WO2007064846A2 - COMPOSITIONS AND METHODS OF USING siRNA TO KNOCKDOWN GENE EXPRESSION AND TO IMPROVE SOLID ORGAN AND CELL TRANSPLANTATION - Google Patents

COMPOSITIONS AND METHODS OF USING siRNA TO KNOCKDOWN GENE EXPRESSION AND TO IMPROVE SOLID ORGAN AND CELL TRANSPLANTATION Download PDF

Info

Publication number
WO2007064846A2
WO2007064846A2 PCT/US2006/045933 US2006045933W WO2007064846A2 WO 2007064846 A2 WO2007064846 A2 WO 2007064846A2 US 2006045933 W US2006045933 W US 2006045933W WO 2007064846 A2 WO2007064846 A2 WO 2007064846A2
Authority
WO
WIPO (PCT)
Prior art keywords
organ
sirna
targeting
targeting polynucleotide
targets
Prior art date
Application number
PCT/US2006/045933
Other languages
French (fr)
Other versions
WO2007064846A3 (en
Inventor
Marie Denise Parker
Julian Roy Pratt
Yijia Liu
Yang Lu
Martin Woodle
Yuefeng Xie
Original Assignee
Intradigm Corporation
University Of Leeds
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 Intradigm Corporation, University Of Leeds filed Critical Intradigm Corporation
Priority to EP06838740A priority Critical patent/EP1963508A2/en
Priority to JP2008543476A priority patent/JP2009518008A/en
Priority to CA002670801A priority patent/CA2670801A1/en
Priority to US12/085,873 priority patent/US20100028848A1/en
Publication of WO2007064846A2 publication Critical patent/WO2007064846A2/en
Publication of WO2007064846A3 publication Critical patent/WO2007064846A3/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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/31Combination therapy
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific

