US10385343B2 - Methods and compositions for the treatment of cancer - Google Patents

Methods and compositions for the treatment of cancer Download PDF

Info

Publication number
US10385343B2
US10385343B2 US15/506,010 US201515506010A US10385343B2 US 10385343 B2 US10385343 B2 US 10385343B2 US 201515506010 A US201515506010 A US 201515506010A US 10385343 B2 US10385343 B2 US 10385343B2
Authority
US
United States
Prior art keywords
epcam
cells
cancer
cell
tumor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US15/506,010
Other languages
English (en)
Other versions
US20170275629A1 (en
Inventor
Judy Lieberman
Adi GILBOA-GEFFEN
Lee Adam Wheeler
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Childrens Medical Center Corp
Original Assignee
Childrens Medical Center Corp
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 Childrens Medical Center Corp filed Critical Childrens Medical Center Corp
Priority to US15/506,010 priority Critical patent/US10385343B2/en
Assigned to CHILDREN'S MEDICAL CENTER CORPORATION reassignment CHILDREN'S MEDICAL CENTER CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WHEELER, LEE ADAM, LIEBERMAN, JUDY, GILBOA-GEFFEN, Adi
Publication of US20170275629A1 publication Critical patent/US20170275629A1/en
Application granted granted Critical
Publication of US10385343B2 publication Critical patent/US10385343B2/en
Assigned to THE GOVERNMENT OF THE UNITED STATES, AS REPRESENTED BY THE SECRETARY OF THE ARMY reassignment THE GOVERNMENT OF THE UNITED STATES, AS REPRESENTED BY THE SECRETARY OF THE ARMY CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: BOSTON CHILDREN'S HOSPITAL
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/115Aptamers, i.e. nucleic acids binding a target molecule specifically and with high affinity without hybridising therewith ; Nucleic acids binding to non-nucleic acids, e.g. aptamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • 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
    • C12N15/1137Non-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 enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5152Tumor cells
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/16Aptamers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3519Fusion with another nucleic acid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • C12N2310/3533Halogen

