecule according f the preceding claims.
142. A method of disrupting cellular processes in a nematode comprising the steps of:
(a) 'providing a composition comprising a compound according to any of the preceding claims; and
(b) contacting a nematode with said composition.
143. An isolated promoter comprising the following nucleotide sequence:
aacagcccaagataacagaaaagtcaaaggtgttcgaaa gaccacttgtgactaaggatcatttcatccataat atctggtagca cagactcatgataaetgcgaggaacacaagttctttacagtcgattc aaagacactttctctttacggtttcattgaaggagccgacccagaat atgtcagagaagcttttcactgtgggttaatttcattaatctatcca ggtgaaaacctcaaggagatctctcttctcccaaaagacctctacag ggcaatcaaaaactacagaaccagagtttgtagtgcacagagtagac caatctacctgagaatcacgagtaccttcctagagtgggaaaatgat gacatccttattccataccactggattgaggtaggactatccaatgg aaaaattccatgggacaagtcatataagaagaccgcaacagtcgagt atcttccagagataactgcactcagacctaaaaggataaaagcagta tataatcagtgtactaagatcttcgcagattcaaagaagaagcttaa ctatgctgatgacaagataattctaataagcaattattcagaattaa tcaaggagaaagaattaataactctttcagaa-tatgaagcccgcttt acaagtggccagctagc atcactgaaaagacagcaagacaatggtg tctcgatgcaccagaaccacatct gcagcagatgtgaagcagcca gagtggtccacaagacgcactcagaaaaggcatcttctaccgacaca gaaaaagacaaccacagctca catccaacatgtagactgtcgttat gcgtcggctgaagataagactgaccccaggccagcactaaagaagaa ataatgcaagtggtcctagctccactttagctttaataattatgttt cattattattctctgcttttgctctc atataaagagcttgtatttt catttgaaggcagaggcgaacacacacacagaacctccctgcttaca aaccatgtattgtagctaaacctcttaggag! .
144. An isolated promoter comprising the following nucleotide sequence:
tggtggggacaatggatccggtctgcgtagcaacaaggctg aaaaagattaaacagaaacctgtgatcat agcgttggaccaccacc aaaacctcctgagccaccaaagcctccagagcctgaaaaaccaaagc ctccaccagcacctgaaccaccaaagcatgta gcaagccaccttac tgcaacagttgtgatgttgtgtctg actacc atgaaagtggaag cggctgcaccattctttgagtcatatatcgcgtaccatagccttcat gttaagtcctgta tagccaatactaattcatcatgttctcatgct tttttgtt atttctttttctcaaatatgaatctctgttgtttgtcc ctcccctgtttataattagtcgcttctttgacacaagaagtctcatg agttcatgctaaagaaaataaaagttcaaattaaaacaccaaatgtt tgattaatttccataaacctgtgaagcagaaagttagtcatgttgac ctgaacagagcttaggaagtccttgaaggaca atcttcaagtgcta ttgggtcgtagcactcttaggcccattaacttcattgagcccattaa a tatgcaaaacaagaaatgagacatatggaaacattagggttctta caggaaaaaataggaaaaagcagggacaactaaacaaaaattcagaa acaagaggcaagtggacgaccacggcgtaagatcaacatgtggtgat gtgcatgagaccaagaccattttttctcgttcttcaacgcacacttg gtcttttcttatgtttgttgcatttctttattaggcagaccctctct cttttttaataggatagtaaaaaatatatgattttattttgttgaaa cattttgagttaaaacctaaacttatagtaagcatttgtagagtgaa tttccta acgacatcta caacatgacctctaaccaaaaaaatatt gatgaaactact taagtagtaaaacctaaagcaattaaaatttcct ttaaattagtagtttgtgtaaattaattgacatgattgcgtcgaaag aaatcaaaacag atatcgtgaacttaggagaatgttttatatcgt gtttcaacacatgattgctagcatatgtgtaggtgtcgtagacgtta cataacaatcatcactcgtaaatatcaaagtggtttctgagagaaac aaagggttatgattttcccaactgcactagttgtgtattgtttcttt cacacg a gct ctgagttctgcccaaagtggaaattaaagcagag ttgggagagatcataat tattagggttcgttatgctcaagtcatga cgtaaaatgaaaatttgtttttattctttcaccaacacaaagaatag ctagttatctctttttttatatataacaattcatgaagttgatcagc tttatacacatcatccaatcgaattgctaatctagagatggaaa at caggatagagccaataaga atcaaatccaatggacccattttctcc a gtgctaattcatacaatctgtttttgtctgcttta ttgatgatg atgctgagcgtttttaagtgtgaactaagatctagctaaccaaacaa aagatggtctcttctgtctttgtcgtataagagcaagagagtggttt gattcaatttttaaaattctaaataaaactccaaccgtgaatccagc catgaaactctttttagaaaatccttttttataacaaataattcctc tgcttcttcttcttcttcgtttatttcaccttttttggtttctttag ctcagaaaaagcccattctttttttctattcttgtttattttaatca tactgtgcgtttctacaaagtttgttcctttcttcttcaactctctc actcacagtcacagagatctgtttcttttctttttttgctttcactc ttctcttccagt*.
145. An isolated promoter comprising the following nucleotide sequence:
agcaaagcaagaacaccagagaagaagaaaagcactacaga gaaaaatgtgagcttaagcgctctccaacaacacttctctgggagtc taaaggatgctgcaaaaagccttggtggtgagacttccgca atttc caagcatgggtttatttttgttagcacacaaactatctgaccctcga cttggattttcttctgcagtttgtccaactacattgaaacggatatg caggcaacatggga catgaggtggccatctcgtaagattaacaaag tgaacaggtcactaaggaaaatacagacggtactggactcggtccaa ggtgtagaaggaggactaaagttcgactcagcaactggcgaa tcat tgcagttagaccttttattcaagaaattgatacccaaaagggtctgt cgtctcttgataatgatgcacatgcaagaagaagtcaggaggatatg cctgacgatacttcattcaagctccaggaagctaaatctgtcgacaa tgccattaagttagaggaggatacaaccatgaatcaagcaagaccag gtaagaacttctctatccataaaccatagatggagcgattagaatct taatccattttcagtttttgcaggatcattcatggaggttaatgcta gtggtcagccatgggcttggatggccaaagagtctggcttgaatggc agtgaaggaataaagagcgtttgcaacttaagctctgtggaaatttc agatggaatggatccaacaatccgatgcagtggcagta tgttgaac ctaaccaatccatgtcatgcagcatatcagattcatcaaatggc ca ggcgcagttctgcgtggaagctcatctacttccatggaagattggaa ccaaatgagaacccacaacagtaatagcagcgagagtggatcaacaa cgctgatcgtaaaggccagttatagagaagacactgtacgtttcaag ttcgagccatcagttgggtgtcctcagctctacaaagaagttggaaa • acgt ttaaactgcaggacgggtcgtttcagctgaagtac ggatg atgaagaagaatgggtgatgctggttacagattctgatctccaagaa tgtttggagatattacatggtatgggaaaacactcggtgaagtttct cgttcgtgatttgtctgcccctctaggtagttctggtggcagtaatg g tatcttggaacaggcttatgacgtcgtaagacatagacacacaca gttatgtattcccagtgaaagaatgttgt a tctctagatatta gtatgc tataaataggcatgaaggagaaagacaattttgg atagt ggagttcagcagaaaatgtatatgttttttcgttttatatgaatcag agaataaaagttggatgttatatctacgttgctaatgttgtacctgc tcacccatctttcatataagaaaagagaacacttttagttatccctg tgatgcagaatcgtattctttgttatctctccattcctgtggaaacc aacaaagtcaactaaatttcggtttaattggttggtttttaagtcaa cgaggact gattttagttgggcttgggccta aattgtg tca ca ttgggttttttcccccttatcagtttaacgtccatatccatatcttt ttctttttttaacggcaaagttcatatccatatcttatgatgtgcct aaaagagggagaagatgcgaagacagaattttcatatttgaaagggt tcgatatcgatattgggaaacgaatcaaggtcaaaaaactcagtcta atagttgaaatttaaaaatt tat aattcaatccgattggtttcgt tttgttatggttcggttctatatcatcaaaccaatcggtttggtcct aaagataattat aatattcaccaacaccagtgttaaacacata ca acaaacctaaagttagataaacaaagaga; .
An isolated promoter comprising the following nucleotide sequence:
-aattggcactcttcttctgctgggttccaaaagaaacgaat caatatgtgcaacaagaagagctccagaagcagtca cttctaaaat cttaatctaacaacagctcaagaagaaaaaaattccatagctagaga gaacacaaagtcacaagacgacgtcgtagaggcacaaagtcaaacct gaatggcttaagccgaactgagtggttttgactagaccatcatcaga aaagtctccaagacggtagtcggatgttagatcgctcaagtaatttt tggttttgttggtctcacgttttcagctgcccatttgatttcagttt gggctt tcct a ctctaaaggcccaat tcatttaggtttagttt atttgatcattatccttactataaaggcttcgcctttcgagaaattt agggtttcttctgtctgtctcgtcactcaggtttgtgcctcaacgac tgcttcacttctagcttgattcttcttcttcgtttatatgtatactg tacattagat attcttgtttctcgagcttctgctatagattttgat tcttt ttttgttgtctttgtttcgtttccaggatcagatcttagct aaattgagacaagctcaaaatgaggtacttgacgcatctc taoatt cactgtttaattagagaacaatapgtctctgaatcgtgattcagaga cgtattgttcttctgtcatatgcaataagtttaattagagaacaata cgtctctgaatcgtgattgttttttggatgtgcgttattgatagctt tatgatgttaatagtctaggattgacacgaagttgttctgcagtttt gcataaatgctctttactaaggcctctaaatttggatgacaaatcta aatcttgcc cataaaaatttaggtgtattaagataagattat ttg tatggtagtgtctataatgtgggttgttcatgttgaggttgtcaatg ttgtgtatttttgtttgtttagttaatttgcttaactctgttctttg tgggttaatacagtaagcttcagagtgaggccgttcgtgaagccatc actactatcacagggaaatccgaggcaaagaaacgtaactttgtcga gactattgagctccagatcggtctgaagaactatgaccctcaaaagg acaagcgtttcagtggatctgtcaagttaccacatatcccccgtcct aaaatgaagatctgcatgctcggagatgcccagcatgttgaagaggt gata a cttttcatggaaattgatcattttgtgctctgtttcttgt ataatggttttgtgctcatttcatttggtggctctattagtttca t tgatgttgtatatgtcttctgaatgtagatgcatgatgttttcggaa tttggtcattgt tat taggcttca ttcttgcataattaaata t tgctta tcatct gtatct ttcgtaggctgagaagatggggttg gaaaacatggatgttgagtctctaaaaaagct aacaagaacaagaa actcgtcaagaagcttgcaaagaaataccatgctttcttggcctctg agtctgtcattaagcagattcctcgtcttcttggtcctggtcttaac aaggcaggcaagttctggctacagctaatattccattgttcttcttt acatccgttttgattttggataggttttagtagtctatttcttttgt caatgtctttttgatacaatgccaatcctt atcctgtgagattatg cttctttgatgattcttaagtaacattcctttgctttactttacaca ggaaaattcccaactcttgtgagccaccaggaatccttggagtcaaa ggtgaatgaaacaaaggcaacagtgaagttccagctgaagaaggttc tgtgcatgggagttgcagttggtaaccttt .
7. An isolated promoter comprising the following nucleotide sequence:
•tggcaaactgagatataagagggaaggtgattttcatgcaa attttttttttatttttttttgaatgaatgcaaaatttattcaaaaa aaaaaaacctgggctacatcaagtacttcatttctgagtttttgaaa aatctaaagacaacaaaagactttacaatttaataaaaaaataataa aaatactttatcactctcaacgaaattgttgatttaataacgtatct cttggtaaaacagcgttttat tgacgaaattgttataaatgaataa aatgataatagaaactagtgtggtacgtaaaatacctctcatttggc aaaataacggttatg atcatgagtat gcatacgacagcgtgctta aatagtgtgctttcaggagaaaatatataccaag at tgctgaaa ttaccacgcaaatctgaggttcgaatggcaaaataaaaaaccaatgt catttccttaatgtattaaggtcatttaaataaaattgtacactttt ttcacctgtaagcgttccaaagtgtagaatggataactagaagggtc aaaggta aata taataagcgaactcactttttgcccaagtgattt cacttcttacatttgcttgatatagttacccaaaagtgtatatatat tcccttatacaattgttctattttctggattataaggggaataagaa aaaagaaaagagagagtatataataatactttt taaagtgatgtta gattctaatttgtaacgaaaagttcaaagtgaaagaaaaaacgaaaa agttt tctgttttgttttatatctatagccaagaaagtttctcaga tttacaagaagttaactgagaaaaacaaaaaaaaaacttatgaagca tgaaagactaattaacgaggtgattaattttgagacaaattaaacat cgaattaaaagtaacatttggagggtttatatgtta atatgtgaca tgataagtccgattcatgactaatgtatatctggaatctaacatgga agaatagagaacgaagcagagccaaggtcaacttgccagacacgaat caacagattgtgaatgagaccaaatcaatggtcataaaccggttggg tttaaaccggcaagtcatccttggctcaattccattcg tattcctt catgcaagaccctctgatacaaccaaagactcccattacaatattct ttcgatcacgagctacttat ttcaaatgtgttacctctttcgtgac tcttgtgttgtgtggtaaagcctagtcgagatgtgtcggtata ata ggcatacatatacaaatgcgacaaaataagtatattatattgtttaa tttctatattccatttctatatgcatggctgggatttttgaccaaaa ccctaattcaagaatagaatccaaaagatgggatcaaagaatataat ctaatgggctgaccacattttccgatttaattcgcatagttaa at ctttccactactttatgccgcagaaatttgtaattaagtaagacaaa gaaatacagatataagatggtcgtagaaaccagtagaggaatttcat ttttcgtggataagtggaatattaataagagaatggtctttactct tacagtgggaaatgggaatagtagcccattataatttca cagattc tatatatgcatgtttgtataagctaaaataaatacgtttaagcattc ttcaaaaaaatttacaagttctagagactctcttaacgtcggcaatt tatattctactttacatgacactttcaggaaaagaaaactatactca ctagcagatcattaaattttctttttctttttttgaatgaaccttag ttgtggtttttatttttgttagctagaaacttcagtgttttttttcc gccaatggtagtgctttgatgatggtccgg' .
148. An isolated promoter comprising the following nucleotide sequence:
caatcaaggtaacgaaggaggatcagcgaaaggatgggcta ta tggagttttttcctgcgtgtaagtaatgctttgtgatcttcca tgcggacata aactgaagaataaactcaactcattgtgttctggtg tgtttcttctgatcaga tcctcgttgcatctgcacttttctgctgt gggggctt at tataaaacaagagtagagcgtgtggtaatcttcat atctttctacaattccacttccattctctaattattctctcacgtga tatacacacactcaatcactgatgtactcgtatggatgcagcgtgga actgatgcattgccggggatgtcacttctatcgggcttactagaaac tgtaagta tacaagaaaactcaaaaggattccatttatgcaaaatc taagagaaagctcactgtggtctttggttacaatttatggatc ctc aagagacaaatgctatgtaagctaattgattttggtcttgataaaca ggtgagtggaagtggacaaagctactcaagaactgaagacatcaaca atgcttttgccaatgaagtctcatgggaccgctcttccgcatcttct actcaagcgacaacaacacagagaccaagtgaaagaacata ggtgc gatctaattttgtcaagtgcctcacaagaggtactgtttcaagccat ggtatggcacgcttgtgatctgcgatttctggattttgctttgtatg tttattttctacc tctagaaagaggtcaaaaagttaatagc tcac cgtgagaatgttgttttcaccagattcatgtgctatgatagaaaaag acaaagcaaacaagagttctttctttgcttaggttacaagaacaaga gtatcgttataaagtcaacaaagattgaaacatatttttgtcaaggg agtggttagaatctcttcctactctcttgcctttctcactaagacaa aaaaaagacttggactttgtctaaggttttgtggatattat aacca agtccttttgcaaaaagtaatattgttttttcgcattcctcttttag aatttagt taatctaggctttatattggttattactttcttgaaaa atgatctgtttattctattcatacttggttacctcgctttttatctt act ctacaaaaggattatcagtgaaagttagtctcttactctcacc ttccgaaaataaaacaaaaatatcgatacttctagatcaaaccaagt tgattaaaacatccctattccctacgattctgatcttgagatatatt atcatgttaagatctaaattgacaagaaaactgatttttcatttcta gtaggaaaaataattactattagtgatcatgattgtcgaccgtaaga ggtggtttagttactctccatctttct tgaagaagtcagaaagtca gaaattatatcaaattaaacatcaatattgaacacata atctgtat ggttttatgtttagaaaattccaatatttatatattcctagggaaaa agaagcttattcttcaaat attgttatgagtcgttaaaatatggat aaaaatataaagtctaaatattaaaaactcagtttgctttgctttta cctctccaagtctccaaagtcaaattaattttagttaattaaaccaa aaaaggtttattagtcaaacttagcatgcaatgctgggtaccaaacc caagcattagtctcttttaatcttctttttctccaataagtttttac aatttttaattgtttgcatttcccttgattatttatcttcatcccaa tttagctaataccaactccgtttcttattcttccaagtcttttccta taaatacgttcttcttcccctcttatttcatatcactcaccacaaag tcttctcatttcctcat .
. An isolated promoter comprising the following nucleotide sequence:
atgttgtgagtgaaggagaagaagagggaaacaaaggtatt tatttgtagcgagttttgttttgtgacgcggttttgtctgtgttcaa tgttgacgaaacgagtgagagagtgtctgattattaaagaaaaccct aattaagtcagacccgccggttataaaaatagtcaaaaagtaggaaa acgcgtgtgtgagtgagacagagacagcccattgtttgctttatggg cttataagcgagacgtgttaattgggctttttcctttatggccgaaa acaaaagaaacgtcgcctgagagattcgaactctcgcgggcagagcc catgtacttagcaggcacacgccttaaccactcggccaaagcgactt gttgctatgagttagacaaaatcattaaaattctctattatgatttc tcatagtgtgtgtgtatattgtggatctactaaaaattctttgttat tattactttat ttgtgaattagtttgatataggtaagtacaaagtt aactttattatttactcaaaatttatcagattaactgattttatatt gtttcctttggtatatagacgtactatagtttttagaaaaaccataa gattcct atatttcatagagtgaagagatgagatgagatcttggc tggagaagaaataagtttccacgaggaggactctttttttttggtga agacgaggaggaggactcttggttgatccagtctttacgttagacat cgacccctacatttatttgcctttctctatcaacatggcaggtaaaa atcttcattcaaccgaaccaaccaaagtctcttcccaataa attca agcaccatcctttgggaaactcatacatactacagtctacactcttt cattttctttcaacgctcaacttaacaaatgatatagtctagttgtc aattatatgttttaattagtgttttcacatcaaattctggtttgata tttgatgactattttcggaaacatctcaatgtcccgcaaatacaatc gtctatcatatataatcccgtacgttgtat cttatagatagaataa tatggcgtgatctttataatataacatatagaatcgtgtagat tat tttattttatttttatatatcgcataaattgcaaaatacttatatat gtttgttatatatgatacccattttatagttacttaaaaaaagttaa gcgataatatatatatatcaactttttataacaaaaaagtataacac atggtaaagaaaaataaaaatgaagacatggtgtgacacgaaaatgg cactaaatatacatatataatagatagc acaata ccca cataca cacttttttaattgactaatacataacttacacacttttttaattga ctaattcataactttttatcattgtcaacatgcaaattcatatttcc gttgaactattattcttattttgtttttaaaagaagggcttcctggt aataaaaatatgatttccaaatgacgttagagcaaaaaaaaaaaaag gttgtctggtctggtaaaatgaaaaagcaaagcgtcttggtatagaa aagtaatatactgcctcctaatttcttcgtccttctaccgaagaatc tctccactcttgccctctttcgaaaccctaaaccagaagcaccagat tttttcaactttttcccagagaacaatagaaaacccaacttgtgctc tctagggttttctttattccttctcatctttggattttcttgggtca tcattttggaagcttacccaccagcgaaaaaattataacttccatcg attcctggcttctctctctcgctctctctgcatgtgctaaatcgccg gactgatcctcactgtcacctctgtt .
. An isolated promoter comprising the following nucleotide sequence:
ga aggggtttgagttgtcactggaaagaggtttgattgt gagtgatgatggagagattatgaaggagtttgtgtgtatttatagag gagttagggttttgaggtttgatgagaagtaggtttgaagaagtttt gttgttgcaactta ttagagttacttgttccacaaccacaagtaag attggtcacttctaagttctaactagaaacaaccatgacacatggag atttcagctaacctagtttaatgtatatgtattatattttatttaaa tattataaaataaaataaattttcacaaataaaagaactacaaaaaa gtgagaaaaataatttgataaacaaatttagaaaattagtatatcaa taaataaatttataatccgatggttttgccttttggtttggcctttg tttgaacttcgatgagtgactatgtatagcgaaaacaattcggtttg tttttggtttaattttaaaaaatacaagcgacaatatctgatgagaa taggtgaaaagcaaataatatcagtttaattggaaatat act tt ttacaactaatattttgtttggtcaaccaacaaatagatttaattaa ttatggtttatgagcttttatttgttgcgacagtatatatatgttaa aatagtgata tgcatggcggaaggtccggaagcaacacatatctcc tttttaattttttttttaacaagaataacatgttaatttttttttga aattaataaagaatacatatttctaatttttgcgtcagatagatgat taaagagtgtgtgttttttttaacaaacaaggaatacattatacata tttcatatttctctcgacattgtttgtttttttaaaaaatagattaa agagtctacgaagctaagtagctaacgaagacttgaaatgagaagaa gacgagaatcttttaatattttttgttaagcgataatattttgaaaa ttaataaatatagattaaggaaataacaataacgcagatatcgg aa gtcatagaaaaaaagaaacaacacaaacttacataaacatgtttcct aatttgtaatggagtaaaattccttcttttttttttttttttgattt ggattccaattagtaaagaactcaatgactataaataacctttaacc ctctcattatttcttactatcaattgattaagctctcgttcc aaga aagcaatagacgaacaagaacccatcgaagaacacaaatctctcttt gaagttgtcgataatgttagtacaccgttacttcgtccaagactttt ttgccgttccgtttcttacaaaacaaggatttggttacca tacttt tgtcgtaactcctttttacatgtacgtcaaaaagtggttcctcgctc cggcttgaagaaacgaccttcttacccacaaaaagcttattttaaac cgtctaaaaccggaaaatctcaatctaaaccggatacggttea gag aaaccgattcaaacaccgagtgaagaagtagaattttttgatggttc cgtcacaatgtgtgctgctccttcgccaagacatgtaccgattccga tattttgtggtgtaaagatgatcaaagagtcttcaaagctaagcacg acttgaatgagaagaagaagaccaattactcaattagattttgtttt gtggagcaattattgtctatttatctttgtttttagcaaataatctg tatccactaatcttcacagtacttgactaacaagaagtaaagagttt tcttatttccaattgttttttaatctgatacttttttcataatttta caatgtttgatgaaaaaaaacattcaaacctaaattttctttttttg gtatgaattcaaacctgaattact ttgacgaggacccgacggtata aatagggtgatctcccaacaaacaaaaagggt .