Definitions

  • the present invention provides compositions and methods for the prevention of allograft rejection or xenograft rejection and ischemia/reperfusion injury in solid organ or tissue transplantation using siRNA- mediated down regulation of gene expression.
  • Transplantation immunology refers to an extensive sequence of events that occurs after an allograft or a xenograft is removed from a donor and then transplanted into a recipient . Tissue is damaged at both the graft and the transplantation sites. An inflammatory reaction follows immediately, as does activation of biochemical cascades . A series of specific and nonspecific cellular responses ensues as antigens are recognized. Eventually, the damage is controlled through tissue repair and reinforcement; if damage is nonpathologic, the graft survives. [0006] Antigen-independent causes of tissue damage (i.e., ischemia, hypothermia, reperfusion injury) are the result of mechanical trauma as well as disruption of the blood supply as the graft is harvested.
  • tissue damage i.e., ischemia, hypothermia, reperfusion injury
  • cytokines e.g., tumour necrosis factor, interleukin-1
  • endothelial changes help recruit large numbers of T cells to the transplantation site.
  • Damaged tissues release proinflammatory mediators (e.g., Hageman factor [factor XII]) that trigger several biochemical cascades.
  • the clotting cascade induces fibrin and several related fibrinopeptides, which promote local vascular permeability and attract neutrophils and macrophages .
  • the kinin cascade principally produces bradykinin, which promotes vasodilation, smooth muscle contraction, and increased vascular permeability.
  • an antibody-antigen complex i.e., immune complex
  • CIq triggers the activation process when it docks onto antibodies within the immune complexes via the classical pathway, whilst complement factor C3 can recognize damaged cell surfaces as acceptors for alternative pathway activation.
  • Activated complement causes damage through the deposition of the membrane attack complex (e.g., C5b, C6, C7, C8, C9) and cell -bound ligands, such as C4b and C3b, which activate leukocytes bearing complement receptors.
  • the membrane attack complex e.g., C5b, C6, C7, C8, C9
  • cell -bound ligands such as C4b and C3b
  • production of bioactive anaphylatoxins C5a and C3a causes the influx and activation of inflammatory cells.
  • These chemoattractants also initiate mast cell degranulation, which releases several mediators. Histamine and 5- hydroxytryptamine increase vascular permeability.
  • Prostaglandin E2 promotes vasodilation and vascular permeability.
  • Leukotrienes B4 and D2 promote leukocyte accumulation and vascular permeability. Another means - A -
  • complement by which complement is activated is through tissue ischemia and reperfusion, which exposes phospholipids and mitochondrial proteins. These by-products activate complement directly through binding CIq or mannose- binding lectin or factor C3b.
  • RNA interference (RNAi) compounds the intermediate short interfering RNA oligonucleotides (siRNAs)
  • siRNAs the intermediate short interfering RNA oligonucleotides
  • RNA interference compounds provide a unique strategy for using a combination of multiple siRNA duplexes to target multiple disease-causing genes in the same treatment, since all siRNA duplexes are chemically homogenous with the same source of origin and the same manufacturing process (5, 6, 7, 8) .
  • siRNA inhibitors are expected to have much better clinical efficiency with minimum toxicity and safety concerns.
  • Genetic modification is a promising therapeutic strategy for organ transplantation. Based on the attractive technology of RNA interference for silencing a particular gene expression (9, 10) , siRNA therapy may represent an attractive and powerful approach in preventing ischemia/reperfusion injury as well as organ rejection in transplant recipients.
  • This invention provides targeting polynucleotides that target immunomodulatory or immunoeffector genes present in cells of an organ to be donated to a recipient .
  • Targets for these polynucleotides can be derived from sequences of immunomodulatory and immunoeffector genes listed in Tables 1-15 (see below) .
  • the targeting polynucleotide may target sequences in the C3 , ICAMl, VCAM-I, IFN-Y, IL-I, IL-6, IL-8, TNF- ⁇ , CD80, CD86, MHC-II, MHC-I, CD28, CTLA-4, or PV-B19 genes.
  • the targeting polynucleotides can comprise siRNA duplexes that target one or more of the sequences listed in Tables 1-15.
  • the targeting polynucleotide may be a single-stranded linear polynucleotide, a double- stranded linear polynucleotide, or a hairpin polynucleotide .
  • This invention also provides a method of suppressing rejection of a transplanted organ by contacting the organ with a composition comprising the targeting polynucleotide of the invention before transplanting the organ into a recipient.
  • the method can be effective in down-regulating or inhibiting the expression of a target immunomodulatory or immunoeffector gene in an organ or a cell of an organ during storage before transplantation.
  • the organ is perfused with a composition comprising a targeting polynucleotide of the invention.
  • the organ is bathed or submerged in the composition comprising a targeting polynucleotide of the invention.
  • the composition can also be administered to an organ recipient.
  • the organ may be the recipient's own organ.
  • the recipient of the said organ can be human.
  • Organs, tissues, and cells contacted with the composition comprising a targeting polynucleotide of the invention include the kidney, liver, lung, pancreas, heart, small bowel, cornea, epithelial cells, vascular endothelium, vascular smooth muscle cells, myocardium and passenger leukocytes resident in the organ at the time of transplantation.
  • the composition comprising the targeting polynucleotide of the invention can also comprise a carrier, including, but not limited to, perfusion fluid, Hyper Osmolar Citrate solution, PolyTran polymer solution, TargeTran nanoparticle solution, or University of Wisconsin solution.
  • the composition can also comprise small molecule drugs, monoclonal antibody drugs , and other immune modulators .
  • the composition comprises a plurality of the targeting polynucleotide of the invention.
  • a composition can contain a plurality of targeting polynucleotides of the invention that can target a plurality of gene sequences.
  • the targeting polynucleotides are a cocktail that targets the C3, TNF- ⁇ , and. IL-8 gene sequences.
  • Figure 1 is a bar graph that shows the relative expression of C3 mRNA in rat renal cells.
  • the cells were stimulated with IL-I and IL-6 to increase C3 expression.
  • Three candidate C3 siRNA sequences (C3-1, C3-2, C3-3) or FITC-labelled scrambled siRNA were transfected into the cells at various concentrations.
  • One set of cells was treated with Lipofectamine and no siRNA (+ lipofectamine) while another set was stimulated to produce C3 and treated with neither
  • Lipofectamine nor siRNA (- lipofectamine) .
  • C3 mRNA levels were measured in the cells by Real Time PCR 48 hours after transfection. The dotted line indicates unstimulated cell C3 expression. The experiment showed the feasibility and efficacy of gene knockdown by siRNA. The C3-3 siRNA was selected as the candidate to use in further experiments .
  • Figure 2 is a bar graph showing the relative expression of C3 mRNA in rat renal cells stimulated with IL-I and IL-6 to increase C3 expression. These cells were also transfected with various concentrations of the C3-3 candidate sequence. Real Time PCR for C3 mRNA expression after 48 hours of stimulation indicated that this siRNA sequence produced a reduction in C3 expression compared to stimulated cells treated with no siRNA. Measurements were normalized to unstimulated C3 mRNA expression in cells (dotted line) .
  • Figure 3 is a bar graph that shows the relative expression of C3 mRNA levels in transplanted rat kidneys. The kidneys were untreated or treated with nanoparticles containing various amounts of scrambled or C3 specific siRNA before transplantation.
  • Each data point contains data from 4 separate kidneys, and each PCR reaction was performed in triplicate.
  • C3 raRNA levels in these experimental conditions were compared to C3 mRNA levels in normal non-transplanted kidneys (NKC, normal kidney control) and transplanted kidneys untreated with siRNA (ISCH, ischaemic control) .
  • the figure demonstrates that C3 mRNA levels are lower in kidneys treated with C3 specific siRNA before transplantation as compared to C3 mRNA levels in normal non-transplanted kidneys and transplanted kidneys untreated with C3 specific siRNA.
  • the C3 specific siRNA was packaged with various ratios of PolyTran, labelled in Figure 1 as follows: C3 , lO ⁇ g C3 siRNA in PolyTran at 1:4.5; C3 naked, lO ⁇ g C3 siRNA with no PolyTran; C3 3:1, lO ⁇ g C3 siRNA in PolyTran at 1:3; C3 1.5:1, lO ⁇ g C3 siRNA in PolyTran at 1:1.5.
  • two sets of kidneys were treated with scrambled siRNA before transplantation: FITC, lO ⁇ g scrambled FITC-labeled siRNA; SCRAM CON, lO ⁇ g scrambled non-labeled siRNA.
  • FIG 4 is a set of two panels showing histological analysis of transplanted rat kidneys.
  • the upper panel shows a non-treated kidney 48 hours after transplantation.
  • the histopathology reveals widespread tubular attenuation and tubule dilation indicative of acute tubular necrosis (ATN) .
  • ATN acute tubular necrosis
  • This particular pathology is linked to the initial non-function of transplanted tissue after transplantation.
  • the lower panel depicts a kidney pre-treated with C3 siRNA (in 1:4.5 ratio with PolyTran) at 48 hours after transplantation.
  • the histopathology of this kidney exhibits less ATN.
  • Figure 5 shows two bar graphs presenting the results of an experiment serving to identify short peptides that can be used to target siRNA-comprising nanoparticles to specific organs. Phage display was used to identify candidate peptides that are concentrated in the transplanted kidney.
  • the upper panel of Figure 5 shows illustrative data for one experiment, with increasing concentrations of phage (in plague forming units per gram of tissue) retrieved from the kidneys after three rounds of phage library injection, retrieval, and expansion.
  • streptavidin was used as a target for phage binding (R3vsStrep) .
  • the lower panel of Figure 5 shows the number of phage retrieved after the third round of biopanning in the recipient's transplanted kidney (Tx kidney), normal kidney (N kidney), pancreas, heart, and lungs.
  • the data shows selectivity in phage horning into the transplanted kidney compared to the numbers of phage retrieved from other organs .
  • oligonucleotides and similar terms based on this relate to short polymers composed of naturally occurring nucleotides as well as to polymers composed of synthetic or modified nucleotides, as described in the immediately preceding paragraph. Oligonucleotides may be 10 or more nucleotides in length, or 15, or 16, or 17, or 18, or
  • An oligonucleotide that is an siRNA may have any number of nucleotides between 15 and 30 nucleotides. In many embodiments an siRNA may 006/045933
  • nucleotides between 21 and 25 nucleotides .
  • an siRNA may have two blunt ends, or two sticky ends, or one blunt end with one sticky end, or one end with over hang.
  • the over hang nucleotides can be ranged from one to four or more .
  • RNA interference RNA interference
  • gene expression of immunomodulatory or immunoeffector gene targets is attenuated by RNA interference.
  • Expression products of a immunomodulatory or immunoeffector gene are targeted by specific double stranded siRNA nucleotide sequences that are complementary to at least a segment of the immunomodulatory or immunoeffector gene target sequence that contains any number of nucleotides between 15 and 30, or in many cases, contains anywhere between 21 and 25 nucleotides, or more.
  • the target may occur in the 5' untranslated (UT) region, in a coding sequence, or in the 3' UT region.
  • a targeting polynucleotide according to the invention includes an siRNA oligonucleotide.
  • siRNA can also be prepared by chemical synthesis of nucleotide sequences identical or similar to an intended sequence.
  • a targeting siRNA can be obtained using a targeting polynucleotide sequence, for example, by digesting an immunomodulatory or immunoeffector ribopolynucleotide sequence in a cell-free system, such as, but not limited to, a Drosophila extract, or by transcription of recombinant double stranded cRNA.
  • siRNA duplexes composed of a 16-30 nt sense strand and a 16-30 nt antisense strand of the same length.
  • each strand of an siRNA paired duplex has in addition a 2-nt overhang at the 3' end.
  • the sequence of the 2-nt 3 ' overhang makes an additional small contribution to the specificity of siRNA target recognition.
  • the nucleotides in the 3' overhang are ribonucleotides.
  • the nucleotides in the 3' overhang are deoxyribonucleotides . Use of 3' deoxynucleotides provides enhanced intracellular stability.
  • a recombinant expression vector of the invention when introduced within a cell, is processed to provide an RNA that comprises an siRNA sequence targeting an immunomodulatory or immunoeffector gene within the organ.
  • a vector may be a DNA molecule cloned into an expression vector comprising operatively-linked regulatory sequences flanking the immunomodulatory or immunoeffector gene targeting sequence in a manner that allows for expression.
  • an RNA molecule that is antisense to the target RNA is transcribed by a first promoter (e.g., a promoter sequence 3 ' of the cloned DNA) and an RNA molecule that is the sense strand for the RNA target is transcribed by a second promoter (e.g., a promoter sequence 5' of the cloned DNA) .
  • the sense and antisense strands then hybridize in vivo to generate siRNA constructs targeting an immunomodulatory or immunoeffector gene sequence.
  • two constructs can be utilized to create the sense and anti-sense strands of an siRNA construct.
  • cloned DNA can encode a transcript having secondary structure, wherein a single transcript has both the sense and complementary antisense sequences from the target gene or genes.
  • a hairpin RNAi product is similar to all or a portion of the target gene.
  • a hairpin RNAi product is an siRNA.
  • the regulatory sequences flanking the immunomodulatory or immunoeffector gene sequence may be identical or may be different, such that their expression may be modulated independently, or in a temporal or spatial manner.
  • siRNAs are transcribed intracellularly by cloning the immunomodulatory or immunoeffector gene sequences into a vector containing, e.g., an RNA pol III transcription unit from the smaller nuclear RNA (snRNA) U6 or the human RNase P RNA Hl.
  • a vector system is the GeneSuppressorTM RNA Interference kit (Imgenex Corp.) .
  • the U6 and Hl promoters are members of the type III class of Pol III promoters.
  • the +1 nucleotide of the U6-like promoters is always guanosine, whereas the +1 for Hl promoters is adenosine.
  • the termination signal for these promoters is defined by five consecutive thymidines.
  • the transcript is typically cleaved after the second uridine. Cleavage at this position generates a 3 ' UU overhang in the expressed siRNA, which is similar to the 3 ' overhangs of synthetic siRNAs.
  • Any sequence less than 400 nucleotides in length can be transcribed by these promoter, therefore they are ideally suited for the expression of around 21-nucleotide siRNAs in, e.g., an approximately 50 nucleotide RNA stem loop transcript.
  • the characteristics of RNAi and of factors affecting siRNA efficacy have been studied (See, e.g., Elbashir, Lendeckel and Tuschl (2001). Genes & Dev. 15: 188- 200) .
  • the targeting polynucleotide is generally 300 nucleotides in length or less, and includes a first nucleotide sequence that targets a gene sequence present in cells of the donated organ, or in passenger cells accompanying the donated organ once removed from the donor, and that is implicated in immunomodulatory or immunoeffector responses when a donated organ is introduced within a recipient subject.
  • a first nucleotide sequence that targets a gene sequence present in cells of the donated organ, or in passenger cells accompanying the donated organ once removed from the donor, and that is implicated in immunomodulatory or immunoeffector responses when a donated organ is introduced within a recipient subject.
  • any T (thymidine) or any U (uridine) may optionally be substituted by the other.
  • the first nucleotide sequence consists of a) a sequence whose length is any number of nucleotides from 15 to 30, or more, or b) a complement of a sequence given in a) .
  • polynucleotide may be termed a linear polynucleotide herein.
  • a single stranded polynucleotide frequently is one strand of a double stranded siRNA.
  • the polynucleotide described above further includes a second nucleotide sequence separated from the first nucleotide sequence by a loop sequence, such that the second nucleotide sequence . a) has substantially the same length as the first nucleotide sequence, and b) is substantially complementary to the first nucleotide sequence.
  • hairpin polynucleotide the first nucleotide sequence hybridizes with the second nucleotide sequence to form a hairpin whose complementary sequences are linked by the loop sequence.
  • A. hairpin polynucleotide is digested intracellularly to form a double stranded siKNA.
  • the targets of the linear polynucleotide and of the hairpin polynucleotide are a gene sequence present in cells of the donated organ, or in passenger cells accompanying the donated organ, and the first nucleotide sequence is either.
  • the length of the first nucleotide sequence is any number of nucleotides from 21 to 25.
  • a linear polynucleotide or a hairpin polynucleotide consists of a targeting sequence that targets a sequence chosen from Tables 1- 15, and optionally includes a dinucleotide overhang bound to the 3' of the chosen sequence.
  • the dinucleotide sequence at the 3' end of the first nucleotide sequence is TT, TU, UT, or UU and includes either ribonucleotides or deoxyribonucleotides or both.
  • a linear or hairpin polynucleotide may be a DNA, or it may be an RNA, or it may be composed of both deoxyribonucleotides and ribonucleotides.
  • siRNA oligos specific to particular human genes are listed in Tables Ia to 15b below.
  • the tables include both 21 mers with overhang and 25 mers with blunt ends for all the genes listed.