Definitions

  • the technology described herein relates to chimeric molecules comprising an EpCAM binding-molecule and an inhibitory nucleic acid and methods of using such compositions for the treatment of cancer, e.g. epithelial cancer.
  • RNA interference has been explored for therapeutic use in reducing gene expression in the liver.
  • the liver is unique in being easy to transfect with RNAi molecules. Delivery of small RNAs and resulting gene knockdown in other tissues continues to be inefficient and ultimately ineffective.
  • the delivery roadblock is a major obstacle to harnessing RNAi to treat cancer.
  • AsiCs novel chimeric aptamer-siRNA molecules
  • a chimeric molecule comprising a cancer marker-binding aptamer domain and an inhibitory nucleic acid domain.
  • the cancer marker is EpCAM or EphA2.
  • the inhibitory nucleic acid specifically binds to a gene product upregulated in a cancer cell.
  • the inhibitory nucleic acid inhibits the expression of a gene selected from the group consisting of: Plk1; MCL1; EphA2; PsmA2; MSI1; BMI1; XBP1; PRPF8; PFPF38A; RBM22; USP39; RAN; NUP205; and NDC80.
  • the cancer marker is EpCAM and the inhibitory nucleic acid domain inhibits the expression of Plk1.
  • the molecule is an aptamer-siRNA chimera (AsiC).
  • the cancer marker-binding aptamer domain comprises the sequence of SEQ ID NO: 33.
  • the cancer marker-binding aptamer domain consists essentially of the sequence of SEQ ID NO: 33.
  • the inhibitory nucleic acid domain comprises the sequence of SEQ ID NO: 2.
  • the inhibitory nucleic acid domain consists essentially of the sequence of SEQ ID NO: 2.
  • the molecule comprises the sequence of one of SEQ ID NOs: 1-3. In some embodiments, the molecule consists essentially of the sequence of one of SEQ ID NOs: 1-3.
  • the 3′ end of the molecule comprises dTdT. In some embodiments, the molecule comprises at least one 2′-F pyrimidine.
  • a pharmaceutical composition comprising a chimeric molecule as described herein and a pharmaceutically acceptable carrier.
  • the composition comprises at least two chimeric molecules as described herein wherein the chimeric molecules have different aptamer domains and/or inhibitory nucleic acid domains.
  • the different apatmer or inhibitory nucleic acid domains recognize different targets.
  • the different apatmer or inhibitory nucleic acid domains have sequences and recognize the same target.
  • described herein is a method of treating cancer, the method comprising administering a chimeric molecule and/or composition as described herein.
  • the cancer is an epithelial cancer or breast cancer.
  • the breast cancer is triple-negative breast cancer.
  • the administration is subcutaneous.
  • the subject is further administered an additional cancer treatment.
  • the cancer treatment is paclitaxel.
  • FIGS. 1A-1H demonstrate that EpCAM aptamer specifically targets Basal A breast cancer cells.
  • Design of EpCAM-AsiC containing an EpCAM aptamer and a PLK1 siRNA (sense strand disclosed as SEQ ID NO: 1 and antisense strand disclosed as SEQ ID NO: 2) ( FIG. 1C ).
  • Epithelial breast cancer cell line (BPLER) over express EpCAM protein compared to normal breast epithelial cell line (BPE) ( FIGS. 1A-1B ).
  • EpCAM-AsiC targeting GFP was Alexa647 or Cy3 labeled at the 3′ end of the antisense siRNA strand and incubated with BPLER and BPE cells. Uptake was assessed 24 hours later by flow cytometry ( FIG.
  • EpCAM-AsiC targeting AKT1 selectively knocks-down AKT1 mRNA ( FIG. 1E ) and protein ( FIG. 1F ) expression in basal A and luminal breast cancer cell lines and not in basal B or human fibroblasts (hFb).
  • Transfection with siRNA targeting AKT1 induces gene knockdown in all cell lines, while treatment with EpCAM-AsiC targeting GFP doesn't effect AKT1 mRNA and protein levels (* p ⁇ 0.05, p ⁇ 0.01).
  • Plots of AKT1 Protein and gene Knockdown comparing the effect of EpCAM-AsiC to siRNA transfection.
  • FIGS. 2A-2E demonstrate that EpCAM AsiC targeting PLK1 specifically inhibits cell proliferation in Basal A breast cancer cells.
  • the effect of EpCAM-AsiC targeting PLK1 on cell proliferation was tested on 10 breast cancer cell lines representative of basal A, B and luminal cell lines using cell-titer-glo assay (CTG).
  • CCG cell-titer-glo assay
  • EpCAM-AsiC targeting PLK1 decreased cell proliferation in both basal A and luminal cell lines while having no effect on basal B cells ( FIGS. 2A, 2C ).
  • a correlation was seen between EpCAM expression levels and cell viability ( FIG. 2B ).
  • Basal A (EpCAM+GFP ⁇ ) cell were co-cultured with BPE (EpCAM-GFP+) cells and treated with EpCAM-AsiC targeting PLK1 or untreated.
  • Untreated co-culture displayed a similar ration of cells following EpCAM-AsiC targeting PLK1 treatment the ratio of EpCAM+ cells decreased and EpCAM ⁇ cells increased.
  • a representative flow cytometry plot FIG. 2D
  • FIGS. 3A-3D demonstrate that human TNBC tissue specifically takes up Cy3-EpCAM aptamers.
  • Experimental design Cy3-EpCAM-AsiC targeting GFP, Alexa647-siRNA-GFP or Alexa647-chol-siRNA-GFP (2 ⁇ M of each) were added to breast cancer and control explants and incubated for 24 h before tissue was digested with collagenase to a single cell suspension and analyzed by flow cytometry ( FIG. 3A ).
  • FIG. 3B Representative histograms from one of three independent experiments show that siRNA and chol-siRNA penetrated both tumor and healthy tissue with similar efficacy while EpCAM-AsiC was selectively uptaken by the tumor tissue biopsy and not by the healthy control tissue sample ( FIG. 3C ).
  • the uptake experiment was repeated in tumors from three different patients, each biopsy receive was tested 3 times for each treatment.
  • FIGS. 4A-4C demonstrate that EpCAM AsiC targeting PLK1 specifically inhibits tumor initiation in Basal A breast cancer cells.
  • Colony assays of breast cancer cell lines were treated with EpCAM-AsiC targeting PLK1 or GFP (4 uM) or paclitaxel (100 nM) for 24 hr and cultured for 8 days in drug-free medium.
  • Treatment with paclitaxel decreased colony formation in all cells lines while treatment with EpCAM-AsiC targeting PLK1 only eliminated colony formation in luminal (MCF7) and basal A (HCC1954) cells, treatment with EpCAM-AsiC targeting GFP had no effect ( FIG. 4A ).
  • the assay was repeated in 3 more cells lines and results were reproducible ( FIG. 4B ).
  • EpCAM-AsiC targeting PLK1 decreased the number of spheres only in basal A and luminal cells and had no effect on basal B cells ( FIG. 4C ).
  • MB468-luc cells were treated for 24 h with EpCAM-AsiC targeting either GFP or PLK1 and injected s.c. to the flank of nude mice. Mice were imaged every 5 days for 20 days. Untreated mice and mice treated with EpCAM-AsiC targeting GFP, displayed increase in tumor initiation while mice injected with cell pretreated with EpCAM-AsiC targeting PLK1 has no tumor initiation.
  • FIGS. 5A-5C demonstrate the selective uptake of Alexa750-EpCAM-AsiCs into EpCAM+ tumors.
  • FIG. 5A depicts the experimental setup; nude mice were injected with MB468-luc (left flank) and MB231-luc-mCherry (right flank) cells, 5 days post injection Alexa750 labeled EpCAM-AsiC targeting GFP (0.5 mg/kg) was injected s.c. in the neck area. The mice were imaged immediately after injection and again after 24, 48 hr and 5 days.
  • FIG. 5C depicts a graph of Alexa750 uptake rates.
  • FIGS. 6A-6B demonstrate the EpCAM AsiC targeting PLK1 specifically inhibits tumor growth in Basal A breast cancer cells.
  • FIG. 6A depicts the experimental design. Nude mice injected with either MB231-luc-mCherry cells (5 ⁇ 10 5 ) or MB468-luc cells (5 ⁇ 10 6 ) were treated with 5 mg/Kg of either EpCAM AsiC targeting PLK1 or GFP every 72 h or left untreated.
  • FIG. 6B MB468-luc tumors treated with EpCAM-AsiC targeting PLK1 shrunk in size as early as 6 days post treatment and in many mice completely disappeared after 14 days, Untreated tumors both EpCAM+ and EpCAM-increased in size over the 14 days.
  • FIG. 7 demonstrates that EpCAM AsiCs are stable in human and mouse serum.
  • eGFP EpCAM-AsiCs synthesized using 2′-fluoro-pyrimidines, chemically-stabilized cholesterol-conjugated eGFP siRNAs (chol-siRNA), or unmodified eGFP siRNAs were incubated with an equal volume of human or mouse serum. Aliquots were removed at regular intervals and resuspended in gel loading buffer and stored at ⁇ 80° C. before electrophoresis on denaturing PAGE gels. The average intensity (+S.E.M.) of bands from 2 independent experiments quantified by densitometry after staining is shown.
  • FIGS. 8A-8B demonstrate that injection of EpCAM AsiCs does not stimulate innate immunity in mice.
  • FIG. 8A Serum samples, collected at baseline and 6 and 16 hr after treatment were assessed for IFN ⁇ , IL-6 and IP-10 by multiplex immunoassay. * p ⁇ 0.05. ** p ⁇ 0.01, *** p ⁇ 0.001, compared to baseline.
  • FIG. 9 depicts a table of sequences. (SEQ ID NOS 1-2 and 23-32, respectively, in order of appearance).
  • FIGS. 10A-10B depict aptamers-siRNA chimera (AsiC).
  • FIG. 10A depicts a diagram of the AsiC (aptamer covalently linked to one strand of an siRNA) specifically recognizing a cancer cell surface receptor, being endocytosed and then released to the cytosol, where it is processed like endogenous pre-miRNAs to knockdown a target gene. Bars indicate the 2 delivery hurdles—cell uptake and release from endosomes to the cytosol where Dicer and the RNA induced silencing complex (RISC) are located.
  • FIG. 10B depicts the design of the EpCAM AsiC targeting PLK1. (sense strand disclosed as SEQ ID NO: 1 and antisense strand disclosed as SEQ ID NO: 2).
  • FIGS. 11A-11D demonstrate that EpCAM-AsiC knockdown and antitumor effect correlates with EpCAM levels and inhibits epithelial breast tumor T-ICs.
  • FIGS. 11A-11B Representative experiment ( FIG. 11A ) and AKT1 knockdown comparing EpCAM-AsiC with lipid siRNA transfection ( FIG. 11B ).
  • FIG. 11C Anti-proliferative effect of EpCAM-AsiCs knocking down PLK1 only in EpCAM+ cell lines. D PLK1 EpCAM-AsiCs inhibit colony formation in luminal MCF and basal-A TNBC HCC1143, but not in mesenchymal basal-B MB231 cells.
  • FIGS. 12A-12B demonstrate the identification of a functional EphA2 aptamer
  • FIG. 12A Incubation of EphA2+ basal-B MB231 cells with EphA2 aptamer (EphA2apt) leads to EphA2 degradation and a transient decrease in active Akt (pAkt).
  • FIG. 12B EphA2+ breast cancer cells incubated for 2 h with EphA2apt (0 to 100 nM), but not control nonbinding aptamer (ctl), show reduced EphA2. Addition of Ephrin A was used as a positive control for EphA2 degradation.
  • FIGS. 13A-13C EpCAM-AsiCs knockdown GFP protein ( FIG. 13A ) and AKT1 mRNA ( FIGS. 13B-13C ) only in EpCAM+ cell lines, but not in immortalized breast epithelial cell line (BPE) or mesenchymal basal B TNBC or human fibroblasts. A transfected siRNA is nonspecific in its knockdown. *, P ⁇ 0.05
  • FIG. 14 Normal breast tissue and basal-A TNBC tumor biopsies from the same subject were incubated with Cy3-labeled EpCAM-AsiC and single cell suspensions were analyzed 3 d later for uptake by flow cytometry. Naked siRNAs were not taken up by either, cholesterol-conjugated siRNAs were equally taken up, but EpCAM-AsiCs were specifically taken up by the tumor. Representative tissues are shown at left.
  • FIGS. 15A-15C Treatment of EpCAM+, but with not EpCAM ⁇ , breast cancer lines with PLK1 EpCAM-AsiCs inhibits colony ( FIGS. 15A, 15B ) and mammosphere ( FIG. 15C ) function, in vitro assays of T-IC function.
  • FIG. 16 demonstrates that ex vivo treatment of MB468 cells with PLK1 EpCAM-AsiCs eliminated their ability to form tumors in nude mice. An equal number of viable cells were implanted the day after treatment.
  • FIGS. 17A-17B demonstrate that EpCAM-AsiCs are selectively taken up into EpCAM+, but not EpCAM ⁇ , TNBC tumors.
  • FIG. 17A depicts the experimental scheme.
  • FIG. 17B depicts the concentration of EpCAM-AsiCs in excised tumors at sacrifice.
  • FIG. 18A-18B demonstrate that PLK1 EpCAM-AsiCs caused complete tumor regression of EpCAM+ TNBC xenografts, but had no effect on EpCAM ⁇ basal-B xenografts.
  • FIG. 18A depicts the experimental design. Imaging of luciferase activity of left and right flank tumors was performed sequentially over 2 wks.
  • FIG. 18B depicts a graph of tumor size by luciferase activity. All the EpCAM+ tumors in mice treated with PLK1 AsiCs rapidly regressed, while the other tumors continued to grow.
  • FIGS. 19A-19C demonstrate that basal dependency genes include 4 tri-snRNP spliceosome complex genes (PFPF8, PRPF38A, RBM22, USP39), 2 nuclear export genes (NUP205, RAN), and a kinetochore gene (NDC80).
  • FIG. 19A depicts cell viability, 3 d after knockdown, normalized to control siRNA.
  • FIG. 19B depicts colony formation assessed by plating viable cells 2 d after knockdown.
  • FIG. 19C depicts caspase activation 2 d after knockdown is specific for MB468 and does not occur in BPE cells.
  • FIG. 20 depicts some possible designs for multimerized EpCAM-AsiCs to improve endocytosis. In these designs the sense and antisense strands could be exchanged and the linkers could be varied.
  • FIGS. 21A-21D demonstrate that EpCAM aptamer specifically targets Basal A breast cancer cells.
  • FIG. 21A depicts the design of EpCAM-AsiC, containing an EpCAM aptamer and a PLK1 siRNA (sense strand disclosed as SEQ ID NO: 1 and antisense strand disclosed as SEQ ID NO: 2).
  • FIG. 21B depicts graphs demonstrateing that epithelial breast cancer cell line (BPLER) over express EpCAM protein compared to normal breast epithelial cell line (BPE).
  • BPLER epithelial breast cancer cell line
  • BPE normal breast epithelial cell line
  • EpCAM-AsiC targeting GFP was Alexa647 or Cy3 labeled at the 3′ end of the antisense siRNA strand and incubated with BPLER and BPE cells. Uptake was assessed 24 hours later by flow cytometry ( FIG.
  • FIG. 22 depicts graphs demonstrating that EpCAM aptamers do not bind mouse EpCAM.
  • Mouse ESA EpCAM
  • 4T1 cell an epithelial mouse breast cancer cell line displayed high expression levels of EpCAM.
  • RAW mouse monocyte cell line
  • MB468 human basal A cell line
  • a mouse mesanchymal cancer cell line (67NR) displayed a minimal increase in EpCAM expression. Uptake experiments demonstrated that EpCAM-Aptamer was not taken up by neither 4T1 nor 67NR cells.
  • FIG. 23 depicts graphs demonstrating that EpCAM is over expressed in basal A and luminal but not basal B breast cancer cell lines. Representative FACS plots of 8 different breast cancer cell lines, testing EpCAM expression levels by flow cytometery using a hEpCAM Antibody. EpCAM is over expressed in all basal A and luminal cells lines and not in basal B. (mock, shaded gray EpCAM, black)
  • FIGS. 24A-24F demonstrate that EpCAM AsiC specifically silences gene expression in Basal A breast cancer cells.
  • EpCAM-AsiC targeting AKT1 selectively knocks-down AKT1 mRNA ( FIG. 24A ) and protein ( FIGS. 24B, 24C ) expression in basal A and luminal breast cancer cell lines and not in basal B or human fibroblasts (hFb).
  • Transfection with siRNA targeting AKT1 induces gene knockdown in all cell lines, while treatment with EpCAM-AsiC targeting GFP doesn't effect AKT1 mRNA and protein levels (* p ⁇ 0.05, p ⁇ 0.01).
  • Plots of AKT1 Protein and gene Knockdown comparing the effect of EpCAM-AsiC to siRNA transfection.
  • EpCAM-AsiC induced knockdown correlates with EpCAM expression ( FIG. 24D, 24E ). (n 3; mean ⁇ SEM normalized to mock; *P ⁇ 0.05, **P ⁇ 0.01, 2-tailed t test).
  • FIGS. 25A-25E demonstrate that human TNBC tissue specifically takes up Cy3-EpCAM aptamers.
  • FIG. 25A depicts the experimental design; Cy3-EpCAM-AsiC targeting GFP, Alexa647-siRNA-GFP or Alexa647-chol-siRNA-GFP (2 ⁇ M of each) were added to breast cancer and control explants and incubated for 24 h before tissue was digested with collagenase to a single cell suspension and analyzed by flow cytometry.
  • FIG. 25B depicts graphs demonstrating that tumor biopsies over express EpCAM and cytokeratin, an epithelial cell marker.
  • 25C depicts representative histograms from one of three independent experiments show that siRNA and chol-siRNA penetrated both tumor and healthy tissue with similar efficacy while EpCAM-AsiC was selectively uptaken by the tumor tissue biopsy and not by the healthy control tissue sample. The uptake experiment was repeated in tumors from three different patients, each biopsy received was tested 3 times for each treatment.
  • FIG. 26 depicts graphs demonstrating that EpCAM-AsiC is taken up by both healthy and colon cancer biopsies.
  • Cy3-EpCAM-AsiC targeting GFP, Alexa647-siRNA-GFP or Alexa647-chol-siRNA-GFP (2 ⁇ M of each) were added to colon cancer and control explants and incubated for 24 h before tissues were digested with collagenase to a single cell suspension and analyzed by flow cytometry.
  • Representative histograms show that EpCAM-AsiC, siRNA and chol-siRNA penetrated both tumor and healthy tissue with similar efficacy.
  • FIGS. 27A-27D demonstrate that EpCAM AsiC targeting PLK1 specifically inhibits cell proliferation in Basal A breast cancer cells.
  • the effect of EpCAM-AsiC targeting PLK1 on cell proliferation was tested on 10 breast cancer cell lines representative of basal A, B and luminal cell lines using cell-titer-glo assay (CTG).
  • CCG cell-titer-glo assay
  • EpCAM-AsiC targeting PLK1 decreased cell proliferation in both basal A and luminal cell lines while having no effect on basal B cells ( FIG. 27A ).
  • a correlation was seen between EpCAM expression levels and cell viability ( FIG. 27B ).
  • Basal A (EpCAM+GFP ⁇ ) cell were co-cultured with BPE (EpCAM-GFP+) cells and treated with EpCAM-AsiC targeting PLK1 or untreated.
  • FIG. 27C depicts representative flow cytometry plots
  • FIG. 28 depicts a graph demonstrating specific decrease in cell viability in Basal A breast cancer cell lines is PLK1 dependent.
  • Ten different breast cancer cell lines representing basal A, B and luminal cells were treated with either EpCAM-AsiC targeting PLK1 or just the EpCAM-aptamer and compared to untreated controls. None of the cell lines treated with EpCAM-aptamer displayed decrease in cell viability, while basal A and luminal cell lines displayed a decrease in cell viability following treatment with EpCAM-AsiC targeting PLK1.
  • FIGS. 29A-29C demonstrate that EpCAM AsiC targeting PLK1 specifically inhibits tumor initiation in Basal A breast cancer cells.
  • Colony assays of breast cancer cell lines were treated with EpCAM-AsiC targeting PLK1 or GFP (4 uM) or paclitaxel (100 nM) for 24 hr and cultured for 8 days in drug-free medium.
  • Treatment with paclitaxel decreased colony formation in all cells lines while treatment with EpCAM-AsiC targeting PLK1 only eliminated colony formation in luminal (MCF7) and basal A (HCC1954) cells, treatment with EpCAM-AsiC targeting GFP had no effect.
  • FIG. 29A depicts images of the assay results.
  • FIG. 29C depicts a graph demonstrating that sphere formation assay indicated similar results, EpCAM-AsiC targeting PLK1 decreased the number of spheres only in basal A and luminal cells and had no effect on basal B cells.
  • MB468-luc cells were treated for 24 h with EpCAM-AsiC targeting either GFP or PLK1 and injected s.c. to the flank of nude mice. Mice were imaged every 5 days for 20 days. Untreated mice and mice treated with EpCAM-AsiC targeting GFP, displayed increase in tumor initiation while mice injected with cell pretreated with EpCAM-AsiC targeting PLK1 has no tumor initiation.
  • FIGS. 30A-30B demonstrate that EpCAM AsiC is stable in human and mouse serum for 36 hours.
  • 20 ⁇ L was removed, and resuspended in gel loading buffer and frozen at ⁇ 80° C. before being electrophoresed on a denaturing PAGE gel.
  • FIG. 30A depicts representative PAGE gels and FIG.
  • 30B depicts graphs of the average intensity (+S.E.M.) of bands from two independent experiments analyzed by densitometry. Both the stabilized cholesterol-conjugated siRNA and the EpCAM-AsiC are stable over the 36 h of the experiment.
  • FIGS. 31A-31B demonstrate selective uptake of Alexa750-EpCAM-AsiCs into EpCAM+ tumors.
  • FIG. 31A depicts the experimental setup; nude mice were injected with MB468-luc (left flank) and MB231-luc-mCherry (right flank) cells, 5 days post injection Alexa750 labeled EpCAM-AsiC targeting GFP (0.5 mg/kg) was injected s.c. in the neck area. The mice were imaged immediately after injection and again after 24, 48 hr and 5 days.
  • FIGS. 32A-32B demonstrate that EpCAM AsiC targeting PLK1 specifically inhibits tumor growth in Basal A breast cancer cells.
  • FIG. 32A depicts the experimental setup; nude mice injected with either MB231-luc-mCherry cells (5 ⁇ 10 5 ) or MB468-luc cells (5 ⁇ 10 6 ) were treated with 5 mg/Kg of either EpCAM AsiC targeting PLK1 or GFP every 72 h or left untreated. Mice were imaged using the IVIS Spectra imaging system every 72 h for 14 days.
  • FIG. 32A depicts the experimental setup; nude mice injected with either MB231-luc-mCherry cells (5 ⁇ 10 5 ) or MB468-luc cells (5 ⁇ 10 6 ) were treated with 5 mg/Kg of either EpCAM AsiC targeting PLK1 or GFP every 72 h or left untreated. Mice were imaged using the IVIS Spectra imaging system every 72 h for 14 days.
  • 32B depicts a graph demonstrating that MB468-luc tumors treated with EpCAM-AsiC targeting PLK1 shrunk in size as early as 6 days post treatment and in many mice completely disappeared after 14 days, Untreated tumors both EpCAM+ and EpCAM ⁇ increased in size over the 14 days.
  • FIG. 33 depicts graphs of tumor growth demonstrating that MB468 tumors regress only after treatment with PLK1 EpCAM-AsiC.
  • Mice with sc MB468 tumors were treated with 5 mg/kg RNA 2 ⁇ /wk beginning when tumors became palpable.
  • PLK1 EpCAM-AsiC, GFP SpCAM-AsiC, EpCAM aptamer, PLK1 siRNA, and mock treated samples were analyzed as indicated.
  • FIG. 34 demonstrates that PLK1 siRNA associates with Argonaute (AGO) in cells treated with PLK1 EpCAM-AsiCs.
  • MB-468 cells treated with PLK1 EPCAM-AsiC or siRNA for 2 days, were lysed, and cell lysates were immunoprecipitated with pan-AGO antibody or IgG isotype control.
  • the amount of PLK1 siRNA in the immunoprecipitates was quantified by Taqman qRT-PCR, presented as log 2 mean with SEM, relative to miR-16. **, P ⁇ 0.01 by Student's t-test relative to siRNA-treated cells. ND, not detectable.
  • PLK1 siRNA was found in the RISC after treatment with PLK1 EpCAM-AsiCs. However, the Ago immunoprecipitation did not significantly deplete PLK1 siRNAs from the supernatant. This is likely because most RNAs that are taken up by cells are not released from endosomes to the cytosol (A. Wittrup et al., Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown. Nature Biotechnology 2015, in press).
  • FIG. 35 demonstrates that PLK EpCAM AsiC suppresses MCF10CA1a (CA1a) tumor growth.
  • the top panel depicts the experimental scheme. In this experiment the AsiCs were injected sc in the flank near the tumor, but not into the tumor.
  • the inventors have demonstrated the suprising efficacy of AsiCs (aptamer-siRNA chimeric molecules) in treating cancer.
  • the AsiC's described herein utilize an aptamer that targets the chimeric molecule specifically to cancer cells, providing effective and on-target suppression of the gene targeted by the siRNA.
  • the aptamers described herein permit the therapy to target tumor-initiating cells (also referred to as cancer stem cells). These cells are responsible not only for tumor initiation, replapse, and metastasis, but are also relatively resistant to conventional cytotoxic therapy.
  • tumor-initiating cells also referred to as cancer stem cells.
  • the compositions and methods described herein permit effective treatment of the underlying pathology in a way that existing therapies fail to do.
  • the success of the AsiC's described herein is particularly suprising in that direct targeting of EpCAM with antibodies has been previously investigated and found to lack effectiveness.
  • the AsiC's described herein are demonstrated to be surprisingly efficacious in the treatment of epithelial cancers, e.g. breast cancer (e.g. triple negative breast cancer (TNBC)).
  • epithelial cancers e.g. breast cancer (e.g. triple negative breast cancer (TNBC)).
  • TNBC triple negative breast cancer
  • the AsiC's described herein demonstrated effective gene knockdown specifically in luminal and basal-A TNBC cells as compared to healthy cells, suppressed colony and mammosphere formation in vitro and abrogated tumor initiation ex vivo. In vitro treatment with the AsiC's resulted in targeted delivery of the therapeutic and rapid tumor regression.
  • cancer marker-binding domain refers to a domain and/or molecule that can bind specifically to a molecule more highly expressed on the surface of a cancer cell as compared to a healthy cell of the same type (a cancer marker).
  • the cancer marker can be a protein and/or polypeptide.
  • the cancer marker can be selected from EpCAM or EphA2.
  • the cancer marker-binding domain can be an aptamer.
  • EpCAM or “epithelial cell adhesion molecule” refers to a transmembrane glycoprotein mediating Ca2+-independent homotypic cell-cell adhesion in epithelial cells. Sequences for EpCAM are known for a variety of species, e.g., human EpCAM (see, e.g., NCBI Gene ID:4072; protein sequence: NCBI Ref Seq: NP_002345.2).
  • EphA2 or “EPH receptor A2” refers to a ephirin type protein-tyrosine kinase receptor. EphA2 binding ephrin-A ligands and permits entry of Kaposi sarcoma-associated herpesvirus into host cells. Sequences for EphA2 are known for a variety of species, e.g., human EphA2 (see, e.g., NCBI Gene ID:1969; protein sequence: NCBI Ref Seq: NP_004422.2).
  • inhibitory nucleic acid domain refers to a domain comprising an inhibitory nucleic acid.
  • the inhibitory nucleic acid can be a siRNA.
  • the inhibitory nucleic acid domain can inhibit, e.g., can target, the expression of a gene product that is upregulated in a cancer cell and/or the expression of a gene that is required for cell growth and/or survival.
  • the inhibitory nucleic acid domain can inhibit the expression of a gene selected from Plk1 (e.g. “polo-like kinase 1”; NCBI Gene ID: 5347); MCL1 (e.g. myeloid cell leukemia 1; NCBI Gene ID: 4170); EphA2 (NCBI Gene ID: 1969); PsmA2 (e.g.
  • NCBI Gene ID: 5683 MSI1 (e.g., musashi RNA-binding protein 1; NCBI Gene ID: 4440); BMI1 (e.g., B lymphoma Mo-MLV insertion 1, NCBI Gene ID: 648); XBP1 (X-boxn binding protein 1; NCBI Gene ID: 7494); PRPF8 (e.g., pre-mRNA processing factor 8; NCBI Gene ID:10594), PFPF38A (e.g., pre-mRNA processing factor 38A; NCBI Gene ID: 84950), RBM22 (e.g., RNA binding motif protein 22; NCBI Gene ID: 55696), USP39 (e.g., ubiquitin specific peptidase 39; NCBI Gene ID: 10713); RAN (e.g., ras-related nuclear protein; NCBI Gene ID: 5901); NUP205 (e.g., nucleoporin 205 kDa; NCBI
  • Sequences of these genes are readily obtained from the NCBI database and can be used by one of skill in the art to design inhibitory nucleic acids.
  • exemplary inhibitory nucleic acid domains e.g. a nuleic acid having the sequence of SEQ ID NO: 2.
  • a composition as described herein can comprise a cancer marker-binding domain comprising an aptamer and an inhibitory nucleic acid domain comprising an siRNA, e.g. the composition can comprise an aptamer-siRNA chimera (AsiC).
  • a cancer marker-binding domain comprising an aptamer and an inhibitory nucleic acid domain comprising an siRNA
  • the composition can comprise an aptamer-siRNA chimera (AsiC).
  • the methods described herein relate to treating a subject having or diagnosed as having cancer with a composition as described herein.
  • Subjects having cancer can be identified by a physician using current methods of diagnosing cancer.
  • Symptoms and/or complications of cancer which characterize these conditions and aid in diagnosis are well known in the art and include but are not limited to, for example, in the case of breast cancer a lump or mass in the breast tissue, swelling of all or part of a breast, skin irritation, dimpling of the breast, pain in the breast or nipple, nipple retraction, redness, scaliness, or irritation of the breast or nipple, and nipple discharge.
  • breast cancer include, but are not limited to, mammograms, x-rays, MRI, ultrasound, ductogram, a biopsy, and ductal lavage.
  • a family history of cancer or exposure to risk factors for cancer e.g. smoke, radiation, pollutants, BRCA1 mutation, etc.
  • malignancy refers to an uncontrolled growth of cells which interferes with the normal functioning of the bodily organs and systems.
  • cancer relates generally to a class of diseases or conditions in which abnormal cells divide without control and can invade nearby tissues. Cancer cells can also spread to other parts of the body through the blood and lymph systems.
  • a “cancer cell” or “tumor cell” refers to an individual cell of a cancerous growth or tissue.
  • a tumor refers generally to a swelling or lesion formed by an abnormal growth of cells, which may be benign, pre-malignant, or malignant. Most cancer cells form tumors, but some, e.g., leukemia, do not necessarily form tumors. For those cancer cells that form tumors, the terms cancer (cell) and tumor (cell) are used interchangeably.
  • a subject that has a cancer or a tumor is a subject having objectively measurable cancer cells present in the subject's body. Included in this definition are malignant, actively proliferative cancers, as well as potentially dormant tumors or micrometastatses. Cancers which migrate from their original location and seed other vital organs can eventually lead to the death of the subject through the functional deterioration of the affected organs. Hemopoietic cancers, such as leukemia, are able to out-compete the normal hemopoietic compartments in a subject, thereby leading to hemopoietic failure (in the form of anemia, thrombocytopenia and neutropenia) ultimately causing death.
  • cancer examples include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, leukemia, basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and CNS cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma (GBM); hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); lymphoma including Hodgkin's and non-Hodgkin's lymphoma; mela
  • a “cancer cell” is a cancerous, pre-cancerous, or transformed cell, either in vivo, ex vivo, or in tissue culture, that has spontaneous or induced phenotypic changes that do not necessarily involve the uptake of new genetic material.
  • transformation can arise from infection with a transforming virus and incorporation of new genomic nucleic acid, or uptake of exogenous nucleic acid, it can also arise spontaneously or following exposure to a carcinogen, thereby mutating an endogenous gene.
  • Transformation/cancer is associated with, e.g., morphological changes, immortalization of cells, aberrant growth control, foci formation, anchorage independence, malignancy, loss of contact inhibition and density limitation of growth, growth factor or serum independence, tumor specific markers, invasiveness or metastasis, and tumor growth in suitable animal hosts such as nude mice. See, e.g., Freshney, C ULTURE A NIMAL C ELLS : M ANUAL B ASIC T ECH . (3rd ed., 1994).
  • compositions and methods described herein can be administered to a subject having or diagnosed as having cancer.
  • the methods described herein comprise administering an effective amount of compositions described herein, to a subject in order to alleviate a symptom of a cancer.
  • “alleviating a symptom of a cancer” is ameliorating any condition or symptom associated with the cancer. As compared with an equivalent untreated control, such reduction is by at least 5%, 10%, 20%, 40%, 50%, 60%, 80%, 90%, 95%, 99% or more as measured by any standard technique.
  • a variety of means for administering the compositions described herein to subjects are known to those of skill in the art.
  • Such methods can include, but are not limited to oral, parenteral, intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), pulmonary, cutaneous, topical, injection, or intratumoral administration.
  • Administration can be local or systemic.
  • the administration is subcutaneous.
  • the administration of an AsiC as described herein is subcutaneous.
  • an effective amount refers to the amount of of a composition needed to alleviate at least one or more symptom of the disease or disorder, and relates to a sufficient amount of pharmacological composition to provide the desired effect.
  • the term “therapeutically effective amount” therefore refers to an amount that is sufficient to provide a particular anti-cancer effect when administered to a typical subject.
  • An effective amount as used herein, in various contexts, would also include an amount sufficient to delay the development of a symptom of the disease, alter the course of a symptom disease (for example but not limited to, slowing the progression of a symptom of the disease), or reverse a symptom of the disease. Thus, it is not generally practicable to specify an exact “effective amount”. However, for any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using only routine experimentation.
  • Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dosage can vary depending upon the dosage form employed and the route of administration utilized.
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50.
  • Compositions and methods that exhibit large therapeutic indices are preferred.
  • a therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of a composition) which achieves a half-maximal inhibition of symptoms) as determined in cell culture, or in an appropriate animal model.
  • IC50 i.e., the concentration of a composition
  • Levels in plasma can be measured, for example, by high performance liquid chromatography.
  • the effects of any particular dosage can be monitored by a suitable bioassay, e.g., assay for tumor size, among others. The dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
  • the technology described herein relates to a pharmaceutical composition as described herein, and optionally a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers and diluents include saline, aqueous buffer solutions, solvents and/or dispersion media. The use of such carriers and diluents is well known in the art.
  • materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as e
  • wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation.
  • excipient e.g. as described herein.
  • carrier e.g. as described herein.
  • the pharmaceutical composition as described herein can be a parenteral dose form. Since administration of parenteral dosage forms typically bypasses the patient's natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. In addition, controlled-release parenteral dosage forms can be prepared for administration of a patient, including, but not limited to, DUROS®-type dosage forms and dose-dumping.
  • Suitable vehicles that can be used to provide parenteral dosage forms as disclosed within are well known to those skilled in the art. Examples include, without limitation: sterile water; water for injection USP; saline solution; glucose solution; aqueous vehicles such as but not limited to, sodium chloride injection, Ringer's injection, dextrose Injection, dextrose and sodium chloride injection, and lactated Ringer's injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and propylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • Compounds that alter or modify the solubility of a pharmaceutically acceptable salt can also be incorporated into the parenteral dosage forms of the disclosure, including conventional and controlled-release parenteral dosage forms.
  • compositions can also be formulated to be suitable for oral administration, for example as discrete dosage forms, such as, but not limited to, tablets (including without limitation scored or coated tablets), pills, caplets, capsules, chewable tablets, powder packets, cachets, troches, wafers, aerosol sprays, or liquids, such as but not limited to, syrups, elixirs, solutions or suspensions in an aqueous liquid, a non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil emulsion.
  • Such compositions contain a predetermined amount of the pharmaceutically acceptable salt of the disclosed compounds, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington: The Science and Practice of Pharmacy, 21st Ed., Lippincott, Williams, and Wilkins, Philadelphia Pa. (2005).
  • Conventional dosage forms generally provide rapid or immediate drug release from the formulation. Depending on the pharmacology and pharmacokinetics of the drug, use of conventional dosage forms can lead to wide fluctuations in the concentrations of the drug in a patient's blood and other tissues. These fluctuations can impact a number of parameters, such as dose frequency, onset of action, duration of efficacy, maintenance of therapeutic blood levels, toxicity, side effects, and the like.
  • controlled-release formulations can be used to control a drug's onset of action, duration of action, plasma levels within the therapeutic window, and peak blood levels.
  • controlled- or extended-release dosage forms or formulations can be used to ensure that the maximum effectiveness of a drug is achieved while minimizing potential adverse effects and safety concerns, which can occur both from under-dosing a drug (i.e., going below the minimum therapeutic levels) as well as exceeding the toxicity level for the drug.
  • the composition can be administered in a sustained release formulation.
  • Controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled release counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include: 1) extended activity of the drug; 2) reduced dosage frequency; 3) increased patient compliance; 4) usage of less total drug; 5) reduction in local or systemic side effects; 6) minimization of drug accumulation; 7) reduction in blood level fluctuations; 8) improvement in efficacy of treatment; 9) reduction of potentiation or loss of drug activity; and 10) improvement in speed of control of diseases or conditions. Kim, Cherng-ju, Controlled Release Dosage Form Design, 2 (Technomic Publishing, Lancaster, Pa.: 2000).
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body.
  • Controlled-release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, ionic strength, osmotic pressure, temperature, enzymes, water, and other physiological conditions or compounds.
  • a variety of known controlled- or extended-release dosage forms, formulations, and devices can be adapted for use with the salts and compositions of the disclosure. Examples include, but are not limited to, those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548; 5,073,543; 5,639,476; 5,354,556; 5,733,566; and 6,365,185 B1; each of which is incorporated herein by reference.
  • dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View, Calif. USA)), or a combination thereof to provide the desired release profile in varying proportions.
  • active ingredients for example, hydroxypropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems (such as OROS® (Alza Corporation, Mountain View, Calif. USA)), or a combination thereof to provide the desired release profile in varying proportions.
  • OROS® Alza Corporation, Mountain View, Calif. USA
  • the methods described herein can further comprise administering a second agent and/or treatment to the subject, e.g. as part of a combinatorial therapy.
  • a second agent and/or treatment can include radiation therapy, surgery, gemcitabine, cisplastin, paclitaxel, carboplatin, bortezomib, AMG479, vorinostat, rituximab, temozolomide, rapamycin, ABT-737, PI-103; alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethi
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epi
  • the methods of treatment can further include the use of radiation or radiation therapy. Further, the methods of treatment can further include the use of surgical treatments.
  • a chimeric molecule as described herein can be administered in combination with a taxane (e.g. docetaxel or paclitaxel). In some embodiments of any of the aspects described herein, a chimeric molecule as described herein can be administered in combination with paclitaxel. In some embodiments of any of the aspects described herein, an AsiC as described herein can be administered in combination with a taxane. In some embodiments of any of the aspects described herein, an AsiC as described herein can be administered in combination with paclitaxel.
  • a taxane e.g. docetaxel or paclitaxel
  • a chimeric molecule as described herein can be administered in combination with paclitaxel.
  • an AsiC as described herein can be administered in combination with a taxane. In some embodiments of any of the aspects described herein, an AsiC as described herein can be administered in combination with paclitaxel.
  • an effective dose of a composition as described herein can be administered to a patient once. In certain embodiments, an effective dose of a composition can be administered to a patient repeatedly.
  • subjects can be administered a therapeutic amount of a composition comprising such as, e.g. 0.1 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 5 mg/kg, 10 mg/kg, 15 mg/kg, 20 mg/kg, 25 mg/kg, 30 mg/kg, 40 mg/kg, 50 mg/kg, or more.
  • the treatments can be administered on a less frequent basis. For example, after treatment biweekly for three months, treatment can be repeated once per month, for six months or a year or longer.
  • Treatment according to the methods described herein can reduce levels of a marker or symptom of a condition, e.g. by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80% or at least 90% or more.
  • the dosage of a composition as described herein can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment. With respect to duration and frequency of treatment, it is typical for skilled clinicians to monitor subjects in order to determine when the treatment is providing therapeutic benefit, and to determine whether to increase or decrease dosage, increase or decrease administration frequency, discontinue treatment, resume treatment, or make other alterations to the treatment regimen.
  • the dosing schedule can vary from once a week to daily depending on a number of clinical factors, such as the subject's sensitivity to the composition.
  • the desired dose or amount of activation can be administered at one time or divided into subdoses, e.g., 2-4 subdoses and administered over a period of time, e.g., at appropriate intervals through the day or other appropriate schedule.
  • administration can be chronic, e.g., one or more doses and/or treatments daily over a period of weeks or months.
  • dosing and/or treatment schedules are administration daily, twice daily, three times daily or four or more times daily over a period of 1 week, 2 weeks, 3 weeks, 4 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, or 6 months, or more.
  • a composition can be administered over a period of time, such as over a 5 minute, 10 minute, 15 minute, 20 minute, or 25 minute period.
  • “decrease”, “reduced”, “reduction”, or “inhibit” are all used herein to mean a decrease by a statistically significant amount. In some embodiments, “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease by at least 10% as compared to a reference level (e.g.
  • “reduction” or “inhibition” does not encompass a complete inhibition or reduction as compared to a reference level.
  • “Complete inhibition” is a 100% inhibition as compared to a reference level.
  • a decrease can be preferably down to a level accepted as within the range of normal for an individual without a given disorder.
  • the terms “increased”, “increase”, “enhance”, or “activate” are all used herein to mean an increase by a statically significant amount.
  • the terms “increased”, “increase”, “enhance”, or “activate” can mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • a “increase” is a statistically significant increase in such level.
  • a “subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters.
  • Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, canine species, e.g., dog, fox, wolf, avian species, e.g., chicken, emu, ostrich, and fish, e.g., trout, catfish and salmon.
  • the subject is a mammal, e.g., a primate, e.g., a human.
  • the terms, “individual,” “patient” and “subject” are used interchangeably herein.
  • the subject is a mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but is not limited to these examples. Mammals other than humans can be advantageously used as subjects that represent animal models of cancer.
  • a subject can be male or female.
  • a subject can be one who has been previously diagnosed with or identified as suffering from or having a condition in need of treatment (e.g. cancer) or one or more complications related to such a condition, and optionally, have already undergone treatment for cancer or the one or more complications related to cancer.
  • a subject can also be one who has not been previously diagnosed as having cancer or one or more complications related to cancer.
  • a subject can be one who exhibits one or more risk factors for cancer or one or more complications related to cancer or a subject who does not exhibit risk factors.
  • a “subject in need” of treatment for a particular condition can be a subject having that condition, diagnosed as having that condition, or at risk of developing that condition.
  • protein and “polypeptide” are used interchangeably herein to designate a series of amino acid residues, connected to each other by peptide bonds between the alpha-amino and carboxy groups of adjacent residues.
  • protein and “polypeptide” refer to a polymer of amino acids, including modified amino acids (e.g., phosphorylated, glycated, glycosylated, etc.) and amino acid analogs, regardless of its size or function.
  • Protein and “polypeptide” are often used in reference to relatively large polypeptides, whereas the term “peptide” is often used in reference to small polypeptides, but usage of these terms in the art overlaps.
  • polypeptide proteins and “polypeptide” are used interchangeably herein when referring to a gene product and fragments thereof.
  • exemplary polypeptides or proteins include gene products, naturally occurring proteins, homologs, orthologs, paralogs, fragments and other equivalents, variants, fragments, and analogs of the foregoing.
  • nucleic acid or “nucleic acid sequence” refers to any molecule, preferably a polymeric molecule, incorporating units of ribonucleic acid, deoxyribonucleic acid or an analog thereof.
  • the nucleic acid can be either single-stranded or double-stranded.
  • a single-stranded nucleic acid can be one nucleic acid strand of a denatured double-stranded DNA. Alternatively, it can be a single-stranded nucleic acid not derived from any double-stranded DNA.
  • the nucleic acid can be DNA.
  • nucleic acid can be RNA.
  • Suitable nucleic acid molecules are DNA, including genomic DNA or cDNA. Other suitable nucleic acid molecules are RNA, including mRNA.
  • Inhibitors of the expression of a given gene can be an inhibitory nucleic acid or inhibitory oligonucleotide.
  • the inhibitory nucleic acid is an inhibitory RNA (iRNA).
  • the inhibitory nucleic acid is an inhibitory DNA (iDNA).
  • dsRNA Double-stranded RNA molecules have been shown to block gene expression in a highly conserved regulatory mechanism known as RNA interference (RNAi).
  • the inhibitory nucleic acids described herein can include an RNA or DNA strand (the antisense strand) having a region which is 30 nucleotides or less in length, i.e., 8-30 nucleotides in length, generally 19-24 nucleotides in length, which region is substantially complementary to at least part of a precursor or mature form of a target gene's transcript.
  • the use of these inhibitory oligonucleotides enables the targeted degradation of the target gene, resulting in decreased expression and/or activity of the target gene.
  • inhibitory oligonucleotide As used herein, the term “inhibitory oligonucleotide,” “inhibitory nucleic acid,” or “antisense oligonucleotide” (ASO) refers to an agent that contains an oligonucleotide, e.g. a DNA or RNA molecule which mediates the targeted cleavage of an RNA transcript. In one embodiment, an inhibitory oligonucleotide as described herein effects inhibition of the expression and/or activity of a target gene.
  • ASO antisense oligonucleotide
  • Inhibitory nucleic acids useful in the present methods and compositions include antisense oligonucleotides, ribozymes, external guide sequence (EGS) oligonucleotides, siRNA compounds, single- or double-stranded RNA interference (RNAi) compounds such as siRNA compounds, modified bases/locked nucleic acids (LNAs), antagomirs, peptide nucleic acids (PNAs), and other oligomeric compounds or oligonucleotide mimetics which hybridize to at least a portion of the target nucleic acid and modulate its function.
  • EGS external guide sequence
  • siRNA compounds single- or double-stranded RNA interference (RNAi) compounds
  • siRNA compounds single- or double-stranded RNA interference (RNAi) compounds
  • siRNA compounds single- or double-stranded RNA interference (RNAi) compounds
  • LNAs locked nucleic acids
  • PNAs peptide nucleic acids
  • the inhibitory nucleic acids include antisense RNA, antisense DNA, chimeric antisense oligonucleotides, antisense oligonucleotides comprising modified linkages, interference RNA (RNAi), short interfering RNA (siRNA); a micro, interfering RNA (miRNA); a small, temporal RNA (stRNA); or a short, hairpin RNA (shRNA); small RNA-induced gene activation (RNAa); small activating RNAs (saRNAs), or combinations thereof.
  • RNAi interference RNA
  • siRNA short interfering RNA
  • miRNA micro, interfering RNA
  • shRNA small, temporal RNA
  • shRNA short, hairpin RNA
  • RNAa small RNA-induced gene activation
  • saRNAs small activating RNAs
  • inhibitory nucleic acids please see US2010/0317718 (antisense oligos); US2010/0249052 (double-stranded ribonucleic acid (dsRNA)); US2009/0181914 and US2010/0234451 (LNAs); US2007/0191294 (siRNA analogues); US2008/0249039 (modified siRNA); and WO2010/129746 and WO2010/040112 (inhibitory nucleic acids).
  • contacting a cell with the inhibitor results in a decrease in the target RNA level in a cell by at least about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, up to and including 100% of the target mRNA level found in the cell without the presence of the inhibitory oligonucleotide.
  • RNA refers to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript via an RNA-induced silencing complex (RISC) pathway.
  • RISC RNA-induced silencing complex
  • an iRNA as described herein effects inhibition of the expression and/or activity of the target gene.
  • an RNA interference agent includes a single stranded RNA that interacts with a target RNA sequence to direct the cleavage of the target RNA.
  • Dicer Type III endonuclease known as Dicer (Sharp et al., Genes Dev.
  • Dicer a ribonuclease-III-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3′ overhangs (Bernstein, et al., (2001) Nature 409:363).
  • the siRNAs are then incorporated into an RNA-induced silencing complex (RISC) where one or more helicases unwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, et al., (2001) Cell 107:309).
  • RISC RNA-induced silencing complex
  • an RNA interference agent Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleaves the target to induce silencing (Elbashir, et al., (2001) Genes Dev. 15:188).
  • an RNA interference agent relates to a double stranded RNA that promotes the formation of a RISC complex comprising a single strand of RNA that guides the complex for cleavage at the target region of a target transcript to effect silencing of the target gene.
  • the inhibitory oligonucleotide can be a double-stranded nucleic acid (e.g. a dsRNA).
  • a double-stranded nucleic acid includes two nucleic acid strands that are sufficiently complementary to hybridize to form a duplex structure under conditions in which the double-stranded nucleic acid will be used.
  • One strand of a double-stranded nucleic acid (the antisense strand) includes a region of complementarity that is substantially complementary, and generally fully complementary, to a target sequence.
  • the target sequence can be derived from the sequence of an mRNA and/or the mature miRNA formed during the expression of the target gene.
  • the other strand includes a region that is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions.
  • the duplex structure is between 8 and 30 inclusive, more generally between 18 and 25 inclusive, yet more generally between 19 and 24 inclusive, and most generally between 19 and 21 base pairs in length, inclusive.
  • the region of complementarity to the target sequence is between 8 and 30 inclusive, more generally between 18 and 25 inclusive, yet more generally between 19 and 24 inclusive, and most generally between 19 and 21 nucleotides in length, inclusive.
  • the dsRNA is between 15 and 20 nucleotides in length, inclusive, and in other embodiments, the dsRNA is between 25 and 30 nucleotides in length, inclusive.
  • the targeted region of an RNA targeted for cleavage will most often be part of a larger RNA molecule, often an mRNA molecule.
  • a “part” of an mRNA and/or miRNA target is a contiguous sequence of an mRNA target of sufficient length to be a substrate for antisense-directed cleavage (e.g., cleavage through a RISC pathway).
  • Double-stranded nucleic acids having duplexes as short as 8 base pairs can, under some circumstances, mediate antisense-directed RNA cleavage. Most often a target will be at least 15 nucleotides in length, preferably 15-30 nucleotides in length.
  • the duplex region is a primary functional portion of a double-stranded inhibitory nucleic acid, e.g., a duplex region of 8 to 36, e.g., 15-30 base pairs.
  • a functional duplex of e.g., 15-30 base pairs that targets a desired RNA for cleavage an inhibitory nucleic acid molecule or complex of inhibitory nucleic acid molecules having a duplex region greater than 30 base pairs is a double-stranded nucleic acid.
  • a miRNA is a dsRNA.
  • a dsRNA is not a naturally occurring miRNA.
  • an inhibitory nucleic acid agent useful to target the target gene expression is not generated in the target cell by cleavage of a larger double-stranded nucleic acid molecule.
  • target sequence is generally 15-30 nucleotides in length, there is wide variation in the suitability of particular sequences in this range for directing cleavage of any given target RNA.
  • target sequence can be as short as 8 nucleotides, including the “seed” region (e.g. nucleotides 2-8)).
  • RNA sequence a “window” or “mask” of a given size (as a non-limiting example, 21 nucleotides) is literally or figuratively (including, e.g., in silico) placed on the target RNA sequence to identify sequences in the size range that may serve as target sequences.
  • a “window” or “mask” of a given size as a non-limiting example, 21 nucleotides
  • figuratively including, e.g., in silico
  • This process coupled with systematic synthesis and testing of the identified sequences (using assays as described herein or as known in the art) to identify those sequences that perform optimally can identify those RNA sequences that, when targeted with an inhibitory nucleic acid agent, mediate the best inhibition of target gene expression.
  • a double-stranded inhibitory nucleic acid as described herein can further include one or more single-stranded nucleotide overhangs.
  • the double-stranded inhibitory nucleic acid can be synthesized by standard methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied Biosystems, Inc.
  • the antisense strand of a double-stranded inhibitory nucleic acid has a 1-10 nucleotide overhang at the 3′ end and/or the 5′ end.
  • the sense strand of a double-stranded inhibitory nucleic acid has a 1-10 nucleotide overhang at the 3′ end and/or the 5′ end. In one embodiment, at least one end of a double-stranded inhibitory nucleic acid has a single-stranded nucleotide overhang of 1 to 4, generally 1 or 2 nucleotides. Double-stranded inhibitory nucleic acids having at least one nucleotide overhang have unexpectedly superior inhibitory properties relative to their blunt-ended counterparts.
  • one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.
  • nucleotide overhang refers to at least one unpaired nucleotide that protrudes from the duplex structure of an inhibitory nucleic acid, e.g., a dsRNA. For example, when a 3′-end of one strand of a double-stranded inhibitory nucleic acid extends beyond the 5′-end of the other strand, or vice versa, there is a nucleotide overhang.
  • a double-stranded inhibitory nucleic acid can comprise an overhang of at least one nucleotide; alternatively the overhang can comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more.
  • a nucleotide overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside.
  • the overhang(s) may be on the sense strand, the antisense strand or any combination thereof.
  • the nucleotide(s) of an overhang can be present on the 5′ end, 3′ end or both ends of either an antisense or sense strand of a double-stranded inhibitory nucleic acid.
  • blunt or “blunt ended” as used herein in reference to a double-stranded inhibitory nucleic acid mean that there are no unpaired nucleotides or nucleotide analogs at a given terminal end of a dsRNA, i.e., no nucleotide overhang.
  • One or both ends of a double-stranded inhibitory nucleic acid can be blunt. Where both ends of a double-stranded inhibitory nucleic acid are blunt, the double-stranded inhibitory nucleic acid is said to be blunt ended.
  • a “blunt ended” double-stranded inhibitory nucleic acid is a double-stranded inhibitory nucleic acid that is blunt at both ends, i.e., no nucleotide overhang at either end of the molecule. Most often such a molecule will be double-stranded over its entire length.
  • one of the two strands is complementary to the other of the two strands, with one of the strands being substantially complementary to a sequence of a the target gene precursor or mature miRNA.
  • a double-stranded inhibitory nucleic acid will include two oligonucleotides, where one oligonucleotide is described as the sense strand and the second oligonucleotide is described as the corresponding antisense strand of the sense strand.
  • the complementary sequences of a double-stranded inhibitory nucleic acid can also be contained as self-complementary regions of a single nucleic acid molecule, as opposed to being on separate oligonucleotides.
  • inhibitory nucleic acid having a duplex structure of between 20 and 23, but specifically 21, base pairs have been hailed as particularly effective in inducing antisense-mediated inhibition (Elbashir et al., EMBO 2001, 20:6877-6888).
  • others have found that shorter or longer inhibitory nucleic acids can be effective as well.
  • optimized sequences can be adjusted by, e.g., the introduction of modified nucleotides as described herein or as known in the art, addition or changes in overhang, or other modifications as known in the art and/or discussed herein to further optimize the molecule (e.g., increasing serum stability or circulating half-life, increasing thermal stability, enhancing transmembrane delivery, targeting to a particular location or cell type, increasing interaction with silencing pathway enzymes, increasing release from endosomes, etc.) as an expression inhibitor.
  • modified nucleotides as described herein or as known in the art, addition or changes in overhang, or other modifications as known in the art and/or discussed herein to further optimize the molecule (e.g., increasing serum stability or circulating half-life, increasing thermal stability, enhancing transmembrane delivery, targeting to a particular location or cell type, increasing interaction with silencing pathway enzymes, increasing release from endosomes, etc.) as an expression inhibitor.
  • An inhibitory nucleic acid as described herein can contain one or more mismatches to the target sequence.
  • an inhibitory nucleic acid as described herein contains no more than 3 mismatches. If the antisense strand of the inhibitory nucleic acid contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity. If the antisense strand of the inhibitory nucleic acid contains mismatches to the target sequence, it is preferable that the mismatch be restricted to be within the last 5 nucleotides from either the 5′ or 3′ end of the region of complementarity.
  • the strand generally does not contain any mismatch within the central 13 nucleotides.
  • the methods described herein or methods known in the art can be used to determine whether an inhibitory nucleic acid containing a mismatch to a target sequence is effective in inhibiting the expression of the target gene. Consideration of the efficacy of inhibitory nucleic acids with mismatches in inhibiting expression of the target gene is important, especially if the particular region of complementarity in the target gene is known to have polymorphic sequence variation within the population.
  • nucleic acid of an inhibitory nucleic acid is chemically modified to enhance stability or other beneficial characteristics.
  • the nucleic acids featured in the invention may be synthesized and/or modified by methods well established in the art, such as those described in “Current protocols in nucleic acid chemistry,” Beaucage, S. L. et al. (Edrs.), John Wiley & Sons, Inc., New York, N.Y., USA, which is hereby incorporated herein by reference.
  • Modifications include, for example, (a) end modifications, e.g., 5′ end modifications (phosphorylation, conjugation, inverted linkages, etc.) 3′ end modifications (conjugation, DNA nucleotides, inverted linkages, etc.), (b) base modifications, e.g., replacement with stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners, removal of bases (abasic nucleotides), or conjugated bases, (c) sugar modifications (e.g., at the 2′ position or 4′ position) or replacement of the sugar, as well as (d) backbone modifications, including modification or replacement of the phosphodiester linkages.
  • end modifications e.g., 5′ end modifications (phosphorylation, conjugation, inverted linkages, etc.) 3′ end modifications (conjugation, DNA nucleotides, inverted linkages, etc.
  • base modifications e.g., replacement with stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners
  • nucleic acid compounds useful in the embodiments described herein include, but are not limited to nucleic acids containing modified backbones or no natural internucleoside linkages.
  • Nucleic acids having modified backbones include, among others, those that do not have a phosphorus atom in the backbone.
  • modified nucleic acids that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.
  • the modified nucleic acid will have a phosphorus atom in its internucleoside backbone.
  • Modified backbones can include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those) having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3′-5′ to 5′-3′ or 2′-5′ to 5′-2′.
  • Various salts, mixed salts and free acid forms are also included.
  • Modified backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatoms and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • alkene containing backbones sulfamate backbones
  • sulfonate and sulfonamide backbones amide backbones; and others having mixed N, O, S and CH2 component parts.
  • both the sugar and the internucleoside linkage, i.e., the backbone, of the nucleotide units are replaced with novel groups.
  • the base units are maintained for hybridization with an appropriate nucleic acid target compound.
  • One such oligomeric compound, a nucleic acid mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA).
  • PNA peptide nucleic acid
  • the sugar backbone of a nucleic acid is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative U.S. patents that teach the preparation of PNA compounds include, but are not limited to, U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Further teaching of PNA compounds can be found, for example, in Nielsen et al., Science, 1991, 254, 1497-1500.
  • Some embodiments featured in the invention include nucleic acids with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH2-NH—CH2-, —CH2-N(CH3)-O—CH2-[known as a methylene (methylimino) or MMI backbone], —CH2-O—N(CH3)-CH2-, —CH2-N(CH3)-N(CH3)-CH2- and —N(CH3)-CH2-CH2-[wherein the native phosphodiester backbone is represented as —O—P—O—CH2-] of the above-referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above-referenced U.S. Pat. No. 5,602,240.
  • the inhibitory nucleic acids featured herein have morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.
  • Modified nucleic acids can also contain one or more substituted sugar moieties.
  • the inhibitory nucleic acids featured herein can include one of the following at the 2′ position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl.
  • Exemplary suitable modifications include O[(CH2)nO] mCH3, O(CH2).nOCH3, O(CH2)nNH2, O(CH2) nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10.
  • dsRNAs include one of the following at the 2′ position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an inhibitory nucleic acid, or a group for improving the pharmacodynamic properties of an inhibitory nucleic acid, and other substituents having similar properties.
  • the modification includes a 2′ methoxyethoxy (2′-O—CH2CH2OCH3, also known as 2′-O-(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group.
  • 2′-O—CH2CH2OCH3 also known as 2′-O-(2-methoxyethyl) or 2′-MOE
  • 2′-dimethylaminooxyethoxy i.e., a O(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, as described in examples herein below
  • 2′-dimethylaminoethoxyethoxy also known in the art as 2′-O-dimethylaminoethoxyethyl or 2′-DMAEOE
  • 2′-O—CH2-O—CH2-N(CH2)2 also described in examples herein below.
  • modifications include 2′-methoxy (2′-OCH3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2) and 2′-fluoro (2′-F). Similar modifications can also be made at other positions on the nucleic acid of an inhibitory nucleic acid, particularly the 3′ position of the sugar on the 3′ terminal nucleotide or in 2′-5′ linked dsRNAs and the 5′ position of 5′ terminal nucleotide. Inhibitory nucleic acids may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos.
  • nucleobase can also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
  • base nucleobase
  • “unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl anal other 8-substituted adenines and guanines, 5-halo, particularly 5-bromo, 5-trifluoromethyl and other 5-substi
  • nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in Modified Nucleosides in Biochemistry, Biotechnology and Medicine, Herdewijn, P. ed. Wiley-VCH, 2008; those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990, these disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chapter 15, dsRNA Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., Ed., CRC Press, 1993.
  • nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds featured in the invention.
  • These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.
  • 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., dsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are exemplary base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.
  • the nucleic acid of an inhibitory nucleic acid can also be modified to include one or more locked nucleic acids (LNA).
  • LNA locked nucleic acids
  • a locked nucleic acid is a nucleotide having a modified ribose moiety in which the ribose moiety comprises an extra bridge connecting the 2′ and 4′ carbons. This structure effectively “locks” the ribose in the 3′-endo structural conformation.
  • the addition of locked nucleic acids to siRNAs has been shown to increase siRNA stability in serum, and to reduce off-target effects (Elmen, J. et al., (2005) Nucleic Acids Research 33(1):439-447; Mook, O R. et al., (2007) Mol Canc Ther 6(3):833-843; Grunweller, A. et al., (2003) Nucleic Acids Research 31(12):3185-3193).
  • Another modification of the nucleic acid of an inhibitory nucleic acid featured in the invention involves chemically linking to the nucleic acid one or more ligands, moieties or conjugates that enhance the activity, cellular distribution, pharmacokinetic properties, or cellular uptake of the inhibitory nucleic acid.
  • moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556), cholic acid (Manoharan et al., Biorg. Med. Chem.
  • a thioether e.g., beryl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306-309; Manoharan et al., Biorg. Med. Chem. Let., 1993, 3:2765-2770), a thiocholesterol (Oberhauser et al., Nucl.
  • Acids Res., 1990, 18:3777-3783 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651-3654), a palmityl moiety (Mishra et al., Biochim Biophys. Acta, 1995, 1264:229-237), or an octadecylamine or hexylamino-carbonyloxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923-937).
  • a ligand alters the distribution, targeting or lifetime of an inhibitory nucleic acid agent into which it is incorporated.
  • a ligand provides an enhanced affinity for a selected target, e.g, molecule, cell or cell type, compartment, e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand.
  • Preferred ligands will not take part in duplex pairing in a duplexed nucleic acid.
  • Ligands can include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), or globulin); carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); or a lipid.
  • the ligand may also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid.
  • polyamino acids examples include polylysine (PLL), poly L aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine.
  • PLL polylysine
  • poly L aspartic acid poly L-glutamic acid
  • styrene-maleic acid anhydride copolymer examples include poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copoly
  • polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
  • Ligands can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as an hepatopcyte or a macrophage, among others.
  • a cell or tissue targeting agent e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as an hepatopcyte or a macrophage, among others.
  • a targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-gulucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, vitamin A, biotin, or an RGD peptide or RGD peptide mimetic.
  • ligands include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g.
  • intercalating agents e.g. acridines
  • cross-linkers e.g. psoralene, mitomycin C
  • porphyrins TPPC4, texaphyrin, Sapphyrin
  • polycyclic aromatic hydrocarbons e.g., phenazine, dihydrophenazine
  • artificial endonucleases e.g.
  • EDTA lipophilic molecules, e.g, cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid,O3-(oleoyl)lithocholic acid, O3-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted al
  • biotin e.g., aspirin, vitamin E, folic acid
  • transport/absorption facilitators e.g., aspirin, vitamin E, folic acid
  • synthetic ribonucleases e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+ complexes of tetraazamacrocycles), dinitrophenyl, HRP, or AP
  • Ligands can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a hepatocyte or macrophage.
  • Ligands may also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-gulucosamine multivalent mannose, or multivalent fucose.
  • the ligand can be a substance, e.g, a drug, which can increase the uptake of the inhibitory nucleic acid agent into the cell, for example, by disrupting the cell's cytoskeleton, e.g., by disrupting the cell's microtubules, microfilaments, and/or intermediate filaments.
  • the drug can be, for example, taxon, vincristine, vinblastine, cytochalasin, nocodazole, japlakinolide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.
  • a ligand attached to an inhibitory nucleic acid as described herein acts as a pharmacokinetic (PK) modulator.
  • PK modulator refers to a pharmacokinetic modulator.
  • PK modulators include lipophiles, bile acids, steroids, phospholipid analogues, peptides, protein binding agents, PEG, vitamins etc.
  • Examplary PK modulators include, but are not limited to, cholesterol, fatty acids, cholic acid, lithocholic acid, dialkylglycerides, diacylglyceride, phospholipids, sphingolipids, naproxen, ibuprofen, vitamin E, biotin etc.
  • Oligonucleotides that comprise a number of phosphorothioate linkages are also known to bind to serum protein, thus short oligonucleotides, e.g., oligonucleotides of about 5 bases, 10 bases, 15 bases or 20 bases, comprising multiple of phosphorothioate linkages in the backbaone are also amenable to the present invention as ligands (e.g. as PK modulating ligands).
  • ligands e.g. as PK modulating ligands
  • aptamers that bind serum components are also suitable for use as PK modulating ligands in the embodiments described herein.
  • Membrane-destabilizing polyanions interaction with lipid bilayers and endosomal escape of biomacromolecules. Adv. Drug Deliv. Rev. 56, 999-1021; Oliveira, S., van Rooy, I. et al. (2007). Fusogenic peptides enhance endosomal escape improving inhibitory nucleic acid-induced silencing of oncogenes. Int. J. Pharm. 331, 211-4. They have generally been used in the context of drug delivery systems, such as liposomes or lipoplexes.
  • a pH-sensitive fusogenic peptide For folate receptor-mediated delivery using liposomal formulations, for instance, a pH-sensitive fusogenic peptide has been incorporated into the liposomes and shown to enhance the activity through improving the unloading of drug during the uptake process (Turk, M. J., Reddy, J. A. et al. (2002). Characterization of a novel pH-sensitive peptide that enhances drug release from folate-targeted liposomes at endosomal pHs is described in Biochim Biophys. Acta 1559, 56-68).
  • the endosomolytic components can be polyanionic peptides or peptidomimetics which show pH-dependent membrane activity and/or fusogenicity.
  • a peptidomimetic can be a small protein-like chain designed to mimic a peptide.
  • a peptidomimetic can arise from modification of an existing peptide in order to alter the molecule's properties, or the synthesis of a peptide-like molecule using unnatural amino acids or their analogs. In certain embodiments, they have improved stability and/or biological activity when compared to a peptide.
  • the endosomolytic component assumes its active conformation at endosomal pH (e.g., pH 5-6).
  • the “active” conformation is that conformation in which the endosomolytic component promotes lysis of the endosome and/or transport of the modular composition of the invention, or its any of its components (e.g., a nucleic acid), from the endosome to the cytoplasm of the cell.
  • Exemplary endosomolytic components include the GALA peptide (Subbarao et al., Biochemistry, 1987, 26: 2964-2972), the EALA peptide (Vogel et al., J. Am. Chem. Soc., 1996, 118: 1581-1586), and their derivatives (Turk et al., Biochem. Biophys. Acta, 2002, 1559: 56-68).
  • the endosomolytic component can contain a chemical group (e.g., an amino acid) which will undergo a change in charge or protonation in response to a change in pH.
  • the endosomolytic component may be linear or branched.
  • Exemplary primary sequences of endosomolytic components include H2N-(AALEALAEALEALAEALEALAEAAAAGGC)-CO2H (SEQ ID NO: 16); H2N-(AALAEALAEALAEALAEALAAAAGGC)-CO2H (SEQ ID NO: 17); and H2N-(ALEALAEALEALAEA)-CONH2 (SEQ ID NO: 18).