151. A transgenic plant or transgenic plant tissue comprising an isolated promoter according to any of claims 143 through 150.
54 APPENDIX 1
Figure imgf000055_0001
Figure imgf000056_0001
56
Figure imgf000057_0001
57
Figure imgf000058_0001
58
Figure imgf000059_0001
59
Appendix 2:
Exemplary genes used for RNAi vectors. Promoters:
Constitutive:
Super Ubiquitin from Pine
CCCGGGAAAACCCCT CACAAATACATAAAAAAAATTCTTTATTTAATTATCAAACTCTCCACTACCTT TCCCACCAACCGTTACAATCCTGAATGTTGGAAAAAACTAACTACATTGATATAAAAAAACTACATTA CTTCCTAAATCATAT CAAAATTGTATAAATATATCCACTCAAAGGAGTCTAGAAGATCCACTTGGACA AATTGCCCATAGTTGGAAAGATGTTCACCAAGTCAACAAGATTTATCAATGGAAAAATCCATCTACCA AACTTACTTTCAAGAAAATCCAAGGATTATAGAGTAAAAAATCTATGTATTATTAAGTCAAAAAGAAA ACCAAAGTGAACAAATATTGATGTACAAGTTTGAGAGGATAAGACATTGGAATCGTCTAACCAGGAGG CGGAGGAATTCCCTAGACAGTTAAAAGTGGCCGGAATCCCGGTAAAAAAGATTAAAATTTTTTTGTAG AGGGAGTGCTTGAATCATGTTTTTTATGATGGAAATAGATTCAGCACCATCAAAAACATTCAGGACAC CTAAAATTTTGAAGTTTAACAAAAATAACTTGGATCTACAAAAATCCGTATCGGATTTTCTCTAAATA TAACTAGAATTTTCATAACTTTCAAAGCAACTCCTCCCCTAACCGTAAAACTTTTCCTACTTCACCGT TAATTACATTCCTTAAGAGTAGATAAAGAAATAAAGTAAATAAAAGTATTCACAAACCAACAATTTAT TTCTTTTATTTACTTAAAAAAACAAAAAGTTTATTTATTTTACTTAAATGGCATAATGACATATCGGA GATCCCTCGAACGAGAATCTTTTATCTCCCTGGTTTTGTATTAAAAAGTAATTTATTGTGGGGTCCAC GCGGAGTTGGAATCCTACAGACGCGCTTTACATACGTCT CGAGAAGCGTGACGGATGTGCGAC CGGAT GACCCTGTATAACCCACCGACACAGCCAGCGCACAGTATACACGTGTCATTTCTCTATTGGAAAATGT CGTTGTTATCCCCGCTGGTACGCAACCACCGATGGTGACAGGTCGTCTGTTGTCGTGTCGCGTAGCGG GAGAAGGGTCTCATCCAACGCTATTAAATACTCGCCTTCACCGCGTTACTT CTCATCTTTTCT CTTGC GTTGTATAATCAGTGCGATATTCTCAGAGAGCTTTTCATTCAACCCGGG
Strawberry Banding Vein Virus 1 aagcttttcactgtgggttaatttcattaatctatccaggtgaaaacctcaaggaga tctctcttctcccaaaagacctctacagggcaatcaaaaactacagaaccagagttt gtagtgcacagagtagaccaatctacctgagaatcacgagtaccttcctagagtggg aaaatgatgacatccttattccataccactggattgaggtaggactatccaatggaa aaattccatgggacaagtcatataagaagaccgcaacagtcgagtatcttccagaga taactgcactcagacctaaaaggataaaagcagtatataatcagtgtactaagatct tcgcagattcaaagaagaagctt
Strawberry Banding Vein Virus 2
Gtttaaacaacagcccaagataacagaaaagtcaaaggtgttcgaaagaccacttgt gactaaggatcatttcatccataattatctggtagcacagactcatgataactgcga ggaacacaagttctttacagtcgattcaaagacactttctctttacggtttcattga aggagccgacccagaatatgtcagagaagcttttcactgtgggttaatttcattaat ctatccaggtgaaaacctcaaggagatctctcttctcccaaaagacctctacagggc aatcaaaaactacagaaccagagtttgtagtgcacagagtagaccaatctacctgag aatcacgagtaccttcctagagtgggaaaatgatgacatccttattccataccactg gattgaggtaggactatccaatggaaaaattccatgggaσaagtcatataagaagac cgcaacagtcgagt tcttccagagataactgcactcagacctaaaaggataaaagc agtatataatcagtgtactaagatcttcgcagattcaaagaagaagcttaactatgc tgatgacaagataattctaataagcaattattcagaattaatcaaggagaaagaatt aataactctttcagaatatgaagcccgctttacaagtggccagctagctatcactga aaagacagcaagacaatggtgtctcgatgcaccagaaccacatctttgcagcagatg tgaagcagccagagtggtccacaagacgcactcagaaaaggcatcttctaccgacac agaaaaagacaaccacagctcatcatccaacatgtagactgtcgttatgcgtcggct gaagataagactgaccccaggccagcactaaagaagaaataatgcaagtggtcctag ctccactttagctttaataattatgtttcattattattctctgcttttgctctctat ataaagagcttgtattttcatttgaaggcagaggcgaacacacacacagaacctccc tgcttacaaaccatgtattgtagctaaacctcttaggaggatatc 60
Nematode Inducible:
Trypsin Inhibitor from Arabidopsis (clone#6598343) cccgggagcaaagcaagaacaccagagaagaagaaaagcactacagagaaaaatgtg agcttaagcgctctccaacaacacttctctgggagtctaaaggatgctgcaaaaagc cttggtggtgagacttccgcatatttccaagcatgggtttatttttgttagcacaca aactatctgaccctcgacttggattttcttctgcagtttgtccaactacattgaaac ggatatgcaggcaacatgggatcatgaggtggccatctcgtaagattaacaaagtga acaggtcactaaggaaaatacagacggtactggactcggtccaaggtgtagaaggag gactaaagttcgactcagcaactggcgaattcattgcagttagaccttttattcaag aaattgatacccaaaagggtctgtcgtctcttgataatgatgcacatgcaagaagaa gtcaggaggatatgcctgacgatacttcattcaagctccaggaagctaaatctgtcg acaatgccattaagttagaggaggatacaaccatgaatcaagcaagaccaggtaaga acttctctatccataaaccatagatggagcgattagaatcttaatccattttcagtt tttgcaggatcattcatggaggttaatgctagtggtcagccatgggcttggatggcc aaagagtctggcttgaatggcagtgaaggaataaagagcgtttgcaacttaagctct gtggaaatttcagatggaatggatccaacaatccgatgcagtggcagtattgttgaa cctaaccaatccatgtcatgcagcatatcagattcatcaaatggctcaggcgcagtt ctgcgtggaagctcatctacttccatggaagattggaaccaaatgagaacccacaac agtaatagcagcgagagtggatcaacaacgctgatcgtaaaggccagttatagagaa gacactgtacgtttcaagttcgagccatcagttgggtgtcctcagctctacaaagaa gttggaaaacgttttaaactgcaggacgggtcgtttcagctgaagtacttggatgat gaagaagaatgggtgatgctggttacagattctgatctccaagaatgtttggagata ttacatggtatgggaaaacactcggtgaagtttctcgttcgtgatttgtctgcccct ctaggtagttctggtggcagtaatggttatcttggaacaggcttatgacgtcgtaag acatagacacacacagttatgtattcccagtgaaagaatgttgtttatttctctaga tattagtatgcttataaataggcatgaaggagaaagacaattttggtatagtggagt tcagcagaaaatgt tatgttttttcgttttatatgaatcagagaataaaagttgga tgttatatctacgttgctaatgttgtacctgctcacccatctttcatataagaaaag agaacacttttagttatccctgtgatgcagaatcgtattctttgttatctctccatt cctgtggaaaccaacaaagtcaactaaatttcggtttaattggttggtttttaagtc aacgaggacttgattttagttgggcttgggcctataattgtgttcatcattgggttt tttcccccttatcagtttaacgtccatatccatatctttttctttttttaacggcaa agttcatatccatatcttatgatgtgcctaaaagagggagaagatgcgaagacagaa ttttcatatttgaaagggttcga atcgatattgggaaacgaatcaaggtcaaaaaa ctcagtctaatagttgaaatttaaaaattttatt attcaatccgattggtttcgtt ttgttatggttcggttctatatcatcaaaccaatcggtttggtcctaaagataatta taaatattcaccaacaccagtgttaaacacat tcaacaaacctaaagttagataaa caaagagacccggg
Arabidopsis Transmembrane Protein from Arabidopsis (clone#6468048) cccgggaattggcactcttcttctgctgggttccaaaagaaacgaatcaatatgtgc aacaagaagagctccagaagcagtcatcttctaaaatcttaatctaacaacagctca agaagaaaaaaattccatagctagagagaacacaaagtcacaagacgacgtcgtaga ggcacaaagtcaaacctgaatggcttaagccgaactgagtggttttgactagaccat catcagaaaagtctccaagacggtagtcggatgttagatcgctcaagtaatttttgg ttttgttggtctcacgttttcagctgcccatttgatttcagtttgggcttttcctta tctctaaaggcccaatttcatttaggtttagtttatttgatcattatccttactata aaggcttcgcctttcgagaaatttagggtttcttctgtctgtctcgtcactcaggtt tgtgcctcaacgactgcttcacttctagcttgattcttcttcttcgtttatatgtat actgtacattagattattcttgtttctcgagcttctgctatagattttgattctttt ttttgttgtctttgtttcgtttccaggatcagatcttagctaaattgagacaagctc aaaatgaggtacttgacgcatctcttacattcactgtttaattagagaacaatacgt ctctgaatcgtgattcagagacgtattgttcttctgtcatatgcaataagtttaatt agagaacaatacgtctctgaatcgtgattgttttttggatgtgcgttattgatagct 61 ttatgatgttaatagtctaggattgacacgaagttgttctgcagttttgcataaatg ctctttactaaggcctctaaatttggatgacaaatctaaatcttgcctcataaaaat ttaggtgtattaagataagattattttgtatggtagtgtctataatgtgggttgttc atgttgaggttgtcaatgttgtgtatttttgtttgtttagttaatttgcttaactct gttctttgtgggttaatacagtaagcttcagagtgaggccgttcgtgaagccatcac tactatcacagggaaatccgaggcaaagaaacgtaactttgtcgagactattgagct ccagatcggtctgaagaactatgaccctcaaaaggacaagcgtttcagtggatctgt caagttaccacatatcccccgtcctaaaatgaagatctgcatgctcggagatgccca gcatgttgaagaggtgatatatcttttcatggaaattgatcattttgtgctctgttt cttgtataatggttttgtgctcatttcatttggtggctctattagtttcatttgatg ttgtatatgtcttctgaatgtagatgcatgatgttttcggaatttggtcattgttta tttaggcttcatttcttgcataattaaatatttgcttatttca cttgtatcttttc gtaggctgagaagatggggttggaaaacatggatgttgagtctctaaaaaagcttaa caagaacaagaaactcgtcaagaagcttgcaaagaaataccatgctttcttggcctc tgagtctgtcattaagcagattcctcgtcttcttggtcctggtcttaacaaggcagg caagttctggctacagctaatattccattgttcttctttacatccgttttgattttg gataggttttagtagtctatttcttttgtcaatgtctttttgatacaatgccaatcc tttatcctgtgagattatgcttctttgatgattcttaagtaacattcctttgcttta ctttacacaggaaaattcccaactcttgtgagccaccaggaatccttggagtcaaag gtgaatgaaacaaaggcaacagtgaagttccagctgaagaaggttctgtgcatggga gttgcagttggtaacctttcccggg
Diaminopimelate Decarboxylase from Arabidopsis (clone#4159709) cccgggtggcaaactgagatataagagggaaggtgattttcatgcaaattttttttt tatttttttttgaatgaatgcaaaatttattcaaaaaaaaaaaacctgggctacatc aagtacttcatttctgagtttttgaaaaatctaaagacaacaaaagactttacaatt taataaaaaaataataaaaatactttatcactctcaacgaaattgttgatttaataa cgtatctcttggtaaaacagcgttttatttgacgaaattgttataaatgaataaaat gataatagaaactagtgtggtacgtaaaatacctctcatttggcaaaataacggtta tgtatcatgagtattgcatacgacagcgtgcttaaatagtgtgctttcaggagaaaa tatataccaagttatttgctgaaattaccacgcaaatctgaggttcgaatggcaaaa taaaaaaccaatgtcatttccttaatgtattaaggtcatttaaataaaattgtacac ttttttcacctgtaagcgttccaaagtgtagaatggataactagaagggtcaaaggt ataatattaataagcgaactcactttttgcccaagtgatttcacttcttacatttgc ttgatatagttacccaaaagtgtatatatattcccttatacaattgttctattttct ggattataaggggaataagaaaaaagaaaagagagagtatataataatacttttata aagtgatgttagattctaatttgtaacgaaaagttcaaagtgaaagaaaaaacgaaa aagtttttctgttttgttttatatctatagccaagaaagtttctcagatttacaaga agttaactgagaaaaacaaaaaaaaaacttatgaagcatgaaagactaattaacgag gtgattaattttgagacaaattaaacatcgaattaaaagtaacatttggagggttta tatgttatatatgtgacatgataagtccgattcatgactaatgtatatctggaatct aacatggaagaatagagaacgaagcagagccaaggtcaacttgccagacacgaatca acagattgtgaatgagaccaaatcaatggtcataaaccggttgggtttaaaccggca agtcatccttggctcaattccattcgttattccttcatgcaagaccctctgatacaa ccaaagactcccattaςaatattctttcgatcacgagctacttattttcaaatgtgt tacctctttcgtgactcttgtgttgtgtggtaaagcctagtcgagatgtgtcggtat atataggcatacatatacaaatgcgacaaaataagtatattatattgtttaatttct atattccatttctatatgcatggctgggatttttgaccaaaaccctaattcaagaat agaatccaaaagatgggatcaaagaatataatctaatgggctgaccacattttccga tttaattcgcatagttaatattctttccactactttatgccgcagaaatttgtaatt aagtaagacaaagaaatacagatataagatggtcgtagaaaccagtagaggaatttc atttttcgtggataagtggaat ttaataagagaatggtctttactctttacagtgg gaaatgggaatagtagcccattataatttcatcagattctatatatgcatgtttgta taagctaaaataaatacgtttaagcattcttcaaaaaaatttacaagttctagagac tctcttaacgtcggcaatttatattctactttacatgacactttcaggaaaagaaaa ctatactcactagcagatcattaaattttctttttctttttttgaatgaaccttagt tgtggtttttatttttgttagctagaaacttcagtgttttttttccgccaatggtag 62 tgctttgatgatggtccggcccggg
Peroxidase from Arabidopsis (clone#4006885) cccgggcaatcaaggtaacgaaggaggatcagcgaaaggatgggctatatttggagt tttttcctgcgtgtaagtaatgctttgtgatcttccatgcggacatataactgaaga ataaactcaactcattgtgttctggtgtgtttcttctgatcagattcctcgttgcat ctgcacttttctgctgtgggggctttatttataaaacaagagtagagcgtgtggtaa tcttcatatctttctacaattccacttccattctctaattattctctcacgtgatat acacacactcaatcactgatgtactcgtatggatgcagcgtggaactgatgcattgc cggggatgtcacttctatcgggcttactagaaactgtaagtattacaagaaaactca aaaggattccatttatgcaaaatctaagagaaagctcactgtggtctttggttacaa tttatggatctctcaagagacaaatgctatgtaagctaattgattttggtcttgata aacaggtgagtggaagtggacaaagctactcaagaactgaagacatcaacaatgctt ttgccaatgaagtctcatgggaccgctcttccgcatcttctactcaagcgacaacaa cacagagaccaagtgaaagaacatatggtgcgatctaattttgtcaagtgcctcaca agaggtactgtttcaagccatggtatggcacgcttgtgatctgcgatttctggattt tgctttgtatgtttattttctaccttctagaaagaggtcaaaaagttaatagcttca ccgtgagaatgttgttttcaccagattcatgtgctatgatagaaaaagacaaagcaa acaagagttctttctttgcttaggttacaagaacaagagtatcgttataaagtcaac aaagattgaaacatatttttgtcaagggagtggttagaatctcttcctactctcttg cctttctcactaagacaaaaaaaagacttggactttgtctaaggttttgtggatatt attaaccaagtccttttgcaaaaagtaatattgttttttcgcattcctcttttagaa tttagtttaatctaggctttatattggttattactttcttgaaaaatgatctgttta ttctattcatacttggttacctcgctttttatcttacttctacaaaaggattatcag tgaaagttagtctcttactctcaccttccgaaaataaaacaaaaatatcgatacttc tagatcaaaccaagttgattaaaacatccctattccctacgattctgatcttgagat atatta catgttaagatctaaattgacaagaaaactgatttttcatttctagtagg aaaaataattactattagtgatcatgattgtcgaccgtaagaggtggtttagttact ctccatctttctttgaagaagtcagaaagtcagaaattatatcaaattaaacatcaa tattgaacacatatatctgtatggttttatgtttagaaaattccaatatttatat t tcctagggaaaaagaagcttattcttcaaattattgttatgagtcgttaaaatatgg ataaaaatataaagtctaaatattaaaaactcagtttgctttgcttttacctctcca agtctccaaagtcaaattaattttagttaattaaaccaaaaaaggtttattagtcaa acttagcatgcaatgctgggtaccaaacccaagcattagtctcttttaatcttcttt ttctccaataagtttttacaatttttaattgtttgcatttcccttgattatttatct tcatcccaatttagctaataccaactccgtttcttattcttccaagtcttttcctat aaatacgttcttcttcccctcttatttcatatcactcaccacaaagtcttctcattt cctcatcccggg
Mitochondrial ϋncoupler from Arabidopsis (clone#4220510) cccgggatgttgtgagtgaaggagaagaagagggaaacaaaggtat tatttgtagc gagttttgttttgtgacgcggttttgtctgtgttcaatgttgacgaaacgagtgaga gagtgtctgattattaaagaaaaccctaattaagtcagacccgccggttataaaaat agtcaaaaagtaggaaaacgcgtgtgtgagtgagacagagacagcccattgtttgct ttatgggcttataagcgagacgtgttaattgggctttttcctttatggccgaaaaca aaagaaacgtcgcctgagagattcgaactctcgcgggcagagcccatgtacttagca ggcacacgccttaaccactcggccaaagcgacttgttgctatgagttagacaaaatc attaaaattctctattatgatttctcatagtgtgtgtgtatattgtggatctactaa aaattctttgttattattactttattttgtgaattagtttgatataggtaagtacaa agttaactttattatttactcaaaatttatcagattaactgattttatattgtttcc tttggtatatagacgtactatagtttttagaaaaaccataagattcctttatatttc atagagtgaagagatgagatgagatcttggctggagaagaaataagtttccacgagg aggactctttttttttggtgaagacgaggaggaggactcttggttgatccagtcttt acgttagacatcgacccctacatttatttgcctttctctatcaacatggcaggtaaa aatcttcattcaaccgaaccaaccaaagtctcttcccaataatat caagcaccatc ctttgggaaactcatacatactacagtctacactctttcattttctttcaacgctca acttaacaaatgatatagtctagttgtcaattatatgttttaattagtgttttcaca 63 tcaaattctggtttgatatttgatgactattttcggaaacatctcaatgtcccgcaa atacaatcgtctatcatatataatcccgtacgttgtattcttatagatagaataata tggcgtgatctttataatataacatatagaatcgtgtagatttattttattttattt ttatatatcgcataaattgcaaaatacttatatatgtttgttatatatgatacccat tttatagttacttaaaaaaagttaagcgataatatatatatatcaactttttataac aaaaaagtataacacatggtaaagaaaaataaaaatgaagacatggtgtgacacgaa aatggcactaaatatacatatataatagatagctacaatatcccatcatacacactt ttttaattgactaatacataacttacacacttttttaattgactaattcataacttt ttatcattgtcaacatgcaaattcatatttccgttgaactattattcttattttgtt tttaaaagaagggcttcctggtaataaaaatatgatttccaaatgacgttagagcaa aaaaaaaaaaaggttgtctggtctggtaaaatgaaaaagcaaagcgtcttggtatag aaaagtaatatactgcctcctaatttcttcgtccttctaccgaagaatctctccact cttgccctctttcgaaaccctaaaccagaagcaccagattttttcaactttttccca gagaacaatagaaaacccaacttgtgctctctagggttttctttattccttctcatc tttggattttcttgggtcatcattttggaagcttacccaccagcgaaaaaattataa cttccatcgattcctggcttctctctctcgctctctctgcatgtgctaaatcgccgg actgatcctcactgtcacctctgttcccggg
Stress protein from Arabidopsis (clone#6598614) cccggggattaggggtttgagttgtcactggaaagaggtttgattgtgagtgatgat ggagagattatgaaggagtttgtgtgtatttatagaggagttagggttttgaggttt gatgagaagtaggtttgaagaagttttgttgttgcaacttatttagagttacttgtt ccacaaccacaagtaagattggtcacttctaagttctaactagaaacaaccatgaca catggagatttcagctaacctagtttaatgtatatgtattatattttatttaaatat tataaaataaaataaattttcacaaataaaagaactacaaaaaagtgagaaaaataa tttgataaacaaatttagaaaattagtatatcaataaataaatttataatccgatgg ttttgccttttggtttggcctttgtttgaacttcgatgagtgactatgtatagcgaa aacaattcggtttgtttttggtttaattttaaaaaatacaagcgacaatatctgatg agaataggtgaaaagcaaataatatcagtttaattggaaatatttac tttttacaa ctaatattttgtttggtcaaccaacaaatagatttaattaattatggtttatgagct tttatttgttgcgacagtata atatgttaaaatagtgatattgcatggcggaaggt ccggaagcaacacatatctcctttttaattttttttttaacaagaataacatgttaa tttttttttgaaattaataaagaatacatatttctaatttttgcgtcagatagatga ttaaagagtgtgtgttttttttaacaaacaaggaatacattatacatatttcatatt tctctcgacattgtttgtttttttaaaaaatagattaaagagtctacgaagctaagt agctaacgaagacttgaaatgagaagaagacgagaatcttttaatattttttgttaa gcgataatattttgaaaattaataaatatagattaaggaaataacaataacgcagat atcggtaagtcatagaaaaaaagaaacaacacaaacttacataaacatgtttcctaa tttgtaatggagtaaaattccttcttttttttttttttttgatttggattccaatta gtaaagaactcaatgactataaataacctttaaccctctcattatttcttactatca attgattaagctctcgttcctaagaaagcaatagacgaacaagaacccatcgaagaa cacaaatctctctttgaagttgtcgataatgttagtacaccgttacttcgtccaaga cttttttgccgttccgtttcttacaaaacaaggatttggttaccattacttttgtcg taactcctttttacatgtacgtcaaaaagtggttcctcgctccggcttgaagaaacg accttcttacccacaaaaagcttattttaaaccgtctaaaaccggaaaatctcaatc taaaccggatacggttcatgagaaaccgattcaaacaccgagtgaagaagtagaatt ttttgatggttccgtcacaatgtgtgctgctccttcgccaagacatgtaccgattcc gatattttgtggtgtaaagatgatcaaagagtcttcaaagctaagcacgacttgaat gagaagaagaagaccaattactcaattagattttgttttgtggagcaattattgtct at tatctttgtttttagcaaataatctgtatccactaatcttcacagtacttgact aacaagaagtaaagagttttcttatttccaattgttttttaatctgatacttttttc ataattttacaatgtttgatgaaaaaaaacattcaaacctaaattttctttttttgg tatgaattcaaacctgaattacttttgacgaggacccgacggtataaatagggtgat ctcccaacaaacaaaaagggtcccggg
Pectinacetylesterase from Arabidopsis (clone#6671954) cccgggtggtggggacaatggatccggtctgcgtagcaacaaggctgaaaaagatta
Figure imgf000065_0001
Figure imgf000065_0002
1/8
PCT/US2001/018911 2000-06-12 2001-06-12 Materials and methods for the control of nematodes WO2001096584A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001275474A AU2001275474A1 (en) 2000-06-12 2001-06-12 Materials and methods for the control of nematodes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US21091700P 2000-06-12 2000-06-12
US60/210,917 2000-06-12