  • sequences of potential siRNA oligos specific to genes of other mammalian animals that are the transplantation donors should be designed in reference to the corresponding human genes but with the gene sequences of those animals in mind.
  • C3 gene Homo sapiens complement component 3 (C3) , Accession: NM_000064, Gene ID: 4557384, 25 siRNA candidates were selected targeting the following gene sequences :
  • ICAMl gene Homo sapiens intercellular adhesion molecule 1 (CD54) , human rhinovirus receptor (ICAMl) , Accession: NM_000201, Gene ID: 4557877, 19 siRNA candidates were selected targeting the following gene sequences : Table 2a. 23 mer DNA sense strand sequences:
  • VCAMl gene Homo sapiens vascular cell adhesion molecule 1 (VCAMl) , Transcript variant 2, mRNA.
  • VCAM-I Human mRNA for vascular cell adhesion molecule 1
  • X53051.11 HSVCAMl [37648]
  • siRNA candidates were selected to target the following gene sequences:
  • siRNA sequences targeting human IFN-gamma (Accession, NM_000619) :
  • siRNA sequences targeting human IL-I (Accession: NM_033292) :
  • siRNA sequences targeting human IL-6 (Accession NM_000600) :
  • siRNA sequences targeting human IL-8 (Accession: NM_000584) :
  • Table 8 siRNA sequences targeting human TNF- ⁇ (Accession: NM_004862) :
  • siRNA sequences targeting human CD86 (Accession: NM__175862) :
  • siRNA sequences targeting human MHC-II (Accession: NM_002119) :
  • siRNA sequences targeting human CD28 (Accession: NM_006139) :
  • siRNA sequences targeting human parvovirus B19 (Accession: AY903437) :
  • siRNA duplexes of 25 basepair with blunt ends exhibit more potent gene knockdown efficacy than 19 basepair with overhang at both 3' ends, both in vitro and in vivo.
  • the invention provides a double stranded polynucleotide that includes a first linear polynucleotide strand described above and a second polynucleotide strand that is complementary to at least the first nucleotide sequence of the first strand and is hybridized thereto to form a double stranded siRNA composition.
  • a variety of carriers serve to prepare formulations or pharmaceutical compositions containing siRNAs.
  • the siRNA polynucleotides of the invention are delivered into cells in culture or into cells of an organ awaiting transplantation by liposome-mediated transfection, for example by using commercially available reagents or techniques, e.g., OligofectamineTM, LipofectAmineTM reagent, LipofectAmine 2000TM (Invitrogen) , as well as by electroporation, and similar techniques.
  • the pharmaceutical compositions containing the siRNAs include additional components that protect the stability of siRNA, prolong siRNA lifetime, potentiate siRNA function, or target siRNA to specific tissues/cells.
  • biodegradable polymers include a variety of biodegradable polymers, cationic polymers (such as polyethyleneimine) , cationic copolypeptides such as histidine-lysine (HK) polypeptides see, for example, PCT publications WO 01/47496 to Mixson et al . , WO 02/096941 to Biomerieux, and WO 99/42091 to Massachusetts Institute of Technology) , PEGylated cationic polypeptides, and ligand- incorporated polymers, etc.
  • cationic polymers such as polyethyleneimine
  • cationic copolypeptides such as histidine-lysine (HK) polypeptides see, for example, PCT publications WO 01/47496 to Mixson et al . , WO 02/096941 to Biomerieux, and WO 99/42091 to Massachusetts Institute of Technology
  • PEGylated cationic polypeptides and ligand- incorporated polymers, etc.
  • PolyTran solutions saline or aqueous solution of HK polymers and polysaccharides such as natural polysaccharides, also known as scleroglucan
  • TargeTran a saline or aqueous suspension of nano-particle composed of conjugated RGD-PEG-PEI polymers including a targeting ligand
  • surfactants Infasurf ; Forest
  • Infasurf ® calfactant
  • calfactant is a natural lung surfactant isolated from calf lung for use in intratracheal instillation; it contains phospholipids, neutral lipids, and hydrophobic surfactant-associated proteins B and C.
  • the polymers can either be uni-dimensional or multi-dimensional, and also could be microparticles or nanoparticles with diameters less than 20 microns, between 20 and 100 microns, or above 100 micron.
  • the said polymers could carry ligand molecules specific for receptors or molecules of special tissues or cells, thus be used for targeted delivery of siRNAs.
  • the siRNA polynucleotides are also delivered by cationic liposome based carriers, such as DOTAP, DOTAP/Cholesterol (Qbiogene, Inc.) and other types of lipid aqueous solutions.
  • low percentage (5-10%) glucose aqueous solution, and Infasurf are effective carriers for airway delivery of siRNA (Li B.J.
  • a carrier may include Hyper Osmolar Citrate solution (560 m ⁇ sm/kg solution of meglumine hydrochloride, 560 m ⁇ sm/kg meglumine ioxaglate, and 600 m ⁇ sm/kg sodium ioxaglate, and so forth) .
  • University of Wisconsin solution has the potential to enhance and extend heart, kidney, lung and liver preservation. University of Wisconsin solution is widely accepted for the cold storage and transport of human donor pancreata destined for islet isolation.
  • the composition may further comprise a polymeric carrier.
  • the polymeric carrier may comprise a cationic polymer that binds to the RNA molecule.
  • the cationic polymer may be an amino acid copolymer, comprising, for example, histidine and lysine residues.
  • the polymer may comprise a branched polymer.
  • the composition may comprise a targeted synthetic vector.
  • the synthetic vector may comprise a cationic polymer, a hydrophilic polymer, and a targeting ligand.
  • the polymer may comprise a polyethyleneimine
  • the hydrophilic polymer may comprise a polyethylene glycol or a polyacetal
  • the targeting ligand may comprise a peptide comprising an RGD sequence .
  • the siRNA/carrier may be formulated in either the storage solution or the perfusion medium in a nonspecific manner, or via the systemic circulation in a targeted delivery system.
  • the present invention provides methods for prevention of allograft rejection and ischemia/reperfusion injury in solid organ transplantation by silencing or down-regulation of a target gene expression by introducing RNA interference (siRNA) .
  • siRNA is applied to an organ intended for transplantation in the form of an organ-storage solution, i.e., after removal from the donor and while it is being transported to the recipient.
  • the donor or recipient of the transplanted organ, tissues, and/or cells can be a mammal, including, but not limited to, human, non- human mammal, non-human primate, rat, mouse, pig, dog, cow, and horse.
  • the organs destined for transplantation are maintained by an organ storage solution comprising one siRNA oligonucleotide or multiple siRNA oligonucleotides as a cocktail.
  • siRNA can access the donor organ and cells easily and selectively, which facilitates the reduction of potentially harmful systemic side effects.
  • donor organs are subjected to flushing and storage in static or recirculating systems, in hypothermic conditions (less than 37 0 C for humans, e.g. 4 0 C) or normothermic conditions (37 0 C for humans) , in specially formulated solutions (organ preservation solutions) in order to wash out debris and to decrease damage during transportation.
  • the methods of the present invention include siRNA transfection of the donor organ and cells during organ preservation.
  • siRNA transfection formulation is used for flushing the solid donor organ in situ and/or ex vivo, and for static or machine perfusion organ storage.
  • the formulated solution is useful for both local injection into the solid organ and to bathe the entire solid organ by submerging it in the siRNA formulation.
  • the siRNA agent can be used as either single or multiple duplexes, targeting single or multiple genes, with or without transfection carriers for the treatment of the transplanted organs (tissues) and cells.
  • the transfection agents include but are not limited to synthetic polymers, liposomes and sugars, etc.
  • the siRNA agents can also be used with other agents such as small molecule and monoclonal antibody inhibitors, immune modulators and other types of oligonucleotides.
  • the injection of and submerging of organs for transplantation with the siRNA/carrier solution will minimize tissue damage and host rejection, and therefore, will enhance the success of the transplanted organ in terms of organ function and survival and the minimization of co-morbidities.
  • organs and cells can be treated by siRNA/carrier formulation during the process of transplantation. All solid organ transplantations essentially require surgical preparation of the donor, which may include flush perfusion of the body, or of specific organs to be used in transplantation. Perfusion may be with one or more fluids.
  • the organ (s) are removed for storage during transportation to the recipient, and the organ is surgically implanted into the recipient .
  • Organs useful in the methods of the invention include, but are not limited to, kidney, liver, heart, pancreas, pancreatic islets, small bowel, lung, cornea, limb, and skin, as well as cells in culture corresponding to each of those organs.
  • hepatocyte cell lines are beginning to be developed as universal donors for isolated liver cell transplantation, which is a less invasive method than orthotopic liver transplantation for treatment of metabolic liver disease. Costimulation via pathways such as CD28/B7 or CD40/CD40L is a major concern for the success of such transplantation (2) . Therefore, using siRNA/carrier formulation to silence both CD28 or CD40 pathways will be a good strategy to improve the success rate of the transplant.
  • Another example for renal transplant failure is the infection of parvovirus B19 (PV-B19) after solid organ transplantation which may cause pure red cell aplasia (PRCA) . PV-B19 infection in immunosuppressed transplant recipients is associated with significant morbidity (1) .
  • siRNA to inhibit PV-B19 or any other viral infection and replication is an adjunct therapy for improvement in renal transplant by treatment of both donor organ and transplant recipient during the initial phase of the transplantation.
  • the present invention provides compositions comprising one or more siRNA duplexes in which siRNA can simultaneously target several genes involved in allograft or xenograft rejection or ischemia/reperfusion injury.
  • a combination of multiple siRNA duplexes could be more effective for inhibition of allograft rejection or ischemia/reperfusion injury.
  • the process of immune modulation offers a plethora of molecular targets for siRNA silencing using the methods of the invention such as (1) molecules on lymphocytes associated with activation; (2) molecules on antigen presenting cells (APCs) which stimulate lymphocytes such as MHC class II and costimulatory molecules; (3) soluble molecular signals such as cytokines such as TNF- ⁇ , IFN- ⁇ , IL-I, IL-6, IL-8; (4) molecules associated with lymphocyte extravasation and homing such as Vascular Cell Adhesion Molecule-1, Intercellular Adhesion Molecular-1; and (5) effector molecules of immunity such as but not limited to complement factor C3.
  • Additional candidate target genes include Intercellular Adhesion Molecule-1, Major Histocompatibility Complex Class I, Major Histocompatibility Complex Class II, IFN- ⁇ , CD80, CD86, CD40 and CD40L.
  • the present invention also provides methods and compositions for using siRNA oligo cocktail (siRNA- OC) as therapeutic agent useful in the methods of the invention or to achieve more potent antiangiogenesis efficacy for treatment of cancer and inflammations .
  • This siRNA oligo cocktail comprises at least three duplexes targeting at least three mRNA targets.
  • the siRNA oligo cocktail may comprise any of the siRNA sequences listed in tables 1-15.
  • the siRNA oligo cocktail comprises the siRNAs specific for complement C3 , MHC-II, and IFN ⁇ .
  • the present invention is based on two important aspects: first, the siRNA duplex is a very potent gene expression inhibitor, and each siRNA molecule is made of short double-stranded RNA oligo (21-23nt, or 24-25nt, or 26- 29nt) with the same chemistry property; Second, allograft or xenograft rejection and ischemia/reperfusion injury relate, in part, to overexpressions of endogenous genes. Therefore, using siRNA-OC targeting multiple genes represents an advantageous therapeutic approach, due to the chemical uniformity of siRNA duplexes and synergistic effect from down regulation of multiple disease- or injury- causing genes.
  • the invention defines that siRNA-OC is a combination of siRNA duplexes targeting at lease three genes, at various proportions, at various physical forms, and being applied through the same route at the same time, or different route and time into disease tissues.
  • siRNA-mediated silencing can be applied with either single siRNA targeting one such gene or a combination of multiple siRNAs targeting several target sequences within the same gene, or targeting various genes from different categories such as those identified in this paragraph.
  • a composition comprising multiple siRNA duplexes may have each present with the same or different ratios.
  • duplex I, duplex II and duplex III may either each be present at 33.3% (w/w) of total siRNA agent each, or at 20%, 45% and 35% respectively, by way of nonlimiting example.
  • RNA interference blocks gene expression according to small unique segments of their sequence. This natural process can be exploited to reduce transcription of specific genes.
  • donor derived complement C3 is rapidly upregulated in ischemia/reperfusion injury (I/RI) , contributing to tissue damage.
  • I/RI ischemia/reperfusion injury
  • Complement C3 is described as a local mediator of various forms of injury and immune regulation and is a valid target for gene knockdown after transplant ischemia/reperfusion injury that may well assist in the regulation of allo- immunity as well .
  • This study sought to exploit siRNA to knock-down C3 gene expression in donor organs.
  • Rat renal epithelial cell lines were stimulated with 10 ⁇ g/ml IL-I and 0.1 ⁇ g/ml IL-6 to upregulate C3 gene expression. 72 hours after stimulation, the cells were transfected with one of a panel of C3-specific siRNAs.
  • C3 expression was determined by Real Time PCR. Results showed that C3 expression was upregulated in non-transfected cells after stimulation ( Figure 1) . Cells treated with siRNA showed up to a 60% reduction of C3 expression as compared to control cells that were not treated with siRNA. These experiments identified the most effective C3 siRNA sequence from the panel that did not non- specifically induce IFN ⁇ upregulation, a potential off- target effect of siRNA (labelled as C3-3 siRNA in Figure 1) .
  • the candidate C3 siRNA obtained in the previous experiment was transfected into rat renal epithelial cells stimulated to express C3 , as described above.
  • Example 2 siRNA mediated C3 expression knockdown in vivo
  • HHHKHHHKHHHKHHH 2KH4NH4.
  • the polymers were selected because of their in vitro or in vivo efficacy for different nucleic acid forms.
  • the branched HK polymer was dissolved in aqueous solution and then mixed with siRNA aqueous solution at the listed ratios by mass, forming nanoparticles of average size of 150- 200 nm in diameter.
  • the HKP-siRNA aqueous solutions were semi-transparent without noticeable aggregation of precipitate. These solutions can be stored at 4°C for at least three months .
  • the nanoparticles were added to Hyper Osmolar Citrate perfusion fluid and administered to donor rat kidneys. After 4 hours of cold ischemia, the kidneys were transplanted into syngeneic hosts. Two days later the kidneys were harvested and C3 gene expression was determined by Real-Time PCR. Non-transplanted, non- treated kidneys served as a negative control (labelled NKC in Figure 3) , while perfused, transplanted kidneys not treated with siRNA served as a positive control (labelled as ISCH in Figure 3) . The levels in the siRNA-treated kidneys were normalized to mRNA levels in non-transplanted, non-treated kidneys. Results are shown in Figure 3.
  • the FITC-labelled scrambled siRNA controls exhibited a greater upregulation of C3 gene expression than the untreated kidneys, suggestive of off-target effects.
  • siRNA inhibition of C3 gene expression effectively reduced local C3 activity compared to controls.
  • the nanoparticle strategy appears to overcome the problem of effective siRNA delivery. It now appears possible to develop arrays of specific siRNA to diminish pro-inflammatory gene expression in donor organs as adjunct therapies to conventional immunosuppression or tolerance induction.
  • Example 3 Determination of peptide sequences concentrated in transplanted kidneys by phage display [0061]
  • peptides concentrated in the organ of interest can be identified by phage display. This method was used to identify candidate target peptides in the rat model of kidney transplantation described above. Donor kidneys were flushed with Hyper Osmolar Citrate and stored at 4°C for 4 hours before transplantation into a syngeneic host. After 48 hours, recipients were anaesthetized and injected via the tail vein with the prepared cysteine-constrained 7mer phage library (New England Biolabs) .
  • the transplanted kidneys were harvested and phage extracted from the kidney, in a first round of "in vivo biopanning" .
  • the extracted phage were expanded in E. coli bacteria before being injected into another kidney transplant recipient. This biopanning was repeated for a total of three rounds. After each round, a sample of phage was taken to estimate the numbers present in the transplanted kidney. After each expansion, a sample of phage was grown in bacterial colonies on agar plates so that phage could be isolated and the DNA sequence of the expressed library peptide could be determined.
  • Figure 5 shows increasing numbers of phage retrieved from transplanted kidneys after each round of biopanning (random phage) , as compared to a control targeting streptavidin (R3vsStrep) .
  • Examples of identified peptide sequences concentrated in the kidney are C-LPSPKRT-C, C-LPSPKKT-C, C-PTSVPKT-C.
  • phage are concentrated in the transplanted kidney and are found in much lower numbers in other organs of the recipient ( Figure 5, lower panel) .
  • the candidate peptides can be incorporated into TargeTran nanoparticles to provide specificity for siRNA targeting to transplanted organs.