  • more than one endosomolytic component can be incorporated into the inhibitory nucleic acid agent of the invention. In some embodiments, this will entail incorporating more than one of the same endosomolytic component into the inhibitory nucleic acid agent. In other embodiments, this will entail incorporating two or more different endosomolytic components into inhibitory nucleic acid agent.
  • endosomolytic components can mediate endosomal escape by, for example, changing conformation at endosomal pH.
  • the endosomolytic components can exist in a random coil conformation at neutral pH and rearrange to an amphipathic helix at endosomal pH. As a consequence of this conformational transition, these peptides may insert into the lipid membrane of the endosome, causing leakage of the endosomal contents into the cytoplasm. Because the conformational transition is pH-dependent, the endosomolytic components can display little or no fusogenic activity while circulating in the blood (pH ⁇ 7.4).
  • Fusogenic activity is defined as that activity which results in disruption of a lipid membrane by the endosomolytic component.
  • fusogenic activity is the disruption of the endosomal membrane by the endosomolytic component, leading to endosomal lysis or leakage and transport of one or more components of the modular composition of the invention (e.g., the nucleic acid) from the endosome into the cytoplasm.
  • Suitable endosomolytic components can be tested and identified by a skilled artisan.
  • the ability of a compound to respond to, e.g., change charge depending on, the pH environment can be tested by routine methods, e.g., in a cellular assay.
  • a test compound is combined with or contacted with a cell, and the cell is allowed to internalize the test compound, e.g., by endocytosis.
  • An endosome preparation can then be made from the contacted cells and the endosome preparation compared to an endosome preparation from control cells.
  • a change, e.g., a decrease, in the endosome fraction from the contacted cell vs. the control cell indicates that the test compound can function as a fusogenic agent.
  • the contacted cell and control cell can be evaluated, e.g., by microscopy, e.g., by light or electron microscopy, to determine a difference in the endosome population in the cells.
  • the test compound and/or the endosomes can labeled, e.g., to quantify endosomal leakage.
  • an inhibitory nucleic acid agent described herein is constructed using one or more test or putative fusogenic agents.
  • the inhibitory nucleic acid agent can be labeled for easy visulization.
  • the ability of the endosomolytic component to promote endosomal escape, once the inhibitory nucleic acid agent is taken up by the cell, can be evaluated, e.g., by preparation of an endosome preparation, or by microscopy techniques, which enable visualization of the labeled inhibitory nucleic acid agent in the cytoplasm of the cell.
  • the inhibition of gene expression, or any other physiological parameter may be used as a surrogate marker for endosomal escape.
  • circular dichroism spectroscopy can be used to identify compounds that exhibit a pH-dependent structural transition.
  • a two-step assay can also be performed, wherein a first assay evaluates the ability of a test compound alone to respond to changes in pH, and a second assay evaluates the ability of a modular composition that includes the test compound to respond to changes in pH.
  • a ligand or conjugate is a lipid or lipid-based molecule.
  • a lipid or lipid-based molecule preferably binds a serum protein, e.g., human serum albumin (HSA).
  • HSA binding ligand allows for distribution of the conjugate to a target tissue, e.g., a non-kidney target tissue of the body.
  • Other molecules that can bind HSA can also be used as ligands. For example, neproxin or aspirin can be used.
  • a lipid or lipid-based ligand can (a) increase resistance to degradation of the conjugate, (b) increase targeting or transport into a target cell or cell membrane, and/or (c) can be used to adjust binding to a serum protein, e.g., HSA.
  • a serum protein e.g., HSA.
  • the ligand is a cell-permeation agent, preferably a helical cell-permeation agent.
  • a cell-permeation agent preferably a helical cell-permeation agent.
  • such agent is amphipathic.
  • An exemplary agent is a peptide such as tat or antennopedia. If the agent is a peptide, it can be modified, including a peptidylmimetic, invertomers, non-peptide or pseudo-peptide linkages, and use of D-amino acids.
  • the helical agent is preferably an alpha-helical agent, which preferably has a lipophilic and a lipophobic phase.
  • Peptides suitable for use with the present invention can be a natural peptide, e.g., tat or antennopedia peptide, a synthetic peptide, or a peptidomimetic.
  • the peptide can be a modified peptide, for example peptide can comprise non-peptide or pseudo-peptide linkages, and D-amino acids.
  • a peptidomimetic also referred to herein as an oligopeptidomimetic is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide.
  • the attachment of peptide and peptidomimetics to inhibitory nucleic acid agents can affect pharmacokinetic distribution of the inhibitory nucleic acid, such as by enhancing cellular recognition and absorption.
  • the peptide or peptidomimetic moiety can be about 5-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
  • a peptide or peptidomimetic can be, for example, a cell permeation peptide, cationic peptide, amphipathic peptide, or hydrophobic peptide (e.g., consisting primarily of Tyr, Trp or Phe).
  • the peptide moiety can be a dendrimer peptide, constrained peptide or crosslinked peptide.
  • the peptide moiety can include a hydrophobic membrane translocation sequence (MTS).
  • An exemplary hydrophobic MTS-containing peptide is RFGF having the amino acid sequence AAVALLPAVLLALLAP (SEQ ID NO: 19).
  • An RFGF analogue e.g., amino acid sequence AALLPVLLAAP (SEQ ID NO: 20)
  • a hydrophobic MTS can also be a targeting moiety.
  • the peptide moiety can be a “delivery” peptide, which can carry large polar molecules including peptides, oligonucleotides, and protein across cell membranes.
  • sequences from the HIV Tat protein GRKKRRQRRRPPQ (SEQ ID NO: 21)
  • the Drosophila Antennapedia protein RQIKIWFQNRRMKWKK (SEQ ID NO: 22)
  • a peptide or peptidomimetic can be encoded by a random sequence of DNA, such as a peptide identified from a phage-display library, or one-bead-one-compound (OBOC) combinatorial library (Lam et al., Nature, 354:82-84, 1991).
  • OBOC one-bead-one-compound
  • the peptide or peptidomimetic tethered to a dsRNA agent via an incorporated monomer unit is a cell targeting peptide such as an arginine-glycine-aspartic acid (RGD)-peptide, or RGD mimic
  • RGD arginine-glycine-aspartic acid
  • a peptide moiety can range in length from about 5 amino acids to about 40 amino acids.
  • the peptide moieties can have a structural modification, such as to increase stability or direct conformational properties. Any of the structural modifications described below can be utilized.
  • a “cell permeation peptide” is capable of permeating a cell, e.g., a microbial cell, such as a bacterial or fungal cell, or a mammalian cell, such as a human cell.
  • a microbial cell-permeating peptide can be, for example, an ⁇ -helical linear peptide (e.g., LL-37 or Ceropin P1), a disulfide bond-containing peptide (e.g., ⁇ -defensin, ⁇ -defensin or bactenecin), or a peptide containing only one or two dominating amino acids (e.g., PR-39 or indolicidin).
  • a cell permeation peptide can also include a nuclear localization signal (NLS).
  • NLS nuclear localization signal
  • a cell permeation peptide can be a bipartite amphipathic peptide, such as MPG, which is derived from the fusion peptide domain of HIV-1 gp41 and the NLS of SV40 large T antigen (Simeoni et al., Nucl. Acids Res. 31:2717-2724, 2003).
  • the inhibitory nucleic acid oligonucleotides described herein further comprise carbohydrate conjugates.
  • the carbohydrate conjugates are advantageous for the in vivo delivery of nucleic acids, as well as compositions suitable for in vivo therapeutic use, as described herein.
  • carbohydrate refers to a compound which is either a carbohydrate per se made up of one or more monosaccharide units having at least 6 carbon atoms (which may be linear, branched or cyclic) with an oxygen, nitrogen or sulfur atom bonded to each carbon atom; or a compound having as a part thereof a carbohydrate moiety made up of one or more monosaccharide units each having at least six carbon atoms (which may be linear, branched or cyclic), with an oxygen, nitrogen or sulfur atom bonded to each carbon atom.
  • Representative carbohydrates include the sugars (mono-, di-, tri- and oligosaccharides containing from about 4-9 monosaccharide units), and polysaccharides such as starches, glycogen, cellulose and polysaccharide gums.
  • Specific monosaccharides include C5 and above (preferably C5-C8) sugars; di- and trisaccharides include sugars having two or three monosaccharide units (preferably C5-C8).
  • the carbohydrate conjugate further comprises other ligand such as, but not limited to, PK modulator, endosomolytic ligand, and cell permeation peptide.
  • the conjugates described herein can be attached to the inhibitory nucleic acid oligonucleotide with various linkers that can be cleavable or non cleavable.
  • linker or “linking group” means an organic moiety that connects two parts of a compound.
  • Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NR8, C(O), C(O)NH, SO, SO2, SO2NH or a chain of atoms, such as, but not limited to, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocyclylalkyl, heterocyclylalkenyl, heterocyclylalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl, alkylarylalkyl, alkylarylalkenyl, alkylarylalkynyl, alkenylarylalkyl, alkenylarylalkenyl, alkeny
  • a cleavable linking group is one which is sufficiently stable outside the cell, but which upon entry into a target cell is cleaved to release the two parts the linker is holding together.
  • the cleavable linking group is cleaved at least 10 times or more, preferably at least 100 times faster in the target cell or under a first reference condition (which can, e.g., be selected to mimic or represent intracellular conditions) than in the blood of a subject, or under a second reference condition (which can, e.g., be selected to mimic or represent conditions found in the blood or serum).
  • Cleavable linking groups are susceptible to cleavage agents, e.g., pH, redox potential or the presence of degradative molecules. Generally, cleavage agents are more prevalent or found at higher levels or activities inside cells than in serum or blood. Examples of such degradative agents include: redox agents which are selected for particular substrates or which have no substrate specificity, including, e.g., oxidative or reductive enzymes or reductive agents such as mercaptans, present in cells, that can degrade a redox cleavable linking group by reduction; esterases; endosomes or agents that can create an acidic environment, e.g., those that result in a pH of five or lower; enzymes that can hydrolyze or degrade an acid cleavable linking group by acting as a general acid, peptidases (which can be substrate specific), and phosphatases.
  • redox agents which are selected for particular substrates or which have no substrate specificity, including, e.g.,
  • a cleavable linkage group such as a disulfide bond can be susceptible to pH.
  • the pH of human serum is 7.4, while the average intracellular pH is slightly lower, ranging from about 7.1-7.3.
  • Endosomes have a more acidic pH, in the range of 5.5-6.0, and lysosomes have an even more acidic pH at around 5.0.
  • Some linkers will have a cleavable linking group that is cleaved at a preferred pH, thereby releasing the cationic lipid from the ligand inside the cell, or into the desired compartment of the cell.
  • a linker can include a cleavable linking group that is cleavable by a particular enzyme.
  • the type of cleavable linking group incorporated into a linker can depend on the cell to be targeted.
  • Further examples of cleavable linking groups include but are not limited to, redox-cleavable linking groups (e.g. a disulphide linking group (—S—S—)), phosphate-based cleavable linkage groups, ester-based cleavable linking groups, and peptide-based cleavable linking groups.
  • Representative U.S. patents that teach the preparation of RNA conjugates include, but are not limited to, U.S. Pat. Nos.
  • the suitability of a candidate cleavable linking group can be evaluated by testing the ability of a degradative agent (or condition) to cleave the candidate linking group. It will also be desirable to also test the candidate cleavable linking group for the ability to resist cleavage in the blood or when in contact with other non-target tissue.
  • a degradative agent or condition
  • the candidate cleavable linking group for the ability to resist cleavage in the blood or when in contact with other non-target tissue.
  • the evaluations can be carried out in cell free systems, in cells, in cell culture, in organ or tissue culture, or in whole animals.
  • useful candidate compounds are cleaved at least 2, 4, 10 or 100 times faster in the cell (or under in vitro conditions selected to mimic intracellular conditions) as compared to blood or serum (or under in vitro conditions selected to mimic extracellular conditions).
  • the present invention also includes inhibitory nucleic acid compounds that are chimeric compounds.
  • “Chimeric” inhibitory nucleic acid compounds or “chimeras,” in the context of this invention, are inhibitory nucleic acid compounds, e.g. dsRNAs, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of a dsRNA compound.
  • inhibitory nucleic acid typically contain at least one region wherein the nucleic acid is modified so as to confer upon the inhibitory nucleic acid increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the inhibitory nucleic acid may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
  • RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of inhibitory nucleic acid inhibition of gene expression.
  • RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • the nucleic acid of an inhibitory nucleic acid can be modified by a non-ligand group.
  • a number of non-ligand molecules have been conjugated to inhibitory nucleic acids in order to enhance the activity, cellular distribution or cellular uptake of the inhibitory nucleic acid, and procedures for performing such conjugations are available in the scientific literature.
  • Such non-ligand moieties have included lipid moieties, such as cholesterol (Kubo, T. et al., Biochem. Biophys. Res. Comm, 2007, 365(1):54-61; Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med.
  • a thioether e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3:2765), a thiocholesterol (Oberhauser et al., Nucl.
  • Acids Res., 1990, 18:3777 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923).
  • Typical conjugation protocols involve the synthesis of an nucleic acid bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the nucleic acid still bound to the solid support or following cleavage of the nucleic acid, in solution phase. Purification of the nucleic acid conjugate by HPLC typically affords the pure conjugate.
  • aptamer refers to a nucleic acid molecule that is capable of binding to a target molecule, such as a polypeptide.
  • a target molecule such as a polypeptide.
  • an aptamer of the invention can specifically bind to a target molecule, or to a molecule in a signaling pathway that modulates the expression and/or activity of a target molecule.
  • the generation and therapeutic use of aptamers are well established in the art. See, e.g., U.S. Pat. No. 5,475,096.
  • specific binding refers to a chemical interaction between two molecules, compounds, cells and/or particles wherein the first entity binds to the second, target entity with greater specificity and affinity than it binds to a third entity which is a non-target.
  • specific binding can refer to an affinity of the first entity for the second target entity which is at least 10 times, at least 50 times, at least 100 times, at least 500 times, at least 1000 times or greater than the affinity for the third nontarget entity.
  • a reagent specific for a given target is one that exhibits specific binding for that target under the conditions of the assay being utilized.
  • the terms “treat,” “treatment” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder, e.g. cancer.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder associated with a cancer.
  • Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease is reduced or halted.
  • treatment includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment).
  • the term “pharmaceutical composition” refers to the active agent in combination with a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry.
  • pharmaceutically acceptable is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • administering refers to the placement of a compound as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site.
  • Pharmaceutical compositions comprising the compounds disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject.
  • statically significant or “significantly” refers to statistical significance and generally means a two standard deviation (2SD) or greater difference.
  • compositions, methods, and respective component(s) thereof are used in reference to compositions, methods, and respective component(s) thereof, that are essential to the method or composition, yet open to the inclusion of unspecified elements, whether essential or not.
  • compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
  • the term “consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment.
  • Example 1 Gene Knockdown by EpCAM Aptamer-siRNA Chimeras Inhibits Basal-Like Triple Negative Breast Cancers and their Tumor-Initiating Cells
  • EpCAM is a tumor-associated antigen highly expressed on common epithelial cancers and their tumor-initiating cells (T-IC, also known as cancer stem cells). It is demonstrated herein that aptamer-siRNA chimeras (AsiC, an EpCAM aptamer linked to an siRNA sense strand and annealed to the siRNA antisense strand) are selectively taken up and knockdown gene expression in EpCAM+ cancer cells in vitro and in human cancer biopsy tissues.
  • AsiC an EpCAM aptamer linked to an siRNA sense strand and annealed to the siRNA antisense strand
  • EpCAM-AsiCs inhibit colony and mammosphere formation (in vitro T-IC assays) and tumor initiation by EpCAM+ luminal and basal-A triple negative breast cancer (TNBC) cell lines, but not EpCAM ⁇ mesenchymal basal-B TNBCs, in nude mice.
  • Subcutaneously administered EpCAM-AsiCs concentrate in EpCAM+ Her2+ and TNBC tumors and suppress their growth.
  • EpCAM-AsiCs provide an attractive approach for treating epithelial cancer.
  • RNA interference offers the opportunity to treat disease by knocking down disease-causing genes.
  • RNAi RNA interference
  • 1 Recent early phase clinical trials have shown vigorous (75-95%), sustained (lasting for several weeks or up to several months) and safe knockdown of a handful of gene targets in the liver using lipid nanoparticle-encapsulated or GalNAc-conjugated siRNAs.
  • 2-5 The liver, the body's major filtering organ, traps particles and, hence, is relatively easy to transfect.
  • the major obstacle to harnessing RNAi for treating most diseases however has yet to be solved—namely efficient delivery of small RNAs and gene knockdown in cells beyond the liver.
  • the delivery roadblock is a major obstacle to harnessing RNAi to treat cancer. 6
  • TNBC Triple negative breast cancers
  • Most TNBCs have epithelial properties and are classified as basal-like or belong to the basal-A subtype, although a sizable minority are mesenchymal (basal-B subtype).
  • TNBC afflicts younger women and is the subtype associated with BRCA1 genetic mutations. No targeted therapy is available. Although most TNBC patients respond to chemotherapy, within 3 years about a third develop metastases and eventually die. Thus new approaches are needed.
  • RNA aptamer-linked siRNAs known as aptamer-siRNA chimeras (AsiC) have been used in small animal models to treat prostate cancer and prevent HIV infection.
  • EpCAM for targeting basal-like TNBC because EpCAM is highly expressed on epithelial cancers.
  • a high affinity EpCAM aptamer was previously identified.
  • EpCAM also marks tumor-initiating cells (T-ICs, also known as cancer stem cells). 20-27 These cells are thought responsible not only for initiating tumors, but are also relatively resistant to conventional cytotoxic therapy and are thought responsible for tumor relapse and metastasis. Devising therapies to eliminate T-ICs is an important unmet goal of cancer research. 28
  • EpCAM In normal epithelia, EpCAM is only weakly expressed on basolateral gap junctions, where it may not be accessible to drugs. 29 In epithelial cancers it is not only more abundant (by orders of magnitude), but is also distributed along the cell membrane. Ligation of EpCAM promotes adhesion and enhances cell proliferation and invasivity. Proteolytic cleavage of EpCAM releases an intracellular fragment that increases stem cell factor transcription. 30,31 EpCAM's oncogenic properties may make it difficult for tumor cells to develop resistance by down-modulating EpCAM. In one study about 2 ⁇ 3 of TNBCs, presumably the basal-A subtype, stained strongly for EpCAM. 25 The number of EpCAM+ circulating cells is linked to poor prognosis in breast cancer.
  • EpCAM antibody has been evaluated clinically for epithelial cancers, but had limited effectiveness on its own. 37-39 EpCAM expression identifies circulating tumor cells in an FDA-approved method for monitoring metastatic breast, colon and prostate cancer treatment 32-36 . Moreover, about 97% of human breast cancers and virtually 100% of other common epithelial cancers, including lung, colon, pancreas and prostate, stain brightly for EpCAM, suggesting that the platform developed here could be adapted for RNAi-based therapy of common cancers.
  • EpCAM-AsiCs caused targeted gene knockdown in luminal and basal-A TNBC cancer cells and human breast cancer tissues in vitro, but not in normal epithelial cells, mesenchymal tumor cells or normal human breast tissues. Knockdown was proportional to EpCAM expression.
  • EpCAM-AsiC-mediated knockdown of PLK1 a gene required for mitosis, suppressed in vitro T-IC functional assays (colony and mammosphere formation) of epithelial breast cancer lines. Ex vivo treatment specifically abrogated tumor initiation.
  • Subcutaneously injected PLK1 EpCAM-AsiCs were taken up specifically by EpCAM+ basal-A triple negative breast cancer (TNBC) orthotopic xenografts of poor prognosis basal-A and Her2 breast cancers and caused rapid tumor regression.
  • TNBC triple negative breast cancer
  • EpCAM is Highly Expressed on Epithelial Breast Cancer Cell Lines
  • EpCAM expression was examined in breast cancer cell lines. Based on gene expression data in the Cancer Cell Line Encyclopedia40,EpCAM mRNA is highly expressed in basal-A TNBC and luminal breast cancer cell lines, but poorly in basal-B (mesenchymal) TNBCs ( FIG. 1A ). Surface EpCAM staining, assessed by flow cytometry, was 2-3 logs brighter in all luminal and basal-like cell lines tested, than in normal epithelia immortalized with hTERT (BPE) 41 , fibroblasts or mesenchymal TNBCs ( FIG. 1B ). Thus EpCAM is highly expressed in epithelial breast cancer cell lines compared to normal cells or mesenchymal tumors.
  • EpCAM-AsiCs Selectively Knock Down Gene Expression in EpCAM+ Breast Cancer Cells
  • nt nucleotide (nt) aptamer that binds to human EpCAM with 12 nM affinity 19 was identified by SELEX. 42,43 It does not bind to mouse EpCAM (data not shown).
  • EpCAM-AsiCs that linked either the sense or antisense strand of the siRNA to the 3′-end of the aptamer by several linkers were designed and synthesized with 2′-fluoropyrimidine substitutions and 3′-dTdT overhangs to enhance in vivo stability, avoid off-target knockdown of partially complementary genes bearing similar sequences, and limit innate immune receptor stimulation.
  • siRNAs were incorporated to knockdown eGFP (as a useful marker gene); AKT1, an endogenous gene expressed in all the cell lines studied, whose knockdown is not lethal; and PLK1, a kinase required for mitosis, whose knockdown is lethal ( FIG. 9 ).
  • the AsiC that performed best in dose response studies of gene knockdown joined the 19 nt EpCAM aptamer to the sense (inactive) strand of the siRNA via a U-U-U linker ( FIG. 1C ).
  • the EpCAM-AsiC was produced by annealing the chemically synthesized ⁇ 42-44 nt long strand (19 nt aptamer+linker+20-22 nt siRNA sense strand) to a 20-22 nt antisense siRNA strand.
  • EpCAM+ tumor cells To verify selective uptake by EpCAM+ tumor cells, confocal fluorescence microscopy was used to compare internalization of the EpCAM aptamer, fluorescently labeled at the 5′-end with Cy3, in EpCAM+ MDA-MB-468 TNBC cells and BPE, EpCAM dim immortalized breast epithelial cells (data not shown).
  • AsiCs contain only one aptamer, they do not crosslink the receptor they recognize. As a consequence, cellular internalization is slow since it likely occurs via receptor recycling, rather than the more rapid process of activation-induced endocytosis.
  • EpCAM-AsiCs were specifically taken up by EpCAM bright cell lines.
  • EpCAM+ BPLER a basal-A TNBC cell line transformed from BPE by transfection with human TERT, SV40 early region and H-RASV12, took up Alexa-647 EpCAM-AsiCs when analyzed after a 24 hr incubation, but BPE cells did not ( FIG. 1D ).
  • TNBC cells took up Alexa-467 EpCAM-AsiCs, but no uptake was detectable in BPE cells ( FIG. 1E ).
  • eGFP knockdown was compared in these same cell lines, which stably express eGFP, by eGFP EpCAM-AsiCs and lipid transfection of eGFP siRNAs ( FIG. 1D ).
  • transfection of eGFP siRNAs knocked down gene expression equivalently in BPE and BPLER
  • Incubation with EpCAM-AsiCs in the absence of any transfection lipid selectively knocked down expression only in BPLER.
  • AsiC knockdown was uniform and comparable to that achieved with lipid transfection.
  • AKT1 AsiCs was selectively knocked down by EpCAM-AsiCs targeting AKT1 only in EpCAM bright luminal and basal-A TNBCs, but not in mesenchymal basal-B TNBCs, fibroblasts or BPE ells (data not shown).
  • AsiCs targeting eGFP had no effect on AKT1 levels and transfection of AKT1 siRNAs comparably knocked down expression in all the cell lines studied.
  • EpCAM-AsiC knockdown of AKT1 strongly correlated with EpCAM expression ( FIG. 1G ). Similar results were obtained when AKT1 protein was analyzed by flow cytometry in stained transfected cells ( FIG. 1G, 1H ). Thus in vitro knockdown by EpCAM-AsiCs is effective and specific for EpCAM bright tumor cells.
  • EpCAM-AsiCs could be used as anti-tumor agents in breast cancer.
  • CellTiterGlo assay the effect of AsiCs directed against PLK1, a kinase required for mitosis, on survival of 10 breast cancer cell lines that included 5 basal-A TNBCs, 2 luminal cell lines, and 3 basal-B TNBCs.
  • EpCAM-AsiCs targeting PLK1, but not control AsiCs directed against eGFP decreased cell proliferation in the basal-A and luminal cell lines, but did not inhibit basal-B cells ( FIG. 2A ). Lipid transfection of PLK1 siRNAs suppressed the growth of all the cell lines.
  • ligation of the EpCAM aptamer contributed to the anti-proliferative effect of the EpCAM-AsiC we compared survival of cells that were treated with the PLK1 EpCAM-AsiC with cells treated with the aptamer on its own ( FIG. 2C ).
  • the aptamer by itself did not have a reproducible effect on survival of any breast cancer cell lines, possibly because as a monomeric agent it does not cross-link the EpCAM receptor to alter EpCAM signaling.
  • the PLK1 EpCAM-AsiC asserts its specific anti-tumor effect on EpCAM+ breast cancer cells by gene knockdown.
  • EpCAM-AsiCs specifically target EpCAM+ cells when mixed with EpCAM dim non-transformed epithelial cells
  • PLK1 EpCAM-AsiCs or medium used GFP fluorescence to measure their relative survival by flow cytometry 3 days later.
  • EpCAM-AsiCs targeting PLK1 greatly reduced the proportion of surviving EpCAM+ basal-A tumor cells, but had no effect on survival of an EpCAM ⁇ basal-B cell line.
  • PLK1 EpCAM-AsiCs are selectively cytotoxic for EpCAM+ tumor cells when mixed with normal cells.
  • EpCAM-AsiCs Concentrate in EpCAM+ Breast Tumor Biopsy Specimens
  • EpCAM-AsiCs concentrate in human breast tumors relative to normal breast samples within intact tissues. Paired normal tissue and breast tumor biopsies from 3 breast cancer patients were cut into cubes with ⁇ 3 mm edges and placed in Petri dishes. The tumor sample cells were all EpCAM bright and the normal tissue cells were EpCAM dim ( FIG. 3A ). Fluorescently labeled Alexa647-siRNAs (not expected to be taken up by either normal tissue or tumor), Alexa647-cholesterol-conjugated siRNAs (chol-siRNAs, expected to be taken up by both), or Cy3-EpCAM-AsiCs were added to the culture medium and the tissues were incubated for 24 hr before harvest.
  • the Cy3 signal of the AsiC which could be visualized by the naked eye, concentrated only in the tumor specimens and was not detected in normal tissue ( FIG. 3B ).
  • flow cytometry analysis was performed on washed single cell suspensions of the tissue specimens (representative tumor-normal tissue pair ( FIG. 3C ), mean ⁇ SD of triplicate biospies from 3 EpCAM bright paired breast tumor-normal tissue samples ( FIG. 3D )).
  • the EpCAM-AsiC was significantly taken up by the tumor, but not normal tissue, while neither took up the unconjugated siRNA and both took up the chol-siRNA to some extent.
  • EpCAM-AsiCs are selectively delivered to EpCAM bright tumors relative to normal tissue.
  • EpCAM was chosen for targeting in part because EpCAM marks T-ICs and metastasis-initiating cells (M-IC). 20,22,26,27,31
  • T-IC functional surrogate assays T-IC functional surrogate assays
  • PLK1 EpCAM-AsiCs more strongly inhibited colony and mammosphere formation of EpCAM+ basal-A TNBCs and luminal cell lines than paclitaxel, but were inactive against EpCAM ⁇ basal-B TNBCs ( FIGS. 4A-C ).
  • T-IC inhibition was specific, since eGFP AsiCs had no effect. Incubation with PLK1 EpCAM-AsiCs, but not eGFP AsiCs, also reduced the proportion of cells with the phenotype of T-ICs, namely the numbers of CD44 + CD24 low/ ⁇ and ALDH+ cells specifically in basal-A and luminal breast cancer cell lines (data not shown).
  • EpCAM+ MB468 cells stably expressing luciferase were treated overnight with medium or PLK1 or eGFP EpCAM-AsiCs and equal numbers of viable cells were then implanted sc in nude mice.
  • PLK1 EpCAM-AsiCs completely blocked tumor formation assessed by in vivo tumor cell luminescence (data not shown). In contrast similar treatment of basal-B MB436 cells had no effect on tumor initiation (data not shown). Thus PLK1 EpCAM-AsiCs inhibit in vitro T-IC assays and tumor initiation selectively for EpCAM+ breast cancers.
  • EpCAM-AsiCs are Selectively Taken Up by Distant EpCAM+ TNBCs
  • EpCAM-AsiCs need to be taken up by disseminated tumor cells.
  • Intravenous injection of fluorescent EpCAM-AsiCs in the tail vein of mice did not lead to significant AsiC accumulation within subcutaneous tumors implanted in the flanks of nude mice (data not shown), probably because their size ( ⁇ 25 kDa) is below the threshold for kidney filtration and they are rapidly excreted.
  • Linkage to polyethylene glycol greatly enhanced the circulating half-life, tumor accumulation and antitumor therapeutic effect of PSMA-AsiCs in a mouse xenograft model of prostate cancer.
  • EpCAM+ MB468-luc cells were implanted in Matrigel in one flank of a nude mouse and EpCAM ⁇ MB231-luc-mCherry cells were implanted on the opposite flank. Once the luciferase signal of both tumors was clearly detected above background, groups of 5-6 mice were mock treated or injected sc with 5 mg/kg of EpCAM-AsiCs targeting PLK1 or eGFP every 3 d for 2 wks.
  • Targeted therapy so far has relied on using tumor-specific antibodies or inhibitors to oncogenic kinases. No one before has shown that an unconjugated AsiC can have potent antitumor effects or that AsiCs could be administered sc. There is currently no targeted therapy for TNBC or for T-ICs. Developing targeted therapy for TNBC and developing ways of eliminating T-ICs are important unmet goals of cancer research.
  • EpCAM-AsiCs can be used to knockdown genes selectively in epithelial breast cancer cells and their stem cells, sparing normal epithelial cells and stroma, to cause tumor regression and suppress tumor initiation.
  • the EpCAM-AsiCs caused complete tumor regression after only 3 injections. This is a flexible platform for targeted therapy, potentially for all the common epithelial cancers, which uniformly express high levels of EpCAM.
  • EpCAM-AsiCs targeting PLK1 was used herein, the siRNA can be varied to knockdown any tumor dependency gene that would be customized to the tumor subtype or the molecular characteristics of an individual patient's tumor. AsiC cocktails targeting more than one gene would be ideal for cancer therapeutics to lessen the chances of developing drug resistance. Targeted cancer therapy so far has relied on using tumor-specific antibodies or small molecule inhibitors to oncogenic kinases. Using EpCAM as an AsiC ligand and developing RNAi therapy to target cancer stem cells is novel. No one before has shown that an unconjugated AsiC can have potent antitumor effects or that AsiCs could be administered sc.
  • RNAiCs are a flexible platform that can target different cell surface receptors and knockdown any gene or combination of genes. ⁇ Burnett, 2012 #18447; Zhou, 2011 #18448; Thiel, 2010 #18445 ⁇
  • AsiC platform can tackle the delivery roadblock that has thwarted the application of RNAi-based therapy to most diseases. This approach is ideal for personalized cancer therapy, since the choice of genes to target can be adjusted depending on a tumor's molecular characteristics.
  • RNA cocktails can knockdown multiple genes at once to anticipate and overcome drug resistance.
  • AsiCs are the most attractive method for gene knockdown outside the liver. They are better than complicated liposomal, nanoparticle or conjugated methods of delivering RNAs because they are a single chemical entity that is stable in the blood, easy to manufacture, nonimmunogenic, able to readily penetrate tissues and are not trapped in the filtering organs.
  • T-ICs cancer stem cells
  • T-ICs cancer stem cells
  • AsiCs described herein target (epithelial) T-ICs with high efficiency. As such they may eliminate this aggressive subpopulation within tumors at risk for progressive disease (see FIG. 6A, 6B ).
  • EpCAM aptamer used here is ideal for an AsiC drug, since RNAs ⁇ 60 nt can be efficiently synthesized.
  • EpCAM-AsiCs are also a powerful in vivo research tool for identifying the dependency genes of tumors and T-ICs to define novel drug targets.
  • aptamer chimeras could be designed to deliver not only siRNAs but also miRNA mimics or antagomirs, antisense oligonucleotides that function by other mechanisms besides RNAi, or even longer mRNAs or noncoding RNAs (50, 51). They could also be designed to incorporate more than one aptamer, multiple siRNAs, or even toxins or small molecule anticancer drugs.
  • RNAs ⁇ 60 nt can be efficiently synthesized. Not only is the siRNA targeted to the tumor, but the drug targets can also be chosen to attack the tumor's Achilles' heels by knocking down tumor dependency genes. This flexibility can be used for personalized cancer therapy that targets the molecular vulnerabilities of an individual patient's cancer.
  • Human BPE and BPLER cells were grown in WIT medium (Stemgent). MB468 were transduced with a luciferase reporter. All other human cell lines were obtained from ATCC and grown in MEM (MCF7, BT474), McCoy's 5A (SKBR3), RPMI1640 (HCC1806, HCC1143, HCC1937, HCC1954, HCC1187, MB468, T47D) or DMEM (MB231, BT549, MB436) all supplemented with 10% FBS, 1 mM L-glutamine and penicillin/streptomycin (Gibco) unless otherwise indicated. 4T1 mouse breast cancer cells were grown in 10% FBS DMEM.
  • MB468 cells stably expressing Firefly luciferase (MB468-luc) were used and MB231 cells stably expressing Firefly luciferase and mCherry (MB231-luc-mCherry) were selected after infection with pLV-Fluc-mCherry-Puro lentivirus (provided by Andrew Kung, Columbia University). MB231 Cells were selected with puromycin.
  • cells were plated at low density (10,000 cells/well in 96-well plates) and treated immediately. All AsiC and siRNA treatments were performed in either OptiMEM or WIT medium. Cell viability was assessed by CellTiter-Glo (Promega) or by Trypan-Blue staining in 96-well plates.
  • 1,000 viable cells were treated for 6 h in round bottom 96-well plates and then transferred to 10-cm plates in serum-containing medium. Medium was replaced every 3 d. After 8-14 d, cells were fixed in methanol ( ⁇ 20 C) and stained with crystal violet.
  • 1,000/ml viable cells were treated for 6 h in round bottom 96-well plates and then cultured in suspension in serum-free DMEM/F12 1:1 (Invitrogen), supplemented with EGF (20 ng/ml, BD Biosciences), B27 (1:50, Invitrogen), 0.4% bovine serum albumin (Sigma) and 4 ⁇ g/ml insulin (Sigma). Spheres were counted after 1 or 2 weeks.
  • qRT-PCR analysis was performed as described (Petrocca, F., et al. (2008). E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 13, 272-286). Briefly, total RNA was extracted with Trizol (Invitrogen) and cDNA prepared from 1000 ng total RNA using Thermoscript RT kit (Invitrogen) as per the manufacturer's SYBR Green Master Mix (Applied Biosystems) and a BioRad C1000 Thermal Cycler (Biorad). Relative CT values were normalized to GAPDH and converted to a linear scale.