Publications (2)

Publication Number Publication Date
WO2001096584A2 true WO2001096584A2 (en) 2001-12-20
WO2001096584A3 WO2001096584A3 (en) 2003-01-23

Family

ID=22784842

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/018911 WO2001096584A2 (en) 2000-06-12 2001-06-12 Materials and methods for the control of nematodes

Country Status (2)

Country Link
AU (1) AU2001275474A1 (en)
WO (1) WO2001096584A2 (en)

Cited By (355)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003070886A2 (en) * 2002-02-20 2003-08-28 Sirna Therapeutics, Inc. Inhibition of protein tyrosine phosphatase type iva expression byshort interfering nucleic acid
WO2003093475A1 (en) * 2002-04-30 2003-11-13 Genesis Research And Development Corporation Limited Compositions and methods for the modification of gene expression_using the superubiquitin promoter from pinus radiata
WO2003070918A3 (en) * 2002-02-20 2004-07-08 Ribozyme Pharm Inc Rna interference by modified short interfering nucleic acid
WO2005071091A1 (en) * 2003-12-23 2005-08-04 Bayer Cropscience Sa Method for modifying gene expression of a phytopathogenic fungus
WO2005082932A2 (en) * 2004-02-24 2005-09-09 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes in plants
EP1572902A2 (en) * 2002-02-01 2005-09-14 Ambion, Inc. HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
WO2006020821A2 (en) 2004-08-13 2006-02-23 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes
US7022828B2 (en) 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
WO2006047495A2 (en) 2004-10-21 2006-05-04 Venganza Inc Methods and materials for conferring resistance to pests and pathogens of plants
WO2007104570A2 (en) * 2006-03-16 2007-09-20 Devgen N.V. Nematode control
WO2007117657A2 (en) 2006-04-07 2007-10-18 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
US7294504B1 (en) 2001-12-27 2007-11-13 Allele Biotechnology & Pharmaceuticals, Inc. Methods and compositions for DNA mediated gene silencing
WO2008095910A1 (en) 2007-02-08 2008-08-14 Basf Plant Science Gmbh Compositions and methods using rna interference of opr3-like gene for control of nematodes
WO2008095886A1 (en) 2007-02-06 2008-08-14 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes
WO2008095970A1 (en) 2007-02-09 2008-08-14 Basf Plant Science Gmbh Compositions and methods using rna interference of cdpk-like for control of nematodes
WO2008118324A2 (en) 2007-03-26 2008-10-02 Macrogenics, Inc. Composition and method of treating cancer with an anti-uroplakin ib antibody
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
JP2009523018A (en) * 2006-01-12 2009-06-18 デブジェン エヌブイ Method for controlling pests using RNAi
JP2009523017A (en) * 2006-01-12 2009-06-18 デブジェン エヌブイ Genetically modified plant system method for plant pests using RNAi
US7576261B2 (en) 2004-10-13 2009-08-18 University Of Georgia Research Foundation, Inc. Nematode resistant transgenic plants
US7645734B2 (en) 2003-05-19 2010-01-12 The Trustees Of Columbia University In The City Of New York Compositions and methods for treating and preventing heart tissue degeneration and uses thereof
US7659389B2 (en) 2001-05-18 2010-02-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MYC and/or MYB gene expression using short interfering nucleic acid (siNA)
US7659390B2 (en) 2002-02-20 2010-02-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA)
US7662952B2 (en) 2002-02-20 2010-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of GRB2 associated binding protein (GAB2) gene expression using short interfering nucleic acid (siNA)
US7662951B2 (en) 2000-08-30 2010-02-16 Sirna Therapeutics, Inc. RNA interference mediated treatment of Alzheimer's disease using short interfering nucleic acid (siNA)
US7667029B2 (en) 2002-02-20 2010-02-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of checkpoint kinase-1 (CHK-1) gene expression using short interfering nucleic acid (siNA)
US7667030B2 (en) 2002-02-20 2010-02-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of matrix metalloproteinase 13 (MMP13) gene expression using short interfering nucleic acid (siNA)
US7678897B2 (en) 2002-02-20 2010-03-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
US7691999B2 (en) 2002-02-20 2010-04-06 Sirna Therapeutics, Inc. RNA interference mediated inhibition of NOGO and NOGO receptor gene expression using short interfering nucleic acid (siNA)
US7700760B2 (en) 2002-02-20 2010-04-20 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular cell adhesion molecule (VCAM) gene expression using short interfering nucleic acid (siNA)
WO2010046423A2 (en) 2008-10-22 2010-04-29 Basf Se Use of sulfonylurea herbicides on cultivated plants
WO2010046422A2 (en) 2008-10-22 2010-04-29 Basf Se Use of auxin type herbicides on cultivated plants
US7795422B2 (en) 2002-02-20 2010-09-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
WO2010106163A1 (en) 2009-03-20 2010-09-23 Basf Plant Science Company Gmbh Nematode-resistant transgenic plants
US7803984B2 (en) * 2002-07-10 2010-09-28 Kansas State University Research Foundation Compositions and methods for controlling plant parasitic nematodes
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
US7858769B2 (en) 2004-02-10 2010-12-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using multifunctional short interfering nucleic acid (multifunctional siNA)
US7868159B2 (en) 2005-06-23 2011-01-11 Baylor College Of Medicine Modulation of negative immune regulators and applications for immunotherapy
US7868158B2 (en) 2004-07-19 2011-01-11 Baylor College Of Medicine Modulation of cytokine signaling regulators and applications for immunotherapy
US7888326B2 (en) 2003-04-04 2011-02-15 The Trustees Of Columbia University In The City Of New York Methods for promoting apoptosis and treating tumor cells inhibiting the expression or function of the transcription factor ATF5
US7893248B2 (en) 2002-02-20 2011-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Myc and/or Myb gene expression using short interfering nucleic acid (siNA)
US7897752B2 (en) 2002-02-20 2011-03-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of telomerase gene expression using short interfering nucleic acid (siNA)
US7897757B2 (en) 2002-02-20 2011-03-01 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of protein tyrosine phosphatase-1B (PTP-1B) gene expression using short interfering nucleic acid (siNA)
US7897753B2 (en) 2002-02-20 2011-03-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of XIAP gene expression using short interfering nucleic acid (siNA)
US7910724B2 (en) 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
US7910725B2 (en) 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US7915400B2 (en) 2002-02-20 2011-03-29 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of hepatitis C virus (HCV) gene expression using short interfering nucleic acid (siNA)
US7923549B2 (en) 2002-02-20 2011-04-12 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US7928220B2 (en) 2002-02-20 2011-04-19 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of stromal cell-derived factor-1 (SDF-1) gene expression using short interfering nucleic acid (siNA)
US7928219B2 (en) 2002-02-20 2011-04-19 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (SINA)
US7935812B2 (en) 2002-02-20 2011-05-03 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of hepatitis C virus (HCV) expression using short interfering nucleic acid (siNA)
US7943757B2 (en) 2002-02-20 2011-05-17 Mcswiggen James RNA interference mediated inhibition of intercellular adhesion molecule (ICAM) gene expression using short interfering nucleic acid (siNA)
WO2011062962A2 (en) 2009-11-17 2011-05-26 The Trustees Of The University Of Pennsylvania Smndelta7 degron: novel compositions and methods of use
US7977472B2 (en) 2002-02-20 2011-07-12 Leonid Beigelman RNA interference mediated inhibition of myostatin gene expression using short interfering nucleic acid (siNA)
US7985853B2 (en) 2002-02-20 2011-07-26 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of platelet derived growth factor (PDGF) and platelet derived growth factor receptor (PDGFR) gene expression using short interfering nucleic acid (siNA)
US8008473B2 (en) 2002-02-20 2011-08-30 Mcswiggen James RNA interference mediated inhibition of TNF and TNF receptor gene expression using short interfering nucleic acid (siNA)
US8008472B2 (en) 2001-05-29 2011-08-30 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of human immunodeficiency virus (HIV) gene expression using short interfering nucleic acid (siNA)
WO2011104153A1 (en) 2010-02-23 2011-09-01 Basf Plant Science Company Gmbh Nematode-resistant transgenic plants
US8013143B2 (en) 2002-02-20 2011-09-06 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of CXCR4 gene expression using short interfering nucleic acid (siNA)
US8017761B2 (en) 2001-05-18 2011-09-13 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of Stearoyl-CoA desaturase (SCD) gene expression using short interfering nucelic acid (siNA)
EP2221377B1 (en) 2002-02-01 2011-10-19 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US8067575B2 (en) 2002-02-20 2011-11-29 Merck, Sharp & Dohme Corp. RNA interference mediated inhibition of cyclin D1 gene expression using short interfering nucleic acid (siNA)
WO2012003207A3 (en) * 2010-07-01 2012-02-16 E. I. Du Pont De Nemours And Company Plant seeds with altered storage compound levels, related constructs and methods involving genes encoding pae and pae-like polypeptides
WO2012079000A1 (en) 2010-12-09 2012-06-14 The Trustees Of The University Of Pennsylvania Use of chimeric antigen receptor-modified t cells to treat cancer
US8221740B2 (en) 2005-09-26 2012-07-17 The Trustees Of Columbia University In The City Of New York Side population cells in cardiac repair
US8258288B2 (en) 2002-02-20 2012-09-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of respiratory syncytial virus (RSV) expression using short interfering nucleic acid (siNA)
EP2500429A2 (en) 2005-05-31 2012-09-19 Devgen N.V. RNAi for the control of insects and arachnids
US8324369B2 (en) 2007-11-30 2012-12-04 Baylor College Of Medicine Dendritic cell vaccine compositions and uses of same
WO2013050410A1 (en) 2011-10-04 2013-04-11 Bayer Intellectual Property Gmbh RNAi FOR THE CONTROL OF FUNGI AND OOMYCETES BY INHIBITING SACCHAROPINE DEHYDROGENASE GENE
WO2013055420A2 (en) 2011-07-12 2013-04-18 Philadelphia Health & Education Corporation Novel clostridium difficile dna vaccine
WO2013067492A1 (en) 2011-11-03 2013-05-10 The Trustees Of The University Of Pennsylvania Isolated b7-h4 specific compositions and methods of use thereof
WO2013078230A1 (en) 2011-11-23 2013-05-30 The Trustees Of The University Of Pennsylvania Use of pdl1 expressing cells to convert t cells into regulatory t cells
WO2013126729A1 (en) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Use of the cd2 signaling domain in second-generation chimeric antigen receptors
WO2013126733A1 (en) 2012-02-22 2013-08-29 The Trustees Of University Of Pennsylvania Use of icos-based cars to enhance antitumor activity and car persistence
WO2014053395A1 (en) 2012-10-01 2014-04-10 Basf Se Use of n-thio-anthranilamide compounds on cultivated plants
WO2014090765A1 (en) 2012-12-12 2014-06-19 Bayer Cropscience Ag Use of 1-[2-fluoro-4-methyl-5-(2,2,2-trifluoroethylsulfinyl)phenyl]-5-amino-3-trifluoromethyl)-1 h-1,2,4 tfia zole for controlling nematodes in nematode-resistant crops
WO2014130635A1 (en) 2013-02-20 2014-08-28 Novartis Ag Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
WO2014130657A1 (en) 2013-02-20 2014-08-28 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
WO2014153270A1 (en) 2013-03-16 2014-09-25 Novartis Ag Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
US8865968B2 (en) 2004-10-04 2014-10-21 Devgen Nv Method for down-regulating gene expression in fungi
US8957044B2 (en) 2013-03-01 2015-02-17 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
US8999654B2 (en) 2002-09-09 2015-04-07 Omeros Corporation Method of identifying a compound for the treatment or prevention of obesity
US9044461B2 (en) 2006-04-07 2015-06-02 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
WO2015090230A1 (en) 2013-12-19 2015-06-25 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
WO2015116178A1 (en) 2014-01-31 2015-08-06 Thomas Jefferson University Fusion proteins for modulating regulatory and effector t cells
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015157252A1 (en) 2014-04-07 2015-10-15 BROGDON, Jennifer Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2015168613A2 (en) 2014-05-02 2015-11-05 The Trustees Of The University Of Pennsylvania Compositions and methods of chimeric autoantibody receptor t cells
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
WO2016011432A2 (en) 2014-07-17 2016-01-21 Czerniecki Brian J Identification of immunogenic mhc class ii peptides for immune-based therapy
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
US9267145B2 (en) 2002-05-03 2016-02-23 Duke University Method of regulating gene expression
US9272002B2 (en) 2011-10-28 2016-03-01 The Trustees Of The University Of Pennsylvania Fully human, anti-mesothelin specific chimeric immune receptor for redirected mesothelin-expressing cell targeting
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016057705A1 (en) 2014-10-08 2016-04-14 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
US9365641B2 (en) 2012-10-01 2016-06-14 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
WO2016091674A1 (en) 2014-12-12 2016-06-16 Basf Se Use of cyclaniliprole on cultivated plants
WO2016098078A2 (en) 2014-12-19 2016-06-23 Novartis Ag Dimerization switches and uses thereof
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
WO2016138491A1 (en) 2015-02-27 2016-09-01 Icell Gene Therapeutics Llc Chimeric antigen receptors (cars) targeting hematologic malignancies, compositions and methods of use thereof
WO2016140910A2 (en) 2015-03-04 2016-09-09 University Of Rochester Compositions and methods of using anti-mullerian hormone for treatment of infertility
US9446105B2 (en) 2013-03-15 2016-09-20 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor specific for folate receptor β
WO2016164308A1 (en) 2015-04-06 2016-10-13 Subdomain, Llc De novo binding domain containing polypeptides and uses thereof
WO2016164580A1 (en) 2015-04-07 2016-10-13 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2016162371A1 (en) 2015-04-07 2016-10-13 Basf Agrochemical Products B.V. Use of an insecticidal carboxamide compound against pests on cultivated plants
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2016170484A1 (en) 2015-04-21 2016-10-27 Novartis Ag Rna-guided gene editing system and uses thereof
WO2016174652A1 (en) 2015-04-30 2016-11-03 Technion Research & Development Foundation Limited Chimeric antigen receptors and methods of their use
WO2016178996A1 (en) 2015-05-01 2016-11-10 The Regents Of The University Of California Glycan-dependent immunotherapeutic molecules
WO2016187349A1 (en) 2015-05-18 2016-11-24 Tcr2, Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2016210293A1 (en) 2015-06-25 2016-12-29 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS OF USE THEREOF
WO2017009853A1 (en) 2015-07-16 2017-01-19 Yeda Research And Development Co. Ltd. Genetically modified anti-third party central memory t cells and use of same in immunotherapy
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
US9572836B2 (en) 2012-07-13 2017-02-21 The Trustees Of The University Of Pennsylvania Methods for assessing the suitability of transduced T cells for administration
WO2017040195A1 (en) 2015-08-28 2017-03-09 The Trustees Of The University Of Pennsylvania Methods and compositions for cells expressing a chimeric intracellular signaling molecule
WO2017040930A2 (en) 2015-09-03 2017-03-09 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
US9598489B2 (en) 2012-10-05 2017-03-21 The Trustees Of The Univeristy Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
WO2017070608A1 (en) 2015-10-23 2017-04-27 Eureka Therapeutics, Inc. Antibody/t-cell receptor chimeric constructs and uses thereof
WO2017075421A1 (en) 2015-10-29 2017-05-04 Temple University-Of The Commonwealth System Of Higher Education Modification of 3' terminal ends of nucleic acids by dna polymerase theta
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
WO2017088012A1 (en) 2015-11-27 2017-06-01 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
WO2017095823A1 (en) 2015-11-30 2017-06-08 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
WO2017112741A1 (en) 2015-12-22 2017-06-29 Novartis Ag Mesothelin chimeric antigen receptor (car) and antibody against pd-l1 inhibitor for combined use in anticancer therapy
WO2017117112A1 (en) 2015-12-28 2017-07-06 Novartis Ag Methods of making chimeric antigen receptor -expressing cells
WO2017118726A1 (en) 2016-01-06 2017-07-13 Lonza Ltd Inhibition of protein degradation for improved production
US9708384B2 (en) 2011-09-22 2017-07-18 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US9765156B2 (en) 2012-07-13 2017-09-19 The Trustees Of The University Of Pennsylvania Enhancing activity of CAR T cells by co-introducing a bispecific antibody
WO2017172981A2 (en) 2016-03-29 2017-10-05 University Of Southern California Chimeric antigen receptors targeting cancer
US9790267B2 (en) 2011-11-08 2017-10-17 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
WO2017181101A1 (en) 2016-04-15 2017-10-19 The Trustees Of The University Of Pennsylvania Compositions and methods of chimeric alloantigen receptor t cells
WO2017178526A1 (en) 2016-04-14 2017-10-19 Lonza Ltd Compositions and methods for the detection of host cell proteins
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
WO2017190001A1 (en) 2016-04-29 2017-11-02 The Regents Of The University Of Colorado, A Body Corporate Compounds and compositions useful for treating metabolic syndrome, and methods using same
US9808504B2 (en) 2009-09-22 2017-11-07 Yale University Immunogenic epitopes as targets for universal cancer vaccines
WO2017191165A1 (en) 2016-05-03 2017-11-09 Lonza Ltd Modulation of lipid metabolism for protein production
US9834612B2 (en) 2011-07-22 2017-12-05 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
US9834610B2 (en) 2013-01-31 2017-12-05 Thomas Jefferson University Fusion proteins for modulating regulatory and effector T cells
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2017214333A1 (en) 2016-06-08 2017-12-14 Intrexon Corporation Cd33 specific chimeric antigen receptors
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018014122A1 (en) 2016-07-18 2018-01-25 Helix Biopharma Corp. Car immune cells directed to carcinoembryonic antigen related cell adhesion molecule 6 to treat cancer
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026953A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018026956A1 (en) 2016-08-02 2018-02-08 Lonza Ltd Method and system for providing buffer solutions
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
WO2018031858A1 (en) 2016-08-12 2018-02-15 Lonza Ltd Proteomic analysis of host cell proteins
WO2018053542A1 (en) 2016-09-19 2018-03-22 University Of Southern California Non-radioactive cytotoxicity assays
US9937205B2 (en) 2012-09-04 2018-04-10 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
WO2018067697A1 (en) 2016-10-04 2018-04-12 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
WO2018067993A1 (en) 2016-10-07 2018-04-12 TCR2 Therapeutics Inc. Compositions and methods for t-cell receptors reprogramming using fusion proteins
US9944713B2 (en) 2004-11-24 2018-04-17 Medicinal Bioconvergence Research Center Antibody specific to the AIMP2-DX2
WO2018083606A1 (en) 2016-11-01 2018-05-11 Novartis Ag Methods and compositions for enhancing gene editing
WO2018098365A2 (en) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
WO2018119298A1 (en) 2016-12-21 2018-06-28 TCR2 Therapeutics Inc. Engineered t cells for the treatment of cancer
US10011658B2 (en) 2015-04-03 2018-07-03 Eureka Therapeutics, Inc. Constructs targeting AFP peptide/MHC complexes and uses thereof
EP3346001A1 (en) 2017-01-06 2018-07-11 TXCell Monospecific regulatory t cell population with cytotoxicity for b cells
WO2018127585A1 (en) 2017-01-06 2018-07-12 Txcell Monospecific regulatory t cell population with cytotoxicity for b cells
WO2018132390A1 (en) 2017-01-10 2018-07-19 Christiana Care Health Services, Inc. Methods for in vitro site-directed mutagenesis using gene editing technologies
WO2018132494A1 (en) 2017-01-10 2018-07-19 Intrexon Corporation Modulating expression of polypeptides via new gene switch expression systems
WO2018134824A1 (en) 2017-01-18 2018-07-26 Yeda Research And Development Co. Ltd. Genetically modified veto cells and use of same in immunotherapy
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
US10040846B2 (en) 2012-02-22 2018-08-07 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
WO2018145649A1 (en) 2017-02-08 2018-08-16 西比曼生物科技(上海)有限公司 Construction of chimeric antigen receptor targeting cd20 antigen and activity identification of engineered t cells thereof
WO2018157072A1 (en) 2017-02-27 2018-08-30 Life Technologies Corporation Expansion of populations of t cells by the use of modified serum free media
US10066023B2 (en) 2015-10-30 2018-09-04 Aleta Biotherapeutics Inc. Compositions and methods for tumor transduction
US10106793B2 (en) 2002-02-01 2018-10-23 Life Technologies Corporation Double-stranded oligonucleotides
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018200583A1 (en) 2017-04-26 2018-11-01 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
WO2018208837A1 (en) 2017-05-08 2018-11-15 Precision Biosciences, Inc. Nucleic acid molecules encoding an engineered antigen receptor and an inhibitory nucleic acid molecule and methods of use thereof
WO2018232265A1 (en) 2017-06-16 2018-12-20 Lonza Ltd Universal self-regulating mammalian cell line platform for the production of biologics
WO2018232020A1 (en) 2017-06-13 2018-12-20 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2019007991A1 (en) 2017-07-03 2019-01-10 Universite De Strasbourg Mtmr2-s polypeptide for use in the treatment of myopathies
WO2019047899A1 (en) 2017-09-06 2019-03-14 亘喜生物科技(上海)有限公司 Universal chimeric antigen receptor t-cell preparation technique
WO2019055853A1 (en) 2017-09-15 2019-03-21 Life Technologies Corporation Compositions and methods for culturing and expanding cells
WO2019062817A1 (en) 2017-09-27 2019-04-04 亘喜生物科技(上海)有限公司 Engineered immune cell capable of inducing secretion of anti-cd47 antibody
WO2019070161A2 (en) 2017-10-04 2019-04-11 Opko Pharmaceuticals, Llc Articles and methods directed to personalized therapy of cancer
WO2019070856A1 (en) 2017-10-03 2019-04-11 Precision Biosciences, Inc. Modified epidermal growth factor receptor peptides for use in genetically-modified cells
WO2019069125A1 (en) 2017-10-06 2019-04-11 Oslo Universitetssykehus Hf Chimeric antigen receptors
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019092251A1 (en) 2017-11-11 2019-05-16 Universite De Strasbourg Compositions and method for the treatment of x-linked centronuclear myopathy
WO2019099639A1 (en) 2017-11-15 2019-05-23 Navartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
WO2019099440A1 (en) 2017-11-14 2019-05-23 Arcellx, Inc. Multifunctional immune cell therapies
WO2019099433A2 (en) 2017-11-14 2019-05-23 Arcellx, Inc. D-domain containing polypeptides and uses thereof
WO2019108900A1 (en) 2017-11-30 2019-06-06 Novartis Ag Bcma-targeting chimeric antigen receptor, and uses thereof
WO2019113050A1 (en) 2017-12-05 2019-06-13 Lonza Ltd Methods of assaying tropolone
WO2019129851A1 (en) 2017-12-29 2019-07-04 Cellectis Method for improving production of car t cells
WO2019136432A1 (en) 2018-01-08 2019-07-11 Novartis Ag Immune-enhancing rnas for combination with chimeric antigen receptor therapy
WO2019145711A1 (en) 2018-01-23 2019-08-01 Immetacyte Limited Biomarker predictive of tumour infiltrating lymphocyte therapy and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
WO2019152876A2 (en) 2018-02-02 2019-08-08 Lonza Ltd Methods of cell selection and modifying cell metabolism
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
EP3549611A1 (en) 2011-07-29 2019-10-09 The Trustees of the University of Pennsylvania Switch costimulatory receptors
WO2019196713A1 (en) 2018-04-12 2019-10-17 西比曼生物科技(香港)有限公司 Bcma-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019213180A1 (en) 2018-04-30 2019-11-07 Amicus Therapeutics, Inc. Gene therapy constructs and methods of use
WO2019217512A1 (en) 2018-05-08 2019-11-14 Life Technologies Corporation Compositions and methods for culturing and expanding cells
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
US10508143B1 (en) 2015-10-30 2019-12-17 Aleta Biotherapeutics Inc. Compositions and methods for treatment of cancer
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
WO2019241549A1 (en) 2018-06-15 2019-12-19 A2 Biotherapeutics, Inc. Foxp3-expressing car-t regulatory cells
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019243835A1 (en) 2018-06-21 2019-12-26 Immetacyte Limited Chimeric growth factor receptors
WO2020030979A2 (en) 2018-08-10 2020-02-13 Sangamo Therapeutics France New car constructs comprising tnfr2 domains
WO2020047501A1 (en) 2018-08-30 2020-03-05 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020043899A1 (en) 2018-08-31 2020-03-05 Invectys Chimeric antigen receptors against multiple hla-g isoforms
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
WO2020077114A2 (en) 2018-10-10 2020-04-16 Amicus Therapeutics, Inc. Disulfide bond stabilized polypeptide compositions and methods of use
WO2020081929A1 (en) 2018-10-19 2020-04-23 University Of Rochester Immune modulators in combination with radiation treatment for advanced pancreatic cancer
WO2020086627A1 (en) 2018-10-22 2020-04-30 University Of Rochester Genome editing by directed non-homologous dna insertion using a retroviral integrase-cas9 fusion protein
WO2020092455A2 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Car t cell transcriptional atlas
WO2020102676A1 (en) 2018-11-16 2020-05-22 Celgene Corporation Improved t cell manufacturing process
WO2020102555A1 (en) 2018-11-16 2020-05-22 Memorial Sloan Kettering Cancer Center Antibodies to mucin-16 and methods of use thereof
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
US10689873B2 (en) 2016-03-10 2020-06-23 Lonza Ltd Customizable facility
WO2020140007A1 (en) 2018-12-28 2020-07-02 University Of Rochester Gene therapy for best1 dominant mutations
WO2020150287A1 (en) 2019-01-14 2020-07-23 University Of Rochester Targeted nuclear rna cleavage and polyadenylation with crispr-cas
WO2020152451A1 (en) 2019-01-22 2020-07-30 Immetacyte Limited Receptors providing targeted costimulation for adoptive cell therapy
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US10738099B2 (en) 2015-09-22 2020-08-11 The Trustees Of The University Of Pennsylvania Method of redirecting T cells to treat HIV infection
WO2020161186A1 (en) 2019-02-08 2020-08-13 Biontech Cell & Gene Therapies Gmbh Chimeric antigen receptor-modified cells for the treatment of cldn6-expressing cancers
EP3699284A1 (en) 2012-07-05 2020-08-26 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020185121A2 (en) 2019-03-13 2020-09-17 Общество С Ограниченной Ответственностью "Анабион" Isolated alternative intracellular signalling domain of a chimeric antigen receptor and chimeric antigen receptor comprising said signalling domain
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
WO2020188103A1 (en) 2019-03-20 2020-09-24 Centre National De La Recherche Scientifique Amphiphysin / bin1 for the treatment of autosomal dominant centronuclear myopathy
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
US10808031B2 (en) 2015-01-21 2020-10-20 Cancer Research Technology Limited Inhibitors of the interaction between CLEC14A and multimerin-2 for inhibition of angiogenesis
US10815311B2 (en) 2018-09-25 2020-10-27 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
WO2020224606A1 (en) 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Engineered immune cell targeting bcma and use thereof
US10844134B2 (en) 2016-11-23 2020-11-24 Harpoon Therapeutics, Inc. PSMA targeting trispecific proteins and methods of use
US10849973B2 (en) 2016-11-23 2020-12-01 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
WO2021003442A1 (en) 2019-07-02 2021-01-07 M6P Therapeutics Vector compositions and methods of using same for treatment of lysosomal storage disorders
WO2021016453A1 (en) 2019-07-23 2021-01-28 University Of Rochester Targeted rna cleavage with crispr-cas
WO2021016608A1 (en) 2019-07-25 2021-01-28 Precision Biosciences, Inc. Compositions and methods for sequential stacking of nucleic acid sequences into a genomic locus
WO2021018311A1 (en) 2019-08-01 2021-02-04 上海赛比曼生物科技有限公司 Universal car-t cell and preparation and use thereof
WO2021030182A1 (en) 2019-08-09 2021-02-18 A2 Biotherapeutics, Inc. Bifunctional single variable domain t cell receptors and uses thereof
US10927180B2 (en) 2017-10-13 2021-02-23 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
WO2021035054A1 (en) 2019-08-20 2021-02-25 Precision Biosciences, Inc. Lymphodepletion dosing regimens for cellular immunotherapies
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2021032779A1 (en) 2019-08-19 2021-02-25 Universität Basel Cell therapy methods
US10934337B2 (en) 2019-03-15 2021-03-02 Cartesian Therapeutics, Inc. Anti-BCMA chimeric antigen receptors
US10934336B2 (en) 2017-04-13 2021-03-02 The Trustees Of The University Of Pennsylvania Use of gene editing to generate universal TCR re-directed T cells for adoptive immunotherapy
US10954311B2 (en) 2015-05-21 2021-03-23 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
EP3808766A1 (en) 2019-10-15 2021-04-21 Sangamo Therapeutics France Chimeric antigen receptor specific for interleukin-23 receptor
EP3811970A1 (en) 2014-03-15 2021-04-28 Novartis AG Regulatable chimeric antigen receptor
WO2021096868A1 (en) 2019-11-12 2021-05-20 A2 Biotherapeutics, Inc. Engineered t cell receptors and uses thereof
WO2021094461A1 (en) 2019-11-14 2021-05-20 Lonza Ltd Methods of cell selection
WO2021098882A1 (en) 2019-11-21 2021-05-27 博生吉医药科技(苏州)有限公司 Cd7-car-t cell and preparation and application thereof
WO2021108613A1 (en) 2019-11-26 2021-06-03 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021119489A1 (en) 2019-12-11 2021-06-17 A2 Biotherapeutics, Inc. Lilrb1-based chimeric antigen receptor
WO2021158964A1 (en) 2020-02-07 2021-08-12 University Of Rochester Ribozyme-mediated rna assembly and expression
WO2021158982A2 (en) 2020-02-07 2021-08-12 University Of Rochester Targeted translation of rna with crispr-cas13 to enhance protein synthesis
US11090336B2 (en) 2019-03-27 2021-08-17 The Trustees Of The University Of Pennsylvania Tn-MUC1 chimeric antigen receptor (CAR) T cell therapy
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
WO2021170071A1 (en) 2020-02-28 2021-09-02 Shanghai Henlius Biotech, Inc. Anti-cd137 constructs, multispecific antibody and uses thereof
WO2021170067A1 (en) 2020-02-28 2021-09-02 上海复宏汉霖生物技术股份有限公司 Anti-cd137 construct and use thereof
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173674A1 (en) 2020-02-26 2021-09-02 A2 Biotherapeutics, Inc. Polypeptides targeting mage-a3 peptide-mhc complexes and methods of use thereof
WO2021186246A1 (en) 2020-03-19 2021-09-23 Trizell Ltd. Temperature-responsive virus storage system
WO2021186056A1 (en) 2020-03-20 2021-09-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Chimeric antigen receptor specific for human cd45rc and uses thereof
US11130783B2 (en) 2018-11-13 2021-09-28 Regents Of The University Of Minnesota CD40 targeted peptides and uses thereof
WO2021195519A1 (en) 2020-03-27 2021-09-30 University Of Rochester Targeted destruction of viral rna by crispr-cas13
WO2021195525A1 (en) 2020-03-27 2021-09-30 University Of Rochester Crispr-cas13 crrna arrays
US11136403B2 (en) 2017-10-13 2021-10-05 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
WO2021208750A1 (en) 2020-04-16 2021-10-21 上海赛比曼生物科技有限公司 Cd22-targeted chimeric antigen receptor, preparation method therefor and application thereof
WO2021222576A1 (en) 2020-05-01 2021-11-04 A2 Biotherapeutics, Inc. Pag1 fusion proteins and methods of making and using same
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021238886A1 (en) 2020-05-27 2021-12-02 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Antibodies specifically recognizing nerve growth factor and uses thereof
WO2021252635A1 (en) 2020-06-11 2021-12-16 A2 Biotherapeutics, Inc. Compositions and methods for treating cancers
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
US11219675B2 (en) 2015-03-13 2022-01-11 Syz Cell Therapy Co. Methods of cancer treatment using activated T cells
US11219676B2 (en) 2018-04-13 2022-01-11 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific T cells
WO2022011651A1 (en) 2020-07-16 2022-01-20 上海交通大学 Immunotherapy method of targeted chemokine and cytokine delivery by mesenchymal stem cell
WO2022016112A1 (en) 2020-07-17 2022-01-20 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
WO2022016114A1 (en) 2020-07-17 2022-01-20 Instill Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2022015916A1 (en) 2020-07-15 2022-01-20 University Of Rochester Targeted rna cleavage with dcasl3-rnase fusion proteins
US11236520B2 (en) 2016-03-10 2022-02-01 Lonza Ltd Customizable facility
US11248208B2 (en) 2018-03-30 2022-02-15 Syz Cell Therapy Co. Multiple antigen specific cell therapy methods
WO2022036068A1 (en) 2020-08-13 2022-02-17 A2 Biotherapeutics, Inc. Gene fusions for control of genetically modified cells
US11274157B2 (en) 2017-01-12 2022-03-15 Eureka Therapeutics, Inc. Constructs targeting histone H3 peptide/MHC complexes and uses thereof
WO2022079308A1 (en) 2020-10-16 2022-04-21 Institut Pasteur Chimeric constructs useful in vaccination and cancer therapy
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022105893A1 (en) 2020-11-23 2022-05-27 博生吉医药科技(苏州)有限公司 Preparation method and application of cd7-car-t cells
WO2022123316A1 (en) 2020-12-09 2022-06-16 Takeda Pharmaceutical Company Limited Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
WO2022123307A1 (en) 2020-12-09 2022-06-16 Takeda Pharmaceutical Company Limited Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
WO2022147463A2 (en) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
US11390659B2 (en) 2018-04-13 2022-07-19 Syz Cell Therapy Co. Methods of obtaining tumor-specific T cell receptors
WO2022155421A1 (en) 2021-01-15 2022-07-21 University Of Rochester Staphylococcus aureus antigen-based nucleic acid vaccines
WO2022151960A1 (en) 2021-01-13 2022-07-21 博生吉医药科技(苏州)有限公司 B7-h3 chimeric antigen receptor-modified t cell and use thereof
US11396552B2 (en) 2018-02-12 2022-07-26 Diabetes-Free Inc. Antagonistic anti-human CD40 monoclonal antibodies
EP4036109A2 (en) 2014-12-29 2022-08-03 Novartis AG Methods of making chimeric antigen receptor-expressing cells
WO2022162192A2 (en) 2021-01-29 2022-08-04 Thelper As Therapeutic and diagnostic agents and uses thereof
WO2022165111A1 (en) 2021-01-28 2022-08-04 Precision Biosciences, Inc. Modulation of tgf beta signaling in genetically-modified eukaryotic cells
WO2022166665A1 (en) 2021-02-08 2022-08-11 浙江大学 Chimeric antigen receptor with endogenous protein molecule replacing single domain antibody
US11433100B2 (en) 2020-08-20 2022-09-06 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
US11440958B2 (en) 2016-11-22 2022-09-13 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
US11441146B2 (en) 2016-01-11 2022-09-13 Christiana Care Health Services, Inc. Compositions and methods for improving homogeneity of DNA generated using a CRISPR/Cas9 cleavage system
US11447564B2 (en) 2017-04-26 2022-09-20 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
US11453716B2 (en) 2016-05-20 2022-09-27 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
WO2022226296A2 (en) 2021-04-23 2022-10-27 University Of Rochester Genome editing by directed non-homologous dna insertion using a retroviral integrase-cas fusion protein and methods of treatment
US11485782B2 (en) 2018-03-14 2022-11-01 Beijing Xuanyi Pharmasciences Co., Ltd. Anti-claudin 18.2 antibodies
WO2022254337A1 (en) 2021-06-01 2022-12-08 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
WO2022262764A1 (en) 2021-06-16 2022-12-22 四川大学华西医院 Preparation and application of lox1-based chimeric antigen receptor immune cell
WO2022266660A1 (en) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
WO2022262765A1 (en) 2021-06-16 2022-12-22 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cell constructed on basis of granzyme b
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
WO2022271955A1 (en) 2021-06-23 2022-12-29 Musc Foundation For Research Development Novel targeted shrna nanoparticles for cancer therapy
WO2023278641A1 (en) 2021-06-29 2023-01-05 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
WO2023283611A1 (en) 2021-07-08 2023-01-12 Staidson Biopharma Inc. Antibodies specifically recognizing tnfr2 and uses thereof
WO2023284714A1 (en) 2021-07-14 2023-01-19 舒泰神(北京)生物制药股份有限公司 Antibody that specifically recognizes cd40 and application thereof
US11602544B2 (en) 2020-08-20 2023-03-14 A2 Biotherapeutics, Inc. Compositions and methods for treating EGFR positive cancers
US11602543B2 (en) 2020-08-20 2023-03-14 A2 Biotherapeutics, Inc. Compositions and methods for treating mesothelin positive cancers
US11607453B2 (en) 2017-05-12 2023-03-21 Harpoon Therapeutics, Inc. Mesothelin binding proteins
WO2023056329A1 (en) 2021-09-30 2023-04-06 Akouos, Inc. Compositions and methods for treating kcnq4-associated hearing loss
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2023069979A1 (en) 2021-10-20 2023-04-27 University Of Rochester Isolated glial progenitor cells for use in the competition treatment of age-related white matter loss
WO2023081633A1 (en) 2021-11-02 2023-05-11 University Of Rochester Tcf7l2 mediated remyelination in the brain
WO2023086939A1 (en) 2021-11-12 2023-05-19 Amicus Therapeutics, Inc. Compositions and methods for treating mucopolysaccharidosis iiia
WO2023089556A1 (en) 2021-11-22 2023-05-25 Pfizer Inc. Reducing risk of antigen mimicry in immunogenic medicaments
WO2023105005A1 (en) 2021-12-09 2023-06-15 BioNTech SE Chimeric antigen receptor-modified cells for the treatment of cldn6 expressing cancer
US11679132B2 (en) 2015-02-06 2023-06-20 National University Of Singapore Methods for enhancing efficacy of therapeutic immune cells
US11697677B2 (en) 2021-07-16 2023-07-11 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023144779A1 (en) 2022-01-28 2023-08-03 Pfizer Inc. Coronavirus antigen variants
WO2023144392A1 (en) 2022-01-28 2023-08-03 Thelper As Therapeutic and diagnostic agents and uses thereof
US11730764B2 (en) 2021-02-16 2023-08-22 A2 Biotherapeutics, Inc. Compositions and methods for treating HER2 positive cancers
WO2023177954A1 (en) 2022-03-18 2023-09-21 University Of Rochester Combination therapy for treatment of cancer, methods and systems of delivery thereof
WO2023215860A1 (en) 2022-05-05 2023-11-09 University Of Rochester Modified bacteria and methods of use for bioglass microlenses
US11814432B2 (en) 2017-09-20 2023-11-14 The University Of British Columbia Anti-HLA-A2 antibodies, related chimeric antigen receptors, and uses thereof
WO2024020407A1 (en) 2022-07-19 2024-01-25 Staidson Biopharma Inc. Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024046572A1 (en) 2022-09-01 2024-03-07 BioNTech SE Chimeric antigen receptor-modified cells for the treatment of cldn6 expressing cancer
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy
US11945876B2 (en) 2021-06-16 2024-04-02 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8202979B2 (en) 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017054A1 (en) * 1991-03-26 1992-10-15 Mogen International N.V. Method for the isolation and/or testing of genes and promoters involved in plant-nematode interactions using plants of the genus arabidopsis
WO1993010251A1 (en) * 1991-11-20 1993-05-27 Mogen International N.V. A method for obtaining plants with reduced susceptibility to plant-parasitic nematodes
WO1999032619A1 (en) * 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Genetic inhibition by double-stranded rna
WO2000001846A2 (en) * 1998-07-03 2000-01-13 Devgen N.V. Characterisation of gene function using double stranded rna inhibition
WO2001037654A2 (en) * 1999-11-24 2001-05-31 Dna Plant Technology Corporation METHOD OF EXPRESSING dsRNA IN PLANTS TO INHIBIT INSECT PESTS