Abstract

This invention describes compositions and methods using siRNA to target various genes expressed in cells of transplanted organs or tissues and/or genes expressed in the host to improve the success of the transplantation.

Description

COMPOSITIONS AND METHODS OF USING siRNA
TO KNOCKDOWN GENE EXPRESSION AND TO IMPROVE SOLID ORGAN AND CELL TRANSPLANTATION
[0001] This application claims the benefit of United States provisional application no. 60/741,157, the entire disclosure of which is incorporated herein by- reference .
Field of the Invention
[0002] The present invention provides compositions and methods for the prevention of allograft rejection or xenograft rejection and ischemia/reperfusion injury in solid organ or tissue transplantation using siRNA- mediated down regulation of gene expression.
Background of the Invention [0003] Solid organ transplantation is the only- effective therapy for the treatment of end-stage organ failure (1, 2) . Transplant programs around the world have become increasingly successful and such operations are becoming increasingly routine (3, 4) . Despite the impressive results of one-year survival rates, organ transplantation still faces major problems. The immune system poses the most significant barrier to the long term survival of the transplanted organs. Without life long treatment with powerful immunosuppressive agents to keep the immune response at bay, organ grafts will invariably be rejected. However, current anti- rejection drugs reduce systemic immunity nonselectively and increase the risk of opportunistic infections and tumour development on the long term. Therefore, alternative strategies are being sought. [0004] The advancement of molecular techniques over the past decade has improved our understanding of the signals necessary to elicit both an immune response and ischemia/reperfusion injury. Agents designed to target these novel signals provide hope that they will eventually allow for the long-term, drug-free acceptance of transplanted organs.
[0005] Transplantation immunology refers to an extensive sequence of events that occurs after an allograft or a xenograft is removed from a donor and then transplanted into a recipient . Tissue is damaged at both the graft and the transplantation sites. An inflammatory reaction follows immediately, as does activation of biochemical cascades . A series of specific and nonspecific cellular responses ensues as antigens are recognized. Eventually, the damage is controlled through tissue repair and reinforcement; if damage is nonpathologic, the graft survives. [0006] Antigen-independent causes of tissue damage (i.e., ischemia, hypothermia, reperfusion injury) are the result of mechanical trauma as well as disruption of the blood supply as the graft is harvested.
[0007] In contrast, antigen-dependent causes of tissue damage involve immune-mediated damage. Macrophages release cytokines (e.g., tumour necrosis factor, interleukin-1) , which heighten the intensity of inflammation by stimulating inflammatory endothelial responses; these endothelial changes help recruit large numbers of T cells to the transplantation site. Damaged tissues release proinflammatory mediators (e.g., Hageman factor [factor XII]) that trigger several biochemical cascades. The clotting cascade induces fibrin and several related fibrinopeptides, which promote local vascular permeability and attract neutrophils and macrophages . The kinin cascade principally produces bradykinin, which promotes vasodilation, smooth muscle contraction, and increased vascular permeability. [0008] The formation of an antibody-antigen complex (i.e., immune complex) activates the classic pathway of the complement system. CIq triggers the activation process when it docks onto antibodies within the immune complexes via the classical pathway, whilst complement factor C3 can recognize damaged cell surfaces as acceptors for alternative pathway activation.
Activated complement causes damage through the deposition of the membrane attack complex (e.g., C5b, C6, C7, C8, C9) and cell -bound ligands, such as C4b and C3b, which activate leukocytes bearing complement receptors. In addition, production of bioactive anaphylatoxins C5a and C3a causes the influx and activation of inflammatory cells. These chemoattractants also initiate mast cell degranulation, which releases several mediators. Histamine and 5- hydroxytryptamine increase vascular permeability.
Prostaglandin E2 promotes vasodilation and vascular permeability. Leukotrienes B4 and D2 promote leukocyte accumulation and vascular permeability. Another means - A -
by which complement is activated is through tissue ischemia and reperfusion, which exposes phospholipids and mitochondrial proteins. These by-products activate complement directly through binding CIq or mannose- binding lectin or factor C3b.
[0009] Currently, successful transplantation of allografts requires the systemic use of immunosuppressive drugs. These can cause serious morbidity due to toxicity and increased susceptibility to cancer and infections. Local production of immunosuppressive molecules limited to the graft site would reduce the need for conventional, generalized immunosuppressive therapies and thus educe fewer side effects. This is particularly salient in a disease like type 1 diabetes, which is not immediately life- threatening yet islet allografts can effect a cure. Anti-CD4 strategy may be even more effective when a combination of antibodies are used; similar strategies may also prevent xenograft rejection. Suppressing the host's immune responses also increases the risk of cancer. Attempts to suppress the immune response to avoid graft rejection and graft versus host disease (GVHD) weaken the ability of the body to combat infectious agents (e.g., bacteria, viruses, fungi, etc.) .
[0010] RNA interference (RNAi) compounds, the intermediate short interfering RNA oligonucleotides (siRNAs) , provide a unique strategy for using a combination of multiple siRNA duplexes to target multiple disease-causing genes in the same treatment, since all siRNA duplexes are chemically homogenous with the same source of origin and the same manufacturing process (5, 6, 7, 8) . Such siRNA inhibitors are expected to have much better clinical efficiency with minimum toxicity and safety concerns. Genetic modification is a promising therapeutic strategy for organ transplantation. Based on the attractive technology of RNA interference for silencing a particular gene expression (9, 10) , siRNA therapy may represent an attractive and powerful approach in preventing ischemia/reperfusion injury as well as organ rejection in transplant recipients.
Summary of the Invention
[0011] This invention provides targeting polynucleotides that target immunomodulatory or immunoeffector genes present in cells of an organ to be donated to a recipient . Targets for these polynucleotides can be derived from sequences of immunomodulatory and immunoeffector genes listed in Tables 1-15 (see below) . For example, the targeting polynucleotide may target sequences in the C3 , ICAMl, VCAM-I, IFN-Y, IL-I, IL-6, IL-8, TNF-α, CD80, CD86, MHC-II, MHC-I, CD28, CTLA-4, or PV-B19 genes. The targeting polynucleotides can comprise siRNA duplexes that target one or more of the sequences listed in Tables 1-15. The targeting polynucleotide may be a single-stranded linear polynucleotide, a double- stranded linear polynucleotide, or a hairpin polynucleotide .
[0012] This invention also provides a method of suppressing rejection of a transplanted organ by contacting the organ with a composition comprising the targeting polynucleotide of the invention before transplanting the organ into a recipient. The method can be effective in down-regulating or inhibiting the expression of a target immunomodulatory or immunoeffector gene in an organ or a cell of an organ during storage before transplantation. In one embodiment, the organ is perfused with a composition comprising a targeting polynucleotide of the invention. In another embodiment, the organ is bathed or submerged in the composition comprising a targeting polynucleotide of the invention. The composition can also be administered to an organ recipient. In some embodiments of the invention, the organ may be the recipient's own organ. The recipient of the said organ can be human. Organs, tissues, and cells contacted with the composition comprising a targeting polynucleotide of the invention include the kidney, liver, lung, pancreas, heart, small bowel, cornea, epithelial cells, vascular endothelium, vascular smooth muscle cells, myocardium and passenger leukocytes resident in the organ at the time of transplantation. [0013] The composition comprising the targeting polynucleotide of the invention can also comprise a carrier, including, but not limited to, perfusion fluid, Hyper Osmolar Citrate solution, PolyTran polymer solution, TargeTran nanoparticle solution, or University of Wisconsin solution. The composition can also comprise small molecule drugs, monoclonal antibody drugs , and other immune modulators . In some embodiments the composition comprises a plurality of the targeting polynucleotide of the invention. A composition can contain a plurality of targeting polynucleotides of the invention that can target a plurality of gene sequences. In one embodiment, the targeting polynucleotides are a cocktail that targets the C3, TNF-α, and. IL-8 gene sequences. Brief Description Of The Drawings
[0014] Figure 1 is a bar graph that shows the relative expression of C3 mRNA in rat renal cells. The cells were stimulated with IL-I and IL-6 to increase C3 expression. Three candidate C3 siRNA sequences (C3-1, C3-2, C3-3) or FITC-labelled scrambled siRNA were transfected into the cells at various concentrations. One set of cells was treated with Lipofectamine and no siRNA (+ lipofectamine) while another set was stimulated to produce C3 and treated with neither
Lipofectamine nor siRNA (- lipofectamine) . C3 mRNA levels were measured in the cells by Real Time PCR 48 hours after transfection. The dotted line indicates unstimulated cell C3 expression. The experiment showed the feasibility and efficacy of gene knockdown by siRNA. The C3-3 siRNA was selected as the candidate to use in further experiments .
[0015] Figure 2 is a bar graph showing the relative expression of C3 mRNA in rat renal cells stimulated with IL-I and IL-6 to increase C3 expression. These cells were also transfected with various concentrations of the C3-3 candidate sequence. Real Time PCR for C3 mRNA expression after 48 hours of stimulation indicated that this siRNA sequence produced a reduction in C3 expression compared to stimulated cells treated with no siRNA. Measurements were normalized to unstimulated C3 mRNA expression in cells (dotted line) . [0016] Figure 3 is a bar graph that shows the relative expression of C3 mRNA levels in transplanted rat kidneys. The kidneys were untreated or treated with nanoparticles containing various amounts of scrambled or C3 specific siRNA before transplantation. Each data point contains data from 4 separate kidneys, and each PCR reaction was performed in triplicate. C3 raRNA levels in these experimental conditions were compared to C3 mRNA levels in normal non-transplanted kidneys (NKC, normal kidney control) and transplanted kidneys untreated with siRNA (ISCH, ischaemic control) . The figure demonstrates that C3 mRNA levels are lower in kidneys treated with C3 specific siRNA before transplantation as compared to C3 mRNA levels in normal non-transplanted kidneys and transplanted kidneys untreated with C3 specific siRNA. The C3 specific siRNA was packaged with various ratios of PolyTran, labelled in Figure 1 as follows: C3 , lOμg C3 siRNA in PolyTran at 1:4.5; C3 naked, lOμg C3 siRNA with no PolyTran; C3 3:1, lOμg C3 siRNA in PolyTran at 1:3; C3 1.5:1, lOμg C3 siRNA in PolyTran at 1:1.5. In order to test the requirement for siRNA specificity, two sets of kidneys were treated with scrambled siRNA before transplantation: FITC, lOμg scrambled FITC-labeled siRNA; SCRAM CON, lOμg scrambled non-labeled siRNA. [0017] Figure 4 is a set of two panels showing histological analysis of transplanted rat kidneys. The upper panel shows a non-treated kidney 48 hours after transplantation. The histopathology reveals widespread tubular attenuation and tubule dilation indicative of acute tubular necrosis (ATN) . This particular pathology is linked to the initial non-function of transplanted tissue after transplantation. The lower panel depicts a kidney pre-treated with C3 siRNA (in 1:4.5 ratio with PolyTran) at 48 hours after transplantation. The histopathology of this kidney exhibits less ATN.
[0018] Figure 5 shows two bar graphs presenting the results of an experiment serving to identify short peptides that can be used to target siRNA-comprising nanoparticles to specific organs. Phage display was used to identify candidate peptides that are concentrated in the transplanted kidney. The upper panel of Figure 5 shows illustrative data for one experiment, with increasing concentrations of phage (in plague forming units per gram of tissue) retrieved from the kidneys after three rounds of phage library injection, retrieval, and expansion. In a control experiment, streptavidin was used as a target for phage binding (R3vsStrep) . The lower panel of Figure 5 shows the number of phage retrieved after the third round of biopanning in the recipient's transplanted kidney (Tx kidney), normal kidney (N kidney), pancreas, heart, and lungs. The data shows selectivity in phage horning into the transplanted kidney compared to the numbers of phage retrieved from other organs .
Detailed Description Of The Invention
[0019] As used herein, "oligonucleotides" and similar terms based on this relate to short polymers composed of naturally occurring nucleotides as well as to polymers composed of synthetic or modified nucleotides, as described in the immediately preceding paragraph. Oligonucleotides may be 10 or more nucleotides in length, or 15, or 16, or 17, or 18, or
19, or 20 or more nucleotides in length, or 21, or 22, or 23, or 24 or more nucleotides in length, or 25, or 26, or 27, or 28 or 29, or 30 or more nucleotides in length, 35 or more, 40 or more, 45 or more, up to about 50, nucleotides in length. An oligonucleotide that is an siRNA may have any number of nucleotides between 15 and 30 nucleotides. In many embodiments an siRNA may 006/045933
- 10 -
have any number of nucleotides between 21 and 25 nucleotides .
[0020] In many embodiments, an siRNA may have two blunt ends, or two sticky ends, or one blunt end with one sticky end, or one end with over hang. The over hang nucleotides can be ranged from one to four or more .
RNA interference (RNAi)