  • Fresh breast or colon cancer and control biopsies were received from the UMASS Tissue Bank, samples were cut into 3 ⁇ 3 ⁇ 3 mm samples and placed in a 96 well plate with 100 ul RPMI. Samples were treated with either Alexa647-siRNA-GFP, Alexa647-chol-siRNA-GFP or Cy3-AsiC-GFP for 24 hr. Samples were photographed and digested. Three samples from each treatment were pooled and put in 10 ml RPMI containing 1 mg/ml collagenase II (Sigma-Aldrich) for 30 minutes at 37° C. with shaking.
  • mice were purchased from the Jackson Laboratory.
  • MB468-luc (5 ⁇ 10 6 ) and MB231-luc-mCherry (5 ⁇ 10 5 ) cells trypsinized with Tryple Express (Invitrogen), resuspended in a 1:1 WIT-Matrigel solution and injected subcutaneously in the flank of 8-week old female Nu/J mice (Stock #002019, Jackson Laboratories). Tumors size was analyzed daily using the IVIS Spectra, after 5 days tumors were clearly visible. Mice bearing tumors of comparable size were randomized into 5 groups and treated with 5 mg/kg of EpCAM-AsiC-PLK1, EpCAM-AsiC-GFP, EpCAM-Aptamer, siRNA-PLK1 or untreated. Mice were treated every 72 h for 14 days.
  • mice were injected sc with eGFP EpCAM-AsiCs (5 mg/kg) or ip with Poly(I:C) (5 or 50 mg/kg).
  • Serum samples collected at baseline and 6 and 16 hr after treatment were stored at ⁇ 80° C. before measuring IFN ⁇ , IL-6 and IP-10 using the ProcartaPlex Multiplex Immunoassay (Affymetrix/eBioscience, San Diego, Calif.).
  • Spleens harvested at sacrifice 16 hr post treatment, were stored in RNAlater (Qiagen) before extracting RNA using TRIZOL (Invitrogen) with the gentleMACS Dissociator (MACS Miltenyi Biotec, San Diego, Calif.).
  • cDNA was synthesized using Superscript III and random hexamers (Invitrogen) and PCR was performed using SsoFast EvaGreen Supermix and a Bio-Rad CFX96 Real-Time PCR System (Bio-Rad Laboratories, Hercules, Calif.) using the following primers:
  • EpCAM is over expressed in basal A and luminal but not basal B breast cancer cell lines (data not shown). FACS was performed with 8 different breast cancer cell lines, testing EpCAM expression levels by flow cytometery using a hEpCAM Antibody. EpCAM is over expressed in all basal A and luminal cells lines and not in basal B.
  • RNA delivery aptamer-siRNA chimeras (AsiC)
  • AsiC aptamer-siRNA chimeras
  • chimeric RNAs composed of a structured RNA, called an aptamer, selected for high affinity binding to a cell surface protein, that is covalently linked to an siRNA.
  • AsiCs are taken up by cells expressing a receptor that the aptamer recognizes and are processed within cells to release the active siRNA.
  • This is a flexible platform that can be modified to target different cells by targeting specific cell surface receptors and can be designed to knockdown any gene or combination of genes.
  • the aptamer was selected for high affinity binding to human EpCAM (CD326 or ESA) which is expressed on all epithelial cells, but is much more highly expressed on epithelial cancers including poorly differentiated breast cancers, such as basal-like TNBC. All the common cancers (lung, pancrease, prostate, breast and colon) have high EpCAM expression and can potentially be targeted.
  • human EpCAM CD326 or ESA
  • epithelial breast cancer cells but not mesenchymal or normal epithelial cells, selectively take up EpCAM-AsiCs and undergo gene knockdown in vitro. Moreover, the extent of knockdown strongly correlates with EpCAM levels. Knockdown of PLK1, a gene needed for mitosis, using EpCAM-AsiCs eliminates cancer cell line growth and stem cell properties including colony and mammosphere formation and tumor initiation in xenografts. This platform can be used to eliminate cancer cells and the malignant cancer stem cells within epithelial tumors.
  • EpCAM AsiCs can be delivered specifically to basal-like tumors and inhibit tumor growth. These AsiCs can also be a powerful research tool for identifying the genes that T-IC cells depend on, which could be good targets for either conventional drugs or RNAi-based drugs.
  • RNA interference A ubiquitous mechanism for regulating gene expression is called RNA interference. It uses small RNAs bearing a short complementary sequence to block the translation of genetic information into proteins. Harnessing this endogenous process offers the exciting possibility to treat disease by knocking down expression of disease-causing genes.
  • the major obstacle is delivering small RNAs into cells, where the RNA interference machinery lies.
  • preliminary clinical studies have shown very promising results without significant toxicity in a few diseases caused by aberrant gene expression in the liver.
  • delivery to the liver an organ that traps particles in the blood, is easier to accomplish than delivering drugs to metastatic tumor cells. Described herein is a strategy for targeting RNAs into epithelial cancer cells that is especially good at targeting the most aggressive type of breast cancer, triple negative breast cancer (TNBC).
  • TNBC triple negative breast cancer
  • cancer stem cells These cells are resistant to chemotherapy drugs and are thought responsible for tumor recurrence and metastasis.
  • An important goal of current cancer research is to replace cytotoxic chemotherapy drugs that are toxic for both cancer cells and normally dividing cells (such as the blood forming cells and cells lining the gut) with agents that have selective activity against the tumor, especially against the cancer stem cells within the tumor.
  • Targeted therapy for one type of breast cancer has revolutionized treatment and significantly improved survival. There is currently no targeted therapy for TNBC or for breast cancer stem cells.
  • RNAs that link an interfering RNA to a structured RNA (aptamer) that recognizes a cell surface protein can knockdown gene expression in aggressive breast cancer cells.
  • Aptamers that bind to proteins highly expressed on breast cancer stem cells and most TNBC cells can knock down proteins required for cancer cell division or survival specifically in the most common subtype of TNBC.
  • These RNAs can be tested, e.g., in both tissue culture and in mouse models of TNBC.
  • Described herein is a platform for harnessing RNA-based drugs to treat poor prognosis breast cancer and demonstration in a mouse model of its efficacy.
  • the cocktail of target genes could be nimbly adjusted. Because normal epithelial cells express low levels of the proteins used for targeting, there may be some uptake and toxicity to normal epithelial cells, which is evaluated herein. However, the platform is flexible so that the therapeutic siRNA cargo can be chosen to kill tumor cells with minimal toxicity to normal cells.
  • Described herein are the design and testing in mouse TNBC models of several molecules capable of causing tumor-specific gene knockdown and tumor suppression.
  • TNBC Triple negative breast cancer
  • RNA interference can be harnessed to knockdown disease-causing genes to treat any disease. 5-9
  • converting small RNAs into drugs is challenging. Recent Phase I and II clinical trials have shown dramatic and durable gene knockdown in the liver ( ⁇ 80-95%, lasting for almost a month after a single injection) with no significant toxicity.
  • AsiCs are composed of an RNA aptamer (a structured RNA with high affinity for a receptor)18,19 covalently linked to an siRNA ( FIGS. 10A-10B ).
  • EpCAM the first described tumor antigen
  • T-IC epithelial cancer tumor-initiating cells
  • the EpCAM aptamer has high affinity (12 nM) and is short (19 nt), which is ideal for an AsiC drug, since RNAs ⁇ 60 nt can be cheaply and efficiently synthesized.
  • the EpCAM-AsiCs consist of a long 42-44 nt strand (19 nt aptamer+3 nt linker+20-22 nt siRNA sense strand) annealed to a 20-22 nt antisense (active) siRNA strand ( FIG. 10B ). They are commercially synthesized with 2′-fluoropyrimidines, which enhance serum stability (T1/2>3d) and block innate immune recognition. 28,54-56
  • EpCAM targeting can cause selective gene knockdown in basal-like TNBCs, relative to normal epithelia. Selective knockdown will reduce both the drug dose and normal tissue toxicity.
  • EpCAM In normal epithelia, EpCAM is only expressed on basolateral gap junctions, where it may not be accessible. In epithelial cancers, it's both more abundant and distributed along the whole cell membrane. EpCAM promotes adhesion, and also enhances proliferation and invasiveness. Proteolytic cleavage of EpCAM releases an intracellular fragment that increases transcription of stem cell factors. The oncogenic properties of EpCAM may make it difficult for tumor cells to develop resistance by down-modulating EpCAM. The number of EpCAM+ circulating cells is linked to poor prognosis in breast cancer.
  • EpCAM+ cells enumerating circulating EpCAM+ cells is the basis of an FDA-approved method for monitoring metastatic breast, colon and prostate cancer treatment.
  • 9 of 9 basal-A TNBC and luminal breast cancer cell lines were strongly EpCAM+, while a normal breast cancer epithelial line and mesenchymal TNBCs had close to background levels ( FIG. 1B ).
  • EpCAM-AsiCs most basal-like TNBCs and luminal breast cancers will likely be targeted by EpCAM-AsiCs.
  • EpCAM-AsiCs selectively knocked down expression in EpCAM+ breast and colon cancer cell lines but not in normal epithelial cells or mesenchymal tumor cells; knockdown was uniform and comparable to lipid transfection, but lipid transfection uniformly knocked down gene expression in all the lines.
  • EpCAM-AsiCs AKT1 knockdown and inhibition of cell proliferation by EpCAM-AsiCs against PLK1, a kinase required for mitosis, correlated with EpCAM levels.
  • BPE normal transformed epithelial cells
  • EpCAM-AsiCs caused PLK1-sensitive cell death only in the tumor cells, sparing BPE cells (not shown).
  • tumor biopsies and normal tissue biopsies were coincubated with fluorescent AsiCs, only the tumors took up the AsiCs and fluoresced (not shown).
  • EpCAM also marks T-ICs. 40,45,58 An important goal of cancer research is to develop a way to target T-ICs. Although the stem cell hypothesis is controversial and may not apply to all cancers, there is good evidence that breast cancers contain a T-IC subpopulation. 59-82 T-Ics are relatively resistant to chemotherapy and are also thought responsible for tumor relapse and metastasis. The AsiCs described herein are designed to target (epithelial) T-ICs with high efficiency. As such they may be suitable for eliminating this aggressive subpopulation within patients at risk for relapse.
  • EpCAM-AsiCs inhibit TNBC T-ICs
  • PLK1 EpCAM-AsiCs but not control GFP AsiCs, eliminated mammosphere and colony formation of breast luminal and basal-like TNBC cell lines ( FIG. 11D ).
  • PLK1 EpCAM-AsiCs also reduced CD44+ CD24low and Aldefluor+ cells (not shown).
  • EphA2 is expressed on epithelial and mesenchymal (basal-A and basal-B, respectively) TNBC cell lines, including their T-ICs, but less than EpCAM and only weakly on other breast cancers. Inhibiting EphA2 reduces tumor growth and angiogenesis in multiple cancer models. Furthermore, EphA2 is selectively accessible on cancer cells, but not normal cells.
  • mouse-human cross-reactive AsiCs which will be valuable for future drug development, since they will enable us to evaluate toxicity and effectiveness in spontaneous mouse tumor models.
  • AsiCs targeting EphA2 can produce dual functioning RNAs that both inhibit EphA2 signaling and cell proliferation and knockdown genes.
  • AsiCs are ideal for personalized cancer therapy, since the genes targeted for knockdown can be adjusted to the molecular characteristics of a tumor. Moreover cocktails of RNAs can be assembled to knockdown multiple genes at once for combinatorial therapy to anticipate and overcome drug resistance. AsiCs not only target the drug to the tumor, but the siRNAs can also be chosen to attack the specific Achilles' heels of the tumor. siRNAs also provide a unique opportunity to target “undruggable” genes. AsiCs that knock down tumor dependency genes, required for tumor, but not normal cell, survival, should have reduced toxicity.
  • BPLER are highly malignant and enriched for T-ICs, forming tumors in nude mice with only 50 cells, while HMLER require >105 cells to initiate tumors.
  • the screen identified 154 genes on which BPLER, but not HMLER, depended. Proteasome genes were highly enriched (P ⁇ 10-14).
  • BPLER dependency gene expression correlated with poor prognosis in breast, but not lung or colon, cancer. Because TNBCs are heterogeneous1,3,4,94, to identify shared dependencies in basal-like TNBCs, we did another screen to test 17 breast cancer cell lines for their dependency on the 154 BPLER dependency genes (unpublished).
  • the proteasome inhibitor bortezomib both killed basal-A TNBCs and also blocked T-IC function, assessed by colony and mammosphere formation, again mostly selectively in basal-like TNBCs. Brief exposure to bortezomib also inhibited colony formation and tumor inhibition of a mouse epithelial TNBC line.
  • Bortezomib strongly inhibited tumor growth of multiple human basal-A lines and primary TNBCs that arose spontaneously in Tp53+/ ⁇ mice, but not basal-B or luminal cell lines. Bortezomib also blocked metastatic lung colonization of IV-injected TNBC cells. However, bortezomib does not penetrate well into solid tumors. The maximum tolerated dose was needed to inhibit proteasome activity and suppress tumors. Although tumor penetration may improve with proteasome inhibitors in development, proteasome gene knockdown might provide more sustained and efficient proteasome inhibition.
  • EpCAM- and EphA2-AsiCs can be used for targeted gene knockdown to treat basal-like TNBC cancers, sparing normal cells, and eliminate the T-Ics within them. There may be some uptake in normal epithelial cells that weakly express EpCAM or EphA2, but gene knockdown will be concentrated in aptamer ligandbright tumor cells.
  • EpCAM- and EphA2-AsiCs target and determine how aptamer ligand level affects gene silencing. uptake/knockdown in cancer tissues vs normal epithelium can also be evaluated. EpCAM-AsiCs can be compared with EphA2-AsiCs for effectiveness in causing knockdown in basal-like TNBCs. It can be determined whether EpCAM-AsiCs and EphA2-AsiCs can target T-ICs to inhibit tumor initiation.
  • PK Pharmacokinetics
  • PD pharmacodynamics
  • TNBC-targeting aptamers Selection of TNBC-targeting aptamers.
  • Aptamers that bind to a chosen target are identified by iterative screening of combinatorial nucleic acid sequence libraries of vast complexity (typically 1012-1014 distinct sequences) by a process termed SELEX (Systematic Evolution of Ligands by Exponential enrichment). 95,96 In the classic method, the RNA library is incubated with the protein target and the RNAs that bind are separated and amplified to generate a pool of binding RNAs. These are again applied in multiple cycles to generate increasingly enriched high affinity RNA pools. Identification of the sequences that emerge after multiple rounds of SELEX was previously accomplished by cloning and sequencing ⁇ 100 individual sequences.
  • aptamers useful for incorporation into AsiCs are efficient internalization into cells. Some ligands of cell-surface proteins are efficiently internalized after binding their cell surface protein targets, while others are not. Another strategy (“toggle SELEX”) selects for cross-reactive aptamers that recognize the same ligand from different species, a useful attribute for preclinical development. By toggling cycles between selection with orthologous protein ligands (e.g., mouse and human forms), it is possible to enrich for cross-species reactive aptamers. 105
  • This library of 51 nt long oligonucleotides is designed with a random region of 20 nucleotides flanked by constant regions of known sequence for PCR amplification at each selection round.
  • Previously described methods will be used to select for high affinity RNAs that bind to immobilized C-terminal tagged proteins.37 (This leaves the N-terminal region exposed to facilitate selection of aptamers that recognize the extracellular domain.)
  • a tagged control protein can be used to pre-clear the RNA aptamer library to remove non-specific binders. 7-10 iterative rounds of SELEX can be performed to enrich for specific aptamers. Enrichment after each round can be monitored by Surface Plasmon Resonance. Enriched pools that show specific binding can be sequenced using high-throughput sequencing. Sequences can be chosen for experimental validation using bioinformatics analysis of the enriched library sequences as described. 97,98,106
  • the top 10-15 sequences from the high throughput sequencing and bioinformatics analysis can be evaluated by Surface Plasmon Resonance to assess relative binding affinities as described, 99,106 using the previously characterized human aptamers for comparison.
  • aptamers can also be evaluated for their ability to inhibit tumor cell line proliferation specifically. Aptamers with this property may be receptor antagonists, which will be verified by examining their effect on cell signaling. Given the high homology between the human and mouse EphA2 extracellular domains (>90% identity; >90% structural homology), identifying aptamers that cross-react with human and mouse EphA2 can be as simple as testing the already selected aptamers for cross-reactivity against mouse. The existing set of 20 human EphA2 aptamers can therefore first be evaluated for the ability to bind mouse EphA2. Alternatively, the approach described above can be followed. For a few of the top aptamers, truncated sequences (lacking either or both of the library adapter sequences) can be synthesized to define the minimal sequence required for binding.
  • Aptamers of ⁇ 20-35 nt in length can be identified for each ligand, which can be designed into AsiCs amenable for chemical synthesis.
  • TNBC-targeting AsiCs In vitro assessment of TNBC-targeting AsiCs and their activity against T-Ics. It can be defined which breast cancer subtypes are efficiently transfected with TNBC-targeting AsiCs and evaluated whether tumor knockdown is specific relative to normal tissue cells, first in cell lines and then in 10 tumor specimens to verify that the results for cell lines translate to 10 tissues. We can also evaluate the potential of TNBC-targeting AsiCs to transfect and target breast T-ICs.
  • AsiC design and initial testing The most attractive aptamers identified above (prioritized based on considerations of affinity, selectivity of binding and expression in poor prognosis cancer vs normal cells, truncation to shorter length, the importance of the ligand in oncogenesis and stem cell behavior, receptor antagonism and cross-species reactivity) can be designed into AsiCs by linkage to siRNAs targeting eGFP, AKT1 and PLK1 (vs control scrambled siRNAs) that have been used for the initial EpCAM-AsiCs as described above herein.
  • Basal-like NBC cell lines stably expressing destabilized (d1)EGFP protein T1/2 of ⁇ 1 hr
  • d1EGFP protein T1/2 of ⁇ 1 hr
  • Basal-like NBC cell lines stably expressing destabilized (d1)EGFP protein T1/2 of ⁇ 1 hr
  • GFP expression can be readily quantified by flow and imaging, and its knockdown has no biological consequences.
  • the short T1/2 allows for rapid and sensitive detection of knockdown.
  • AKT1 which is expressed in all cells, is a good endogenous gene to study, since its knockdown does not much affect cell viability.
  • PLK1 is used for its antitumor effect because its knockdown is cytotoxic to dividing cells. Described herein is robust and reproducible gene knockdown with EpCAM-AsiCs targeting each of these genes. AsiCs will be chemically synthesized with 2′-fluoropyrimidines for stability and inhibition of innate immune recognition and dT residues at their 3′-ends to protect against exonuclease digestion. The 2 strands will be annealed to generate the final RNA ( FIG. 10A-10B ).
  • AsiCs can be evaluated and compared to the original EpCAM-AsiC (as positive control) and CD4 ⁇ or PSMA ⁇ AsiCs (as negative control) in in vitro dose response experiments for AsiC uptake (using fluorophores such as AF-647 (which doesn't affect AsiC activity) conjugated to the 3′end of the short strand), gene knockdown and reduced tumor cell line growth and survival.
  • fluorophores such as AF-647 (which doesn't affect AsiC activity) conjugated to the 3′end of the short strand
  • Types of breast cancer responsive to TNBC-targeting AsiCs It can be determined which types of breast cancer can be transfected with the selected AsiCs and how specific gene knockdown is in tumors relative to normal epithelial cells.
  • In vitro knockdown by the selected AsiCs in 20 human breast cancer cell lines that represent the common breast cancer subtypes, but are weighted towards TNBC (14 TNBC lines, plus a sampling of luminal and Her2+ cell lines) can be evaluated.
  • 93 Aptamer ligand expression, uptake of fluorescent-labeled AsiC and gene silencing can be compared to BPE57 and fibroblast lines as negative controls.
  • This large panel of cell lines can permit evaluation of how cell surface EpCAM and EphA2 levels influence RNA uptake and gene silencing and whether there is an expression threshold needed for efficient knockdown.
  • a dose response experiment can permit verification that the high affinity of the aptamers is preserved in the AsiC. Specificity of uptake (versus nonspecific “sticking”) will be verified by using acid washing to remove loosely adhered aptamers and showing that binding is competed by unlabeled aptamers and eliminated when cells are trypsinized prior to treatment.
  • AsiC-mediated transfection will be compared to lipid transfection as positive control and to naked siRNA as negative control. Knockdown will be assessed by flow cytometry and qRT-PCR after 5 d, the optimal time for AsiC-mediated knockdown.
  • TNBC-targeting AsiCs Do epithelial primary breast cancer cells preferentially take up TNBC-targeting AsiCs and show knockdown relative to normal epithelial cells in tissue explants?
  • Tumor typing can be confirmed by histology and immunohistochemistry (IHC) staining for ER, PR, Her2 and E-cadherin. If the aptamer recognizes the mouse ligand, we can also assess potential toxicity to normal epithelia using mouse tumor/normal tissues.
  • IHC immunohistochemistry
  • Biopsies cut into ⁇ 3 ⁇ 3 ⁇ 3 mm3 pieces, can be transfected in microtiter wells, which should mimic in vivo uptake after SQ or IV infusion.
  • Lipofectamine encapsulated siRNAs and cholesterol-conjugated siRNAs are both effective at gene knockdown of normal epithelial cells in polarized columnar and squamous genital tract mucosa108,109, while naked siRNAs are not taken up. Similar results are expected with these controls in normal breast epithelial tissue.
  • siRNAs to target epithelial genes, which we have previously knocked down (such as E-cadherin, cytokeratin (CK)-5 (a good marker of basal cells) and 14, and nectin-1) 93,108,109, whose expression can be readily followed by IHC, fluorescence microscopy (FM) or flow cytometry of isolated cells. Staining of the target gene can be correlated with staining for phenotypic markers and fluorescently labeled siRNAs to determine which cell types are targeted. Pan-CK antibody can distinguish epithelial cells (normal and tumor) from stroma.
  • EpCAM- and EphA2-AsiCs will be taken up by and cause gene silencing in T-ICs and can be used for targeted therapy to eliminate or cripple T-IC capability within tumors.
  • multicolor flow cytometry of EpCAM, EphA2, CD44 and CD24 in a panel of breast cancer lines can be used to identify which breast cell lines have putative T-IC populations that contain cells that stain brightly for EpCAM and/or EphA2.
  • EpCAM/EphA2 staining of mammospheres and Aldefluor+ cells121,123-125 generated from these cell lines can be used to analyze asiC uptake and gene silencing in T-IC subpopulations.
  • AsiCs can be taken up by EpCAM+ or EphA2+ CD44+ CD24 ⁇ /dim Aldefluor+ cells.
  • To assess gene knockdown in T-IC phenotype cells we can monitor GFP expression in the T-IC population and remaining cells by flow cytometry and qRT-PCR after treatment with eGFP or control siRNA-bearing AsiCs. We can also assess knockdown of endogenous PLK1 and AKT1.
  • PSMA2 proteasome component
  • MSI1 Musashi
  • BMI1 RNA binding protein in breast T-ICs that regulates Wnt and Notch signaling126-130 or BMI1
  • knocking down PLK1, MSI1, BMI1 or PSMA2 can reduce T-IC numbers, proliferation and function in the T-ICs from some cell lines, but different genes may be more active for different breast cell lines.
  • proteasome inhibition eliminated T-ICs in basal-like TNBCs, but only in 1 of 3 mesenchymal TNBC cell lines and not in more differentiated non-TNBC tumors.
  • the knockdown approaches that suppress T-IC can be further investigated by experiments using chemical inhibitors where available (such as bortezomib) or by examining whether knocking down other genes in the same pathway (such as NOTCH1, ⁇ -catenin or WNT1 for MSI1) also has anti-T-IC activity.
  • chemical inhibitors where available such as bortezomib
  • knocking down other genes in the same pathway such as NOTCH1, ⁇ -catenin or WNT1 for MSI1
  • AsiCs that look promising in vitro.
  • Cell lines, treated overnight with the chosen AsiCs (and as negative controls AsiCs that use PSMA aptamer or contain eGFP siRNA), will be assessed for viability.
  • ex vivo treated cells will be injected in a range of cell numbers orthotopically into NOD/scid/“c ⁇ / ⁇ (NSG) mice (these mice have the highest take for tumor implantation).
  • NSG NOD/scid/“c ⁇ / ⁇
  • Bortezomib treatment for 24 hr can serve as a positive control.
  • a few of the AsiCs that perform best can next be evaluated in vivo using nude mice bearing mammary fatpad xenografts of an aptamer ligand+ basal-A TNBC line, such as MB468 or HCC1187, on one side compared to ligand-breast cancer cell line, such as basal-B MB231, on the other ( ⁇ 5-8 mice/gp to obtain reproducible statistics based on our experience with these models).
  • basal-A TNBC line such as MB468 or HCC1187
  • Tissue sections can be assessed for tissue damage and the blood can be analyzed for hematological, liver and kidney toxicity by blood counts and serum chemistries. Toxicity associated with induction of innate immunity or inflammation can be assessed by ELISA assays of serum interferons and inflammatory cytokines. The circulating T1/2 and proportion of the injected drug that localizes to the EpCAM+ tumor can be calculated.
  • MTD maximally tolerated dose
  • Control mice can be treated with PBS or naked siRNAs, AsiCs bearing a scrambled siRNA and PLK1 PSMA-AsiCs. Tumor size can be quantified by imaging and calipers. If the antitumor effect is suboptimal, the dosing regimen can be adjusted to the maximally tolerated regimen.
  • mice can also compare the effect of PLK1 knockdown and standard-of-care chemotherapy, administered on their own and in combination to anticipate potential clinical studies. If there is complete tumor regression, we can evaluate decreased doses. Effective regimens can also be evaluated in mice implanted with a few other basal-A TNBC lines to verify the generality of the antitumor response. We can also evaluate AsiC treatment after tumor cells are injected IV to determine effectiveness against distal metastases. At the time of sacrifice, mice can be sacrificed and mammary fatpads can be inspected for residual microscopic or macroscopic tumor by FM, H&E and IHC.
  • Residual tumor cells can also be assessed for EpCAM/EphA2 expression to determine whether tumor resistance may have developed as a consequence of down-regulating the aptamers ligand.
  • Treated mice can also be observed for clinical signs of toxicity and at time of sacrifice can be carefully examined for gut and bone marrow toxicity, by blood counts and pathological examination of gut, bone marrow and spleen.
  • AsiCs designed with the cross-reacting aptamers can be used to evaluate normal epithelial toxicity. Using our best AsiC design, we can next begin to compare PLK1 knockdown with knockdown of TNBC dependency genes (such as PSMA2 or MCL1) identified in our siRNA screen93 tested alone or in combination with PLK1.
  • RNA interference offers the exciting opportunity to treat disease by knocking down disease-causing genes. Recent early phase clinical trials have shown promising and sustained gene knockdown and/or clinical benefit in a handful of diseases caused by aberrant gene expression in the liver.
  • the major obstacle to harnessing RNAi for cancer treatment is delivery of small RNAs to disseminated cancer cells.
  • Targeted gene knockdown in epithelial cancer cells in vitro can be achieved using chimeric RNAs composed of a structured RNA, called an aptamer, selected for high affinity binding to EpCAM, that is covalently linked to an siRNA.
  • EpCAM aptamer-siRNA chimeras are taken up by EpCAM+ cells and selectively cause gene knockdown in epithelial breast cancer cells, but not normal epithelial cells.
  • knockdown of PLK1 with EpCAM-AsiCs suppresses colony and mammosphere formation of epithelial breast cancer lines, in vitro assays of tumor-initiating potential, and tumor initiation.
  • EpCAM-AsiCs are taken up specifically by EpCAM+ basal-A triple negative breast cancer (TNBC) orthotopic xenografts and cause rapid tumor regression.
  • TNBC has the worst prognosis of any breast cancer and there is no targeted therapy for it.
  • EpCAM-AsiCs can be used for targeted gene knockdown to treat epithelial (basal-like) TNBC cancers, sparing normal cells, and eliminate the T-ICs within them.
  • It can be defined which breast cancer subtypes can be targeted by EpCAM-AsiCs and determine how EpCAM level affects uptake and gene silencing. Relative uptake/knockdown in cancer cells expressing EpCAM and normal epithelium can be evaluated in human breast cancer tissue explants. It can also be determined whether EpCAM-AsiCs can target breast T-ICs to disrupt tumor initiation.
  • EpCAM-AsiCs can be optimized for cell uptake, endosomal release, systemic delivery and in vivo gene knockdown.
  • Pharmacokinetics (PK) and pharmacodynamics (PD) of EpCAM-AsiC uptake and gene silencing and tumor suppression can be evaluated using live animal imaging in TNBC cell line xenograft models.
  • PK Pharmacokinetics
  • PD pharmacodynamics
  • the antitumor effect of knockdown of PLK1 which is needed for cell proliferation can be evaluated.
  • knockdown of novel gene targets identified in a genome-wide siRNA screen for TNBC genetic dependencies will be evaluated in mouse xenograft models.
  • An optimized EpCAM-AsiC and knowledge of its PK, PD and possible toxicity can be used in experiments for further toxicity and other preclinical studies.
  • EpCAM aptamer-siRNA chimeras as a method for targeted gene knockdown in basal-like triple negative breast cancer and other epithelial cancers and the tumor-initiating cells within them.
  • These RNAs provide a versatile and flexible platform for RNA-based drugs to treat poor prognosis breast cancers.
  • EpCAM aptamer on its own does not affect cell growth or viability of EpCAM+ breast tumor cell lines (not shown); (2) when normal breast biopsies are mixed with EpCAM+ TNBC human breast tumor tissues in vitro, fluorescent EpCAM-AsiCs only concentrate in the tumor ( FIG. 14 ); (3) treatment of EpCAM+ luminal and basal-A TNBC cells, but not mesenchymal TNBCs, with PLK1 EpCAM-AsiCs blocks in vitro assays of tumor-initiating cells (T-IC, colony and mammosphere formation) and in vivo tumor initiation ( FIGS.
  • T-ICs are heterogeneous and plastic in epithelial/mesenchymal gene expression. Although mesenchymal traits may facilitate initial tissue invasion, formation of clinically significant metastases (colonization) may require epithelial properties. EpCAM-mediated delivery of siRNA effectively blocks tumor initiation, but only for epithelial (basal-A TNBC, luminal) breast cancers.
  • EpCAM-AsiCs are not taken up by normal human breast biopsies.
  • TNBC Triple negative breast cancer
  • RNAi RNA interference
  • An ideal therapy would selectively knockdown genes in cancer cells, while sparing most normal cells to minimize toxicity.
  • RNAs that use an aptamer (a structured nucleic acid selected for high affinity binding to a target molecule against EpCAM (also known as CD326 or ESA)“+, the first described tumor antigen.
  • EpCAM is highly expressed on epithelial breast cancers (including basal-like TNBC)—on average 400-fold more than on normal breast tissue. It is also highly expressed on other epithelial cancers and is a marker of “cancer stem cells” (also called tumor-initiating cells (T-IC)).
  • Aptamer-siRNA chimeras (AsiC) covalently link a targeting aptamer to an siRNA ( FIG. 10B ). Dicer cleaves the siRNA from the aptamer inside cells.
  • Epithelial breast cancer cells but not mesenchymal or normal epithelial cells, selectively take up EpCAM-AsiCs and undergo gene knockdown in vitro. Moreover, knockdown strongly correlates with EpCAM levels. Knockdown of PLK1, a gene needed for mitosis, using EpCAM-AsiCs eliminates colony and mammosphere formation (in vitro assays that correlate with self renewal and tumor initiation) and tumor initiation in vivo, suggesting that EpCAM-AsiCs might be used to target T-ICs. Sc injection of PLK1 EpCAM-AsiCs caused complete regression of EpCAM+ TNBC xenografts, but had no effect on EpCAM ⁇ mesenchymal TNBCs.
  • EpCAM-AsiCs can be used for targeted gene knockdown to treat basallike TNBC cancers, sparing normal cells, and eliminate the T-ICs within them.
  • AsiCs have important advantages for cancer treatment compared to RNA delivery by nanoparticles, liposomes or RNA-binding proteins—(1) they bypass liver and lung trapping and concentrate in tumors; (2) as a single RNA molecule they are simpler and cheaper to manufacture than multicomponent drugs; (3) they have virtually no toxicity and do not stimulate innate immunity or inflammation or cause significant off-target effects; (4) because they do not elicit antibodies, they can be used repeatedly; (5) they are stable in serum and other body fluids.
  • EpCAM-AsiCs It can be defined which breast cancer subtypes can be targeted by EpCAM-AsiCs and determine how EpCAM level affects uptake and gene silencing.
  • the relative uptake/knockdown in cancer tissues vs normal epithelium can be evaluated. It can also be determined whether EpCAM-AsiCs can target breast T-ICs to inhibit tumor initiation.
  • An important aim is to optimize EpCAM-AsiCs for uptake, endosomal release, systemic delivery and in vivo knockdown.
  • Pharmacokinetics (PK) and pharmacodynamics (PD) of EpCAM-AsiC uptake, gene silencing and tumor suppression will be evaluated by live animal imaging in TNBC orthotopic xenografts.
  • Described herein is: the verification of selective EpCAM-AsiC activity in epithelial breast cancers compared with normal epithelia and evaluate the potential of EpCAM-AsiCs to transfect and eliminate breast T-ICs (i.e., cancer stem cells); optimization of EpCAM-AsiCs to transfect and knockdown genes in epithelial TNBC cells in vitro and for systemic delivery and tumor concentration in vivo, and define PK and PD and maximally tolerated dose; evaluation of the antitumor effect of optimized EpCAM-AsiCs targeting PLK1 and novel dependency genes of basal-like TNBC in human epithelial TNBC models of primary and metastatic cancer in mice
  • TNBCs are heterogeneous, poorly differentiated tumors that may need to be treated by subtype or with individualized therapy. 1,3,4,72 Most TNBCs are basal-like or belong to the basal-A subtype. Described herein is a flexible, targeted platform for treating basal-like TNBCs that is suitable for personalized therapy. Not only will the drug be targeted to the tumor, but the drug targets can also be chosen to attack the tumor's Achilles' heels by knocking down tumor dependency genes.
  • This present approach delivers small interfering RNAs (siRNA) into epithelial cancer cells by linking them to an RNA aptamer that binds to EpCAM ( FIG. 10B ), a cell surface receptor over-expressed on epithelial cancers, including basal-like TNBCs.
  • EpCAM is highly expressed on epithelial cancers and their T-Ics.
  • EpCAM targeting can cause selective gene knockdown in basal-like TNBCs, but not normal epithelia. Selective knockdown will both reduce the drug dose and reduce tissue toxicity.
  • RNAi When RNAi was found in mammals, small RNAs were hailed as the next new drug class. Soon investigators realized that getting RNAi to work as a drug was not simple., however, after addressing the main obstacle to RNA therapy (cellular uptake), there is now optimism about RNAi-based drugs. Recent phase I/II studies have shown 80-95% gene knockdown in hypercholesterolemia, transthyretin-related amyloidosis, hepatitis C, hemophilia and liver metastasis, caused by aberrant liver gene expression. However, applying RNAi for cancer therapy is still a dream. The major obstacle to harnessing RNAi for cancer is delivering small RNAs into disseminated cells. Described herein are methods and compositions that overcome this problem, e.g., by the use of AsiCs.
  • AsiCs are a flexible platform that can target different cell surface receptors and knockdown any gene or combination of genes.
  • the AsiC platform can tackle the delivery roadblock that has thwarted the application of RNAi-based therapy to most diseases. This approach is ideal for personalized cancer therapy, since the choice of genes to target can be adjusted depending on a tumor's molecular characteristics.