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017054A1 (en) * 1991-03-26 1992-10-15 Mogen International N.V. Method for the isolation and/or testing of genes and promoters involved in plant-nematode interactions using plants of the genus arabidopsis
WO1993010251A1 (en) * 1991-11-20 1993-05-27 Mogen International N.V. A method for obtaining plants with reduced susceptibility to plant-parasitic nematodes
WO1999032619A1 (en) * 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Genetic inhibition by double-stranded rna
WO2000001846A2 (en) * 1998-07-03 2000-01-13 Devgen N.V. Characterisation of gene function using double stranded rna inhibition
WO2001037654A2 (en) * 1999-11-24 2001-05-31 Dna Plant Technology Corporation METHOD OF EXPRESSING dsRNA IN PLANTS TO INHIBIT INSECT PESTS

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BASS BRENDA L: "Double-stranded RNA as a template for gene silencing." CELL, vol. 101, no. 3, 28 April 2000 (2000-04-28), pages 235-238, XP002194756 ISSN: 0092-8674 cited in the application *
NIEBEL ET AL: "induction of cdc2a and cyc1at expression in Arabidopsis thaliana during early phases of nematode-induced feeding cell formation" PLANT JOURNAL, BLACKWELL SCIENTIFIC PUBLICATIONS, OXFORD, GB, vol. 10, no. 6, 1996, pages 1037-1043, XP002086054 ISSN: 0960-7412 *
WANG YONGZENG ET AL: "Identification of a novel plant virus promoter using a potyvirus infectious clone." VIRUS GENES, vol. 20, no. 1, February 2000 (2000-02), pages 11-17, XP008002062 ISSN: 0920-8569 *