[0021] According to the invention, gene expression of immunomodulatory or immunoeffector gene targets is attenuated by RNA interference. Expression products of a immunomodulatory or immunoeffector gene are targeted by specific double stranded siRNA nucleotide sequences that are complementary to at least a segment of the immunomodulatory or immunoeffector gene target sequence that contains any number of nucleotides between 15 and 30, or in many cases, contains anywhere between 21 and 25 nucleotides, or more. The target may occur in the 5' untranslated (UT) region, in a coding sequence, or in the 3' UT region. See, e.g., PCT applications WO00/44895, WO99/32619, WOOl/75164, WOOl/92513, WO 01/29058, WO01/89304, WO02/16620, and WO02/29858, each incorporated by reference herein in their entirety. [0022] According to the methods of the present invention, immunomodulatory or immunoeffector gene expression, and thereby ischemia/reperfusion injury or organ transplant rejection due to an adverse immunological reaction, is suppressed using siRNA. A targeting polynucleotide according to the invention includes an siRNA oligonucleotide. Such an siRNA can also be prepared by chemical synthesis of nucleotide sequences identical or similar to an intended sequence. See, e.g., Tuschl, Zamore, Lehmann, Bartel and Sharp (1999) , Genes & Dev. 13: 3191-3197, incorporated herein by reference in its entirety. Alternatively, a targeting siRNA can be obtained using a targeting polynucleotide sequence, for example, by digesting an immunomodulatory or immunoeffector ribopolynucleotide sequence in a cell-free system, such as, but not limited to, a Drosophila extract, or by transcription of recombinant double stranded cRNA. [0023] Efficient silencing is generally observed with siRNA duplexes composed of a 16-30 nt sense strand and a 16-30 nt antisense strand of the same length. In many embodiments each strand of an siRNA paired duplex has in addition a 2-nt overhang at the 3' end. The sequence of the 2-nt 3 ' overhang makes an additional small contribution to the specificity of siRNA target recognition. In one embodiment, the nucleotides in the 3' overhang are ribonucleotides. In an alternative embodiment, the nucleotides in the 3' overhang are deoxyribonucleotides . Use of 3' deoxynucleotides provides enhanced intracellular stability. [0024] A recombinant expression vector of the invention, when introduced within a cell, is processed to provide an RNA that comprises an siRNA sequence targeting an immunomodulatory or immunoeffector gene within the organ. Such a vector may be a DNA molecule cloned into an expression vector comprising operatively-linked regulatory sequences flanking the immunomodulatory or immunoeffector gene targeting sequence in a manner that allows for expression. From the vector, an RNA molecule that is antisense to the target RNA is transcribed by a first promoter (e.g., a promoter sequence 3 ' of the cloned DNA) and an RNA molecule that is the sense strand for the RNA target is transcribed by a second promoter (e.g., a promoter sequence 5' of the cloned DNA) . The sense and antisense strands then hybridize in vivo to generate siRNA constructs targeting an immunomodulatory or immunoeffector gene sequence. Alternatively, two constructs can be utilized to create the sense and anti-sense strands of an siRNA construct. Further, cloned DNA can encode a transcript having secondary structure, wherein a single transcript has both the sense and complementary antisense sequences from the target gene or genes. In an example of this embodiment, a hairpin RNAi product is similar to all or a portion of the target gene. In another example, a hairpin RNAi product is an siRNA. The regulatory sequences flanking the immunomodulatory or immunoeffector gene sequence may be identical or may be different, such that their expression may be modulated independently, or in a temporal or spatial manner. [0025] In certain embodiments, siRNAs are transcribed intracellularly by cloning the immunomodulatory or immunoeffector gene sequences into a vector containing, e.g., an RNA pol III transcription unit from the smaller nuclear RNA (snRNA) U6 or the human RNase P RNA Hl. One example of a vector system is the GeneSuppressor™ RNA Interference kit (Imgenex Corp.) . The U6 and Hl promoters are members of the type III class of Pol III promoters. The +1 nucleotide of the U6-like promoters is always guanosine, whereas the +1 for Hl promoters is adenosine. The termination signal for these promoters is defined by five consecutive thymidines. The transcript is typically cleaved after the second uridine. Cleavage at this position generates a 3 ' UU overhang in the expressed siRNA, which is similar to the 3 ' overhangs of synthetic siRNAs. Any sequence less than 400 nucleotides in length can be transcribed by these promoter, therefore they are ideally suited for the expression of around 21-nucleotide siRNAs in, e.g., an approximately 50 nucleotide RNA stem loop transcript. The characteristics of RNAi and of factors affecting siRNA efficacy have been studied (See, e.g., Elbashir, Lendeckel and Tuschl (2001). Genes & Dev. 15: 188- 200) .
[0026] The targeting polynucleotide is generally 300 nucleotides in length or less, and includes a first nucleotide sequence that targets a gene sequence present in cells of the donated organ, or in passenger cells accompanying the donated organ once removed from the donor, and that is implicated in immunomodulatory or immunoeffector responses when a donated organ is introduced within a recipient subject. In the polynucleotide any T (thymidine) or any U (uridine) may optionally be substituted by the other. Additionally, in the polynucleotide the first nucleotide sequence consists of a) a sequence whose length is any number of nucleotides from 15 to 30, or more, or b) a complement of a sequence given in a) . Such a polynucleotide may be termed a linear polynucleotide herein. A single stranded polynucleotide frequently is one strand of a double stranded siRNA. [0027] In a related aspect, the polynucleotide described above further includes a second nucleotide sequence separated from the first nucleotide sequence by a loop sequence, such that the second nucleotide sequence . a) has substantially the same length as the first nucleotide sequence, and b) is substantially complementary to the first nucleotide sequence. [0028] In this latter structure, termed a hairpin polynucleotide, the first nucleotide sequence hybridizes with the second nucleotide sequence to form a hairpin whose complementary sequences are linked by the loop sequence. A. hairpin polynucleotide is digested intracellularly to form a double stranded siKNA.
[0029] In many embodiments the targets of the linear polynucleotide and of the hairpin polynucleotide are a gene sequence present in cells of the donated organ, or in passenger cells accompanying the donated organ, and the first nucleotide sequence is either. a) a targeting sequence that targets a sequence chosen from the sequences given in Tables 1-15 appended hereto; b) a targeting sequence longer than the sequence given in item a) wherein the targeting sequence targets a sequence chosen from Tables 1-15, c) a fragment of a sequence given in a) or b) wherein the fragment consists of a sequence of contiguous bases at least 15 nucleotides in length and at most one base shorter than the chosen sequence, d) a targeting sequence wherein up to 5 nucleotides differ from a sequence given in a) -c) , or e) a complement of any sequence given in a) to d) .
[0030] In various embodiments of a linear polynucleotide or a hairpin polynucleotide the length of the first nucleotide sequence is any number of nucleotides from 21 to 25.
[0031] In many embodiments a linear polynucleotide or a hairpin polynucleotide consists of a targeting sequence that targets a sequence chosen from Tables 1- 15, and optionally includes a dinucleotide overhang bound to the 3' of the chosen sequence. In yet additional embodiments of a linear polynucleotide or a hairpin polynucleotide the dinucleotide sequence at the 3' end of the first nucleotide sequence is TT, TU, UT, or UU and includes either ribonucleotides or deoxyribonucleotides or both. In various further embodiments a linear or hairpin polynucleotide may be a DNA, or it may be an RNA, or it may be composed of both deoxyribonucleotides and ribonucleotides.
[0032] Exemplary sequences of siRNA oligos specific , to particular human genes are listed in Tables Ia to 15b below. The tables include both 21 mers with overhang and 25 mers with blunt ends for all the genes listed. The sequences of potential siRNA oligos specific to genes of other mammalian animals that are the transplantation donors should be designed in reference to the corresponding human genes but with the gene sequences of those animals in mind.
Table 1 : siRNA targeted sequences in C3 gene:
C3 gene: Homo sapiens complement component 3 (C3) , Accession: NM_000064, Gene ID: 4557384, 25 siRNA candidates were selected targeting the following gene sequences :
Table Ia. 23 mer sequences :
# Position Sequence GC% Thermodynamic Values
1 1858-1880 AAGGGCGTGTTCGTGCTGAATAA 58 -6.9 { -13.5, -6.6 )
2 2797-2819 AAGGCTGCCGTCTACCATCATTT 58 -5.3 ( -12.1, -6.8 )
3 3053-3075 AACGGCTGAAGCACCTCATTGTG 58 -4.9 ( -11.7, -6.8 )
4 586-608 ΆAGCAGGACTCCTTGTCTTCTCA 53 -4.6 ( -12.1, -7.5 )
5 4163-4185 AACCAGCACCGGAAACAGAAAAG 53 -4.6 ( -11.5, -6.9 )
6 851-873 AΆGTGGAGGGAACTGCCTTTGTC 58 -4.5 ( -11.2, -6.7 )
7 805-827 AAGGGCCTGGAGGTCACCATCAC 68 -4.4 ( -14.4, -10. 0)
8 4903-4925 AAGCCCAACCTCAGCTACATCAT 58 -4.2 ( -13.2, -9.0 )
9 3572-3594 AAGCAGGAGACTTCCTTGAAGCC 53 -4.0 ( -12.1, -8.1 )
10 1161-1183 AATGCCCTTTGACCTCATGGTGT 53 -3.9 ( -12.7, -8.8 )
11 4118-4140 AΆGATCAACTCACCTGTAATAAA 37 -3.8 { -9.1, -5.3 )
12 4663-4685 AAGGCCTGTGAGCCAGGAGTGGA 68 -3.8 ( -13.2, -9.4 )
13 2598-2620 AATCCGAGCCGTTCTCTACAATT 53 -3.7 ( -10.9, -7.2 )
14 925-947 AAGCGCATTCCGATTGAGGATGG 53 -3.6 ( -12.5, -8.9 )
15 2848-2870 AAGGTCGTGCCGGAAGGAATCAG 63 -3.5 ( -11.4, -7.9 )
16 2770-2792 AAGACCGGCCTGCAGGAAGTGGA 68 -3.4 ( -11.4, -8.0 )
17 4843-4865 AAGCTGGAGGAGAAGAAACACTA 53 -3.4 ( -12.1, -8.7 )
18 2097-2119 AATGGACAAAGTCGGCAAGTACC 47 -3.4 ( -10.6, -7.2 )
19 4549-4571 AAGGAGGATGGAAAGCTGAACAA 53 -3.3 ( -12.1, -8.8 )
20 4183-4205 AAGAGGCCTCAGGATGCCAΆGAΆ 63 -3.3 ( -12.3, -9.0 )
21 337-359 AACAGGGAGTTCAAGTCAGAAAA 47 -3.2 ( -11.3, -8.1 )
22 1135-1157 AAGACACCCAAGTACTTCAAACC 42 -3.2 ( -10.1, -6.9 )
23 673-695 AAGATCCGAGCCTACTATGAΆΆA 47 -3.2 ( -10.3, -7.1 )
24 3890-3912 AAGCCTTGGCTCAATACCAAAAG 47 -3.1 ( -10.9, -7.8 )
25 4570-4592 AAGCTCTGCCGTGATGAACTGTG 58 -3.1 ( -11.1, -8.0 )
Table Ib. 25 mer siRNA sense strand sequences
Figure imgf000018_0001
Table 2 : siRNA targeted sequences in ICAMl gene:
ICAMl gene: Homo sapiens intercellular adhesion molecule 1 (CD54) , human rhinovirus receptor (ICAMl) , Accession: NM_000201, Gene ID: 4557877, 19 siRNA candidates were selected targeting the following gene sequences : Table 2a. 23 mer DNA sense strand sequences:
Figure imgf000019_0001
Table 2b. 25 mer siRNA sense strand sequences;
Figure imgf000019_0002
Table 3 : siRNA targeted sequences in VCAMl gene :
VCAMl gene: Homo sapiens vascular cell adhesion molecule 1 (VCAMl) , Transcript variant 2, mRNA.
ACCESSION NM_080682. GI: 18201908; transcript variant
1, mRNA. ACCESSION NM__001078, GI : 18201907 ; Human vascular cell adhesion molecule 1 mRNA, complete cds gi 1179885 |gb|M30257.11 HUMCAMlV [179885] , Human vascular cell adhesion molecule 1 mRNA, complete cds, gi I 340193 I gb|M60335.l|HUMVCAMl [340193] , Human vascular cell adhesion molecule-1 (VCAMl) gene, complete CDS, gi|340195|gb|M73255.l|
HUMVCAMlA [340195] , Human mRNA for vascular cell adhesion molecule 1 (VCAM-I) , gi I 37648 | emb|X53051.11 HSVCAMl [37648]
25 siRNA candidates were selected to target the following gene sequences:
Table 3a. 23 mer DNA sense strand sequences :
Figure imgf000020_0001
Table 3b. 25 tner siRNA sense strand sequences
Figure imgf000021_0001
Table 4 : siRNA sequences targeting human IFN-gamma (Accession, NM_000619) :
Table 4a. 19 mer siRNA sense strand sequences :
Figure imgf000022_0001
Table 4b. 25 nαer siRNA sense strand sequences
Figure imgf000022_0002
Table 5 : siRNA sequences targeting human IL-I (Accession: NM_033292) :
Table 5a. 19 mer siRNA sense strand sequences:
Figure imgf000023_0001
Table 5b. 25 mer siRNA sense strand sequences
Figure imgf000023_0002
Table 6 : siRNA sequences targeting human IL-6 (Accession NM_000600) :
Table 6a. 19 mer siRNA sense strand sequences
Figure imgf000024_0001
Table 6b. 25 mer siRNA sense strand sequences
Figure imgf000024_0002
Table 7 : siRNA sequences targeting human IL-8 (Accession: NM_000584) :
Table 7a. 19 mer siRNA sense strand sequences
Figure imgf000025_0001
Table 7a. 25 mer siRNA sense strand sequences
Figure imgf000025_0002
Table 8 : siRNA sequences targeting human TNF-α (Accession: NM_004862) :
Table 8a. 19 mer siRNA sense strand sequences
Figure imgf000026_0001
Table 8b. 25 mer siRNA sense strand sequences
Figure imgf000026_0002
Table 9 : siRNA sequences targeting human CD80 (Accession: NM_005191) :
Table 9a. 19 mer siRNA sense strand sequences
Figure imgf000027_0001
Table 9b. 25 mer siRNA sense strand sequences
Figure imgf000027_0002
Table 10 : siRNA sequences targeting human CD86 (Accession: NM__175862) :
Table 10a. 19 mer siRNA sense strand sequence
Figure imgf000028_0001
Table 10b. 25 mer siRNA sense strand sequence
Figure imgf000028_0002
Table 11 : siRNA sequences targeting human MHC-II (Accession: NM_002119) :
Table 11a. 19 mer siRNA sense strand sequences
Figure imgf000029_0001
Table lib. 25 mer siRNA sense strand sequences
Figure imgf000029_0002
Table 12 : siRNA sequences targeting human MHC-I (Accession: NM_005516)
Table 12a. 19 mer siRNA sense strand sequences:
Figure imgf000030_0001
Table 12b. 25 mer siRNA sense strand sequences:
Figure imgf000030_0002
Table 13 : siRNA sequences targeting human CD28 (Accession: NM_006139) :
Table 13a. 19 mer siRNA sense strand sequences
Figure imgf000031_0001
Table 13b. 25 mer siRNA sense strand sequences
Figure imgf000031_0002
Table 14 : siRNA sequences targeting human CTLA4 (Accession: AF414120) :
Table 14a. 19 raer siRNA sense strand sequences
Figure imgf000032_0001
Table 14b. 25 mer siRNA sense strand sequences
Figure imgf000032_0002
Table 15 : siRNA sequences targeting human parvovirus B19 (Accession: AY903437) :
Table 15a. 19 mer siRNA sense strand sequences
Figure imgf000033_0001
Table 15b. 25 mer siRNA sense strand sequences
Figure imgf000033_0002
[0033] In one embodiment, siRNA duplexes of 25 basepair with blunt ends exhibit more potent gene knockdown efficacy than 19 basepair with overhang at both 3' ends, both in vitro and in vivo. [0034] In an additional aspect the invention provides a double stranded polynucleotide that includes a first linear polynucleotide strand described above and a second polynucleotide strand that is complementary to at least the first nucleotide sequence of the first strand and is hybridized thereto to form a double stranded siRNA composition. Formulations
[0035] A variety of carriers serve to prepare formulations or pharmaceutical compositions containing siRNAs. In several embodiments the siRNA polynucleotides of the invention are delivered into cells in culture or into cells of an organ awaiting transplantation by liposome-mediated transfection, for example by using commercially available reagents or techniques, e.g., Oligofectamine™, LipofectAmine™ reagent, LipofectAmine 2000™ (Invitrogen) , as well as by electroporation, and similar techniques. [0036] The pharmaceutical compositions containing the siRNAs include additional components that protect the stability of siRNA, prolong siRNA lifetime, potentiate siRNA function, or target siRNA to specific tissues/cells. These include a variety of biodegradable polymers, cationic polymers (such as polyethyleneimine) , cationic copolypeptides such as histidine-lysine (HK) polypeptides see, for example, PCT publications WO 01/47496 to Mixson et al . , WO 02/096941 to Biomerieux, and WO 99/42091 to Massachusetts Institute of Technology) , PEGylated cationic polypeptides, and ligand- incorporated polymers, etc. positively charged polypeptides, PolyTran solutions (saline or aqueous solution of HK polymers and polysaccharides such as natural polysaccharides, also known as scleroglucan) , TargeTran (a saline or aqueous suspension of nano-particle composed of conjugated RGD-PEG-PEI polymers including a targeting ligand) , surfactants (Infasurf ; Forest
Laboratories, Inc.; ONY Inc.), and cationic polymers (such as polyethyleneimine) . Infasurf® (calfactant) is a natural lung surfactant isolated from calf lung for use in intratracheal instillation; it contains phospholipids, neutral lipids, and hydrophobic surfactant-associated proteins B and C.
[0037] The polymers can either be uni-dimensional or multi-dimensional, and also could be microparticles or nanoparticles with diameters less than 20 microns, between 20 and 100 microns, or above 100 micron. The said polymers could carry ligand molecules specific for receptors or molecules of special tissues or cells, thus be used for targeted delivery of siRNAs. The siRNA polynucleotides are also delivered by cationic liposome based carriers, such as DOTAP, DOTAP/Cholesterol (Qbiogene, Inc.) and other types of lipid aqueous solutions. In addition, low percentage (5-10%) glucose aqueous solution, and Infasurf are effective carriers for airway delivery of siRNA (Li B.J. et al, 2005, Nature Medicine, 11, 944-951). [0038] In addition, a carrier may include Hyper Osmolar Citrate solution (560 mθsm/kg solution of meglumine hydrochloride, 560 mθsm/kg meglumine ioxaglate, and 600 mθsm/kg sodium ioxaglate, and so forth) . University of Wisconsin solution has the potential to enhance and extend heart, kidney, lung and liver preservation. University of Wisconsin solution is widely accepted for the cold storage and transport of human donor pancreata destined for islet isolation. [0039] The composition may further comprise a polymeric carrier. The polymeric carrier may comprise a cationic polymer that binds to the RNA molecule. The cationic polymer may be an amino acid copolymer, comprising, for example, histidine and lysine residues. The polymer may comprise a branched polymer. [0040] The composition may comprise a targeted synthetic vector. The synthetic vector may comprise a cationic polymer, a hydrophilic polymer, and a targeting ligand. The polymer may comprise a polyethyleneimine, the hydrophilic polymer may comprise a polyethylene glycol or a polyacetal, and the targeting ligand may comprise a peptide comprising an RGD sequence .
[0041] The siRNA/carrier may be formulated in either the storage solution or the perfusion medium in a nonspecific manner, or via the systemic circulation in a targeted delivery system.
Improving Solid Organ And Cell Transplantation