  • RNA cocktails can knockdown multiple genes at once to anticipate and overcome drug resistance.
  • Described herein is the development of an optimized EpCAM-AsiC with well defined PK/PD.
  • T-ICs cancer stem cells
  • AsiCs AsiCs
  • EpCAM-AsiCs can also be a powerful in vivo research tool for identifying the dependency genes of tumors and T-ICs to define novel drug targets.
  • Described herein is a novel targeted therapy for epithelial cancers, and the T-ICs within them by targeting EpCAM, a tumor antigen widely over-expressed in epithelial cancers and their T-ICs.
  • Targeted therapy so far has relied on using tumor-specific antibodies or inhibitors to oncogenic kinases. No one before has shown that an unconjugated AsiC can have potent antitumor effects or that AsiCs could be administered sc.
  • the methods described herein are targeted in 2 ways—the aptamer specifically delivers the therapeutic RNA to tumor cells, while the genes chosen for knockdown can be selected based on the specific molecular dependencies of the targeted tumor.
  • basal-like TNBCs and their T-ICs are selectively dependent on the proteasome, MCL1 and the U4/U6-U5 tri-snRNP splicing complex. This work can identify a new set of drug targets, suitable for both conventional and RNAi-based drugs.
  • the trafficking of siRNAs in transfected cells can be examined and each step of RNA processing in cells be systematically optimized to improve the drug features of an siRNA.
  • CD4-AsiCs durably knockdown gene expression in CD4+ T lymphocytes and macrophages and inhibit HIV transmission to humanized mice.
  • CD4-AsiCs specifically suppressed gene expression in CD4+ T cells and macrophages in polarized human cervicovaginal tissue explants and in the female genital tract of humanized mice. Because they are monomeric and don't cross-link the receptor, CD4-AsiCs did not activate the targeted cells. They also did not stimulate innate immunity Intravaginal application of only 80 pmol of CD4-AsiCs directed against HIV genes and/or CCR5 to humanized mice completely blocked HIV sexual transmission. RNAi-mediated gene knockdown in vivo lasted several weeks.
  • CD4-AsiCs are promising for use in an HIV microbicide.
  • PEG polyethylene glycol
  • EpCAM-AsiCs selectively knockdown gene expression in EpCAM+ cancer cells
  • the EpCAM-AsiCs have a ⁇ 42-44 nt long strand (19 nt aptamer+linker+20-22 nt siRNA strand) annealed to a 20-22 nt complementary siRNA strand ( FIG. 10B ).
  • EpCAM was high in all luminal and basal-like cell lines tested, but close to background in normal epithelia immortalized with hTERT (BPE) 94, fibroblasts and mesenchymal TNBCs ( FIG. 1B ).
  • BPE hTERT
  • fibroblasts fibroblasts and mesenchymal TNBCs
  • FIG. 10B Gene knockdown of eGFP and AKT1 by EpCAM-AsiCs was uniform and selective for EpCAM+ cells and as effective as siRNA lipid transfection, which was not selective ( FIG. 13A-13C ).
  • AKT1 knockdown and inhibition of cell proliferation by PLK1 EpCAM-AsiCs strongly correlated with EpCAM levels ( FIG. 11B-11C ).
  • EpCAM ⁇ BPE cells were mixed with epithelial TNBC cell lines, EpCAM-AsiCs knocked down AKT1 and caused PLK1-sensitive cell death only in tumor cells, sparing the normal epithelial cells (not shown). The proportion of surviving tumor cells decreased 7-fold after 3 d.
  • EpCAM-AsiCs are specific for epithelial tumor cells.
  • EpCAM-AsiCs inhibit T-ICs of EpCAM+ tumors.
  • EpCAM was chosen for targeting partly because EpCAM marks T-ICs and metastasis-initiating cells (M-IC).
  • M-IC metastasis-initiating cells
  • PLK1 EpCAM-AsiCs more strongly inhibited colony and mammosphere formation of multiple EpCAM+ basal-like TNBCs and a luminal cell line than paclitaxel, but was inactive against EpCAM ⁇ basal-B TNBCs ( FIG. 15A-15C ).
  • EpCAM-AsiC's effect on tumor initiation viable luc+ EpCAM+ MB468 and EpCAM ⁇ MB231 cells, treated overnight with medium or PLK1 or GFP EpCAM-AsiCs, were implanted sc in nude mice.
  • PLK1 EpCAM-AsiCs blocked tumor formation, but only in EpCAM+ tumors ( FIG. 16 and data not shown).
  • EpCAM-AsiCs inhibit tumor initiation in EpCAM+ breast cancers.
  • EpCAM-AsiCs are selectively taken up by EpCAM+ TNBCs and cause tumor regression
  • EpCAM-AsiCs were delivered to EpCAM+ TNBCs and cause tumor regression
  • FIG. 17A-17B EpCAM-AsiCs concentrated only in the EpCAM+ tumor.
  • Mice bearing bilateral tumors were mock treated or injected biweekly with PLK1 or GFP EpCAM-AsiCs and tumor growth was followed by luminescence.
  • the EpCAM+ tumors rapidly completely regressed only in mice that received the PLK1-targeting AsiCs ( FIG.
  • RNA-containing late endosomes released a small fraction of their cargo RNA, which diffused rapidly to fill the cytosol (data not shown). Release occurred during a narrow time frame, ⁇ 15-20 min after endocytosis. ⁇ 104 siRNAs were released in a typical event.
  • GFP siRNAs In HeLa cells, stably expressing eGFP-dl, GFP siRNAs caused GFP expression to decrease rapidly after endosomal release with a T1/2 of ⁇ 2.5 h. Only 1000 cytosolic siRNAs were needed for efficient gene silencing. Release triggered autophagy, which sequestered the RNA-containing endosome within a double autophagic membrane. No release occurred after that.
  • BDGs basal-like TNBC dependency genes
  • Proteasome inhibitor sensitivity was a shared feature of basal-A TNBCs and correlated with MCL1 dependency. Normal breast epithelial cells, luminal breast cancer lines and mesenchymal TNBC lines did not depend on the proteasome or MCL1. Proteasome inhibition not only killed basal-A TNBCs, it also blocked T-IC function by colony and mammosphere assays, again mostly selectively in basal-like TNBCs. Brief exposure to bortezomib also inhibited tumor initiation of a mouse basallike TNBC line.
  • TNBCs are heterogeneous1,3,4,72, we rescreened the 154 BPLER dependency genes in 4 basal-A TNBC and 3 luminal human cancer lines. Our goal was to identify additional shared dependencies of basal-like TNBC cell lines as potential EpCAM-AsiC targets. Only 21 of the 154 BPLER dependency genes reduced viability by at least 2-fold in 3 of 4 basal-A cell lines tested. These putative BDGs clustered in 4 functional groups—4 proteasome genes and MCL1 (previously validated), 10 genes implicated in RNA splicing, 2 genes implicated in mitosis and 2 genes required for nuclear export.
  • TNBCs are known to be particularly susceptible to antimitotic agents.
  • USP39 is overexpressed in breast cancer cells vs normal breast tissue and USP39 knockdown inhibited proliferation and colony formation of luminal MCF7 cells.
  • USP39 mutation leads to splicing defects of tumor suppressor genes like rb1 and p21.
  • PRPF8 4 spliceosome tri-snRNP complex BDGs (PRPF8, PRPF38A, RBM22, USP39) in 6 basallike cell lines and in luminal MCF7 cells ( FIG. 19 ).
  • EpCAM-AsiCs can cause targeted gene knockdown in EpCAM+ tumors and the T-ICs within them. Although there may be some uptake in normal epithelial cells that weakly express EpCAM, gene knockdown will be concentrated in EpCAMbright tumor cells, especially in T-ICs.
  • EpCAM-AsiCs can be optimized, as described herein, for favorable PK/PD to suppress tumor growth and metastasis of basal-like TNBCs with acceptable toxicity in mouse models.
  • EpCAM-AsiCs targeting eGFP, AKT1 and PLK1 are used herein as models for assessing gene knockdown and optimizing AsiC design.
  • Cell lines stably expressing destabilized (d1)EGFP, with a protein T1/2 of ⁇ 1 hr, can be generated using lentiviruses. GFP expression can be readily quantified by flow and imaging, and its knockdown has no biological consequences. The short T1/2 allows for rapid and sensitive detection of knockdown.
  • AKT1 which is expressed in all the cells we test, is a good endogenous gene to study, since its knockdown in TNBCs doesn't affect cell viability much.
  • PLK1 is used as proof-of-concept for its antitumor effect because its knockdown is cytotoxic to all dividing cells.
  • EpCAM-AsiCs can be purchased, e.g., as non-GMP RNAs from TriLink or NITTO Avecia. Each strand of the EpCAM-AsiC was synthesized with 2′-fluoropyrimidines and dT residues at their 3′-ends to protect against exonuclease digestion and then annealed to generate the final RNA ( FIG. 10B ). As we optimize the AsiC, other chemical modifications can be substituted and tested to determine if they confer improved activity. The aptamer alone and AsiCs bearing a nontargeting siRNA can serve as controls.
  • EpCAM-AsiCs can also be annealed to an antisense strand modified at the 3′-end with a fluorophore (which doesn't affect AsiC activity (not shown)) to quantify AsiC uptake and trafficking within cells and in vivo.
  • a fluorophore which doesn't affect AsiC activity (not shown)
  • EpCAM-AsiC knockdown in epithelial breast cancers and breast cancer T-ICs vs normal epithelial cells. It can be determined which breast cancer subtypes are transfected with EpCAM-AsiCs and evaluate whether tumor knockdown is specific to cancer cells, first in cell lines and then in 10 tumor tissues to verify that the results for cell lines translate to tissues in situ. Because EpCAM-AsiCs might also transfect normal tissue stem cells, knockdown and toxicity to these rare basal cells will be assessed in the tissue experiments. We can also evaluate the potential of EpCAM-AsiCs to transfect and target breast T-ICs.
  • EpCAM-AsiC-mediated transfection can be compared to lipid transfection and naked siRNAs as controls.
  • EpCAM-AsiCs Do epithelial breast cancer cells preferentially take up EpCAM-AsiCs and show knockdown relative to normal epithelial cells in tissue explants?
  • epithelial breast cancers can undergo efficient gene knockdown.
  • Tissues cut into 3 ⁇ 3 ⁇ 3 mm3 samples can be transfected in Optimem solution in microtiter wells.
  • Lipoplexed siRNA and chol-siRNAs both knockdown genes in normal columnar and squamous genital tract epithelia, while naked siRNAs are not taken up.
  • siRNAs to target epithelial genes, which we have previously knocked down (such as E-cadherin, claudin3, cytokeratin (CK)-5 (a good marker of basal cells), and nectin-1), whose expression can be readily followed by IHC, fluorescence microscopy (FM) or flow cytometry of separated cells. Staining of the target gene product can be correlated with staining for phenotypic markers and fluorescent siRNAs to determine which cell types within the tissue are targeted. Pan-CK antibody can be used to distinguish epithelial cells (normal and tumor) from stroma. We can also compare knockdown of collagenase-digested 10 cells to tissue knockdown.
  • T-ICs transfect T-ICs
  • cancer stem cells T-ICs
  • T-ICs are drug resistant and thought responsible for tumor initiation, relapse and metastasis.
  • Breast T-ICs are not uniquely defined by phenotype, making experiments challenging, since T-ICs are defined functionally by their ability to initiate tumors that can be serially transplanted. Staining for CD44, CD24, EpCAM, CD133, CD49f or ALDH1 in different combinations enriches for T-ICs. 49,61,67,107-111
  • T-ICs are heterogeneous and show plasticity in their epithelial vs mesenchymal features (and in fact may have some features of both states). 28,95,112-118 Some breast T-ICs are mesenchymal and don't express EpCAM. However, there is increasing evidence that the ability of basal-like TNBCs to colonize distant tissues and form macroscopic metastases—arguably the most clinically important function of T-ICs—depends on epithelial properties. Moreover our new data ( FIGS.
  • EpCAM-AsiCs have anti-T-IC activity for basal-A TNBCs.
  • EpCAM-AsiCs are taken up by basal-like TNBC T-ICs and can be used for targeted therapy to cripple T-IC capability within them.
  • EpCAM-AsiC uptake and gene silencing in T-ICs we can first stain a panel of breast cancer lines with EpCAM, CD44 and CD24 to identify breast cell lines whose putative T-IC populations contain cells that stain brightly for EpCAM. We can also examine EpCAM staining of mammospheres and Aldefluor+ cells111,123,124 generated from these cell lines. We can select ⁇ 4-5 lines with the most uniform EpCAM expression within T-ICs as the most attractive cell lines to study in this subaim (and as controls, 1-2 basal-B cell lines whose T-ICs might lack EpCAM staining) and can produce stable eGFP-expressing variants.
  • Knocking down PLK1, MSI1, BMI1 or PSMA2 can reduce T-IC numbers, proliferation and function in some breast cancer subtypes, but different genes may be more active for different breast cell lines (i.e. proteasome inhibition eliminated T-ICs in basal-like TNBCs, but not non-TNBC tumors and in only 1 of 3 basal-B TNBCs95).
  • the knockdown approaches that suppress T-IC can be further investigated by experiments using available chemical inhibitors and/or by knocking down other genes in the same pathway (such as NOTCH1, ⁇ -catenin or WNT1 for MSI1).
  • the effect on T-ICs of EpCAM-AsiCs can be compared with the EpCAM aptamer on its own and the EpCAM antibody, adecatumumab (Amgen).
  • EpCAM-AsiCs follow the following steps: (1) cell receptor binding, (2) endocytosis, (3) endosomal release, (4) Dicer processing, (5) incorporation into the RNA-induced silencing complex (RISC), and (6) target mRNA cleavage.
  • RISC RNA-induced silencing complex
  • the AsiC design variables are the EpCAM aptamer, whose affinity affects steps 1 and 2; the linker sequence between the aptamer and the siRNA, which controls step 4; the siRNA sequence, which controls step 6.
  • each residue used for chemical synthesis from phosphoramidite building blocks can be chemically modified to reduce nuclease digestion, off-target suppression of partially complementary sequences, binding and stimulation of innate immune RNA sensors and improve cell uptake and in vivo PK.
  • 2′-OMe occurs naturally in rRNA and tRNA and is therefore safe, and 2′-F is also well tolerated; heavily Psmodified nucleotides are sticky (and cause binding to serum proteins, which can improve circulating T1/2) and can cause unwanted side effects; lightly modified PS-RNAs are not toxic.
  • Chemical modifications can both inhibit and enhance gene silencing in steps 5 and 6 This can be an iterative process; as modifications are made at one step, the most attractive modified candidates can be optimized for other steps, drawing on lessons learned from previous candidates. We can verify that the modified AsiCs chosen for further development do not stimulate innate immunity or result in cellular toxicity. If they do, we can further modify our designs to avoid these problems.
  • EpCAM binding The EpCAM aptamer has 12 nM affinity, It can be verified that that this affinity is preserved in the EpCAM-AsiC. If the AsiC has lower affinity than the aptamer, we can use bio-layer interferometry (OctetRED System, ICCB-Longwood Core) with recombinant EpCAM to compare the affinity of the aptamer and AsiC. If the AsiC has lower binding affinity, it may not fold properly. To enhance folding into the desired conformation we can try changing the type and length of the linker between the aptamer and the AsiC sense strand (i.e. we can incorporate more 3C linkers or triethylene or hexaethylene glycol spacers).
  • Aptamers can be multimerized by using streptavidin (SA) to bind biotinylated (Bi) aptamers and siRNAs; extending the aptamer with an adapter that binds to an organizing oligonucleotide that contains multiple complementary sequences connected by a flexible linker; or extending the aptamer with complementary adapter sequences to produce a dimer.
  • SA streptavidin
  • Bi biotinylated aptamers and siRNAs
  • multimerization could cause unwanted EpCAM signaling and promote tumor cell proliferation. We can verify that this is not the case using multimerized constructs targeting eGFP.
  • An attractive feature of multimerization is that it could link multiple different siRNAs into a single RNA molecule for combinatorial gene knockdown to produce a cancer “cocktail”.
  • Endosomal release Although fewer than 1000 cytosolic siRNA molecules are estimated to be needed for knockdown (not shown), only a few percent of siRNAs in endocytosed liposomes are released into thecytosol. EpCAM-AsiC endosomal release can be assessed by live cell imaging to measure the efficiency of cytosolic release of endocytosed AsiCs. If this indicates less than desired endosomal release, then improving release should reduce the drug dose substantially. Preincubation and endocytosis of an amphipathic cationic peptide (mellitin) or polymer (butyl vinyl ether) that is reversibly masked, can enhance siRNA escape to the cytosol.
  • mellitin amphipathic cationic peptide
  • polymer butyl vinyl ether
  • Masking means that at neutral pH the peptide or polymer is uncharged and does not interact with the plasma membrane and damage it, but at the negative endosomal pH, a cationic molecule is generated that damages the endosomal membrane and releases coendocytosed oligonucleotides. Iv injection of these masked polymers within 2 hr of siRNA delivery potentiated hepatocyte knockdown by chol-siRNAs as much as 500 fold in mice and nonhuman primates.
  • EpCAM-AsiC and lipoplexed siRNA
  • cytosolic delivery and eGFP knockdown we can also investigate whether incubating EpCAM-AsiCs with basic peptides/polymers can also determine whether inhibition of endosomal acidification using bafilomycin A or concanamycin alters EpCAM-AsiC cytoplasmic release and knockdown, as the proton sponge theory predicts. If these experiments confirm the proton sponge theory, we can investigate strategies for altering EpCAM-AsiCs.
  • EpCAM-AsiCs Dicer processing, RISC incorporation, target mRNA cleavage
  • RISC incorporation target mRNA cleavage
  • Northern blots probed for the sense, antisense and aptamer parts of the EpCAM-AsiC, can analyze EpCAM-AsiC products within cells. Their migration can be compared to that of synthesized sense and antisense strands, aptamer and full length EpCAM-AsiC.
  • RNAs that migrate like the sense and antisense strands (as well as unprocessed EpCAM-AsiCs from endosomes and a band the size of the aptamer joined to its linker). (Dicer dependence can be verified using HCT116 cells expressing hypomorphic Dicer). If the intracellular RNAs are not the expected size, we can clone them to determine where Dicer cuts. If the bands are not cut or are not where we want, we can redesign the linker and double stranded region to produce the desired cleavage.
  • EpCAM-AsiCs targeting additional genes that we evaluate in vivo can be designed with the most active siRNA sequences and best chemical modifications.
  • a small group of siRNA sequences to test for knockdown (without aptamers, by transfection) can be identified by web algorithms.
  • the most efficient siRNAs (pM activity), which also have low predicted melting temperatures (Tm), can be used, since these are processed better. If we need to use sequences with higher Tms, we can add a mismatch at the 3′-end of the sense strand to promote siRNA unwinding and incorporation of the active strand in the RISC.
  • mice bearing mammary fatpad xenografts of Luciferase-mCherry stable transfectants we have generated of EpCAM+ basal-A TNBC lines, such as MB468 or HCC1187, compared to an EpCAM ⁇ mesenchymal basal-B TNBC cell line, such as MB231.
  • EpCAM+ basal-A TNBC lines such as MB468 or HCC1187
  • EpCAM ⁇ mesenchymal basal-B TNBC cell line such as MB231.
  • mice/gp will be used to obtain statistical significance based on our prelim. data in these models.
  • Samples can be analyzed over 5d with frequent sample collection the first day. At each timepoint, blood and urine can be harvested and analyzed by Taqman assay for the antisense strand. Tumor and sample organs can be harvested at fewer timepoints from euthanized animals. Blood can be analyzed for hematological, liver and kidney toxicity by blood counts and serum chemistries. The circulating T1/2 and proportion of the injected drug that localizes to the EpCAM+ tumor can be calculated.
  • the sc and iv PK results will be compared with mCherry knockdown following a single EpCAM-AsiC injection in a range of concentrations, assessed both by in vivo imaging (using the IVIS Spectrum) and by flow cytometry, FM, and qRT-PCR of tumor specimens harvested 4, 7 and 12 d post-treatment.
  • In vivo PK/PD/toxicity evaluation can be performed as above, using the unconjugated AsiC as a positive control (and benchmark) and the conjugated siRNA (without the aptamer) as a negative control.
  • Two or three of the constructs that have the lowest ED75 or ED90 and longest T-KD50 for GFP will be retested using a PLK1 EpCAM-AsiC to determine the corresponding PK/PD parameters, to aid in designing the dosing regimen for antitumor efficacy experiments.
  • We can also determine the maximally tolerated dose (MTD) for these PLK1 constructs.
  • EpCAM-AsiCs Antitumor Effect of EpCAM AsiCs against basal-like TNBCs
  • Our final goal is to test the EpCAM-AsiCs against orthotopic mammary fat pad tumors and metastases.
  • Live animal imaging can be performed using an IVIS Spectrum, sensitive for multicolor fluorescence and bioluminescence.
  • PLK1 EpCAM-AsiCs against orthotopic xenografts We can begin by targeting PLK1/A few PLK1 EpCAM-AsiC designs, optimized as described above, can be injected sc and/or iv in groups of 5-8 mice (size chosen from power calculations based on previous experiments in which this group size gave statistically significant results) using doses and dosing schedules/injection route chosen based on the PK/PD results above. For example if the ED90 is well below the MTD, an initial experiment might investigate administering 2ED90 every T-KD50/2 d. Mice can initially be treated as soon as their tumors become palpable, but in later experiments we can investigate whether larger tumors of fixed diameters regress after multiple administrations.
  • mice bearing representative EpCAM+ basal-A (MB468, HC1187, BPLER) and EpCAM ⁇ basal-B (MB231) tumors will be compared.
  • mice bearing these tumors in each flank but these may require more mice because of intra-animal variations in tumor sizes.
  • Control mice can be treated with PBS or naked siRNAs, the EpCAM aptamer on its own, EpCAM-AsiCs bearing scrambled siRNA sequences and PLK1 PSMA-AsiCs.
  • EpCAM-AsiC treatment with adecatumumab or paclitaxel. Tumor size will be quantified by luminescence and caliper measurements q3d.
  • Treated mice can also be weighed and observed for clinical signs of toxicity and at time of sacrifice can be carefully examined for gut and bone marrow toxicity by blood counts and pathological examination of gut, bone marrow and spleen. Differences between groups can be assessed by one way ANOVA with corrections for multiple comparisons as needed. For AsiCs that are effective, we can also examine the immediate effect of treatment to evaluate the mechanism of antitumor activity and verify that the AsiCs are not activating innate immune responses.
  • Tumor-bearing mice can be sacrificed 1-3 d after a single therapeutic or control injection and the tumors stained for activated caspases to determine if death is by apoptosis and by H&E to look for mitotic spindles to follow the expected effect of PLK1 knockdown.
  • Serum interferons and pro-inflammatory cytokines can be assessed by multiplexed ELISA, and spleen and tumor cells analyzed by qRT-PCR for the corresponding mRNAs. If there is no antitumor effect or the antitumor effect is suboptimal, the dosing regimen can be adjusted to the MTD. If the antitumor effect is complete (complete tumor regression), then we can evaluate decreased doses and/or larger tumors at start of therapy.
  • mice When control mice are sacrificed because untreated tumors have reached the allowed size, the treated mice can be sacrificed and mammary fatpads inspected for residual microscopic or macroscopic tumor by FM, H&E and IHC. Residual tumor cells can also be assessed for EpCAM expression to determine whether tumor resistance, if it occurs, may have developed as a consequence of down-regulating EpCAM. If no residual tumor cells are noted, we can perform an additional experiment to determine whether tumors are eradicated—mice will be treated for 1-2 weeks after the luciferase measurement has returned to background levels, and then mice can be observed for 1-2 months off treatment to see if tumors regrow or metastases appear. The most effective regimen(s) for basal-A TNBCs can also be evaluated against other breast cancer subtypes (luminal, Her2+) that we expect EpCAM to target.
  • breast cancer subtypes luminal, Her2+
  • PLK1 EpCAM-AsiC activity against metastatic tumors To evaluate the effectiveness of EpCAM-AsiCs against metastatic cancer cells, we can evaluate the PLK1 EpCAM-AsiCs against basal-A TNBC cell lines injected intravenously in NSG mice, which have the best tumor take. We can begin to treat mice as soon as lungs become luciferase+ after tail vein injection of basal-A (or basal-B as control) TNBCs. The treatment dosing can use the effective schedule and mode of administration determined above for primary tumors. Mice can be imaged q3d. The controls can be reduced to a mock-treated group and groups treated with paclitaxel or an EpCAM-AsiC containing a non-targeting siRNA.
  • mice When the control mice need to be sacrificed, all groups can be imaged. Lungs, livers and brains can be dissected, weighed, imaged to quantify tumor burden, sections can be analyzed by H&E and staining for EpCAM, and one lung from each animal will be analyzed by qRT-PCR for relative expression of human/mouse Gapdh to quantify tumor burden independently. If mice treated with PLK1 EpCAM-AsiCs are completely protected from metastases or show a significant advantage compared to control groups, we can determine if mice with greater metastatic burdens are also protected by delaying the beginning of treatment until the tumor burden is greater.
  • proteasome inhibitor sensitivity correlates strongly with MCL1 dependency in vitro (not shown)
  • proteasome gene and MCL1 knockdown will be synergistic.
  • the synergy of different AsiC and AsiC/drug combinations can be formally tested by the isobologram method using different RNA dose combinations or combinations with relevant inhibitor drugs. In particular we will determine whether combining EpCAM-AsiCs with standard of care drugs, such as paclitaxel, is synergistic with the original construct.
  • Human BPE and BPLER cells were grown in WIT medium (Stemgent). MB468 were transduced with a luciferase reporter. All other human cell lines were obtained from ATCC and grown in MEM (MCF7, BT474), McCoy's 5A (SKBR3), RPMI1640 (HCC1806, HCC1143, HCC1937, HCC1954, HCC1187, MB468, T47D) or DMEM (MB231, BT549, MB436) all supplemented with 10% FBS, 1 mM L-glutamine and penicillin/streptomycin (Gibco) unless otherwise indicated. 4T1 mouse breast cancer cells, were grown in 10% FBS DMEM.
  • MB468 cells stably expressing Firefly luciferase (MB468-luc) were used and MB231 cells stably expressing Firefly luciferase and mCherry (MB231-luc-mCherry) were selected after infection with pLV-Fluc-mCherry-Puro lentivirus.
  • MB231 Cells were selected with puromycin.
  • cells were plated at low density (10,000 cells/well in 96-well plates) and treated immediately. All AsiC and siRNA treatments were performed in either OptiMEM or WIT medium. Cell viability was assessed by CellTiter-Glo (Promega) or by Trypan-Blue staining in 96-well plates.
  • 1,000 viable cells were treated for 6 h in round bottom 96-well plates and then transferred to 10-cm plates in serum-containing medium. Medium was replaced every 3 d. After 8-14 d, cells were fixed in methanol ( ⁇ 20 C) and stained with crystal violet.
  • 1,000/ml viable cells were treated for 6 h in round bottom 96-well plates and then cultured in suspension in serum-free DMEM/F12 1:1 (Invitrogen), supplemented with EGF (20 ng/ml, BD Biosciences), B27 (1:50, Invitrogen), 0.4% bovine serum albumin (Sigma) and 4 ⁇ g/ml insulin (Sigma). Spheres were counted after 1 or 2 weeks.
  • qRT-PCR analysis was performed as described (Petrocca, F., et al. (2008). E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 13, 272-286). Briefly, total RNA was extracted with Trizol (Invitrogen) and cDNA prepared from 1000 ng total RNA using Thermoscript RT kit (Invitrogen) as per the manufacturer's SYBR Green Master Mix (Applied Biosystems) and a BioRad C1000 Thermal Cycler (Biorad). Relative CT values were normalized to GAPDH and converted to a linear scale.
  • Fresh breast or colon cancer and control biopsies were received from the UMASS Tissue Bank, samples were cut into 3 ⁇ 3 ⁇ 3 mm samples and placed in a 96 well plate with 100 ul RPMI. Samples were treated with either Alexa647-siRNA-GFP, Alexa647-chol-siRNA-GFP or Cy3-AsiC-GFP for 24 hr. Samples were photographed and digested. Three samples from each treatment were pooled and put in 10 ml RPMI containing 1 mg/ml collagenase II (Sigma-Aldrich) for 30 minutes at 37° C. with shaking.
  • mice were purchased from the Jackson Laboratory.
  • MB468-luc (5 ⁇ 10 6 ) and MB231-luc-mCherry (5 ⁇ 10 5 ) cells trypsinized with Tryple Express (Invitrogen), resuspended in a 1:1 WIT-Matrigel solution and injected subcutaneously in the flank of 8-week old female Nu/J mice (Stock #002019, Jackson Laboratories). Tumors size was analyzed daily using the IVIS Spectra, after 5 days tumors were clearly visible. Mice bearing tumors of comparable size were randomized into 5 groups and treated with 5 mg/kg of EpCAM-AsiC-PLK1, EpCAM-AsiC-GFP, EpCAM-Aptamer, siRNA-PLK1 or untreated. Mice were treated every 72 h for 14 days.
  • Student's t-tests computed using Microsoft Excel, were used to analyze the significance between the treated samples and the controls where the test type was set to one-tail distribution and two-sample equal variance.
  • EpCAM-AsiC Specifically Targets Basal a Breast Cancer Cells
  • EpCAM aptamer was selected by Systematic Evolution of Ligands by Exponential Enrichment (SELEX) for binding to human EpCAM.
  • the optimized aptamer is only 19 nucleotides (nt) long and binds to human EpCAM with 12 nM affinity (Shigdar S. et. al. RNA aptamer against a cancer stem cell marker epithelial cell adhesion molecule affinity Cancer Sci. 2011 May; 102(5):991-8). It does not bind to mouse EpCAM ( FIG. 22 ). Its short length is ideal for an AsiC drug, since RNAs of ⁇ 60 nt or less in length can be cheaply and efficiently chemically synthesized.
  • EpCAM-AsiCs we designed consist of a longer strand of 42-44 nt (19 nt aptamer+3 nt linker+20-22 nt sense (inactive) strand of the siRNA), which is annealed to a 20-22 nt antisense (active) siRNA strand ( FIG. 21A ). Both strands were commercially synthesized with 2′-fluoropyrimidine substitutions, which confer enhanced stability in serum and other bodily fluids (T1/2>>3 d) and prevent stimulation of innate immune RNA sensors.
  • T1/2>>3 d 2′-fluoropyrimidine substitutions
  • BPLER basal A TNBC cell line overexpresses EpCAM, while BPE a control epithelial breast cell line do not ( FIG. 21B ).
  • Both BPLER and BPE cell were treated with the Alexa647-EpCAM-AsiC targeting GFP, only BPLER displayed uptake of the AsiC ( FIG. 21C ).
  • EpCAM-AsiC was treated with EpCAM+ MDA-MB-468 cells and BPE controls with Cy3 labeled EpCAM-Aptamer (the 19 nt aptamer was labeled with Cy3 at the 5′ end). After 22 and 43 hours we clearly saw selective AsiC uptake in EpCAM+ cells (data not shown).
  • EpCAM AsiC to selectively trigger gene knockdown we chose BPLER and BPE cell lines which stably overexpress GFP.
  • Cells were treated with either EpCAM-AsiCs targeting GFP or transfected with GFP-siRNA as a positive control ( FIG. 21D ).
  • FIG. 21D EpCAM-AsiCs knocked down gene expression equivalently in BPE and BPLER, EpCAM-AsiCs selectively knocked down expression in BPLER without any lipid; knockdown was uniform and comparable to that achieved with lipid transfection.
  • EpCAM-AsiC is selectively taken-up by EpCAM+ cell and can induce gene knockdown specifically in these EpCAM+ cells. Also we show that using different fluorophores (Alexa647 or Cy3) at different locations (5′ of aptamer or 3′ of anti-sense strand) did not impact the specific uptake.
  • human epithelial breast cancer tissue can specifically take up EpCAM-AsiC compared to healthy human tissue.
  • EpCAM AsiC Targeting PLK1 Specifically Inhibits Cell Proliferation in Basal a Breast Cancer Cells
  • EpCAM-AsiC can specifically target basal A and luminal breast cancer cells and inhibit proliferation.
  • PLK1 is a known trigger for G2/M transition.
  • the effect of EpCAM-AsiC targeting PLK1 on cell proliferation was tested on 10 breast cancer cells representative of basal A, B and luminal cell lines.
  • EpCAM-AsiC targeting PLK1 decreased cell proliferation in both basal A and luminal cell lines while having no effect on basal B cells ( FIG. 27A ).
  • FIG. 27B A correlation was seen between EpCAM expression levels and cell viability
  • EpCAM-AsiC will specifically target EpCAM+ cells in a mix cell population HCC1937 (EpCAM+GFP ⁇ ) cell were co-cultured with BPE (EpCAM-GFP+) cells and treated with EpCAM-AsiC targeting PLK1 or untreated. Untreated co-culture displayed a similar ration of cells (41% BPE and 59% HCC1937). Following EpCAM-AsiC targeting PLK1 treatment the ratio of EpCAM+ cells decreased to 17% and EpCAM ⁇ cells increased to 83% indicating that the EpCAM-AsiC specifically suppresses proliferation in EpCAM+ cells. The co-culture was repeated with other basal A cell lines (MB468 and HCC1143) similar results were obtained.
  • EpCAM-AsiC targeting PLK1 suppressed cell viability in basal A and luminal cell lines while EpCAM-aptamer didn't effect cell viability in any of the cell lines ( FIG. 28 ).
  • EpCAM-AsiC targeting PLK1 To examine whether pretreatment with EpCAM-AsiC targeting PLK1 will inhibit or delay tumor initiation in-vivo we treat MB-468-luc cell with EpCAM-AsiC targeting PLK1, GFP or untreated for 24 h and injected the cells into the flank of nude mice. Using the IVIS Spectra imaging system we followed tumor growth every 5 days for 20 days. Cells pretreated with EpCAM-AsiC targeting PLK1 did not show any sign of a tumor after 20 days while untreated cells or cells pretreated with EpCAM-AsiC targeting GFP displayed tumors after 5 days and the tumor size grew during the 20 days ( FIG. 29D ).
  • EpCAM AsiC Targeting PLK1 Specifically Inhibits Tumor Initiation and Growth in Basal a Breast Cancer Cells
  • EpCAM-AsiC can specifically target EpCAM+ cell in-vitro, to understand whether this ability is retained in-vivo we first tested the stability of EpCAM-AsiC in mouse and human serum over time. We saw that EpCAM-AsiC is stable for at least 36 h in both mouse and human serum ( FIG. 30A-30B ). We injected nude mice with both MB468-luc and MB231-luc-mCherry cells on opposite flanks. After 5 days when tumors were clearly visible using the IVIS Spectra imaging system, we injected mice s.c.
  • EpCAM-AsiC did have some effect even though it was targeting GFP since basal A tumor treated with GFP AsiC did not increase in size as much as control untreated mice.
  • Treatment with EpCAM-Asic targeting GFP suppress tumor growth in both EpCAM+ and EpCAM ⁇ tumors but didn't eliminate tumors.
  • EpCAM-AsiC Sequence NO EpCAM PLK1 GCG ACU GGU UAC CCG GUC GUU 1 sense UUG AAG AAG AUC ACC CUC CUU AdTdT EpCAM PLK1 UAA GGA GGG UGA UCU UCU UCA 2 anti-sense dTdT EpCAM AKT1 GCG ACU GGU UAC CCG GUC GUU 23 sense GCU GGA GAA CCU CAU GCU GdTdT EpCAM AKT1 CAG CAU GAG GUU CUC CAG CdTdT 24 anti-sense EpCAM GFP GCG ACU GGU UAC CCG GUC GUU 25 sense UGG CUA CGU CCA GGA GCG CAdTdT EpCAM GFP UGC GCU CCU GGA CGU AGC CdTdT 26 anti-sense siGFP sense UGG CUA CGU CCA GGA GCG 27 siGFP antisense UGC GCU CCU GGA
  • EpCAM mean fluorescence intensity (MFI) of human breast cell lines Cell line Subtype EpCAM MFI BPE immortalized normal epithelium 2 BPLER basal-A TNBC 109 HMLER unclassified TNBC (myoepithelial) 72 HCC1143 basal-A TNBC 1068 HCC1937 basal-A TNBC 806 HCC1187 basal-A TNBC 289 HCC1806 basal-A TNBC 558 HCC70 basal-A TNBC 443 MB468 basal-A TNBC 340 MCF7 luminal 583 T47D luminal 799 BT549 basal-B TNBC 2 MB231 basal-B TNBC 31 MB436 basal-B TNBC 4 Human fibroblast Normal tissue 14
  • TNBCs Triple negative breast cancers have the worst prognosis of any breast cancer subtype and there is no targeted TNBC therapy.
  • TNBCs have the phenotype associated with tumor initiating cells (T-IC), also known as cancer stem cells. T-IC are resistant to chemotherapy and thought to be responsible for tumor relapse and metastasis.
  • EpCAM is expressed at gap junctions at low levels on normal epithelial cells, but much more highly expressed (100-1000-fold greater) throughout the membrane of virtually all epithelial cancers and is a known TI-C marker.
  • the aptamer-siRNA chimera (AsiC) platform is adapted to transfect epithelial breast cancer cells while also targeting breast tumor-initiating cells (T-IC).
  • the aptamer binds to EpCAM, highly expressed on cancer cells and cancer stem cells.
  • the siRNA is directed at a kinase required for mitosis in all cells (PLK1).
  • EpCAM-AsiC's are stable in human and mouse.
  • the EpCAM AsiCs can be chemically synthesized with 2′-F pyrimidines and dTdT at the 3′-ends, which makes them resistant to RNases and unlikely to stimulate innate immunity.
  • MB468 tumors regress only after treatment with PLK1 EpCAM-AsiC. Mice with sc MB468 tumors were treated with 5 mg/kg RNA 2 ⁇ /wk beginning when tumors became palpable. PLK1 EpCAM-AsiC, GFP SpCAM-AsiC, EpCAM aptamer, PLK1 siRNA, and mock treated samples were analyzed ( FIG. 33 )
US15/506,010 2014-08-29 2015-08-28 Methods and compositions for the treatment of cancer Active US10385343B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/506,010 US10385343B2 (en) 2014-08-29 2015-08-28 Methods and compositions for the treatment of cancer