Cited By (532)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8017765B2 (en) 2000-08-30 2011-09-13 Merck Sharp & Dohme Corp. RNA interference mediated treatment of alzheimer's disease using short interfering nucleic acid (siNA)
US7662951B2 (en) 2000-08-30 2010-02-16 Sirna Therapeutics, Inc. RNA interference mediated treatment of Alzheimer's disease using short interfering nucleic acid (siNA)
US7022828B2 (en) 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US8017761B2 (en) 2001-05-18 2011-09-13 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of Stearoyl-CoA desaturase (SCD) gene expression using short interfering nucelic acid (siNA)
US7659389B2 (en) 2001-05-18 2010-02-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MYC and/or MYB gene expression using short interfering nucleic acid (siNA)
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
US7964578B2 (en) 2001-05-18 2011-06-21 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US8008472B2 (en) 2001-05-29 2011-08-30 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of human immunodeficiency virus (HIV) gene expression using short interfering nucleic acid (siNA)
US7422896B1 (en) 2001-12-27 2008-09-09 Allele Biotechnology & Pharmaceuticals, Inc. Compositions for DNA mediated gene silencing
US7294504B1 (en) 2001-12-27 2007-11-13 Allele Biotechnology & Pharmaceuticals, Inc. Methods and compositions for DNA mediated gene silencing
US7625750B2 (en) 2001-12-27 2009-12-01 Allele Biotechnology & Pharmaceuticals, Inc. Compositions for DNA mediated gene silencing
EP2221377B1 (en) 2002-02-01 2011-10-19 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10626398B2 (en) 2002-02-01 2020-04-21 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
EP1572902A4 (en) * 2002-02-01 2007-04-11 Ambion Inc HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
EP1572902A2 (en) * 2002-02-01 2005-09-14 Ambion, Inc. HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
US10196640B1 (en) 2002-02-01 2019-02-05 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10106793B2 (en) 2002-02-01 2018-10-23 Life Technologies Corporation Double-stranded oligonucleotides
US10036025B2 (en) 2002-02-01 2018-07-31 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9796978B1 (en) 2002-02-01 2017-10-24 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
EP2221377B2 (en) 2002-02-01 2017-05-17 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US7700760B2 (en) 2002-02-20 2010-04-20 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular cell adhesion molecule (VCAM) gene expression using short interfering nucleic acid (siNA)
US8076472B2 (en) 2002-02-20 2011-12-13 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA)
US10662428B2 (en) 2002-02-20 2020-05-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9732344B2 (en) 2002-02-20 2017-08-15 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9738899B2 (en) 2002-02-20 2017-08-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US10351852B2 (en) 2002-02-20 2019-07-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9771588B2 (en) 2002-02-20 2017-09-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
WO2003070886A2 (en) * 2002-02-20 2003-08-28 Sirna Therapeutics, Inc. Inhibition of protein tyrosine phosphatase type iva expression byshort interfering nucleic acid
US8258288B2 (en) 2002-02-20 2012-09-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of respiratory syncytial virus (RSV) expression using short interfering nucleic acid (siNA)
US8153778B2 (en) 2002-02-20 2012-04-10 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of vascular cell adhesion molecule (VCAM) gene expression using short interfering nucleic acid (siNA)
US7659390B2 (en) 2002-02-20 2010-02-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA)
US7662952B2 (en) 2002-02-20 2010-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of GRB2 associated binding protein (GAB2) gene expression using short interfering nucleic acid (siNA)
WO2003070886A3 (en) * 2002-02-20 2004-04-01 Sirna Therapeutics Inc Inhibition of protein tyrosine phosphatase type iva expression byshort interfering nucleic acid
US7667029B2 (en) 2002-02-20 2010-02-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of checkpoint kinase-1 (CHK-1) gene expression using short interfering nucleic acid (siNA)
US7667030B2 (en) 2002-02-20 2010-02-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of matrix metalloproteinase 13 (MMP13) gene expression using short interfering nucleic acid (siNA)
US7678897B2 (en) 2002-02-20 2010-03-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
US10889815B2 (en) 2002-02-20 2021-01-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7691999B2 (en) 2002-02-20 2010-04-06 Sirna Therapeutics, Inc. RNA interference mediated inhibition of NOGO and NOGO receptor gene expression using short interfering nucleic acid (siNA)
WO2003070918A3 (en) * 2002-02-20 2004-07-08 Ribozyme Pharm Inc Rna interference by modified short interfering nucleic acid
US8067575B2 (en) 2002-02-20 2011-11-29 Merck, Sharp & Dohme Corp. RNA interference mediated inhibition of cyclin D1 gene expression using short interfering nucleic acid (siNA)
US8013146B2 (en) 2002-02-20 2011-09-06 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of matrix metalloproteinase 13 (MMP13) gene expression using short interfering nucleic acid (siNA)
US8013143B2 (en) 2002-02-20 2011-09-06 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of CXCR4 gene expression using short interfering nucleic acid (siNA)
US8008473B2 (en) 2002-02-20 2011-08-30 Mcswiggen James RNA interference mediated inhibition of TNF and TNF receptor gene expression using short interfering nucleic acid (siNA)
US7795422B2 (en) 2002-02-20 2010-09-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
US7985853B2 (en) 2002-02-20 2011-07-26 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of platelet derived growth factor (PDGF) and platelet derived growth factor receptor (PDGFR) gene expression using short interfering nucleic acid (siNA)
US7977472B2 (en) 2002-02-20 2011-07-12 Leonid Beigelman RNA interference mediated inhibition of myostatin gene expression using short interfering nucleic acid (siNA)
US9957517B2 (en) 2002-02-20 2018-05-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7855284B2 (en) 2002-02-20 2010-12-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of checkpoint kinase-1 (CHK-1) gene expression using short interfering nucleic acid (siNA)
US7858771B2 (en) 2002-02-20 2010-12-28 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of muscarinic colinergic receptor gene expression using short interfering nucleic acid (siNA)
GB2397818B (en) * 2002-02-20 2005-03-09 Sirna Therapeutics Inc Rna interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
GB2397818A (en) * 2002-02-20 2004-08-04 Sirna Therapeutics Inc Rna interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
US7956178B2 (en) 2002-02-20 2011-06-07 Mcswiggen James RNA interference mediated inhibition of GRB2 associated binding protein (GAB2) gene expression using short interfering nucleic acid (siNA)
US10000754B2 (en) 2002-02-20 2018-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7893248B2 (en) 2002-02-20 2011-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Myc and/or Myb gene expression using short interfering nucleic acid (siNA)
US7897752B2 (en) 2002-02-20 2011-03-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of telomerase gene expression using short interfering nucleic acid (siNA)
US7897755B2 (en) 2002-02-20 2011-03-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
US7897757B2 (en) 2002-02-20 2011-03-01 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of protein tyrosine phosphatase-1B (PTP-1B) gene expression using short interfering nucleic acid (siNA)
US7897756B2 (en) 2002-02-20 2011-03-01 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of NOGO and NOGO receptor gene expression using short interfering nucleic acid (siNA)
US7897753B2 (en) 2002-02-20 2011-03-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of XIAP gene expression using short interfering nucleic acid (siNA)
US7910724B2 (en) 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
US7910725B2 (en) 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US7915400B2 (en) 2002-02-20 2011-03-29 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of hepatitis C virus (HCV) gene expression using short interfering nucleic acid (siNA)
US7943757B2 (en) 2002-02-20 2011-05-17 Mcswiggen James RNA interference mediated inhibition of intercellular adhesion molecule (ICAM) gene expression using short interfering nucleic acid (siNA)
US7923549B2 (en) 2002-02-20 2011-04-12 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US7928220B2 (en) 2002-02-20 2011-04-19 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of stromal cell-derived factor-1 (SDF-1) gene expression using short interfering nucleic acid (siNA)
US7928219B2 (en) 2002-02-20 2011-04-19 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (SINA)
US7935812B2 (en) 2002-02-20 2011-05-03 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of hepatitis C virus (HCV) expression using short interfering nucleic acid (siNA)
WO2003093475A1 (en) * 2002-04-30 2003-11-13 Genesis Research And Development Corporation Limited Compositions and methods for the modification of gene expression_using the superubiquitin promoter from pinus radiata
US9267145B2 (en) 2002-05-03 2016-02-23 Duke University Method of regulating gene expression
US9856476B2 (en) 2002-05-03 2018-01-02 Duke University Method of regulating gene expression
US9850485B2 (en) 2002-05-03 2017-12-26 Duke University Method of regulating gene expression
US10233451B2 (en) 2002-05-03 2019-03-19 Duke University Method of regulating gene expression
US7803984B2 (en) * 2002-07-10 2010-09-28 Kansas State University Research Foundation Compositions and methods for controlling plant parasitic nematodes
US8999654B2 (en) 2002-09-09 2015-04-07 Omeros Corporation Method of identifying a compound for the treatment or prevention of obesity
US10292374B2 (en) 2002-09-09 2019-05-21 Omeros Corporation G protein coupled receptor 85 and SREB3 knockout mice and uses thereof
US8158420B2 (en) 2003-04-04 2012-04-17 The Trustees Of Columbia University In The City Of New York Methods for inhibiting the differentation of proliferative telencephalic cells in vitro by addition of ATF5
US7888326B2 (en) 2003-04-04 2011-02-15 The Trustees Of Columbia University In The City Of New York Methods for promoting apoptosis and treating tumor cells inhibiting the expression or function of the transcription factor ATF5
US9132167B2 (en) 2003-05-19 2015-09-15 The Trustees Of Columbia University In The City Of New York Compositions and methods for treating and preventing heart tissue degeneration and uses thereof
US7645734B2 (en) 2003-05-19 2010-01-12 The Trustees Of Columbia University In The City Of New York Compositions and methods for treating and preventing heart tissue degeneration and uses thereof
US8217157B2 (en) 2003-05-19 2012-07-10 The Trustees Of Columbia University In The City Of New York Compositions and methods for treating and preventing heart tissue degeneration, and uses thereof
WO2005071091A1 (en) * 2003-12-23 2005-08-04 Bayer Cropscience Sa Method for modifying gene expression of a phytopathogenic fungus
US7741531B2 (en) 2003-12-23 2010-06-22 Bayer S.A.S. Method for modifying gene expression of a phytopathogenic fungus
US7858769B2 (en) 2004-02-10 2010-12-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using multifunctional short interfering nucleic acid (multifunctional siNA)
US7622301B2 (en) 2004-02-24 2009-11-24 Basf Plant Science Gmbh Compositions and methods using RNA interference for control of nematodes
EP2163560A2 (en) * 2004-02-24 2010-03-17 BASF Plant Science GmbH Compositions and methods using RNA interference for control of nematodes in plants
WO2005082932A2 (en) * 2004-02-24 2005-09-09 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes in plants
EP2163560A3 (en) * 2004-02-24 2010-06-02 BASF Plant Science GmbH Compositions and methods using RNA interference for control of nematodes in plants
WO2005082932A3 (en) * 2004-02-24 2005-11-10 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes in plants
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US7868158B2 (en) 2004-07-19 2011-01-11 Baylor College Of Medicine Modulation of cytokine signaling regulators and applications for immunotherapy
WO2006020821A2 (en) 2004-08-13 2006-02-23 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes
WO2006020821A3 (en) * 2004-08-13 2006-05-18 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes
US7659444B2 (en) 2004-08-13 2010-02-09 Basf Plant Science Gmbh Compositions and methods using RNA interference for control of nematodes
US8865968B2 (en) 2004-10-04 2014-10-21 Devgen Nv Method for down-regulating gene expression in fungi
US7915479B2 (en) 2004-10-13 2011-03-29 University Of Georgia Research Foundation, Inc. Nematode resistant transgenic plants
US7576261B2 (en) 2004-10-13 2009-08-18 University Of Georgia Research Foundation, Inc. Nematode resistant transgenic plants
WO2006047495A2 (en) 2004-10-21 2006-05-04 Venganza Inc Methods and materials for conferring resistance to pests and pathogens of plants
US9121034B2 (en) 2004-10-21 2015-09-01 Venganza Inc Methods and materials for conferring resistance to pests and pathogens of corn
US8461416B2 (en) 2004-10-21 2013-06-11 Venganza, Inc. Methods and materials for conferring resistance to pests and pathogens of plants
WO2006047495A3 (en) * 2004-10-21 2006-08-24 Charles L Niblett Methods and materials for conferring resistance to pests and pathogens of plants
EP2336333A1 (en) * 2004-10-21 2011-06-22 Venganza Inc. Methods and materials for conferring resistance to pests and pathogens of plants
JP2008517604A (en) * 2004-10-21 2008-05-29 ヴェンガンザ インコーポレイテッド Methods and materials for conferring resistance to plant pests and pathogens
US8581039B2 (en) 2004-10-21 2013-11-12 Venganza, Inc. Methods and materials for conferring resistance to pests and pathogens of plants
US8148604B2 (en) 2004-10-21 2012-04-03 Venganza Inc. Methods and materials for conferring resistance to pests and pathogens of plants
US9944713B2 (en) 2004-11-24 2018-04-17 Medicinal Bioconvergence Research Center Antibody specific to the AIMP2-DX2
EP2500429A2 (en) 2005-05-31 2012-09-19 Devgen N.V. RNAi for the control of insects and arachnids
US8759306B2 (en) 2005-05-31 2014-06-24 Devgen N.V. RNAi for the control of insects and arachnids
US9290764B2 (en) 2005-05-31 2016-03-22 Devgen Nv RNAi for the control of insects and arachnids
US7868159B2 (en) 2005-06-23 2011-01-11 Baylor College Of Medicine Modulation of negative immune regulators and applications for immunotherapy
US8221740B2 (en) 2005-09-26 2012-07-17 The Trustees Of Columbia University In The City Of New York Side population cells in cardiac repair
JP2009523017A (en) * 2006-01-12 2009-06-18 デブジェン エヌブイ Genetically modified plant system method for plant pests using RNAi
JP2009523018A (en) * 2006-01-12 2009-06-18 デブジェン エヌブイ Method for controlling pests using RNAi
WO2007104570A2 (en) * 2006-03-16 2007-09-20 Devgen N.V. Nematode control
WO2007104570A3 (en) * 2006-03-16 2008-04-17 Devgen Nv Nematode control
WO2007117657A2 (en) 2006-04-07 2007-10-18 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
US9044461B2 (en) 2006-04-07 2015-06-02 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
US8524454B2 (en) 2006-04-07 2013-09-03 The Research Foundation Of State University Of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
WO2008095886A1 (en) 2007-02-06 2008-08-14 Basf Plant Science Gmbh Compositions and methods using rna interference for control of nematodes
WO2008095910A1 (en) 2007-02-08 2008-08-14 Basf Plant Science Gmbh Compositions and methods using rna interference of opr3-like gene for control of nematodes
WO2008095970A1 (en) 2007-02-09 2008-08-14 Basf Plant Science Gmbh Compositions and methods using rna interference of cdpk-like for control of nematodes
WO2008118324A2 (en) 2007-03-26 2008-10-02 Macrogenics, Inc. Composition and method of treating cancer with an anti-uroplakin ib antibody
US8324369B2 (en) 2007-11-30 2012-12-04 Baylor College Of Medicine Dendritic cell vaccine compositions and uses of same
WO2010046422A2 (en) 2008-10-22 2010-04-29 Basf Se Use of auxin type herbicides on cultivated plants
WO2010046423A2 (en) 2008-10-22 2010-04-29 Basf Se Use of sulfonylurea herbicides on cultivated plants
WO2010106163A1 (en) 2009-03-20 2010-09-23 Basf Plant Science Company Gmbh Nematode-resistant transgenic plants
DE112010001772T5 (en) 2009-03-20 2012-10-18 Basf Plant Science Company Gmbh NEMATODRESISTENT TRANSGENIC PLANTS
US9808504B2 (en) 2009-09-22 2017-11-07 Yale University Immunogenic epitopes as targets for universal cancer vaccines
WO2011062962A2 (en) 2009-11-17 2011-05-26 The Trustees Of The University Of Pennsylvania Smndelta7 degron: novel compositions and methods of use
WO2011104153A1 (en) 2010-02-23 2011-09-01 Basf Plant Science Company Gmbh Nematode-resistant transgenic plants
US9574204B2 (en) 2010-07-01 2017-02-21 E I Dupont De Nemours And Company Plant seeds with altered storage compound levels, related constructs and methods involving genes encoding PAE and PAE-like polypeptides
WO2012003207A3 (en) * 2010-07-01 2012-02-16 E. I. Du Pont De Nemours And Company Plant seeds with altered storage compound levels, related constructs and methods involving genes encoding pae and pae-like polypeptides
US9970005B2 (en) 2010-10-29 2018-05-15 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US11932854B2 (en) 2010-10-29 2024-03-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US11193126B2 (en) 2010-10-29 2021-12-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
EP3214091A1 (en) 2010-12-09 2017-09-06 The Trustees of The University of Pennsylvania Use of chimeric antigen receptor-modified t cells to treat cancer
WO2012079000A1 (en) 2010-12-09 2012-06-14 The Trustees Of The University Of Pennsylvania Use of chimeric antigen receptor-modified t cells to treat cancer
EP3660029A1 (en) 2010-12-09 2020-06-03 The Trustees of The University of Pennsylvania Use of chimeric antigen receptor-modified t cells to treat cancer
EP3305798A1 (en) 2010-12-09 2018-04-11 The Trustees of The University of Pennsylvania Use of chimeric antigen receptor-modified t cells to treat cancer
US11180550B2 (en) 2011-01-18 2021-11-23 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
US10457729B2 (en) 2011-01-18 2019-10-29 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
WO2013055420A2 (en) 2011-07-12 2013-04-18 Philadelphia Health & Education Corporation Novel clostridium difficile dna vaccine
US9834612B2 (en) 2011-07-22 2017-12-05 The Research Foundation Of State University Of New York Antibodies to the B12-transcobalamin receptor
EP3549611A1 (en) 2011-07-29 2019-10-09 The Trustees of the University of Pennsylvania Switch costimulatory receptors
EP3915588A1 (en) 2011-07-29 2021-12-01 The Trustees of the University of Pennsylvania Switch costimulatory receptors
US10981969B2 (en) 2011-07-29 2021-04-20 The Trustees Of The University Of Pennsylvania Switch costimulatory receptors
US10266580B2 (en) 2011-09-22 2019-04-23 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
US11041012B2 (en) 2011-09-22 2021-06-22 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
US11912753B2 (en) 2011-09-22 2024-02-27 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
US9708384B2 (en) 2011-09-22 2017-07-18 The Trustees Of The University Of Pennsylvania Universal immune receptor expressed by T cells for the targeting of diverse and multiple antigens
WO2013050410A1 (en) 2011-10-04 2013-04-11 Bayer Intellectual Property Gmbh RNAi FOR THE CONTROL OF FUNGI AND OOMYCETES BY INHIBITING SACCHAROPINE DEHYDROGENASE GENE
US9272002B2 (en) 2011-10-28 2016-03-01 The Trustees Of The University Of Pennsylvania Fully human, anti-mesothelin specific chimeric immune receptor for redirected mesothelin-expressing cell targeting
US9422351B2 (en) 2011-11-03 2016-08-23 The Trustees Of The University Of Pennsylvania Isolated B7-H4 specific compositions and methods of use thereof
WO2013067492A1 (en) 2011-11-03 2013-05-10 The Trustees Of The University Of Pennsylvania Isolated b7-h4 specific compositions and methods of use thereof
US10961288B2 (en) 2011-11-03 2021-03-30 The Trustees Of The University Of Pennsylvania Isolated B7-H4 specific compositions and methods of use thereof
US9279008B2 (en) 2011-11-03 2016-03-08 The Trustees Of The University Of Pennsylvania Isolated B7-H4 specific compositions and methods of use thereof
US10259849B2 (en) 2011-11-03 2019-04-16 The Trustees Of The University Of Pennsylvania Isolated B7-H4 specific compositions and methods of use thereof
US9790267B2 (en) 2011-11-08 2017-10-17 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
US10808225B2 (en) 2011-11-23 2020-10-20 The Trustees Of The University Of Pennsylvania Use of PDL1 expressing cells to convert T cells into regulatory T cells
EP3696262A1 (en) 2011-11-23 2020-08-19 The Trustees Of The University Of Pennsylvania Use of pdl1 expressing cells to convert t cells into regulatory t cells
WO2013078230A1 (en) 2011-11-23 2013-05-30 The Trustees Of The University Of Pennsylvania Use of pdl1 expressing cells to convert t cells into regulatory t cells
US9644179B2 (en) 2011-11-23 2017-05-09 The Trustees Of The University Of Pennsylvania Use of PDL1 expressing cells to convert T cells into regulatory T cells
US10501519B2 (en) 2012-02-22 2019-12-10 The Trustees Of The University Of Pennsylvania Use of the CD2 signaling domain in second-generation chimeric antigen receptors
US11299536B2 (en) 2012-02-22 2022-04-12 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
US10040846B2 (en) 2012-02-22 2018-08-07 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