[0042] The present invention provides methods for prevention of allograft rejection and ischemia/reperfusion injury in solid organ transplantation by silencing or down-regulation of a target gene expression by introducing RNA interference (siRNA) . In a method of the present invention, siRNA is applied to an organ intended for transplantation in the form of an organ-storage solution, i.e., after removal from the donor and while it is being transported to the recipient. The donor or recipient of the transplanted organ, tissues, and/or cells can be a mammal, including, but not limited to, human, non- human mammal, non-human primate, rat, mouse, pig, dog, cow, and horse. The organs destined for transplantation are maintained by an organ storage solution comprising one siRNA oligonucleotide or multiple siRNA oligonucleotides as a cocktail. siRNA can access the donor organ and cells easily and selectively, which facilitates the reduction of potentially harmful systemic side effects. [0043] In current practice, donor organs are subjected to flushing and storage in static or recirculating systems, in hypothermic conditions (less than 37 0C for humans, e.g. 4 0C) or normothermic conditions (37 0C for humans) , in specially formulated solutions (organ preservation solutions) in order to wash out debris and to decrease damage during transportation. The methods of the present invention include siRNA transfection of the donor organ and cells during organ preservation. This is an attractive method, because siRNA applied ex vivo to the organ to be donated would not be administered systemically to organ recipients, and treatment could be delivered specifically to the site of inflammation. This method could be useful to prevent graft failure without systemic adverse effects. [0044] The siRNA transfection formulation is used for flushing the solid donor organ in situ and/or ex vivo, and for static or machine perfusion organ storage. The formulated solution is useful for both local injection into the solid organ and to bathe the entire solid organ by submerging it in the siRNA formulation.
[0045] The siRNA agent can be used as either single or multiple duplexes, targeting single or multiple genes, with or without transfection carriers for the treatment of the transplanted organs (tissues) and cells. The transfection agents include but are not limited to synthetic polymers, liposomes and sugars, etc. The siRNA agents can also be used with other agents such as small molecule and monoclonal antibody inhibitors, immune modulators and other types of oligonucleotides. The injection of and submerging of organs for transplantation with the siRNA/carrier solution will minimize tissue damage and host rejection, and therefore, will enhance the success of the transplanted organ in terms of organ function and survival and the minimization of co-morbidities. [0046] Also in the present invention,, various organs and cells can be treated by siRNA/carrier formulation during the process of transplantation. All solid organ transplantations essentially require surgical preparation of the donor, which may include flush perfusion of the body, or of specific organs to be used in transplantation. Perfusion may be with one or more fluids. The organ (s) are removed for storage during transportation to the recipient, and the organ is surgically implanted into the recipient . Organs useful in the methods of the invention include, but are not limited to, kidney, liver, heart, pancreas, pancreatic islets, small bowel, lung, cornea, limb, and skin, as well as cells in culture corresponding to each of those organs. One example, hepatocyte cell lines, are beginning to be developed as universal donors for isolated liver cell transplantation, which is a less invasive method than orthotopic liver transplantation for treatment of metabolic liver disease. Costimulation via pathways such as CD28/B7 or CD40/CD40L is a major concern for the success of such transplantation (2) . Therefore, using siRNA/carrier formulation to silence both CD28 or CD40 pathways will be a good strategy to improve the success rate of the transplant. [0047] Another example for renal transplant failure is the infection of parvovirus B19 (PV-B19) after solid organ transplantation which may cause pure red cell aplasia (PRCA) . PV-B19 infection in immunosuppressed transplant recipients is associated with significant morbidity (1) . Using siRNA to inhibit PV-B19 or any other viral infection and replication is an adjunct therapy for improvement in renal transplant by treatment of both donor organ and transplant recipient during the initial phase of the transplantation. [0048] In another of its aspects, the present invention provides compositions comprising one or more siRNA duplexes in which siRNA can simultaneously target several genes involved in allograft or xenograft rejection or ischemia/reperfusion injury. A combination of multiple siRNA duplexes could be more effective for inhibition of allograft rejection or ischemia/reperfusion injury. [0049] The process of immune modulation offers a plethora of molecular targets for siRNA silencing using the methods of the invention such as (1) molecules on lymphocytes associated with activation; (2) molecules on antigen presenting cells (APCs) which stimulate lymphocytes such as MHC class II and costimulatory molecules; (3) soluble molecular signals such as cytokines such as TNF-α, IFN- β, IL-I, IL-6, IL-8; (4) molecules associated with lymphocyte extravasation and homing such as Vascular Cell Adhesion Molecule-1, Intercellular Adhesion Molecular-1; and (5) effector molecules of immunity such as but not limited to complement factor C3. Additional candidate target genes include Intercellular Adhesion Molecule-1, Major Histocompatibility Complex Class I, Major Histocompatibility Complex Class II, IFN-γ, CD80, CD86, CD40 and CD40L.
[0050] The present invention also provides methods and compositions for using siRNA oligo cocktail (siRNA- OC) as therapeutic agent useful in the methods of the invention or to achieve more potent antiangiogenesis efficacy for treatment of cancer and inflammations . This siRNA oligo cocktail comprises at least three duplexes targeting at least three mRNA targets. The siRNA oligo cocktail may comprise any of the siRNA sequences listed in tables 1-15. In one embodiment, the siRNA oligo cocktail comprises the siRNAs specific for complement C3 , MHC-II, and IFNγ. The present invention is based on two important aspects: first, the siRNA duplex is a very potent gene expression inhibitor, and each siRNA molecule is made of short double-stranded RNA oligo (21-23nt, or 24-25nt, or 26- 29nt) with the same chemistry property; Second, allograft or xenograft rejection and ischemia/reperfusion injury relate, in part, to overexpressions of endogenous genes. Therefore, using siRNA-OC targeting multiple genes represents an advantageous therapeutic approach, due to the chemical uniformity of siRNA duplexes and synergistic effect from down regulation of multiple disease- or injury- causing genes. The invention defines that siRNA-OC is a combination of siRNA duplexes targeting at lease three genes, at various proportions, at various physical forms, and being applied through the same route at the same time, or different route and time into disease tissues.
[0051] The siRNA-mediated silencing can be applied with either single siRNA targeting one such gene or a combination of multiple siRNAs targeting several target sequences within the same gene, or targeting various genes from different categories such as those identified in this paragraph. For example, a composition comprising multiple siRNA duplexes may have each present with the same or different ratios. Thus, in a mixture of three siRNAs duplex I, duplex II and duplex III may either each be present at 33.3% (w/w) of total siRNA agent each, or at 20%, 45% and 35% respectively, by way of nonlimiting example.
[0052] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Exemplary methods and materials are described below, although methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention. All publications and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control . Although a number of documents are cited herein, this citation does not constitute an admission that any of these documents forms part of the common general knowledge in the art. Throughout this specification and claims, the word "comprise," or variations such as "comprises" or "comprising" will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers. The materials, methods, and examples are illustrative only and not intended to be limiting. Example 1: siRNA mediated C3 expression knockdown in vitro
[0053] RNA interference blocks gene expression according to small unique segments of their sequence. This natural process can be exploited to reduce transcription of specific genes. In transplantation, it is established that donor derived complement C3 is rapidly upregulated in ischemia/reperfusion injury (I/RI) , contributing to tissue damage. Complement C3 is described as a local mediator of various forms of injury and immune regulation and is a valid target for gene knockdown after transplant ischemia/reperfusion injury that may well assist in the regulation of allo- immunity as well . This study sought to exploit siRNA to knock-down C3 gene expression in donor organs. [0054] Rat renal epithelial cell lines were stimulated with 10 μg/ml IL-I and 0.1 μg/ml IL-6 to upregulate C3 gene expression. 72 hours after stimulation, the cells were transfected with one of a panel of C3-specific siRNAs.
Figure imgf000042_0001
[0055] After 48 hours, C3 expression was determined by Real Time PCR. Results showed that C3 expression was upregulated in non-transfected cells after stimulation (Figure 1) . Cells treated with siRNA showed up to a 60% reduction of C3 expression as compared to control cells that were not treated with siRNA. These experiments identified the most effective C3 siRNA sequence from the panel that did not non- specifically induce IFNγ upregulation, a potential off- target effect of siRNA (labelled as C3-3 siRNA in Figure 1) . [0056] The candidate C3 siRNA obtained in the previous experiment was transfected into rat renal epithelial cells stimulated to express C3 , as described above. A range of concentrations of this C3 specific siRNA produced significant (P<0.05) C3 mRNA knockdown, as measured by Real Time PCR (Figure 2) . This experiment demonstrates technical feasibility and efficacy of the C3 siRNA sequence identified for in vivo testing.
Example 2 : siRNA mediated C3 expression knockdown in vivo
[0057] The most effective C3 siRNA, as determined in the previous experiment, was then packaged into synthetic polycationic nanoparticles that facilitate in vivo siRNA transfection. The nanoparticles are composed of PolyTran, a family of branched histidine
(H) and lysine (K) polymers, effective for in vitro, in vivo, and ex vivo siRNA transfer. Their core sequence is as follows: R-KR-KR-KR, where R =
[HHHKHHHKHHHKHHH] 2KH4NH4. For in vivo experiments, the following branched HK polymers were initially tested for their efficacy to deliver siRNA into allograft cells: H3K4b. This branched polymer has the same core and structure described above except the R branches differ: R = KHHHKHHHKHHHKHHHK. The polymers were selected because of their in vitro or in vivo efficacy for different nucleic acid forms. The branched HK polymer was dissolved in aqueous solution and then mixed with siRNA aqueous solution at the listed ratios by mass, forming nanoparticles of average size of 150- 200 nm in diameter. The HKP-siRNA aqueous solutions were semi-transparent without noticeable aggregation of precipitate. These solutions can be stored at 4°C for at least three months .
[0058] The nanoparticles were added to Hyper Osmolar Citrate perfusion fluid and administered to donor rat kidneys. After 4 hours of cold ischemia, the kidneys were transplanted into syngeneic hosts. Two days later the kidneys were harvested and C3 gene expression was determined by Real-Time PCR. Non-transplanted, non- treated kidneys served as a negative control (labelled NKC in Figure 3) , while perfused, transplanted kidneys not treated with siRNA served as a positive control (labelled as ISCH in Figure 3) . The levels in the siRNA-treated kidneys were normalized to mRNA levels in non-transplanted, non-treated kidneys. Results are shown in Figure 3. [0059] Results demonstrate that C3-siRNA reduced post-transplant C3 gene expression by 62.56% (P<0.05, n=4) compared to untreated transplants, to a level below that detected in normal kidney. When compared against scrambled-FITC labelled siRNA control, C3 gene expression was reduced by 73.34% (P<0.05, n=4) . The FITC-labelled scrambled siRNA controls exhibited a greater upregulation of C3 gene expression than the untreated kidneys, suggestive of off-target effects. Histology showed sparing from ischemia/reperfusion injury (I/RI) in kidneys treated with C3 siRNA before transplantation (Figure 4) , but direct fluorescence microscopy of cells and tissues perfused with FITC- labelled scrambled siRNA did not contain any detectable siRNA in tissues.
[0060] In conclusion, siRNA inhibition of C3 gene expression effectively reduced local C3 activity compared to controls. The nanoparticle strategy appears to overcome the problem of effective siRNA delivery. It now appears possible to develop arrays of specific siRNA to diminish pro-inflammatory gene expression in donor organs as adjunct therapies to conventional immunosuppression or tolerance induction.
Example 3 : Determination of peptide sequences concentrated in transplanted kidneys by phage display [0061] In order to provide organ target specificity for siRNA-containing nanoparticles, peptides concentrated in the organ of interest can be identified by phage display. This method was used to identify candidate target peptides in the rat model of kidney transplantation described above. Donor kidneys were flushed with Hyper Osmolar Citrate and stored at 4°C for 4 hours before transplantation into a syngeneic host. After 48 hours, recipients were anaesthetized and injected via the tail vein with the prepared cysteine-constrained 7mer phage library (New England Biolabs) . After 5 minutes, the transplanted kidneys were harvested and phage extracted from the kidney, in a first round of "in vivo biopanning" . The extracted phage were expanded in E. coli bacteria before being injected into another kidney transplant recipient. This biopanning was repeated for a total of three rounds. After each round, a sample of phage was taken to estimate the numbers present in the transplanted kidney. After each expansion, a sample of phage was grown in bacterial colonies on agar plates so that phage could be isolated and the DNA sequence of the expressed library peptide could be determined. Figure 5 (lower panel) shows increasing numbers of phage retrieved from transplanted kidneys after each round of biopanning (random phage) , as compared to a control targeting streptavidin (R3vsStrep) . Examples of identified peptide sequences concentrated in the kidney are C-LPSPKRT-C, C-LPSPKKT-C, C-PTSVPKT-C. After the third round of biopanning, phage are concentrated in the transplanted kidney and are found in much lower numbers in other organs of the recipient (Figure 5, lower panel) . The candidate peptides can be incorporated into TargeTran nanoparticles to provide specificity for siRNA targeting to transplanted organs.
Literature
1. Subtirelu MM et al . Acute renal failure in a pediatric kidney allograft recipient treated with intravenous immunoglobulin for parvovirus Bl9 induced pure red cell aplasia. Pediatr Transplant. 2005 Dec;9 (6) :801-4.
2. Sampietro R, et al . Extension of the adult hepatic allograft pool using split liver transplantation. Acta Gastroenterol BeIg. 2005 Jul-Sep; 68 (3) : 369- 75.
3. Chalermskulrat W, et al . Combined donor-specific transfusion and anti-CD154 therapy achieves airway allograft tolerance. Thorax. 2005 Oct 27; [Epub ahead of print] .
4. Oliveira JG, et al . Humoral immune response after kidney transplantation is enhanced by acute rejection and urological obstruction and is down- regulated by mycophenolate mofetil treatment. Transpl Int. 2005 Nov;18 (11) : 1286-91. 5. McManus, M. T. and P.A. Sharp (2002) Gene silencing in mammals by small interfering RNAs. Nature Review, Genetics. 3 (10) : 737-747.
6. Lu, P.Y. et al. (2003) siRNA-mediated antitumorigenesis for drug target validation and therapeutics. Current Opinion in Molecular Therapeutics. 5 (3) :225-234.
7. Lu, P.Y. et al (2002) Tumor inhibition by RNAi- mediated VEGF and VEGFR2 down regulation in xenograft models. Cancer Gene Therapy. 10 (Supplement) ) S4.
8. Kim, B. et al. (2004) Inhibition of ocular angiogenesis by siRNA targeting vascular endothelial growth factor-pathway genes; therapeutic strategy for herpetic stromal keratitis. Am. J. Pathol. 165 (6): 2177-85.
9. Lu, P. Y. and M. Woodle (2005) Delivering siRNA in vivo For functional genomics can novel therapeutics. In RNA Interference Technology. Cambridge University Press. P 303-317.
10. Lu, P. Y. et al . (2005) Modulation of angiogenesis with siRNA inhibitors for novel therapeutics. TRENDS in Molecular Medicine. 11(3), 104-13.