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462043803P 2014-08-29 2014-08-29
US15/506,010 US10385343B2 (en) 2014-08-29 2015-08-28 Methods and compositions for the treatment of cancer
PCT/US2015/047449 WO2016033472A1 (fr) 2014-08-29 2015-08-28 Méthodes et compositions pour le traitement du cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/047449 A-371-Of-International WO2016033472A1 (fr) 2014-08-29 2015-08-28 Méthodes et compositions pour le traitement du cancer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/409,081 Division US11180762B2 (en) 2014-08-29 2019-05-10 Methods and compositions for the treatment of cancer

Publications (2)

Publication Number Publication Date
US20170275629A1 US20170275629A1 (en) 2017-09-28
US10385343B2 true US10385343B2 (en) 2019-08-20

Family

ID=55400670

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/506,010 Active US10385343B2 (en) 2014-08-29 2015-08-28 Methods and compositions for the treatment of cancer
US16/409,081 Active 2036-05-22 US11180762B2 (en) 2014-08-29 2019-05-10 Methods and compositions for the treatment of cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/409,081 Active 2036-05-22 US11180762B2 (en) 2014-08-29 2019-05-10 Methods and compositions for the treatment of cancer

Country Status (8)

Country Link
US (2) US10385343B2 (fr)
EP (2) EP3185910A4 (fr)
JP (1) JP2017526367A (fr)
KR (1) KR20170070022A (fr)
CN (1) CN107106704A (fr)
AU (1) AU2015308721B2 (fr)
CA (1) CA2959386A1 (fr)
WO (1) WO2016033472A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3625359A4 (fr) * 2017-05-18 2021-03-03 Children's National Medical Center Compositions comprenant des aptamères et des charges utiles d'acide nucléique et procédés pour leur utilisation
CN108559748B (zh) * 2018-04-24 2021-12-28 中山大学孙逸仙纪念医院 一种cd4阳性细胞特异的dna核酸适体及其嵌合体
US20220251561A1 (en) * 2019-06-03 2022-08-11 Fundació Institut D'investigació Biomèdica De Bellvitge (Idibell) Eph2a aptamer and uses thereof
JP2022536984A (ja) * 2019-06-21 2022-08-22 ザ チルドレンズ メディカル センター コーポレーション がんの処置のための方法および組成物
WO2023014196A1 (fr) * 2021-08-06 2023-02-09 인터올리고 주식회사 Composition pharmaceutique pour la prévention ou le traitement du cancer du sein triple négatif, comprenant un oligonucléotide en tant que principe actif

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050147593A1 (en) * 2003-05-22 2005-07-07 Medimmune, Inc. EphA2, EphA4 and LMW-PTP and methods of treatment of hyperproliferative cell disorders
WO2010017319A2 (fr) 2008-08-05 2010-02-11 Mdrna, Inc. Composés d’acide nucléique pour inhiber l’expression du gène plk1 et utilisations de ceux-ci
WO2010019446A1 (fr) 2008-08-09 2010-02-18 University Of Iowa Research Foundation Aptamères d'acide nucléique
CN101766817A (zh) 2008-12-26 2010-07-07 中国科学技术大学 一种药物载体及其应用
WO2011130458A2 (fr) 2010-04-13 2011-10-20 John Rossi Aptamères d'arn contre baff-r en tant qu'agents d'administration spécifiques d'un type cellulaire et leurs méthodes d'utilisation
WO2011142970A2 (fr) 2010-05-14 2011-11-17 University Of Iowa Research Foundation Aptamères d'acide nucléique her2
WO2012078637A2 (fr) * 2010-12-06 2012-06-14 Immune Disease Institute, Inc. Composition et méthode pour l'administration d'oligonucléotides
WO2013025930A1 (fr) 2011-08-16 2013-02-21 Research Foundation of State University of New York at Albany Modulateurs aptamères des récepteurs des oestrogènes
WO2014019025A1 (fr) 2012-08-02 2014-02-06 Deakin University Aptamères epcam pour la détection de cellules souches cancéreuses
WO2014068408A2 (fr) 2012-10-23 2014-05-08 Caris Life Sciences Switzerland Holdings, S.A.R.L. Aptamères et leurs utilisations
WO2014093698A1 (fr) 2012-12-12 2014-06-19 The Methodist Hospital Research Institute Analyses de détection de cellules tumorales en une étape, spécifiques à une cellule et à base de multi-aptamères
CN103977433A (zh) 2014-05-14 2014-08-13 上海中医药大学附属岳阳中西医结合医院 一种前列腺癌超声诊断靶向试剂及制备方法
WO2014126160A1 (fr) 2013-02-13 2014-08-21 国立大学法人東京大学 Procédé d'examen du cancer et kit d'examen
WO2016127216A1 (fr) 2015-02-11 2016-08-18 Deakin University Aptamères d'epcam et conjugués desdits aptamères

Family Cites Families (191)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
JPS5927900A (ja) 1982-08-09 1984-02-14 Wakunaga Seiyaku Kk 固定化オリゴヌクレオチド
FR2540122B1 (fr) 1983-01-27 1985-11-29 Centre Nat Rech Scient Nouveaux composes comportant une sequence d'oligonucleotide liee a un agent d'intercalation, leur procede de synthese et leur application
US4605735A (en) 1983-02-14 1986-08-12 Wakunaga Seiyaku Kabushiki Kaisha Oligonucleotide derivatives
US4948882A (en) 1983-02-22 1990-08-14 Syngene, Inc. Single-stranded labelled oligonucleotides, reactive monomers and methods of synthesis
US4824941A (en) 1983-03-10 1989-04-25 Julian Gordon Specific antibody to the native form of 2'5'-oligonucleotides, the method of preparation and the use as reagents in immunoassays or for binding 2'5'-oligonucleotides in biological systems
US4587044A (en) 1983-09-01 1986-05-06 The Johns Hopkins University Linkage of proteins to nucleic acids
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5118802A (en) 1983-12-20 1992-06-02 California Institute Of Technology DNA-reporter conjugates linked via the 2' or 5'-primary amino group of the 5'-terminal nucleoside
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
FR2567892B1 (fr) 1984-07-19 1989-02-17 Centre Nat Rech Scient Nouveaux oligonucleotides, leur procede de preparation et leurs applications comme mediateurs dans le developpement des effets des interferons
US5430136A (en) 1984-10-16 1995-07-04 Chiron Corporation Oligonucleotides having selectably cleavable and/or abasic sites
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
US5258506A (en) 1984-10-16 1993-11-02 Chiron Corporation Photolabile reagents for incorporation into oligonucleotide chains
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
US4828979A (en) 1984-11-08 1989-05-09 Life Technologies, Inc. Nucleotide analogs for nucleic acid labeling and detection
FR2575751B1 (fr) 1985-01-08 1987-04-03 Pasteur Institut Nouveaux nucleosides de derives de l'adenosine, leur preparation et leurs applications biologiques
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US4762779A (en) 1985-06-13 1988-08-09 Amgen Inc. Compositions and methods for functionalizing nucleic acids
US5317098A (en) 1986-03-17 1994-05-31 Hiroaki Shizuya Non-radioisotope tagging of fragments
JPS638396A (ja) 1986-06-30 1988-01-14 Wakunaga Pharmaceut Co Ltd ポリ標識化オリゴヌクレオチド誘導体
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
DE3851889T2 (de) 1987-06-24 1995-04-13 Florey Howard Inst Nukleosid-derivate.
US5585481A (en) 1987-09-21 1996-12-17 Gen-Probe Incorporated Linking reagents for nucleotide probes
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US5525465A (en) 1987-10-28 1996-06-11 Howard Florey Institute Of Experimental Physiology And Medicine Oligonucleotide-polyamide conjugates and methods of production and applications of the same
DE3738460A1 (de) 1987-11-12 1989-05-24 Max Planck Gesellschaft Modifizierte oligonukleotide
US5082830A (en) 1988-02-26 1992-01-21 Enzo Biochem, Inc. End labeled nucleotide probe
EP0406309A4 (en) 1988-03-25 1992-08-19 The University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5109124A (en) 1988-06-01 1992-04-28 Biogen, Inc. Nucleic acid probe linked to a label having a terminal cysteine
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5262536A (en) 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5457183A (en) 1989-03-06 1995-10-10 Board Of Regents, The University Of Texas System Hydroxylated texaphyrins
US5599923A (en) 1989-03-06 1997-02-04 Board Of Regents, University Of Tx Texaphyrin metal complexes having improved functionalization
IT1229203B (it) 1989-03-22 1991-07-25 Bioresearch Spa Impiego di acido 5 metiltetraidrofolico, di acido 5 formiltetraidrofolico e dei loro sali farmaceuticamente accettabili per la preparazione di composizioni farmaceutiche in forma a rilascio controllato attive nella terapia dei disturbi mentali organici e composizioni farmaceutiche relative.
US5391723A (en) 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5451463A (en) 1989-08-28 1995-09-19 Clontech Laboratories, Inc. Non-nucleoside 1,3-diol reagents for labeling synthetic oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5254469A (en) 1989-09-12 1993-10-19 Eastman Kodak Company Oligonucleotide-enzyme conjugate that can be used as a probe in hybridization assays and polymerase chain reaction procedures
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
EP0942000B1 (fr) 1989-10-24 2004-06-23 Isis Pharmaceuticals, Inc. Oligonucléotides modifiés en position 2'
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5292873A (en) 1989-11-29 1994-03-08 The Research Foundation Of State University Of New York Nucleic acids labeled with naphthoquinone probe
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
CA2029273A1 (fr) 1989-12-04 1991-06-05 Christine L. Brakel Compose a base de nucleotide modifie
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US7037646B1 (en) 1990-01-11 2006-05-02 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US6783931B1 (en) 1990-01-11 2004-08-31 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5852188A (en) 1990-01-11 1998-12-22 Isis Pharmaceuticals, Inc. Oligonucleotides having chiral phosphorus linkages
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
WO1991013080A1 (fr) 1990-02-20 1991-09-05 Gilead Sciences, Inc. Pseudonucleosides, pseudonucleotides et leurs polymeres
US5214136A (en) 1990-02-20 1993-05-25 Gilead Sciences, Inc. Anthraquinone-derivatives oligonucleotides
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
DK0455905T3 (da) 1990-05-11 1998-12-07 Microprobe Corp Dipsticks til nukleinsyrehybridiseringsassays og fremgangsmåde til kovalent immobilisering af oligonukleotider
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5475096A (en) 1990-06-11 1995-12-12 University Research Corporation Nucleic acid ligands
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5688941A (en) 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
ATE154246T1 (de) 1990-07-27 1997-06-15 Isis Pharmaceuticals Inc Nuklease resistente, pyrimidin modifizierte oligonukleotide, die die gen-expression detektieren und modulieren
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
WO1992002534A2 (fr) 1990-08-03 1992-02-20 Sterling Drug, Inc. Composes et procedes d'inhibition de l'expression de genes
US5245022A (en) 1990-08-03 1993-09-14 Sterling Drug, Inc. Exonuclease resistant terminally substituted oligonucleotides
US5512667A (en) 1990-08-28 1996-04-30 Reed; Michael W. Trifunctional intermediates for preparing 3'-tailed oligonucleotides
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
WO1992005186A1 (fr) 1990-09-20 1992-04-02 Gilead Sciences Liaisons internucleosides modifiees
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
KR930702373A (ko) 1990-11-08 1993-09-08 안토니 제이. 페이네 합성 올리고누클레오티드에 대한 다중 리포터(Reporter)그룹의 첨합
GB9100304D0 (en) 1991-01-08 1991-02-20 Ici Plc Compound
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5371241A (en) 1991-07-19 1994-12-06 Pharmacia P-L Biochemicals Inc. Fluorescein labelled phosphoramidites
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
EP0538194B1 (fr) 1991-10-17 1997-06-04 Novartis AG Nucléosides et oligonucléosides bicycliques, leur procédé de préparation et leurs intermédiaires
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US6235887B1 (en) 1991-11-26 2001-05-22 Isis Pharmaceuticals, Inc. Enhanced triple-helix and double-helix formation directed by oligonucleotides containing modified pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
EP1695979B1 (fr) 1991-12-24 2011-07-06 Isis Pharmaceuticals, Inc. Oligonucléotides modifiés interrompus par des séquences ADN
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US6277603B1 (en) 1991-12-24 2001-08-21 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
US5565552A (en) 1992-01-21 1996-10-15 Pharmacyclics, Inc. Method of expanded porphyrin-oligonucleotide conjugate synthesis
US5595726A (en) 1992-01-21 1997-01-21 Pharmacyclics, Inc. Chromophore probe for detection of nucleic acid
FR2687679B1 (fr) 1992-02-05 1994-10-28 Centre Nat Rech Scient Oligothionucleotides.
DE4203923A1 (de) 1992-02-11 1993-08-12 Henkel Kgaa Verfahren zur herstellung von polycarboxylaten auf polysaccharid-basis
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (fr) 1992-07-01 1994-01-05 Ciba-Geigy Ag Nucléosides carbocycliques contenant des noyaux bicycliques, oligonucléotides en dérivant, procédé pour leur préparation, leur application et des intermédiaires
US5272250A (en) 1992-07-10 1993-12-21 Spielvogel Bernard F Boronated phosphoramidate compounds
US6346614B1 (en) 1992-07-23 2002-02-12 Hybridon, Inc. Hybrid oligonucleotide phosphorothioates
US5574142A (en) 1992-12-15 1996-11-12 Microprobe Corporation Peptide linkers for improved oligonucleotide delivery
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
EP0691968B1 (fr) 1993-03-30 1997-07-16 Sanofi Analogues de nucleosides acycliques et sequences d'oligonucleotides contenant ceux-ci
EP0691977B1 (fr) 1993-03-31 1997-11-26 Sanofi Oligonucleotides a liaisons d'amides remplacant les liaisons de phosphodiesters
DE4311944A1 (de) 1993-04-10 1994-10-13 Degussa Umhüllte Natriumpercarbonatpartikel, Verfahren zu deren Herstellung und sie enthaltende Wasch-, Reinigungs- und Bleichmittelzusammensetzungen
US5955591A (en) 1993-05-12 1999-09-21 Imbach; Jean-Louis Phosphotriester oligonucleotides, amidites and method of preparation
US6015886A (en) 1993-05-24 2000-01-18 Chemgenes Corporation Oligonucleotide phosphate esters
US6294664B1 (en) 1993-07-29 2001-09-25 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
EP0729474A4 (fr) 1993-11-16 1998-10-21 Genta Inc Oligomeres synthetiques ayant des liaisons internucleosidyle phosphonate chiralement pures melangees avec des liaisons internucleosidyle non phosphonate
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5599922A (en) 1994-03-18 1997-02-04 Lynx Therapeutics, Inc. Oligonucleotide N3'-P5' phosphoramidates: hybridization and nuclease resistance properties
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
IT1270594B (it) 1994-07-07 1997-05-07 Recordati Chem Pharm Composizione farmaceutica a rilascio controllato di moguisteina in sospensione liquida
US5597696A (en) 1994-07-18 1997-01-28 Becton Dickinson And Company Covalent cyanine dye oligonucleotide conjugates
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US6608035B1 (en) 1994-10-25 2003-08-19 Hybridon, Inc. Method of down-regulating gene expression
US6222025B1 (en) 1995-03-06 2001-04-24 Isis Pharmaceuticals, Inc. Process for the synthesis of 2′-O-substituted pyrimidines and oligomeric compounds therefrom
US6166197A (en) 1995-03-06 2000-12-26 Isis Pharmaceuticals, Inc. Oligomeric compounds having pyrimidine nucleotide (S) with 2'and 5 substitutions
US6160109A (en) 1995-10-20 2000-12-12 Isis Pharmaceuticals, Inc. Preparation of phosphorothioate and boranophosphate oligomers
US6444423B1 (en) 1996-06-07 2002-09-03 Molecular Dynamics, Inc. Nucleosides comprising polydentate ligands
US6576752B1 (en) 1997-02-14 2003-06-10 Isis Pharmaceuticals, Inc. Aminooxy functionalized oligomers
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US6639062B2 (en) 1997-02-14 2003-10-28 Isis Pharmaceuticals, Inc. Aminooxy-modified nucleosidic compounds and oligomeric compounds prepared therefrom
JP3756313B2 (ja) 1997-03-07 2006-03-15 武 今西 新規ビシクロヌクレオシド及びオリゴヌクレオチド類縁体
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US6528640B1 (en) 1997-11-05 2003-03-04 Ribozyme Pharmaceuticals, Incorporated Synthetic ribonucleic acids with RNAse activity
US6617438B1 (en) 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US6320017B1 (en) 1997-12-23 2001-11-20 Inex Pharmaceuticals Corp. Polyamide oligomers
US7273933B1 (en) 1998-02-26 2007-09-25 Isis Pharmaceuticals, Inc. Methods for synthesis of oligonucleotides
US6365185B1 (en) 1998-03-26 2002-04-02 University Of Cincinnati Self-destructing, controlled release peroral drug delivery system
US7045610B2 (en) 1998-04-03 2006-05-16 Epoch Biosciences, Inc. Modified oligonucleotides for mismatch discrimination
US6531590B1 (en) 1998-04-24 2003-03-11 Isis Pharmaceuticals, Inc. Processes for the synthesis of oligonucleotide compounds
US6867294B1 (en) 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6465628B1 (en) 1999-02-04 2002-10-15 Isis Pharmaceuticals, Inc. Process for the synthesis of oligomeric compounds
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
ATE356824T1 (de) 1999-05-04 2007-04-15 Santaris Pharma As L-ribo-lna analoge
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
US7321029B2 (en) 2000-01-21 2008-01-22 Geron Corporation 2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
WO2002028875A2 (fr) 2000-10-04 2002-04-11 Cureon A/S Synthese perfectionnee d'analogues d'acides nucleiques bloques de purine
US6878805B2 (en) 2002-08-16 2005-04-12 Isis Pharmaceuticals, Inc. Peptide-conjugated oligomeric compounds
JP2006507841A (ja) * 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド 機能的siRNAおよび超機能的siRNA
DK1606406T4 (da) 2003-03-21 2013-12-16 Santaris Pharma As Short Interfering RNA (siRNA) Analogues
US7427672B2 (en) 2003-08-28 2008-09-23 Takeshi Imanishi Artificial nucleic acids of n-o bond crosslinkage type
WO2005073378A1 (fr) 2004-01-30 2005-08-11 Santaris Pharma A/S Arn a interference breve modifies (arnsi modifies)
US7618947B2 (en) 2004-08-25 2009-11-17 Isis Pharmaceuticals, Inc. Modulation of HIF-1 beta expression
MX2007005557A (es) 2004-11-09 2008-02-15 Santaris Pharma As Oligonucleotidos lna y el tratamiento de cancer.
EP2314594B1 (fr) 2006-01-27 2014-07-23 Isis Pharmaceuticals, Inc. Analogues d'acide nucléique bicyclique modifiés en position 6
PE20090064A1 (es) 2007-03-26 2009-03-02 Novartis Ag Acido ribonucleico de doble cadena para inhibir la expresion del gen e6ap humano y composicion farmaceutica que lo comprende
CN101932709B (zh) 2007-11-26 2014-09-10 桑塔里斯制药公司 靶向雄激素受体的lna拮抗剂
JP6128732B2 (ja) 2008-10-03 2017-05-17 クルナ・インコーポレーテッド アポリポタンパク質−a1に対する天然アンチセンス転写物の抑制によるアポリポタンパク質−a1関連疾患の治療
ES2609655T3 (es) 2009-05-06 2017-04-21 Curna, Inc. Tratamiento de enfermedades relacionadas con tristetraprolina (TTP) mediante inhibición de transcrito antisentido natural para TTP

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050147593A1 (en) * 2003-05-22 2005-07-07 Medimmune, Inc. EphA2, EphA4 and LMW-PTP and methods of treatment of hyperproliferative cell disorders
WO2010017319A2 (fr) 2008-08-05 2010-02-11 Mdrna, Inc. Composés d’acide nucléique pour inhiber l’expression du gène plk1 et utilisations de ceux-ci
US20120014875A1 (en) * 2008-08-09 2012-01-19 University Of Iowa Research Foundation Nucleic acid aptamers
WO2010019446A1 (fr) 2008-08-09 2010-02-18 University Of Iowa Research Foundation Aptamères d'acide nucléique
CN101766817A (zh) 2008-12-26 2010-07-07 中国科学技术大学 一种药物载体及其应用
WO2011130458A2 (fr) 2010-04-13 2011-10-20 John Rossi Aptamères d'arn contre baff-r en tant qu'agents d'administration spécifiques d'un type cellulaire et leurs méthodes d'utilisation
WO2011142970A2 (fr) 2010-05-14 2011-11-17 University Of Iowa Research Foundation Aptamères d'acide nucléique her2
WO2012078637A2 (fr) * 2010-12-06 2012-06-14 Immune Disease Institute, Inc. Composition et méthode pour l'administration d'oligonucléotides
WO2013025930A1 (fr) 2011-08-16 2013-02-21 Research Foundation of State University of New York at Albany Modulateurs aptamères des récepteurs des oestrogènes
WO2014019025A1 (fr) 2012-08-02 2014-02-06 Deakin University Aptamères epcam pour la détection de cellules souches cancéreuses
WO2014068408A2 (fr) 2012-10-23 2014-05-08 Caris Life Sciences Switzerland Holdings, S.A.R.L. Aptamères et leurs utilisations
WO2014093698A1 (fr) 2012-12-12 2014-06-19 The Methodist Hospital Research Institute Analyses de détection de cellules tumorales en une étape, spécifiques à une cellule et à base de multi-aptamères
WO2014126160A1 (fr) 2013-02-13 2014-08-21 国立大学法人東京大学 Procédé d'examen du cancer et kit d'examen
CN103977433A (zh) 2014-05-14 2014-08-13 上海中医药大学附属岳阳中西医结合医院 一种前列腺癌超声诊断靶向试剂及制备方法
WO2016127216A1 (fr) 2015-02-11 2016-08-18 Deakin University Aptamères d'epcam et conjugués desdits aptamères

Non-Patent Citations (31)

* Cited by examiner, † Cited by third party
Title
Aliabadi et al., "Effective response of doxorubicin-sensitive and-resistant breast cancer cells to combinational siRNA therapy." Journal of Controlled Release 172(1):219-228 (2013).
Burnett et al., "RNA-based therapeutics: current progress and future prospects." Chemistry & Biology 19(1):60-71 (2012).
Dassie et al., "Systemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumors." Nature Biotechnology 27(9):839-849 (2009).
Fox et al., "Invasiveness of breast carcinoma cells and transcript profile: Eph receptors and ephrin ligands as molecular markers of potential diagnostic and prognostic application." Biochemical and Biophysical Research Communications 318(4):882-892 (2004).
Gilboa-Geffen et al., "Abstract CN01-02: Targeting basal-like TNBCs and epithelial tumor-initiating cells with aptamer-siRNA chimeras", Molecular Cancer Therapeutics 12(11 Suppl): (2013). (2 pages).
Gilboa-Geffen et al., "Gene knockdown by EpCAM aptamer-siRNA chimeras suppresses epithelial breast cancers and their tumor-initiating cells." Molecular Cancer Therapeutics 14(10):2279-2291 (2015).
Kim et al., "In Vitro Selection of RNA Aptamer and Specific Targeting of ErbB2 in Breast Cancer Cells." Nucleic Acid Therapeutics 21(3):173-178 (2011).
Machine translation of CN 101766817, pp. 1-7 (Year: 2010). *
McNamara et al., "Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras." Nature Biotechnology 24 (8):1005-1015 (2006).
McNamara et al., "Multivalent 4-1BB binding aptamers costimulate CD8+ T cells and inhibit tumor growth in mice." The Journal of Clinical Investigation 118(1):376-386 (2008).
McNamara et al., "Cell type—specific delivery of siRNAs with aptamer-siRNA chimeras." Nature Biotechnology 24 (8):1005-1015 (2006).
Neff et al., "An aptamer-siRNA chimera suppresses HIV-1 viral loads and protects from helper CD4+ T cell decline in humanized mice." Science Translational Medicine 3(66ra6):1-27 (2011).
Pastor et al. "Targeting 4-1BB costimulation to disseminated tumor lesions with bi-specific oligonucleotide aptamers." Molecular Therapy 19(10):1878-1886 (2011).
Pastor et al., "Induction of tumour immunity by targeted inhibition of nonsense-mediated mRNA decay." Nature 465 (7295):227-230 (2010).
Petrocca et al., "A genome-wide siRNA screen identifies proteasome addiction as a vulnerability of basal-like triple-negative breast cancer cells." Cancer Cell 24(2)182-196 (2013).
Rockey et al., "Rational Truncation of an RNA Aptamer to Prostate-Specific Membrane Antigen Using Computational Structural Modeling." Nucleic Acid Therapeutics 21(5)299-314 (2011).
Shigdar et al, RNA aptamer against a cancer stem cell marker epithelial cell adhesion molecule, 2011, Cancer Science, vol. 102, 5: 991-998 (Year: 2011). *
Shigdar et al., "RNA aptamer against a cancer stem cell marker epithelial cell adhesion molecule." Cancer Science 102(5):991-998 (2011).
Subramanian et al., "EpCAM aptamer-siRNA chimera targets and regress epithelial cancer." PloS One 10(7): e0132407 (2015).
Thiel et al., "Delivery of chemo-sensitizing siRNAs to HER2+-breast cancer cells using RNA aptamers." Nucleic Acids Research 40(13):6319-6337 (2012).
Wheeler et al., "Durable knockdown and protection from HIV transmission in humanized mice treated with gel-formulated CD4 aptamer-siRNA chimeras." Molecular Therapy 21(7):1378-1389 (2013).
Wheeler et al., "Inhibition of HIV transmission in human cervicovaginal explants and humanized mice using CD4 aptamer-siRNA chimeras." The Journal of Clinical Investigation 121(6):2401-2412 (2011).
Yang et al., "Wnt modulates MCL1 to control cell survival in triple negative breast cancer." BMC Cancer 14(124):1-13(2014).
Zhang et al., "Synergistic effect of the y-secretase inhibitor PF-03084014 and docetaxel in breast cancer models." Stem Cells Translational Medicine 2(3):233-242 (2013).
Zhou et al., "Aptamer-targeted cell-specific RNA interference." Silence 1(4):1-10 (2010).
Zhou et al., "Cell-specific aptamer-mediated targeted drug delivery." Oligonucleotides 21(1):1-10 (2011).
Zhou et al., "Cell-type-specific, aptamer-functionalized agents for targeted disease therapy." Molecular Therapy-Nucleic Acids 3(6):e169 (2014).
Zhou et al., "Novel dual inhibitory function aptamer-siRNA delivery system for HIV-1 therapy." Molecular Therapy 16 (8):1481-1489 (2008).
Zhou et al., "Selection, characterization and application of new RNA HIV gp 120 aptamers for facile delivery of Dicer substrate siRNAs into HIV infected cells." Nucleic Acids Research 37(9):3094-3109 (2009).
Zhou et al., "Cell-type-specific, aptamer-functionalized agents for targeted disease therapy." Molecular Therapy—Nucleic Acids 3(6):e169 (2014).
Zhou et al., "Novel dual inhibitory function aptamer—siRNA delivery system for HIV-1 therapy." Molecular Therapy 16 (8):1481-1489 (2008).

Also Published As

Publication number Publication date
AU2015308721A1 (en) 2017-03-16
AU2015308721B2 (en) 2021-04-01
US11180762B2 (en) 2021-11-23
US20170275629A1 (en) 2017-09-28
EP3185910A4 (fr) 2018-01-10
CA2959386A1 (fr) 2016-03-03
US20190309297A1 (en) 2019-10-10
WO2016033472A1 (fr) 2016-03-03
CN107106704A (zh) 2017-08-29
JP2017526367A (ja) 2017-09-14
KR20170070022A (ko) 2017-06-21
EP3185910A1 (fr) 2017-07-05
EP4043567A1 (fr) 2022-08-17

Similar Documents

Publication Publication Date Title
US11180762B2 (en) Methods and compositions for the treatment of cancer
US20200000881A1 (en) Efficient protein expression in vivo using modified rna (mod-rna)
ZA200407649B (en) Antisense IAP nucleobase oligomers and uses thereof
TW202144571A (zh) 脂蛋白元c3(apoc3)irna組成物及其用途
TW202138559A (zh) 含類PATATIN磷脂酶結構域3(PNPLA3)iRNA組成物及其使用方法
WO2018227116A1 (fr) Procédés de vaccination dans des configurations pré-malignes
US20220340906A1 (en) Methods and compositions for the treatment of cancer
AU2015339063A1 (en) TLR9 targeted cytotoxic agents
WO2023055885A2 (fr) Inhibition de l'ezh2 dans le cancer du pancréas
BR112021015323A2 (pt) Moduladores de expressão de malat1
WO2020106982A1 (fr) Procédés permettant de surmonter la résistance à des inhibiteurs de points de contrôle immunitaires
US20200115712A1 (en) Compositions and methods for cancer immunotherapy
US20170131280A1 (en) Methods, assays, and systems relating to sakt
WO2024011118A2 (fr) Méthodes et compositions pour le traitement du cancer par ciblage d'arn de transfert oncogènes
EP4124656A1 (fr) Micro arn pd-l1
US20210155928A1 (en) Rna-aided immunotherapeutics
Marrufo Expression and Function of Ligands for Natural Killer Cell Receptors on Triple-negative Breast Cancer Cells
Ge et al. Chemically modified siRNA directed against the KDR gene inhibits the proliferation of breast cancer cells
US20160186171A1 (en) Agents and methods for inhibiting mir-148a for the modulation of cholesterol levels

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHILDREN'S MEDICAL CENTER CORPORATION, MASSACHUSET

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIEBERMAN, JUDY;GILBOA-GEFFEN, ADI;WHEELER, LEE ADAM;SIGNING DATES FROM 20150924 TO 20151014;REEL/FRAME:041399/0615

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE AFTER FINAL ACTION FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: THE GOVERNMENT OF THE UNITED STATES, AS REPRESENTE

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:BOSTON CHILDREN'S HOSPITAL;REEL/FRAME:051027/0175

Effective date: 20170905

AS Assignment

Owner name: THE GOVERNMENT OF THE UNITED STATES AS REPRESENTED BY THE SECRETARY OF THE ARMY, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:BOSTON CHILDREN'S HOSPITAL;REEL/FRAME:058648/0982

Effective date: 20170816

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4