EP4230647A1 (en) 2012-02-22 2023-08-23 The Trustees of the University of Pennsylvania Use of icos-based cars to enhance antitumor activity and car persistence
US10800840B2 (en) 2012-02-22 2020-10-13 The Trustees Of The University Of Pennsylvania Compositions and methods for generating a persisting population of T cells useful for the treatment of cancer
US9783591B2 (en) 2012-02-22 2017-10-10 The Trustees Of The University Of Pennsylvania Use of the CD2 signaling domain in second-generation chimeric antigen receptors
WO2013126733A1 (en) 2012-02-22 2013-08-29 The Trustees Of University Of Pennsylvania Use of icos-based cars to enhance antitumor activity and car persistence
EP4275699A2 (en) 2012-02-22 2023-11-15 The Trustees of the University of Pennsylvania Use of the cd2 signaling domain in second-generation chimeric antigen receptors
US11597754B2 (en) 2012-02-22 2023-03-07 The Trustees Of The University Of Pennsylvania Use of the CD2 signaling domain in second-generation chimeric antigen receptors
WO2013126729A1 (en) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Use of the cd2 signaling domain in second-generation chimeric antigen receptors
EP3747898A1 (en) 2012-02-22 2020-12-09 The Trustees of the University of Pennsylvania Use of icos-based cars to enhance antitumor activity and car persistence
EP3699284A1 (en) 2012-07-05 2020-08-26 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
EP4148135A1 (en) 2012-07-05 2023-03-15 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
EP3584256A1 (en) 2012-07-13 2019-12-25 The Trustees Of The University Of Pennsylvania Methods of assessing the suitability of transduced t cells for administration
US9572836B2 (en) 2012-07-13 2017-02-21 The Trustees Of The University Of Pennsylvania Methods for assessing the suitability of transduced T cells for administration
US10696749B2 (en) 2012-07-13 2020-06-30 The Trustees Of The University Of Pennsylvania Enhancing activity of CAR T cells by co-introducing a bispecific antibody
US11795240B2 (en) 2012-07-13 2023-10-24 The Trustees Of The University Of Pennsylvania Enhancing activity of CAR T cells by co-introducing a bispecific antibody
EP3730512A1 (en) 2012-07-13 2020-10-28 The Trustees of the University of Pennsylvania Enhancing activity of car t cells by co-introducing a bispecific antibody
US9765156B2 (en) 2012-07-13 2017-09-19 The Trustees Of The University Of Pennsylvania Enhancing activity of CAR T cells by co-introducing a bispecific antibody
US9937205B2 (en) 2012-09-04 2018-04-10 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
US11931380B2 (en) 2012-09-04 2024-03-19 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
US11718685B2 (en) 2012-10-01 2023-08-08 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
US10329355B2 (en) 2012-10-01 2019-06-25 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
WO2014053395A1 (en) 2012-10-01 2014-04-10 Basf Se Use of n-thio-anthranilamide compounds on cultivated plants
US9365641B2 (en) 2012-10-01 2016-06-14 The Trustees Of The University Of Pennsylvania Compositions and methods for targeting stromal cells for the treatment of cancer
US10844117B2 (en) 2012-10-05 2020-11-24 The Trustees Of The University Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
US9598489B2 (en) 2012-10-05 2017-03-21 The Trustees Of The Univeristy Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
WO2014090765A1 (en) 2012-12-12 2014-06-19 Bayer Cropscience Ag Use of 1-[2-fluoro-4-methyl-5-(2,2,2-trifluoroethylsulfinyl)phenyl]-5-amino-3-trifluoromethyl)-1 h-1,2,4 tfia zole for controlling nematodes in nematode-resistant crops
US9834610B2 (en) 2013-01-31 2017-12-05 Thomas Jefferson University Fusion proteins for modulating regulatory and effector T cells
WO2014130657A1 (en) 2013-02-20 2014-08-28 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
EP3626741A1 (en) 2013-02-20 2020-03-25 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-egfrviii chimeric antigen receptor
EP3744736A1 (en) 2013-02-20 2020-12-02 Novartis AG Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
WO2014130635A1 (en) 2013-02-20 2014-08-28 Novartis Ag Effective targeting of primary human leukemia using anti-cd123 chimeric antigen receptor engineered t cells
US9839677B2 (en) 2013-03-01 2017-12-12 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
US11564977B2 (en) 2013-03-01 2023-01-31 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
US9895426B2 (en) 2013-03-01 2018-02-20 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
US9415120B2 (en) 2013-03-01 2016-08-16 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked MyoTubular Myopathy (XLMTM)
US8957044B2 (en) 2013-03-01 2015-02-17 Wake Forest University Health Sciences Systemic gene replacement therapy for treatment of X-linked myotubular myopathy (XLMTM)
EP3623380A1 (en) 2013-03-15 2020-03-18 Michael C. Milone Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US9446105B2 (en) 2013-03-15 2016-09-20 The Trustees Of The University Of Pennsylvania Chimeric antigen receptor specific for folate receptor β
WO2014153270A1 (en) 2013-03-16 2014-09-25 Novartis Ag Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
EP4067382A1 (en) 2013-03-16 2022-10-05 Novartis AG Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
EP3539986A1 (en) 2013-03-16 2019-09-18 Novartis AG Treatment of cancer using humanized anti-cd19 chimeric antigen receptor
WO2015090230A1 (en) 2013-12-19 2015-06-25 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
EP4026909A1 (en) 2013-12-19 2022-07-13 Novartis AG Human mesothelin chimeric antigen receptors and uses thereof
WO2015090229A1 (en) 2013-12-20 2015-06-25 Novartis Ag Regulatable chimeric antigen receptor
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
EP4303229A2 (en) 2014-01-21 2024-01-10 Novartis AG Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
WO2015116178A1 (en) 2014-01-31 2015-08-06 Thomas Jefferson University Fusion proteins for modulating regulatory and effector t cells
EP3811970A1 (en) 2014-03-15 2021-04-28 Novartis AG Regulatable chimeric antigen receptor
WO2015142675A2 (en) 2014-03-15 2015-09-24 Novartis Ag Treatment of cancer using chimeric antigen receptor
WO2015157252A1 (en) 2014-04-07 2015-10-15 BROGDON, Jennifer Treatment of cancer using anti-cd19 chimeric antigen receptor
EP3888674A1 (en) 2014-04-07 2021-10-06 Novartis AG Treatment of cancer using anti-cd19 chimeric antigen receptor
WO2015168613A2 (en) 2014-05-02 2015-11-05 The Trustees Of The University Of Pennsylvania Compositions and methods of chimeric autoantibody receptor t cells
EP4008725A1 (en) 2014-05-02 2022-06-08 The Trustees of the University of Pennsylvania Compositions and methods of chimeric autoantibody receptor t cells
EP3791891A1 (en) 2014-07-17 2021-03-17 Brian J. Czerniecki Identification of immunogenic mhc class ii peptides for immune-based therapy
WO2016011432A2 (en) 2014-07-17 2016-01-21 Czerniecki Brian J Identification of immunogenic mhc class ii peptides for immune-based therapy
WO2016014553A1 (en) 2014-07-21 2016-01-28 Novartis Ag Sortase synthesized chimeric antigen receptors
EP3722316A1 (en) 2014-07-21 2020-10-14 Novartis AG Treatment of cancer using a cd33 chimeric antigen receptor
WO2016014565A2 (en) 2014-07-21 2016-01-28 Novartis Ag Treatment of cancer using humanized anti-bcma chimeric antigen receptor
WO2016014530A1 (en) 2014-07-21 2016-01-28 Novartis Ag Combinations of low, immune enhancing. doses of mtor inhibitors and cars
EP3660042A1 (en) 2014-07-31 2020-06-03 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
EP4205749A1 (en) 2014-07-31 2023-07-05 Novartis AG Subset-optimized chimeric antigen receptor-containing cells
WO2016025880A1 (en) 2014-08-14 2016-02-18 Novartis Ag Treatment of cancer using gfr alpha-4 chimeric antigen receptor
US10851149B2 (en) 2014-08-14 2020-12-01 The Trustees Of The University Of Pennsylvania Treatment of cancer using GFR α-4 chimeric antigen receptor
EP3712171A1 (en) 2014-08-19 2020-09-23 Novartis AG Treatment of cancer using a cd123 chimeric antigen receptor
WO2016044605A1 (en) 2014-09-17 2016-03-24 Beatty, Gregory Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
EP3967709A1 (en) 2014-09-17 2022-03-16 Novartis AG Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2016057705A1 (en) 2014-10-08 2016-04-14 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
WO2016090034A2 (en) 2014-12-03 2016-06-09 Novartis Ag Methods for b cell preconditioning in car therapy
WO2016091674A1 (en) 2014-12-12 2016-06-16 Basf Se Use of cyclaniliprole on cultivated plants
WO2016098078A2 (en) 2014-12-19 2016-06-23 Novartis Ag Dimerization switches and uses thereof
EP4036109A2 (en) 2014-12-29 2022-08-03 Novartis AG Methods of making chimeric antigen receptor-expressing cells
US11919959B2 (en) 2015-01-21 2024-03-05 Cancer Research Technology Limited Inhibitors of the interaction between CLEC14A and multimerin-2 for inhibition of angiogenesis
US10808031B2 (en) 2015-01-21 2020-10-20 Cancer Research Technology Limited Inhibitors of the interaction between CLEC14A and multimerin-2 for inhibition of angiogenesis
WO2016126608A1 (en) 2015-02-02 2016-08-11 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US11679132B2 (en) 2015-02-06 2023-06-20 National University Of Singapore Methods for enhancing efficacy of therapeutic immune cells
EP4091616A1 (en) 2015-02-27 2022-11-23 iCell Gene Therapeutics LLC Chimeric antigen receptors (car) targeting hematologic malignancies, compositions and methods of use thereof
WO2016138491A1 (en) 2015-02-27 2016-09-01 Icell Gene Therapeutics Llc Chimeric antigen receptors (cars) targeting hematologic malignancies, compositions and methods of use thereof
WO2016140910A2 (en) 2015-03-04 2016-09-09 University Of Rochester Compositions and methods of using anti-mullerian hormone for treatment of infertility
US11219675B2 (en) 2015-03-13 2022-01-11 Syz Cell Therapy Co. Methods of cancer treatment using activated T cells
US11229689B2 (en) 2015-03-13 2022-01-25 Syz Cell Therapy Co. Methods of cancer treatment using activated T cells
US10011658B2 (en) 2015-04-03 2018-07-03 Eureka Therapeutics, Inc. Constructs targeting AFP peptide/MHC complexes and uses thereof
WO2016164305A1 (en) 2015-04-06 2016-10-13 Subdomain, Llc De novo binding domain containing polypeptides and uses thereof
EP3903804A1 (en) 2015-04-06 2021-11-03 Subdomain, LLC De novo binding domain containing polypeptides and uses thereof
WO2016164308A1 (en) 2015-04-06 2016-10-13 Subdomain, Llc De novo binding domain containing polypeptides and uses thereof
WO2016162371A1 (en) 2015-04-07 2016-10-13 Basf Agrochemical Products B.V. Use of an insecticidal carboxamide compound against pests on cultivated plants
WO2016164580A1 (en) 2015-04-07 2016-10-13 Novartis Ag Combination of chimeric antigen receptor therapy and amino pyrimidine derivatives
EP4056588A1 (en) 2015-04-08 2022-09-14 Novartis AG Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car)- expressing cell
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
EP4234685A2 (en) 2015-04-17 2023-08-30 Novartis AG Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016168595A1 (en) 2015-04-17 2016-10-20 Barrett David Maxwell Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016170484A1 (en) 2015-04-21 2016-10-27 Novartis Ag Rna-guided gene editing system and uses thereof
WO2016172583A1 (en) 2015-04-23 2016-10-27 Novartis Ag Treatment of cancer using chimeric antigen receptor and protein kinase a blocker
WO2016174652A1 (en) 2015-04-30 2016-11-03 Technion Research & Development Foundation Limited Chimeric antigen receptors and methods of their use
WO2016178996A1 (en) 2015-05-01 2016-11-10 The Regents Of The University Of California Glycan-dependent immunotherapeutic molecules
EP4088732A1 (en) 2015-05-01 2022-11-16 The Regents of The University of California Glycan-dependent immunotherapeutic molecules
WO2016187349A1 (en) 2015-05-18 2016-11-24 Tcr2, Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10358474B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10358473B2 (en) 2015-05-18 2019-07-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
EP3466967A1 (en) 2015-05-18 2019-04-10 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10442849B2 (en) 2015-05-18 2019-10-15 Tcr2 Therabeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11028142B2 (en) 2015-05-18 2021-06-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
EP3770168A1 (en) 2015-05-18 2021-01-27 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10954311B2 (en) 2015-05-21 2021-03-23 Harpoon Therapeutics, Inc. Trispecific binding proteins and methods of use
WO2016210293A1 (en) 2015-06-25 2016-12-29 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS OF USE THEREOF
WO2017009853A1 (en) 2015-07-16 2017-01-19 Yeda Research And Development Co. Ltd. Genetically modified anti-third party central memory t cells and use of same in immunotherapy
WO2017015427A1 (en) 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
WO2017040195A1 (en) 2015-08-28 2017-03-09 The Trustees Of The University Of Pennsylvania Methods and compositions for cells expressing a chimeric intracellular signaling molecule
WO2017040930A2 (en) 2015-09-03 2017-03-09 The Trustees Of The University Of Pennsylvania Biomarkers predictive of cytokine release syndrome
US10738099B2 (en) 2015-09-22 2020-08-11 The Trustees Of The University Of Pennsylvania Method of redirecting T cells to treat HIV infection
US11421013B2 (en) 2015-10-23 2022-08-23 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
US10822389B2 (en) 2015-10-23 2020-11-03 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
EP3842450A1 (en) 2015-10-23 2021-06-30 Eureka Therapeutics, Inc. Antibody/t-cell receptor chimeric constructs and uses thereof
WO2017070608A1 (en) 2015-10-23 2017-04-27 Eureka Therapeutics, Inc. Antibody/t-cell receptor chimeric constructs and uses thereof
US10098951B2 (en) 2015-10-23 2018-10-16 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
US10464988B2 (en) 2015-10-23 2019-11-05 Eureka Therapeutics, Inc. Antibody/T-cell receptor chimeric constructs and uses thereof
WO2017075421A1 (en) 2015-10-29 2017-05-04 Temple University-Of The Commonwealth System Of Higher Education Modification of 3' terminal ends of nucleic acids by dna polymerase theta
US10508143B1 (en) 2015-10-30 2019-12-17 Aleta Biotherapeutics Inc. Compositions and methods for treatment of cancer
US10066023B2 (en) 2015-10-30 2018-09-04 Aleta Biotherapeutics Inc. Compositions and methods for tumor transduction
US11807691B2 (en) 2015-10-30 2023-11-07 Aleta Biotherapeutics Inc. Compositions and methods for treatment of cancer
US10072094B2 (en) 2015-10-30 2018-09-11 Aleta Biotherapeutics Inc. Compositions and methods for tumor transduction
US10669349B2 (en) 2015-10-30 2020-06-02 Aleta Biotherapeutics Inc. Compositions and methods for treatment of cancer
US11059904B2 (en) 2015-10-30 2021-07-13 Aleta Biotherapeutics Inc. Compositions and methods for tumor transduction
EP3708587A1 (en) 2015-11-27 2020-09-16 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
EP3708588A1 (en) 2015-11-27 2020-09-16 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
WO2017088012A1 (en) 2015-11-27 2017-06-01 Cartherics Pty. Ltd. Genetically modified cells and uses thereof
WO2017095823A1 (en) 2015-11-30 2017-06-08 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
WO2017112741A1 (en) 2015-12-22 2017-06-29 Novartis Ag Mesothelin chimeric antigen receptor (car) and antibody against pd-l1 inhibitor for combined use in anticancer therapy
WO2017117112A1 (en) 2015-12-28 2017-07-06 Novartis Ag Methods of making chimeric antigen receptor -expressing cells
WO2017118726A1 (en) 2016-01-06 2017-07-13 Lonza Ltd Inhibition of protein degradation for improved production
US11441146B2 (en) 2016-01-11 2022-09-13 Christiana Care Health Services, Inc. Compositions and methods for improving homogeneity of DNA generated using a CRISPR/Cas9 cleavage system
WO2017149515A1 (en) 2016-03-04 2017-09-08 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US11613900B2 (en) 2016-03-10 2023-03-28 Lonza Ltd Customizable facility
US10689873B2 (en) 2016-03-10 2020-06-23 Lonza Ltd Customizable facility
US11236520B2 (en) 2016-03-10 2022-02-01 Lonza Ltd Customizable facility
US11377865B2 (en) 2016-03-10 2022-07-05 Lonza Ltd Customizable facility
WO2017172981A2 (en) 2016-03-29 2017-10-05 University Of Southern California Chimeric antigen receptors targeting cancer
WO2017178526A1 (en) 2016-04-14 2017-10-19 Lonza Ltd Compositions and methods for the detection of host cell proteins
EP3683579A1 (en) 2016-04-14 2020-07-22 Lonza Ltd Compositions and methods for the detection of host cell proteins
EP4219721A2 (en) 2016-04-15 2023-08-02 Novartis AG Compositions and methods for selective protein expression
WO2017181119A2 (en) 2016-04-15 2017-10-19 Novartis Ag Compositions and methods for selective protein expression
WO2017181101A1 (en) 2016-04-15 2017-10-19 The Trustees Of The University Of Pennsylvania Compositions and methods of chimeric alloantigen receptor t cells
WO2017190001A1 (en) 2016-04-29 2017-11-02 The Regents Of The University Of Colorado, A Body Corporate Compounds and compositions useful for treating metabolic syndrome, and methods using same
US11434474B2 (en) 2016-05-03 2022-09-06 Lonza Ltd Modulation of lipid metabolism for protein production
US10655111B2 (en) 2016-05-03 2020-05-19 Lonza Ltd. Modulation of lipid metabolism for protein production
WO2017191165A1 (en) 2016-05-03 2017-11-09 Lonza Ltd Modulation of lipid metabolism for protein production
US11453716B2 (en) 2016-05-20 2022-09-27 Harpoon Therapeutics, Inc. Single domain serum albumin binding protein
US11623958B2 (en) 2016-05-20 2023-04-11 Harpoon Therapeutics, Inc. Single chain variable fragment CD3 binding proteins
WO2017210617A2 (en) 2016-06-02 2017-12-07 Porter, David, L. Therapeutic regimens for chimeric antigen receptor (car)- expressing cells
WO2017214333A1 (en) 2016-06-08 2017-12-14 Intrexon Corporation Cd33 specific chimeric antigen receptors
WO2018013918A2 (en) 2016-07-15 2018-01-18 Novartis Ag Treatment and prevention of cytokine release syndrome using a chimeric antigen receptor in combination with a kinase inhibitor
WO2018014122A1 (en) 2016-07-18 2018-01-25 Helix Biopharma Corp. Car immune cells directed to carcinoembryonic antigen related cell adhesion molecule 6 to treat cancer
WO2018023025A1 (en) 2016-07-28 2018-02-01 Novartis Ag Combination therapies of chimeric antigen receptors adn pd-1 inhibitors
WO2018026819A2 (en) 2016-08-01 2018-02-08 Novartis Ag Treatment of cancer using a chimeric antigen receptor in combination with an inhibitor of a pro-m2 macrophage molecule
US11242376B2 (en) 2016-08-02 2022-02-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10669518B2 (en) 2016-08-02 2020-06-02 Lonza Ltd Method and system for providing buffer solutions
WO2018026953A1 (en) 2016-08-02 2018-02-08 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018026956A1 (en) 2016-08-02 2018-02-08 Lonza Ltd Method and system for providing buffer solutions
US11898137B2 (en) 2016-08-02 2024-02-13 Lonza Ltd Method and system for providing buffer solutions
WO2018031858A1 (en) 2016-08-12 2018-02-15 Lonza Ltd Proteomic analysis of host cell proteins
WO2018053542A1 (en) 2016-09-19 2018-03-22 University Of Southern California Non-radioactive cytotoxicity assays
EP4115951A1 (en) 2016-10-04 2023-01-11 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
US10800833B2 (en) 2016-10-04 2020-10-13 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
US11286291B2 (en) 2016-10-04 2022-03-29 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
WO2018067697A1 (en) 2016-10-04 2018-04-12 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
EP3757120A1 (en) 2016-10-04 2020-12-30 Precision Biosciences, Inc. Co-stimulatory domains for use in genetically-modified cells
EP3848392A1 (en) 2016-10-07 2021-07-14 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018067992A1 (en) 2016-10-07 2018-04-12 Novartis Ag Chimeric antigen receptors for the treatment of cancer
US11085021B2 (en) 2016-10-07 2021-08-10 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10208285B2 (en) 2016-10-07 2019-02-19 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018067993A1 (en) 2016-10-07 2018-04-12 TCR2 Therapeutics Inc. Compositions and methods for t-cell receptors reprogramming using fusion proteins
US11377638B2 (en) 2016-10-07 2022-07-05 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018083606A1 (en) 2016-11-01 2018-05-11 Novartis Ag Methods and compositions for enhancing gene editing
US11945865B2 (en) 2016-11-22 2024-04-02 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
US11851491B2 (en) 2016-11-22 2023-12-26 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
WO2018098365A2 (en) 2016-11-22 2018-05-31 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US11440958B2 (en) 2016-11-22 2022-09-13 National University Of Singapore Blockade of CD7 expression and chimeric antigen receptors for immunotherapy of T-cell malignancies
US10849973B2 (en) 2016-11-23 2020-12-01 Harpoon Therapeutics, Inc. Prostate specific membrane antigen binding protein
US10844134B2 (en) 2016-11-23 2020-11-24 Harpoon Therapeutics, Inc. PSMA targeting trispecific proteins and methods of use
WO2018119298A1 (en) 2016-12-21 2018-06-28 TCR2 Therapeutics Inc. Engineered t cells for the treatment of cancer
WO2018127585A1 (en) 2017-01-06 2018-07-12 Txcell Monospecific regulatory t cell population with cytotoxicity for b cells
WO2018127584A1 (en) 2017-01-06 2018-07-12 Txcell Monospecific regulatory t cell population with cytotoxicity for b cells
EP3346001A1 (en) 2017-01-06 2018-07-11 TXCell Monospecific regulatory t cell population with cytotoxicity for b cells
WO2018132494A1 (en) 2017-01-10 2018-07-19 Intrexon Corporation Modulating expression of polypeptides via new gene switch expression systems
WO2018132390A1 (en) 2017-01-10 2018-07-19 Christiana Care Health Services, Inc. Methods for in vitro site-directed mutagenesis using gene editing technologies