Claims

What is Claimed is:
1. A targeting polynucleotide that targets an immunomodulatory or immunoeffector gene present in a cell of an organ to be donated to a subject.
2. The targeting polynucleotide according to claim 1 wherein the polynucleotide is a single stranded linear polynucleotide, a double stranded linear polynucleotide, or a hairpin polynucleotide.
3. The targeting polynucleotide according to claim 1 that comprises a first nucleotide sequence that targets a sequence chosen from the sequences disclosed in Tables 1-15.
4. A method of suppressing rejection of a transplanted organ by a recipient of the organ comprising the step of contacting the organ with a composition comprising the targeting polynucleotide according to claim 1 before transplanting the organ into the recipient .
5. The method according to claim 4 wherein the compositions comprises the targeting polynucleotide according to claim 2.
6. The method according to claim 4 wherein the composition comprises the targeting polynucleotide according to claim 3.
7. The method according to claim 4 wherein the contacting step comprises perfusing the organ with the composition.
8. The method according to claim 4 wherein the contacting step comprises bathing or submerging the organ in the composition.
9. The method according to claim 4 wherein the composition comprises a plurality of the targeting polynucleotide according to claim 1.
10. The method according to claim 4 wherein the contacting step is effective to down-regulate the immunomodulatory or immunoeffeetor target gene during organ storage before transplantation in a subject.
11. The method according to claim 4 wherein the polynucleotide inhibits expression of the target gene in a cell of the organ.
12. The method according to claim 4, wherein said organ is donor's organ.
13. The method according to claim 4, wherein said organ is kidney.
14. The method according to claim 4 , wherein said organ is liver.
15. The method according to claim 4, wherein said transplanted organ is lung.
16. The method according to claim 4, wherein said organ is pancreas .
17. The method according to claim 4 , wherein said organ is heart.
18. The method according to claim 4, wherein said organ is small bowel.
19. The method according to claim 4, wherein said organ is cornea.
20. The method according to claim 4, wherein the organ comprises cells selected from the group consisting of epithelial cells, vascular endothelium, vascular smooth muscle cells, myocardium (heart) and passenger leukocytes resident in the organ at the time of transplantation.
21. The method according to claim 4, wherein said recipient is human.
22. The method according to claim 6, wherein the targeting polynucleotide targets a C3 (complement C3) sequence selected from the sequences listed in Table 1.
23. The method according to claim 6, wherein the targeting polynucleotide targets an ICAMl (Intercellular Adhesion Molecule-1) sequence selected from the sequences listed in Table 2.
24. The method according to claim 6, wherein the targeting polynucleotide targets a VCAM-I (Vascular Cell Adhesion Molecule -1) sequence selected from the sequences listed in Table 3.
25. The method according to claim 6, wherein the targeting polynucleotide targets an IFM-γ (Interferon gamma) sequence selected from the sequences listed in Table 4.
26. The method according to claim 6, wherein the targeting polynucleotide targets an IL-I (Interleukin -1) sequence selected from the sequences listed in Table 5.
27. The method according to claim 6, wherein the targeting polynucleotide targets an IL-6 (Interleukin -6) sequence selected from the sequences listed in Table 6.
28. The method according to claim 6, wherein the targeting polynucleotide targets an IL-8 (Interleukin -8) sequence selected from the sequences listed in Table 7.
29. The method according to claim 6, wherein the targeting polynucleotide targets a TNF-α (Tumour necrosis factor-alpha) sequence selected from the sequences listed in Table 8
30. The method according to claim 6, wherein the targeting polynucleotide targets a CD80 sequence selected from the sequences listed in Table 9.
31. The method according to claim 6, wherein the targeting polynucleotide targets a CD86 sequence selected from the sequences listed in Table 10.
32. The method according to claim 6, wherein the targeting polynucleotide targets a MHC-II (Major Histocompatibilty Complex Class II) sequence selected from the sequences listed in Table 11.
33. The method according to claim 6, wherein the targeting polynucleotide targets a MHC-I (Major Histocompatibilty Complex Class I) sequence selected from the sequences listed in Table 12.
34. The method according to claim 6, wherein the targeting polynucleotide targets a CD28 sequence selected from the sequences listed in Table 13.
35. The method according to claim 6, wherein the targeting polynucleotide targets a CTLA-4 sequence selected from the sequences listed in Table 14.
36. The method according to claim 6, wherein the targeting polynucleotide targets a PV-B19 sequence selected from the sequences listed in Table 15.
37. The method according to claim 4, wherein the composition further comprises a perfusion fluid.
38. The method according to claim 4, wherein the composition further comprises a PolyTran polymer solution.
39. The method according to claim 4, wherein the composition further comprises a TargeTran nanoparticle solution.
40. The method according to claim 37, wherein the perfusion fluid is Hyper Osmolar Citrate solution or University of Wisconsin solution.
41. The method according to claim 4, wherein said targeting polynucleotide comprises one or more siRNA duplexes against one or more gene sequences .
42. The method according to claim 4, wherein the targeting polynucleotide is used in combination with small molecule drugs, monoclonal antibody drugs or other immune modulators .
43. The method according to claim 4 wherein the composition comprises a plurality of targeting polynucleotides, and wherein the polynucleotides target a plurality of gene sequences.
44. The method according to claim 43 wherein the targeting polynucleotides are a cocktail which targets the C3 , TNF-α and IL-8 gene sequences.
45. The method according to claim 4, wherein the composition is further administered to the organ recipient .
PCT/US2006/045933 2005-11-30 2006-11-30 COMPOSITIONS AND METHODS OF USING siRNA TO KNOCKDOWN GENE EXPRESSION AND TO IMPROVE SOLID ORGAN AND CELL TRANSPLANTATION WO2007064846A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP06838740A EP1963508A2 (en) 2005-11-30 2006-11-30 COMPOSITIONS AND METHODS OF USING siRNA TO KNOCKDOWN GENE EXPRESSION AND TO IMPROVE SOLID ORGAN AND CELL TRANSPLANTATION
JP2008543476A JP2009518008A (en) 2005-11-30 2006-11-30 Compositions and methods for using siRNA to knock down gene expression and to improve solid organ and cell transplantation
CA002670801A CA2670801A1 (en) 2005-11-30 2006-11-30 Compositions and methods of using sirna to knockdown gene expression and to improve solid organ and cell transplantation
US12/085,873 US20100028848A1 (en) 2005-11-30 2006-11-30 Compositions and Methods of Using siRNA to Knockdown Gene Expression and to Improve Solid Organ and Cell Transplantation