US11274157B2 (en) 2017-01-12 2022-03-15 Eureka Therapeutics, Inc. Constructs targeting histone H3 peptide/MHC complexes and uses thereof
WO2018134824A1 (en) 2017-01-18 2018-07-26 Yeda Research And Development Co. Ltd. Genetically modified veto cells and use of same in immunotherapy
WO2018140725A1 (en) 2017-01-26 2018-08-02 Novartis Ag Cd28 compositions and methods for chimeric antigen receptor therapy
EP4043485A1 (en) 2017-01-26 2022-08-17 Novartis AG Cd28 compositions and methods for chimeric antigen receptor therapy
EP4194473A1 (en) 2017-02-08 2023-06-14 Cellular Biomedicine Group Inc. Construction of chimeric antigen receptor targeting cd20 antigen and activity identification of engineered t cells thereof
WO2018145649A1 (en) 2017-02-08 2018-08-16 西比曼生物科技(上海)有限公司 Construction of chimeric antigen receptor targeting cd20 antigen and activity identification of engineered t cells thereof
WO2018157072A1 (en) 2017-02-27 2018-08-30 Life Technologies Corporation Expansion of populations of t cells by the use of modified serum free media
US11597911B2 (en) 2017-02-27 2023-03-07 Life Technologies Corporation Expansion of populations of T cells by the use of modified serum free media
US11535668B2 (en) 2017-02-28 2022-12-27 Harpoon Therapeutics, Inc. Inducible monovalent antigen binding protein
US10934336B2 (en) 2017-04-13 2021-03-02 The Trustees Of The University Of Pennsylvania Use of gene editing to generate universal TCR re-directed T cells for adoptive immunotherapy
US11447564B2 (en) 2017-04-26 2022-09-20 Eureka Therapeutics, Inc. Constructs specifically recognizing glypican 3 and uses thereof
US11613573B2 (en) 2017-04-26 2023-03-28 Eureka Therapeutics, Inc. Chimeric antibody/T-cell receptor constructs and uses thereof
WO2018200583A1 (en) 2017-04-26 2018-11-01 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
US10822413B2 (en) 2017-04-26 2020-11-03 Eureka Therapeutics, Inc. Cells expressing chimeric activating receptors and chimeric stimulating receptors and uses thereof
WO2018201056A1 (en) 2017-04-28 2018-11-01 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
WO2018208837A1 (en) 2017-05-08 2018-11-15 Precision Biosciences, Inc. Nucleic acid molecules encoding an engineered antigen receptor and an inhibitory nucleic acid molecule and methods of use thereof
EP4029943A1 (en) 2017-05-08 2022-07-20 Precision Biosciences, Inc. Nucleic acid molecules encoding an engineered antigen receptor and an inhibitory nucleic acid molecule and methods of use thereof
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US11607453B2 (en) 2017-05-12 2023-03-21 Harpoon Therapeutics, Inc. Mesothelin binding proteins
WO2018232020A1 (en) 2017-06-13 2018-12-20 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2018232265A1 (en) 2017-06-16 2018-12-20 Lonza Ltd Universal self-regulating mammalian cell line platform for the production of biologics
WO2019007991A1 (en) 2017-07-03 2019-01-10 Universite De Strasbourg Mtmr2-s polypeptide for use in the treatment of myopathies
WO2019047899A1 (en) 2017-09-06 2019-03-14 亘喜生物科技(上海)有限公司 Universal chimeric antigen receptor t-cell preparation technique
WO2019055853A1 (en) 2017-09-15 2019-03-21 Life Technologies Corporation Compositions and methods for culturing and expanding cells
US11814432B2 (en) 2017-09-20 2023-11-14 The University Of British Columbia Anti-HLA-A2 antibodies, related chimeric antigen receptors, and uses thereof
WO2019062817A1 (en) 2017-09-27 2019-04-04 亘喜生物科技(上海)有限公司 Engineered immune cell capable of inducing secretion of anti-cd47 antibody
WO2019070856A1 (en) 2017-10-03 2019-04-11 Precision Biosciences, Inc. Modified epidermal growth factor receptor peptides for use in genetically-modified cells
EP4269560A2 (en) 2017-10-03 2023-11-01 Precision Biosciences, Inc. Modified epidermal growth factor receptor peptides for use in genetically-modified cells
US11215618B2 (en) 2017-10-04 2022-01-04 Hesperix SA Articles and methods directed to personalized therapy of cancer
WO2019070161A2 (en) 2017-10-04 2019-04-11 Opko Pharmaceuticals, Llc Articles and methods directed to personalized therapy of cancer
WO2019069125A1 (en) 2017-10-06 2019-04-11 Oslo Universitetssykehus Hf Chimeric antigen receptors
US11136403B2 (en) 2017-10-13 2021-10-05 Harpoon Therapeutics, Inc. Trispecific proteins and methods of use
US10927180B2 (en) 2017-10-13 2021-02-23 Harpoon Therapeutics, Inc. B cell maturation antigen binding proteins
WO2019084288A1 (en) 2017-10-25 2019-05-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2019089798A1 (en) 2017-10-31 2019-05-09 Novartis Ag Anti-car compositions and methods
WO2019092251A1 (en) 2017-11-11 2019-05-16 Universite De Strasbourg Compositions and method for the treatment of x-linked centronuclear myopathy
WO2019099440A1 (en) 2017-11-14 2019-05-23 Arcellx, Inc. Multifunctional immune cell therapies
WO2019099433A2 (en) 2017-11-14 2019-05-23 Arcellx, Inc. D-domain containing polypeptides and uses thereof
WO2019099639A1 (en) 2017-11-15 2019-05-23 Navartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
WO2019108900A1 (en) 2017-11-30 2019-06-06 Novartis Ag Bcma-targeting chimeric antigen receptor, and uses thereof
WO2019113050A1 (en) 2017-12-05 2019-06-13 Lonza Ltd Methods of assaying tropolone
WO2019129851A1 (en) 2017-12-29 2019-07-04 Cellectis Method for improving production of car t cells
WO2019136432A1 (en) 2018-01-08 2019-07-11 Novartis Ag Immune-enhancing rnas for combination with chimeric antigen receptor therapy
WO2019145711A1 (en) 2018-01-23 2019-08-01 Immetacyte Limited Biomarker predictive of tumour infiltrating lymphocyte therapy and uses thereof
EP4233883A2 (en) 2018-01-23 2023-08-30 Instil Bio (Uk) Limited Biomarker predictive of tumour infiltrating lymphocyte therapy and uses thereof
WO2019152660A1 (en) 2018-01-31 2019-08-08 Novartis Ag Combination therapy using a chimeric antigen receptor
EP3741853A1 (en) 2018-02-02 2020-11-25 Lonza Ltd. Methods of cell selection and modifying cell metabolism
WO2019152876A2 (en) 2018-02-02 2019-08-08 Lonza Ltd Methods of cell selection and modifying cell metabolism
US11396552B2 (en) 2018-02-12 2022-07-26 Diabetes-Free Inc. Antagonistic anti-human CD40 monoclonal antibodies
WO2019160956A1 (en) 2018-02-13 2019-08-22 Novartis Ag Chimeric antigen receptor therapy in combination with il-15r and il15
US11485782B2 (en) 2018-03-14 2022-11-01 Beijing Xuanyi Pharmasciences Co., Ltd. Anti-claudin 18.2 antibodies
US11248208B2 (en) 2018-03-30 2022-02-15 Syz Cell Therapy Co. Multiple antigen specific cell therapy methods
US11142581B2 (en) 2018-04-12 2021-10-12 Cellular Biomedicine Group Hk Limited BCMA-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
WO2019196713A1 (en) 2018-04-12 2019-10-17 西比曼生物科技(香港)有限公司 Bcma-targeted chimeric antigen receptor as well as preparation method therefor and application thereof
US11390659B2 (en) 2018-04-13 2022-07-19 Syz Cell Therapy Co. Methods of obtaining tumor-specific T cell receptors
US11471519B2 (en) 2018-04-13 2022-10-18 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific T cells
US11219676B2 (en) 2018-04-13 2022-01-11 Syz Cell Therapy Co. Methods of cancer treatment using tumor antigen-specific T cells
WO2019210153A1 (en) 2018-04-27 2019-10-31 Novartis Ag Car t cell therapies with enhanced efficacy
US10874750B2 (en) 2018-04-30 2020-12-29 Amicus Therapeutics, Inc. Gene therapy constructs and methods of use
WO2019213180A1 (en) 2018-04-30 2019-11-07 Amicus Therapeutics, Inc. Gene therapy constructs and methods of use
US11491243B2 (en) 2018-04-30 2022-11-08 Amicus Therapeutics, Inc. Gene therapy constructs and methods of use
WO2019213282A1 (en) 2018-05-01 2019-11-07 Novartis Ag Biomarkers for evaluating car-t cells to predict clinical outcome
WO2019217512A1 (en) 2018-05-08 2019-11-14 Life Technologies Corporation Compositions and methods for culturing and expanding cells
WO2019227003A1 (en) 2018-05-25 2019-11-28 Novartis Ag Combination therapy with chimeric antigen receptor (car) therapies
WO2019241426A1 (en) 2018-06-13 2019-12-19 Novartis Ag Bcma chimeric antigen receptors and uses thereof
WO2019241549A1 (en) 2018-06-15 2019-12-19 A2 Biotherapeutics, Inc. Foxp3-expressing car-t regulatory cells
WO2019243835A1 (en) 2018-06-21 2019-12-26 Immetacyte Limited Chimeric growth factor receptors
WO2020030979A2 (en) 2018-08-10 2020-02-13 Sangamo Therapeutics France New car constructs comprising tnfr2 domains
WO2020047501A1 (en) 2018-08-30 2020-03-05 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
WO2020047452A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020047449A2 (en) 2018-08-31 2020-03-05 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020043899A1 (en) 2018-08-31 2020-03-05 Invectys Chimeric antigen receptors against multiple hla-g isoforms
US10815311B2 (en) 2018-09-25 2020-10-27 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
US11807692B2 (en) 2018-09-25 2023-11-07 Harpoon Therapeutics, Inc. DLL3 binding proteins and methods of use
WO2020069405A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies
WO2020069409A1 (en) 2018-09-28 2020-04-02 Novartis Ag Cd19 chimeric antigen receptor (car) and cd22 car combination therapies
US11097015B2 (en) 2018-10-10 2021-08-24 Amicus Therapeutics, Inc. Disulfide bond stabilized polypeptide compositions and methods of use
WO2020077114A2 (en) 2018-10-10 2020-04-16 Amicus Therapeutics, Inc. Disulfide bond stabilized polypeptide compositions and methods of use
WO2020081929A1 (en) 2018-10-19 2020-04-23 University Of Rochester Immune modulators in combination with radiation treatment for advanced pancreatic cancer
WO2020086627A1 (en) 2018-10-22 2020-04-30 University Of Rochester Genome editing by directed non-homologous dna insertion using a retroviral integrase-cas9 fusion protein
WO2020092455A2 (en) 2018-10-29 2020-05-07 The Broad Institute, Inc. Car t cell transcriptional atlas
US11130783B2 (en) 2018-11-13 2021-09-28 Regents Of The University Of Minnesota CD40 targeted peptides and uses thereof
WO2020102676A1 (en) 2018-11-16 2020-05-22 Celgene Corporation Improved t cell manufacturing process
WO2020102555A1 (en) 2018-11-16 2020-05-22 Memorial Sloan Kettering Cancer Center Antibodies to mucin-16 and methods of use thereof
EP4151722A1 (en) 2018-11-16 2023-03-22 Celgene Corporation Improved t cell manufacturing process
WO2020140007A1 (en) 2018-12-28 2020-07-02 University Of Rochester Gene therapy for best1 dominant mutations
WO2020150287A1 (en) 2019-01-14 2020-07-23 University Of Rochester Targeted nuclear rna cleavage and polyadenylation with crispr-cas
WO2020152451A1 (en) 2019-01-22 2020-07-30 Immetacyte Limited Receptors providing targeted costimulation for adoptive cell therapy
WO2020161186A1 (en) 2019-02-08 2020-08-13 Biontech Cell & Gene Therapies Gmbh Chimeric antigen receptor-modified cells for the treatment of cldn6-expressing cancers
WO2020172553A1 (en) 2019-02-22 2020-08-27 Novartis Ag Combination therapies of egfrviii chimeric antigen receptors and pd-1 inhibitors
WO2020185121A2 (en) 2019-03-13 2020-09-17 Общество С Ограниченной Ответственностью "Анабион" Isolated alternative intracellular signalling domain of a chimeric antigen receptor and chimeric antigen receptor comprising said signalling domain
US10934337B2 (en) 2019-03-15 2021-03-02 Cartesian Therapeutics, Inc. Anti-BCMA chimeric antigen receptors
US11220535B2 (en) 2019-03-15 2022-01-11 Cartesian Therapeutics, Inc. Anti-BCMA chimeric antigen receptors
WO2020188103A1 (en) 2019-03-20 2020-09-24 Centre National De La Recherche Scientifique Amphiphysin / bin1 for the treatment of autosomal dominant centronuclear myopathy
US11090336B2 (en) 2019-03-27 2021-08-17 The Trustees Of The University Of Pennsylvania Tn-MUC1 chimeric antigen receptor (CAR) T cell therapy
WO2020210678A1 (en) 2019-04-12 2020-10-15 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
WO2020224606A1 (en) 2019-05-07 2020-11-12 亘喜生物科技(上海)有限公司 Engineered immune cell targeting bcma and use thereof
US11840575B2 (en) 2019-05-07 2023-12-12 Gracell Biotechnologies (Shanghai) Co., Ltd. Engineered immune cells targeting BCMA and their uses thereof
WO2021003442A1 (en) 2019-07-02 2021-01-07 M6P Therapeutics Vector compositions and methods of using same for treatment of lysosomal storage disorders
WO2021016453A1 (en) 2019-07-23 2021-01-28 University Of Rochester Targeted rna cleavage with crispr-cas
WO2021016608A1 (en) 2019-07-25 2021-01-28 Precision Biosciences, Inc. Compositions and methods for sequential stacking of nucleic acid sequences into a genomic locus
WO2021018311A1 (en) 2019-08-01 2021-02-04 上海赛比曼生物科技有限公司 Universal car-t cell and preparation and use thereof
WO2021030182A1 (en) 2019-08-09 2021-02-18 A2 Biotherapeutics, Inc. Bifunctional single variable domain t cell receptors and uses thereof
WO2021032779A1 (en) 2019-08-19 2021-02-25 Universität Basel Cell therapy methods
WO2021035054A1 (en) 2019-08-20 2021-02-25 Precision Biosciences, Inc. Lymphodepletion dosing regimens for cellular immunotherapies
WO2021035170A1 (en) 2019-08-21 2021-02-25 Precision Biosciences, Inc. Compositions and methods for tcr reprogramming using fusion proteins
EP3808766A1 (en) 2019-10-15 2021-04-21 Sangamo Therapeutics France Chimeric antigen receptor specific for interleukin-23 receptor
WO2021096868A1 (en) 2019-11-12 2021-05-20 A2 Biotherapeutics, Inc. Engineered t cell receptors and uses thereof
WO2021094461A1 (en) 2019-11-14 2021-05-20 Lonza Ltd Methods of cell selection
WO2021098882A1 (en) 2019-11-21 2021-05-27 博生吉医药科技(苏州)有限公司 Cd7-car-t cell and preparation and application thereof
WO2021108661A2 (en) 2019-11-26 2021-06-03 Novartis Ag Chimeric antigen receptors and uses thereof
WO2021108613A1 (en) 2019-11-26 2021-06-03 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
US11254726B2 (en) 2019-12-11 2022-02-22 A2 Biotherapeutics, Inc. LILRB1-based chimeric antigen receptor
WO2021119489A1 (en) 2019-12-11 2021-06-17 A2 Biotherapeutics, Inc. Lilrb1-based chimeric antigen receptor
WO2021158964A1 (en) 2020-02-07 2021-08-12 University Of Rochester Ribozyme-mediated rna assembly and expression
WO2021158982A2 (en) 2020-02-07 2021-08-12 University Of Rochester Targeted translation of rna with crispr-cas13 to enhance protein synthesis
WO2021163618A1 (en) 2020-02-14 2021-08-19 Novartis Ag Method of predicting response to chimeric antigen receptor therapy
US11180563B2 (en) 2020-02-21 2021-11-23 Harpoon Therapeutics, Inc. FLT3 binding proteins and methods of use
WO2021173674A1 (en) 2020-02-26 2021-09-02 A2 Biotherapeutics, Inc. Polypeptides targeting mage-a3 peptide-mhc complexes and methods of use thereof
WO2021173995A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021173985A2 (en) 2020-02-27 2021-09-02 Novartis Ag Methods of making chimeric antigen receptor-expressing cells
WO2021170071A1 (en) 2020-02-28 2021-09-02 Shanghai Henlius Biotech, Inc. Anti-cd137 constructs, multispecific antibody and uses thereof
WO2021170067A1 (en) 2020-02-28 2021-09-02 上海复宏汉霖生物技术股份有限公司 Anti-cd137 construct and use thereof
WO2021186246A1 (en) 2020-03-19 2021-09-23 Trizell Ltd. Temperature-responsive virus storage system
WO2021186056A1 (en) 2020-03-20 2021-09-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Chimeric antigen receptor specific for human cd45rc and uses thereof
WO2021195519A1 (en) 2020-03-27 2021-09-30 University Of Rochester Targeted destruction of viral rna by crispr-cas13
WO2021195525A1 (en) 2020-03-27 2021-09-30 University Of Rochester Crispr-cas13 crrna arrays
WO2021208750A1 (en) 2020-04-16 2021-10-21 上海赛比曼生物科技有限公司 Cd22-targeted chimeric antigen receptor, preparation method therefor and application thereof
WO2021222576A1 (en) 2020-05-01 2021-11-04 A2 Biotherapeutics, Inc. Pag1 fusion proteins and methods of making and using same
WO2021238886A1 (en) 2020-05-27 2021-12-02 Staidson (Beijing) Biopharmaceuticals Co., Ltd. Antibodies specifically recognizing nerve growth factor and uses thereof
WO2021252635A1 (en) 2020-06-11 2021-12-16 A2 Biotherapeutics, Inc. Compositions and methods for treating cancers
WO2021252920A1 (en) 2020-06-11 2021-12-16 Novartis Ag Zbtb32 inhibitors and uses thereof
WO2022015916A1 (en) 2020-07-15 2022-01-20 University Of Rochester Targeted rna cleavage with dcasl3-rnase fusion proteins
WO2022011651A1 (en) 2020-07-16 2022-01-20 上海交通大学 Immunotherapy method of targeted chemokine and cytokine delivery by mesenchymal stem cell
WO2022016114A1 (en) 2020-07-17 2022-01-20 Instill Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2022016112A1 (en) 2020-07-17 2022-01-20 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
WO2022036068A1 (en) 2020-08-13 2022-02-17 A2 Biotherapeutics, Inc. Gene fusions for control of genetically modified cells
US11602544B2 (en) 2020-08-20 2023-03-14 A2 Biotherapeutics, Inc. Compositions and methods for treating EGFR positive cancers
US11433100B2 (en) 2020-08-20 2022-09-06 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
US11602543B2 (en) 2020-08-20 2023-03-14 A2 Biotherapeutics, Inc. Compositions and methods for treating mesothelin positive cancers
WO2022079308A1 (en) 2020-10-16 2022-04-21 Institut Pasteur Chimeric constructs useful in vaccination and cancer therapy
WO2022104061A1 (en) 2020-11-13 2022-05-19 Novartis Ag Combination therapies with chimeric antigen receptor (car)-expressing cells
WO2022105893A1 (en) 2020-11-23 2022-05-27 博生吉医药科技(苏州)有限公司 Preparation method and application of cd7-car-t cells
WO2022123307A1 (en) 2020-12-09 2022-06-16 Takeda Pharmaceutical Company Limited Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
WO2022123316A1 (en) 2020-12-09 2022-06-16 Takeda Pharmaceutical Company Limited Compositions of guanylyl cyclase c (gcc) antigen binding agents and methods of use thereof
WO2022147463A2 (en) 2020-12-31 2022-07-07 Alamar Biosciences, Inc. Binder molecules with high affinity and/ or specificity and methods of making and use thereof
WO2022151960A1 (en) 2021-01-13 2022-07-21 博生吉医药科技(苏州)有限公司 B7-h3 chimeric antigen receptor-modified t cell and use thereof
WO2022155421A1 (en) 2021-01-15 2022-07-21 University Of Rochester Staphylococcus aureus antigen-based nucleic acid vaccines
WO2022165111A1 (en) 2021-01-28 2022-08-04 Precision Biosciences, Inc. Modulation of tgf beta signaling in genetically-modified eukaryotic cells
WO2022162192A2 (en) 2021-01-29 2022-08-04 Thelper As Therapeutic and diagnostic agents and uses thereof
WO2022166665A1 (en) 2021-02-08 2022-08-11 浙江大学 Chimeric antigen receptor with endogenous protein molecule replacing single domain antibody
US11730764B2 (en) 2021-02-16 2023-08-22 A2 Biotherapeutics, Inc. Compositions and methods for treating HER2 positive cancers
WO2022226296A2 (en) 2021-04-23 2022-10-27 University Of Rochester Genome editing by directed non-homologous dna insertion using a retroviral integrase-cas fusion protein and methods of treatment
WO2022254337A1 (en) 2021-06-01 2022-12-08 Novartis Ag Cd19 and cd22 chimeric antigen receptors and uses thereof
WO2022262764A1 (en) 2021-06-16 2022-12-22 四川大学华西医院 Preparation and application of lox1-based chimeric antigen receptor immune cell
WO2022262765A1 (en) 2021-06-16 2022-12-22 四川大学华西医院 Preparation and application of chimeric antigen receptor immune cell constructed on basis of granzyme b
US11945876B2 (en) 2021-06-16 2024-04-02 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
WO2022266660A1 (en) 2021-06-17 2022-12-22 Amberstone Biosciences, Inc. Anti-cd3 constructs and uses thereof
WO2022271955A1 (en) 2021-06-23 2022-12-29 Musc Foundation For Research Development Novel targeted shrna nanoparticles for cancer therapy
WO2023278641A1 (en) 2021-06-29 2023-01-05 Flagship Pioneering Innovations V, Inc. Immune cells engineered to promote thanotransmission and uses thereof
WO2023283611A1 (en) 2021-07-08 2023-01-12 Staidson Biopharma Inc. Antibodies specifically recognizing tnfr2 and uses thereof
WO2023284714A1 (en) 2021-07-14 2023-01-19 舒泰神(北京)生物制药股份有限公司 Antibody that specifically recognizes cd40 and application thereof
US11697677B2 (en) 2021-07-16 2023-07-11 Instil Bio (Uk) Limited Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2023056329A1 (en) 2021-09-30 2023-04-06 Akouos, Inc. Compositions and methods for treating kcnq4-associated hearing loss
WO2023069979A1 (en) 2021-10-20 2023-04-27 University Of Rochester Isolated glial progenitor cells for use in the competition treatment of age-related white matter loss
WO2023069987A1 (en) 2021-10-20 2023-04-27 University Of Rochester Rejuvenation treatment of age-related white matter loss cross reference to related application
WO2023081633A1 (en) 2021-11-02 2023-05-11 University Of Rochester Tcf7l2 mediated remyelination in the brain
WO2023086939A1 (en) 2021-11-12 2023-05-19 Amicus Therapeutics, Inc. Compositions and methods for treating mucopolysaccharidosis iiia
WO2023089556A1 (en) 2021-11-22 2023-05-25 Pfizer Inc. Reducing risk of antigen mimicry in immunogenic medicaments
WO2023105005A1 (en) 2021-12-09 2023-06-15 BioNTech SE Chimeric antigen receptor-modified cells for the treatment of cldn6 expressing cancer
WO2023144779A1 (en) 2022-01-28 2023-08-03 Pfizer Inc. Coronavirus antigen variants
WO2023144392A1 (en) 2022-01-28 2023-08-03 Thelper As Therapeutic and diagnostic agents and uses thereof
WO2023177954A1 (en) 2022-03-18 2023-09-21 University Of Rochester Combination therapy for treatment of cancer, methods and systems of delivery thereof
WO2023215860A1 (en) 2022-05-05 2023-11-09 University Of Rochester Modified bacteria and methods of use for bioglass microlenses
WO2024020407A1 (en) 2022-07-19 2024-01-25 Staidson Biopharma Inc. Antibodies specifically recognizing b- and t-lymphocyte attenuator (btla) and uses thereof
WO2024046572A1 (en) 2022-09-01 2024-03-07 BioNTech SE Chimeric antigen receptor-modified cells for the treatment of cldn6 expressing cancer
WO2024056809A1 (en) 2022-09-15 2024-03-21 Novartis Ag Treatment of autoimmune disorders using chimeric antigen receptor therapy