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US74115705P 2005-11-30 2005-11-30
US60/741,157 2005-11-30

Publications (2)

Publication Number Publication Date
WO2007064846A2 true WO2007064846A2 (en) 2007-06-07
WO2007064846A3 WO2007064846A3 (en) 2008-02-28

Family

ID=38092807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/045933 WO2007064846A2 (en) 2005-11-30 2006-11-30 COMPOSITIONS AND METHODS OF USING siRNA TO KNOCKDOWN GENE EXPRESSION AND TO IMPROVE SOLID ORGAN AND CELL TRANSPLANTATION

Country Status (6)

Country Link
US (1) US20100028848A1 (en)
EP (1) EP1963508A2 (en)
JP (1) JP2009518008A (en)
CN (1) CN101426913A (en)
CA (1) CA2670801A1 (en)
WO (1) WO2007064846A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8318693B2 (en) 2008-09-02 2012-11-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mutant EGFR gene
DE102011118024A1 (en) * 2011-08-01 2013-02-07 Technische Universität Dresden New procaspase 1 expression inhibitor, useful for preventing and/or treating inflammatory diseases, which are autoinflammatory diseases
US8389474B1 (en) * 2009-07-14 2013-03-05 Alan Anson Wanderer Rationale for IL-1 β targeted therapy to improve harvested organ viability, allograft tolerance, replant success and for conditions characterized by reduced or absent arterial perfusion
AU2006337093B2 (en) * 2005-12-22 2013-03-14 Opko Pharmaceuticals, Llc. Compositions and methods for regulating complement system
WO2019089922A1 (en) * 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
US10465194B2 (en) 2013-12-12 2019-11-05 Alnylam Pharmaceuticals, Inc. Complement component iRNA compositions and methods of use thereof
EP3505629A4 (en) * 2016-10-11 2020-01-01 Shanghai Unicar-therapy Bio-Medicine Technology Co., Ltd Sirna of human interleukin 6, recombinant expression car-t vector, and construction method and use thereof
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8361976B2 (en) 2004-07-09 2013-01-29 University Of Massachusetts Therapeutic alteration of transplantable tissues through in situ or ex vivo exposure to RNA interference molecules
AU2005310131A1 (en) * 2004-11-17 2006-06-08 University Of Maryland, Baltimore Highly branched HK peptides as effective carriers of siRNA
US10652525B2 (en) * 2013-10-31 2020-05-12 3Di Llc Quad view display system
EP3065722A4 (en) * 2013-11-08 2017-11-15 Dana-Farber Cancer Institute, Inc. Nucleic acid nanostructures for in vivo agent delivery
US9994811B2 (en) * 2014-10-09 2018-06-12 Lauren Brasile Reducing the immunogenicity of allografts
CN105664154B (en) * 2014-10-23 2021-02-12 江苏命码生物科技有限公司 Method for reducing tissue and/or organ transplant immune rejection and application thereof
WO2017156264A1 (en) 2016-03-11 2017-09-14 Children's Medical Center Corporation Nucleic acid nanoswitch catenanes
CN109790199B (en) 2016-08-02 2022-10-18 哈佛学院院长及董事 Cross-coordinated self-assembly
EP3406139A1 (en) * 2017-05-26 2018-11-28 Medizinische Hochschule Hannover Method for genetically modifying a vascularised tissue
WO2023186056A1 (en) * 2022-04-02 2023-10-05 上海舶望制药有限公司 Composition and method for inhibiting expression of complement component c3 protein

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003066805A2 (en) * 2001-10-23 2003-08-14 Isis Pharmaceuticals, Inc. Antisense modulation of complement component c3 expression
WO2003104456A1 (en) * 2002-06-10 2003-12-18 London Health Sciences Centre Research Inc. Immunomodulation using altered dendritic cells
WO2005108572A1 (en) * 2004-05-06 2005-11-17 Medizinische Hochschule Hannover Compounds and method for immunosuppression
US20060073127A1 (en) * 2004-07-09 2006-04-06 Umass Medical School Therapeutic alteration of transplantable tissues through in situ or ex vivo exposure to RNA interference molecules
WO2007033475A1 (en) * 2005-09-20 2007-03-29 London Health Sciences Centre Research Inc. THE USE OF siRNAs IN ORGAN STORAGE/REPERFUSION SOLUTIONS

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003066805A2 (en) * 2001-10-23 2003-08-14 Isis Pharmaceuticals, Inc. Antisense modulation of complement component c3 expression
WO2003104456A1 (en) * 2002-06-10 2003-12-18 London Health Sciences Centre Research Inc. Immunomodulation using altered dendritic cells
WO2005108572A1 (en) * 2004-05-06 2005-11-17 Medizinische Hochschule Hannover Compounds and method for immunosuppression
US20060073127A1 (en) * 2004-07-09 2006-04-06 Umass Medical School Therapeutic alteration of transplantable tissues through in situ or ex vivo exposure to RNA interference molecules
WO2007033475A1 (en) * 2005-09-20 2007-03-29 London Health Sciences Centre Research Inc. THE USE OF siRNAs IN ORGAN STORAGE/REPERFUSION SOLUTIONS

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BRADLEY S P ET AL: "Successful incorporation of short-interfering RNA into islet cells by in situ perfusion" TRANSPLANTATION PROCEEDINGS, vol. 37, no. 1, January 2005 (2005-01), pages 233-236, XP004819911 ISSN: 0041-1345 *
ICHIM T E ET AL: "RNA INTERFERENCE: A POTENT TOOL FOR GENE-SPECIFIC THERAPEUTICS" AMERICAN JOURNAL OF TRANSPLANTATION, vol. 4, no. 8, 2004, pages 1227-1236, XP009051639 ISSN: 1600-6135 *
SACKS STEVEN H ET AL: "Locally produced complement and its role in renal allograft rejection." AMERICAN JOURNAL OF TRANSPLANTATION, vol. 3, no. 8, August 2003 (2003-08), pages 927-932, XP002450101 ISSN: 1600-6135 *
ZHENG X ET AL: "Preventing renal ischemia-reperfusion injury using small interfering RNA by targeting complement 3 gene" AMERICAN JOURNAL OF TRANSPLANTATION, vol. 6, no. 9, September 2006 (2006-09), pages 2099-2108, XP003010587 ISSN: 1600-6135 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2006337093B2 (en) * 2005-12-22 2013-03-14 Opko Pharmaceuticals, Llc. Compositions and methods for regulating complement system
EP3208337A1 (en) 2008-09-02 2017-08-23 Alnylam Pharmaceuticals, Inc. Compositions for combined inhibition of mutant egfr and il-6 expression
EP2690175A2 (en) 2008-09-02 2014-01-29 Alnylam Pharmaceuticals Compositions and methods for combined inhibition of mutant EGFR gene and IL-6 expression
US9212364B2 (en) 2008-09-02 2015-12-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mutant EGFR gene
US8318693B2 (en) 2008-09-02 2012-11-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mutant EGFR gene
US9957507B2 (en) 2008-09-02 2018-05-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of mutant EGFR gene
US8389474B1 (en) * 2009-07-14 2013-03-05 Alan Anson Wanderer Rationale for IL-1 β targeted therapy to improve harvested organ viability, allograft tolerance, replant success and for conditions characterized by reduced or absent arterial perfusion
DE102011118024A1 (en) * 2011-08-01 2013-02-07 Technische Universität Dresden New procaspase 1 expression inhibitor, useful for preventing and/or treating inflammatory diseases, which are autoinflammatory diseases
EP3798306A1 (en) * 2013-12-12 2021-03-31 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
US10465194B2 (en) 2013-12-12 2019-11-05 Alnylam Pharmaceuticals, Inc. Complement component iRNA compositions and methods of use thereof
US11186842B2 (en) 2013-12-12 2021-11-30 Alnylam Pharmaceuticals, Inc. Complement component iRNA compositions and methods of use thereof
EP3505629A4 (en) * 2016-10-11 2020-01-01 Shanghai Unicar-therapy Bio-Medicine Technology Co., Ltd Sirna of human interleukin 6, recombinant expression car-t vector, and construction method and use thereof
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
WO2019089922A1 (en) * 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
US11866701B2 (en) 2017-11-01 2024-01-09 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof

Also Published As

Publication number Publication date
CA2670801A1 (en) 2007-06-07
JP2009518008A (en) 2009-05-07
EP1963508A2 (en) 2008-09-03
CN101426913A (en) 2009-05-06
WO2007064846A3 (en) 2008-02-28
US20100028848A1 (en) 2010-02-04

Similar Documents

Publication Publication Date Title
US20100028848A1 (en) Compositions and Methods of Using siRNA to Knockdown Gene Expression and to Improve Solid Organ and Cell Transplantation
Brüggenwirth et al. RNA interference therapeutics in organ transplantation: The dawn of a new era
US10801028B2 (en) Compositions for controlling Varroa mites in bees
DK1799269T3 (en) Oligoribonucleotides and methods of use thereof for treating alopecia, acute renal failure, and other diseases
Ichim et al. RNA interference: a potent tool for gene-specific therapeutics
Sioud et al. Cationic liposome-mediated delivery of siRNAs in adult mice
Thijssen et al. Gene silencing with siRNA (RNA interference): a new therapeutic option during ex vivo machine liver perfusion preservation
Zheng et al. Protection of renal ischemia injury using combination gene silencing of complement 3 and caspase 3 genes
EP2018436A2 (en) Targeted delivery to leukocytes using non-protein carriers
Stepkowski et al. PROTECTION AGAINST ALLOGRAFT REJECTION WITH INTERCELLULAR ADHESION MOLECULE-1 ANTISENSE OLIGODEOXYNUCLEOTIDES1
WO2007033475A1 (en) THE USE OF siRNAs IN ORGAN STORAGE/REPERFUSION SOLUTIONS
JP2024016155A (en) Reduced fratricide of immune cells expressing NKG2D-based receptors
Feng et al. Small interfering RNA targeting RelB protects against renal ischemia-reperfusion injury
Sioud siRNA delivery in vivo
WO2009143371A2 (en) COMPOSITIONS COMPRISING mTOR SIRNA AND METHODS OF USE THEREOF
JP6430945B2 (en) Regulation of RNA activity and vascular permeability
Jing et al. Inhibition of tumor necrosis factor alpha secretion in rat Kupffer cells by siRNA: in vivo efficacy of siRNA-liposomes
Hlawaty et al. Inhibition of MMP-2 gene expression with small interfering RNA in rabbit vascular smooth muscle cells
EP1614751A1 (en) Oligoribonucleotide sequence homologous to a cdna region which codes for the human cd40 receptor and duplex oligoribonucleotides, vectors, pharmaceutical compositions and uses associated thereto
WO2010065437A1 (en) Modulation of olfml-3 mediated angiogenesis
CN112236514A (en) Compositions and methods for improving the persistence of adoptive transfer of cells
Zheng et al. siRNA specific delivery system for targeting dendritic cells
EP2753695A1 (en) Strategies for prevention and/or treatment of diseases based on cd40 silencing
JP7454207B2 (en) Pancreatic cancer cell invasion and metastasis inhibitor
Buabeid et al. Targeting of protein expression in renal disease using siRNA–A review

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2008543476

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006838740

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200680052091.7

Country of ref document: CN

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2670801

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 12085873

Country of ref document: US

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)