Also Published As

Publication number Publication date
AU2001275474A1 (en) 2001-12-24
WO2001096584A3 (en) 2003-01-23

Similar Documents

Publication Publication Date Title
WO2001096584A2 (en) Materials and methods for the control of nematodes
CN107287235B (en) Down-regulating gene expression in insect pests
AU2005299492B2 (en) Methods and materials for conferring resistance to pests and pathogens of plants
RU2671143C2 (en) Plants resistant to insect pests
US5589622A (en) Plant parasitic nematode control
KR102266402B1 (en) Novel pest control methods
CN106413390A (en) Compositions and methods for controlling insect pests
KR20170066404A (en) Copi coatomer beta subunit nucleic acid molecules that confer resistance to coleopteran and hemipteran pests
Youssef et al. Post-transcriptional gene silencing of the gene encoding aldolase from soybean cyst nematode by transformed soybean roots
MX2014000707A (en) Plants resistant to insect pests.
CA2535132A1 (en) Plants resistant to cytoplasm-feeding parasites
TW201321511A (en) Nucleic acid molecules that target RPS6 and confer resistance to coleopteran pests
TW201321509A (en) Nucleic acid molecules that target PP1-87B and confer resistance to coleopteran pests
KR20160093727A (en) Ras opposite (rop) and related nucleic acid molecules that confer resistance to coleopteran and/or hemipteran pests
TW201321508A (en) Nucleic acid molecules that target RPA70 and confer resistance to coleopteran pests
HUE031986T2 (en) Stress-resistant plants and their production
KR20170013885A (en) Sec23 nucleic acid molecules that confer resistance to coleopteran and hemipteran pests
EP0548197B1 (en) Plant parasitic nematode control
JP2018513675A (en) RNA polymerase II33 nucleic acid molecules for controlling pests
WO2007147016A2 (en) Generation of plants with improved pathogen resistance
CN110564740B (en) A gene AtPIP2 for improving disease resistance of plants; 7 and uses thereof
JP2018509150A (en) RNA polymerase II215 nucleic acid molecules for controlling pests
WO2013158966A1 (en) Bronze bug control agents
RU2775717C1 (en) Polynucleotide and method used to control an insect outbreak
KR20170065538A (en) Gho/sec24b2 and sec24b1 nucleic acid molecules to control coleopteran and hemipteran pests

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP