WO2014022811A1 - Isolating traffic-enhancing mutants of drug delivery protein - Google Patents

Isolating traffic-enhancing mutants of drug delivery protein Download PDF

Info

Publication number
WO2014022811A1
WO2014022811A1 PCT/US2013/053493 US2013053493W WO2014022811A1 WO 2014022811 A1 WO2014022811 A1 WO 2014022811A1 US 2013053493 W US2013053493 W US 2013053493W WO 2014022811 A1 WO2014022811 A1 WO 2014022811A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
cells
carrier
phage
cell
Prior art date
Application number
PCT/US2013/053493
Other languages
French (fr)
Inventor
Lali K. Medina-Kauwe
Original Assignee
Cedars-Sinai Medical Center
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
Priority to ES13826083T priority Critical patent/ES2703052T3/en
Priority to DK13826083.1T priority patent/DK2880160T3/en
Priority to RU2015105052A priority patent/RU2015105052A/en
Priority to CN201380041215.1A priority patent/CN104619839A/en
Priority to EP13826083.1A priority patent/EP2880160B1/en
Priority to CA2881582A priority patent/CA2881582A1/en
Priority to PL13826083T priority patent/PL2880160T3/en
Application filed by Cedars-Sinai Medical Center filed Critical Cedars-Sinai Medical Center
Priority to AU2013296218A priority patent/AU2013296218B2/en
Priority to KR1020157005438A priority patent/KR102115630B1/en
Priority to US14/419,233 priority patent/US10036009B2/en
Priority to BR112015002004A priority patent/BR112015002004A2/en
Priority to JP2015525636A priority patent/JP6618360B2/en
Priority to MX2015001366A priority patent/MX366347B/en
Publication of WO2014022811A1 publication Critical patent/WO2014022811A1/en
Priority to IL236955A priority patent/IL236955A0/en
Priority to ZA2015/00739A priority patent/ZA201500739B/en
Priority to IN1196DEN2015 priority patent/IN2015DN01196A/en
Priority to US16/017,866 priority patent/US10752893B2/en
Priority to AU2019204815A priority patent/AU2019204815A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1058Directional evolution of libraries, e.g. evolution of libraries is achieved by mutagenesis and screening or selection of mixed population of organisms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/01DNA viruses
    • C07K14/075Adenoviridae
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1037Screening libraries presented on the surface of microorganisms, e.g. phage display, E. coli display
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector

Definitions

  • the invention provides methods and compositions for cell penetration function to improve drug delivery to specific organelles.
  • Directed evolution has been used to generate pseudotyped adeno-associated virus (AAV) capsids with novel tropism, reduced non-specific delivery, and immune evasion (Kwon and Schaffer, 2008; Maheshri et al., 2006).
  • AAV pseudotyped adeno-associated virus
  • Biopanning of phage display and whole viral libraries in vitro and in vivo with different selective pressures has generated proteins with desired properties such as improved ligand binding and uptake, immune interactions, or enzyme activities (Yuan et al., 2005).
  • this methodology can be a powerful and more effective alternative to rational mutation for creating new protein variants with improved features.
  • Various embodiments include a method of enhancing trafficking to a cell, comprising providing a composition comprising a penton base (PB) protein with one or more mutations that enhance cellular entry and administering an effective dosage of the composition to the cell.
  • the one or more mutations is 111C and/or 333F.
  • the composition targets the cytoplasm and/or nucleus of the cell.
  • the composition further comprises a therapeutic drug.
  • the cell is a tumor cell.
  • the one or more mutations is SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7,SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, and/or SEQ ID NO: 20.
  • Other embodiments include a method for producing a drug delivery molecule that targets an organelle.
  • the method includes the following steps: a) obtaining a polynucleotide encoding the penton base (PB) gene and generating mutants of the polynucleotide, b) cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors, c) transforming cells with the phage library, d) fractioning the transformed cells and harvesting the organelle from the transformed cells, e) amplifying the phages from the harvested organelles, f) transforming cells with the amplified phages from the harvested organelle, g) repeating steps (d), (e), (f), and (g), h) titering the phages from the harvested organelles from each round, i) selecting the phages with the highest titer and obtaining the sequences of the mutant polynucleotide from the phage, and j) producing polypeptides encoded by the sequences, where
  • the organelle is selecting from the group consisting of mitochondrion, Golgi apparatus, endoplasmic reticulum, nucleus, ribosomes, plasma membrane and cytosol.
  • the cells are mammalian or non-mammalian cells.
  • the mutants are generated using any one or more of PCR- based methods, chemical mutagenesis, ultraviolet-induced mutagenesis or a combination thereof.
  • the drug delivery molecule comprises a targeting domain, an endosomolytic ligand domain and a positively charged domain.
  • a carrier for delivering a therapeutic agent to an organelle comprising a polypeptide encoded by one or more penton base (PB) mutations that enhance cellular entry.
  • the one or more penton base (PB) mutations include 111C and/or 333F.
  • the carrier further comprises a polylysine motif.
  • the carrier further comprises a targeting domain of heregulin.
  • the one or more PB mutations comprises a C-terminal deletion.
  • a therapeutic agent comprising a carrier for delivering a therapeutic agent to an organelle comprising a polypeptide encoded by one or more penton base (PB) mutations that enhance cellular entry and a therapeutic drug.
  • the therapeutic drug is a chemotherapeutic agent.
  • Various other embodiments include a method of producing a carrier without proliferative activity, comprising a) obtaining a polynucleotide encoding the receptor binding domain of heregulin (Her) and generating mutants in the polynucleotide, b) cloning the mutant Her polynucleotides into phage vectors and generating a phage library comprising the phage vectors, c) transforming MDA-MB-435 cells with the phage library in the presence of mitotic inhibitors, d) fractioning the transformed cell and extracting the membrane fraction of the MDA-MB-435 cells, e) harvesting membrane phages from the membrane fraction, f) transforming the membrane phages into MDA-MB-435 cells in the presence of mitotic inhibitors, g) repeating steps (d), (e), and (f), h) monitoring MDA-MB-435 cell proliferation in each round and selecting the membrane phages with the lowest MDA-MB-435 cell proliferation, i
  • a carrier for delivering therapeutics to the nucleus comprising a polypeptide encoded by mutant Her sequences.
  • the carrier further comprises a polypeptide encoding penton base (PB) protein and a polylysine motif.
  • PB protein is a mutant penton base protein.
  • a therapeutic agent comprising a carrier and a therapeutic drug.
  • FIG. 1 depicts, in accordance with an embodiment herein, the biopanning strategy.
  • Figure 2 depicts, in accordance with an embodiment herein, a PCR-based random mutagenesis of the penton base gene.
  • the PCR product (just above the 1600bp band) contains lOOng DNA, based on densitometry analysis, giving us a total yield of ⁇ 4800ng.
  • the initial target DNA was 97ng, we have a yield/initial ratio of ⁇ 50 and a duplication number of ⁇ 5.6, which when extrapolated against the standard curve provided in the manufacturer's protocol (Genemorph II, Strategene, La Jolla, CA, USA), corresponds to a mutation rate of ⁇ 8/kb.
  • Figure 3 depicts, in accordance with an embodiment herein, isolation of phage displaying penton base variants with enhanced partitioning in nuclear compartment.
  • a T7 phage library displaying randomly mutagenized PB was added to lxl 0 A 6 HeLa cells at lxl 0 A 8 pfu following the conditions described in the text for cell binding and uptake.
  • Cells were harvested by trypsinization to remove any surface-bound phage, then fractionated using a commercial fractionation kit (Qproteome Cell Compartment Kit; Qiagen Inc., Valencia, CA, USA).
  • Phage was PEG-precipitated overnight from each fraction following standard procedures, then resuspended in TB bacterial media and added to BLT5403 bacteria to amplify the isolated phage. Amplified phage was titered by plaque assay, then re-panned on HeLa using the same titer and conditions as described earlier. The phage obtained from cytosolic fractions underwent 3 rounds of cytosolic biopanning, whereas that obtained from nuclear fractions underwent 2 rounds.
  • Figure 4 depicts, in accordance with an embodiment herein, the alignment of trafficking variants isolated from biopanning.
  • Figure 5 depicts, in accordance with an embodiment herein, full-length mutant, 111C, exhibits enhanced trafficking to cytoplasm and nucleus.
  • Figure 5(A) depicts protein was precipitated from each fraction by acetone precipitation and pellets were resuspended in 40uL desalting buffer, followed by analysis via SDS-PAGE and Western blotting.
  • Figure 5(B) depicts analysis of band densities using Image J show an increase of 111C in both cytosolic and nuclear fractions compared to the wild-type protein.
  • Wild-type (WT) or mutant (111C, 333F) penton base protein 450 ug was added to separate cell aliquots and mixtures were incubated at 4C for 2hrs with agitation to promote receptor binding, followed by incubation at 37C for 2h with agiatation to promote internalization of receptor-bound protein. Control treatments received no protein (NP). Cells were then collected and processed for subcellular fractionation using the Qproteome Cell Compartment assay kit (Qiagen).
  • Figure 6 depicts, in accordance with an embodiment herein, PB mutants, 111C and 333F, exhibit enhanced nuclear entry compared to wild-type PB.
  • Figure 6(A) depicts intracellular trafficking and immunocytochemistry. Procedure followed the established protocols detailed in Gene Therapy (2006) 13, 821-836. Anti-penton base antibody (Ad5 antibody) was used at 1 :500 dilution. Alexa-Fluor 488 Goat Anti-Rabbit was used at 1 :400 dilution (2nd antibody). Phalloidin was used at 1 : 100 dilution, and DAPI used at 300nM. Green, wild-type (WT) or mutant (111C, 333F) PB; Red, actin; Blue, nucleus.
  • WT wild-type
  • mutant 111C, 333F
  • Figure 6(B) depicts quantification of protein trafficking. Sixteen cells from each treatment represented in the left panel was selected for quantification. The counts were based on the histogram of each image in Adobe Photoshop. Bars represent green pixel counts within the 80-255 window in the green channel.
  • Figure 7 depicts, in accordance with an embodiment herein, an updated Alignment of PB Mutants. New peptide lengths are based on amino acid sequences translated from nucleic acid sequences of mutant clones. Sequences are provided in Figures 8-17 herein.
  • Figure 8 depicts, in accordance with an embodiment herein, nucleic Acid and peptide sequence of mutated PB from Fraction 111 Clone A (111 A).
  • Figure 9 depicts, in accordance with an embodiment herein, nucleic acid and predicted amino acid sequence of mutated PB from Fraction 111 Clone C (111C).
  • Figure 10 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 111 Clone G (111G).
  • Figure 11 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 331 Clone E (33 IE).
  • Figure 12 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 331 Clone I (3311).
  • Figure 13 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 331 Clone J (331 J).
  • Figure 14 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 333 Clone A (333A).
  • Figure 15 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 333 Clone D (333D).
  • Figure 16 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 333 Clone E (333E).
  • Figure 17 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Franction 333, clones F, G, and H (333F, 333G, 333H).
  • PB penton base
  • a variety of reagents have been developed to deliver therapeutic genes and drugs to diseased cells, and include liposomes, synthetic polymers, peptides, proteins, viruses, and viral nanoparticles (Medina-Kauwe, 2006; MedinaKauwe et al., 2005).
  • the particles formed by these reagents require modifications to facilitate delivery of gene or drug payloads.
  • modifications include appending targeting ligands or antibodies, membrane penetrating agents, and/or intracellular targeting (such as nuclear targeting) agents.
  • Past and present attempts to improve cell membrane penetration and/or intracellular trafficking have used rational design to conjugate cell penetration or intracellular targeting peptides to drug carriers. Such an approach requires empirical testing of each molecule. Numerous types of cell penetration peptides have been tested for enhanced gene and drug delivery, and include AntP, TAT, GALA, honey bee melittin, and similar peptides (Medina- Kauwe, 2006; Medina-Kauwe et al, 2005). Likewise, nuclear targeting activity has been added to gene delivery agents via appendage of different poly-basic domains such as the SV40 NLS, HMG-1, protamine, and similar peptides or proteins (MedinaKauwe, 2006; Medina- Kauwe et al, 2005).
  • the invention described herein circumvents the time and effort required for rational design and empirical testing of cell penetration/intracellular trafficking proteins/peptides by using selective pressure to isolate protein mutants that have acquired improved cell penetration, intracellular trafficking, and/or subcellular targeting.
  • the protein mutants derived from this process have unique advantages over the parent proteins or existing gene/drug delivery proteins currently in use because of the improved features acquired through artificial evolution/selective pressure.
  • the specific proteins isolated by the process described below will provide an advantage over existing cell penetrating peptides because of their improved membrane lysis and trafficking features. Therefore, they can be used to augment gene and drug delivery, and thus enhance therapeutic efficacy of particles used in nanomedicine.
  • the invention provides a method for producing a drug delivery molecule that targets an organelle.
  • the method includes the steps of (a) obtaining a polynucleotide encoding the penton base gene and generating mutants of the polynucleotide; (b) cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors; (c) transforming cells with the phage library; (d) fractioning the transformed cells and harvesting the organell from the transformed cells; (e) amplifying the phages from the harvested organelles; (f) transforming cells with the amplified phages from the harvested organelle; (g) repeating steps (d), (e), (f), and (g); (h) titering the phages from the harvested organelles from each round; (i) selecting the phages with the highest titer and obtaining the sequences of the mutant polynucleotide from the phage; and (j) producing polypeptides encoded by the sequences, wherein the polypeptid
  • the organelle is selecting from the group consisting of mitochondrion, Golgi apparatus, endoplasmic reticulum, nucleus, ribosomes, plasma membrane and cytosol.
  • the cells may be mammalian or non-mammalian cells.
  • Mutants may be generated using any random mutagenesis methods or targeted mutagenesis methods. Examples of methods that may be used to generate mutants include but are not limited to any one or more of PCR-based methods, chemical mutagenesis, ultraviolet-induced mutagenesis or a combination thereof.
  • Mutations in the penton base gene may be any one or more of insertions, deletions, substitutions or a combination thereof.
  • the drug delivery molecule comprises a targeting domain, an endosomolytic ligand domain and a positively charged domain.
  • the invention also provides a carrier for delivering a therapeutic agent to an organelle, comprising a polypeptide encoded by a mutant penton base gene.
  • the mutation in the penton base gene may be isolated by a) obtaining a polynucleotide encoding the penton base gene and generating mutants of the polynucleotide; (b) cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors; (c) transforming cells with the phage library; (d) fractioning the transformed cells and harvesting the organell from the transformed cells; (e) amplifying the phages from the harvested organelles; (f) transforming cells with the amplified phages from the harvested organelle; (g) repeating steps (d), (e), (f), and (g); (h) titering the phages from the harvested organelles from each round; (i) selecting the phages with the highest t
  • the invention further provides a therapeutic agent comprising the carrier described above and a therapeutic drug.
  • a therapeutic drug may be any drug that, for example, treats, inhibits, prevents, mitigates the effects of, reduce the severity of, reduce the likelihood of developing, slow the progression of and/or cure, a disease.
  • Diseases targeted by the therapeutic agents include but are not limited to carcinomas, sarcomas, lymphomas, leukemia, germ cell tumors, blastomas, antigens expressed on various immune cells, and antigens expressed on cells associated with various hematologic diseases, autoimmune diseases, and/or inflammatory diseases.
  • Therapeutic agents may be a chemotherapeutic agent.
  • the invention also provides a method of producing a carrier without proliferative activity.
  • the method comprises (a) obtaining a polynucleotide encoding the receptor binding domain of heregulin (Her) and generating mutants in the polynucleotide; (b) cloning the mutant Her polynucleotides into phage vectors and generating a phage library comprising the phage vectors; (c) transforming MDA-MB-435 cells with the phage library in the presence of mitotic inhibitors; (for example taxol); (d)fractioning the transformed cell and extracting the membrane fraction of the MDA-MB-435 cells; (e) harvesting membrane phages from the membrane fraction; (f) transforming the membrane phages into MDA-MB-435 cells in the presence of mitotic inhibitors; (g) repeating steps (d), (e), and (f); (h) monitoring MDA-MB- 435 cell proliferation in each round and selecting the membrane phages with the lowest MDA
  • the invention further provides a carrier for delivering therapeutics to the nucleus, comprising a polypeptide encoded by the mutant Her sequences wherein the mutant Her is obtained according to the method described above.
  • the carrier further comprises a polypeptide encoding penton base protein and a polylysine motif.
  • the penton base protein may be a mutant protein.
  • a recombinant adenovirus penton base protein was developed to target and deliver a variety of therapeutic molecules to tumor cells in vitro and in vivo (Agadjanian et al., 2012; Agadjanian et al., 2009; Agadjanian et al., 2006; Medina-Kauwe et al., 2001a; Medina-Kauwe et al, 2001b; Rentsendorj et al, 2008).
  • the penton base recombinant protein (the same one that comprises the 'PB' domain of HerPBKlO, used to target therapeutics to HER2+ tumor cells; (Agadjanian et al., 2012; Agadjanian et al., 2009; Agadjanian et al, 2006; MedinaKauwe et al, 2001b; Rentsendorj et al, 2008) proceeds through multiple cell entry routes after cell binding, some but not all of which support membrane penetration and entry into the cytosol (Rentsendorj et al., 2006).
  • This two-step process involves: 1. Creation of a mutant library through random mutagenesis, and 2. Introduction of a selective pressure to isolate variants with improved function, depending on the screening process.
  • isolation of phage that survive entry into the cytosol and/or nucleus will serve as the selective pressure, which is expected to yield variants with enhanced cell penetration activity (Summarized in Fig. 1).
  • the inventors have randomly mutagenized the penton base gene and generated a library of mutants cloned into a . T7 phage vector.
  • the inventors inserted this product into a T7- Select phage vector and packaged recombinant phage to produce an amplified library titer of 5xl0 10 pfu/mL.
  • the library was panned onto HeLa cells (which express integrin receptors for binding and uptake of the PB protein). Phage were incubated on the cells at 4°C for lh to promote receptor binding but not uptake, then cells were washed and incubated for 2h at 37°C to promoted internalization. After uptake, harvested cells underwent fractionation to isolate cytosolic and nuclear fractions. Phage amplified from each fraction then underwent repeated biopanning, and corresponding sequential fractions were extracted from cell harvests (i.e.
  • nuclear phage isolated from round 1 were amplified and added back to cells, followed by repeat isolation of nuclear fractions to re-obtain nuclear phage). After either two or three rounds of biopanning, phage isolated from each repeat fraction was titered to determine the relative enrichment of nuclear/cytosolic phage from the mutagenized library compared to phage displaying wild-type penton base.
  • cytosolic and nuclear mutants After three rounds of biopanning and isolation of cytosolic and nuclear mutants, the inventors sequenced clones picked randomly from each enriched population and found that the majority of clones isolated from both cytosolic and nuclear fractions encoded carboxy- [C-] terminal truncated protein (Fig. 4).
  • the 287LDV and 340RGD integrin binding motifs located near the middle of the wild-type penton base (wt PB) linear sequence were not retained in most of the truncated clones.
  • One of the truncated mutants contains the LDV but not RGD motif, whereas the remaining truncations lack both LDV and RGD motifs.
  • the full-length clones isolated from the biopanning retain both LDV and RGD motifs but also contain several point mutations that introduce potential function-altering amino acid changes (Fig. 4). Among these are a Leu60Trp replacement in cytosolic fraction clone 111C; and Lys375Glu, Val449Met, and Pro469Ser amino acid changes in nuclear fraction clone 333F. To test the ability of each isolated variant to impart enhanced cytosolic and/or nuclear penetration, the intracellular trafficking of each will be compared to the parent protein by immunofluorescence and confocal microscopy, and confirmed by subcellular fractionation.
  • mutants 11 1C and 333F
  • mutant 331J retain the integrin binding motifs
  • these variants are tested in comparison to wt PB, as they are predicted to enter cells via integrin binding and uptake.
  • the remaining truncated variants lack any receptor-binding motifs, these will be inserted into HerPBKlO to replace the PB domain, and tested in comparison to parental HerPBKlO, which enters cells via human epidermal growth factor receptor (HER) binding and uptake.
  • HER human epidermal growth factor receptor
  • the tumor-targeted cell penetration protein, HerPBKlO is specifically directed to the human epidermal growth factor receptor (HER) via inclusion of the receptor binding domain of heregulin, designated here as the 'Her' domain of HerPBKlO (Medina-Kauwe et al., 2001b).
  • HER human epidermal growth factor receptor
  • heregulin receptors can induce signaling that may result in tumor cell proliferation, differentiation, and in some cases, apoptosis, depending on several factors including receptor heterodimer ratio, ligand subtype, cell type, and presence of certain intracellular molecules (Aguilar et al, 1999; Lewis et al, 1996; Weinstein et al, 1998).
  • the possibility of inducing adverse effects such as tumor progression leads to examining whether the selective pressure introduced by mitotic inhibitors can select receptor binding and endocytosis mutants that lack proliferative signaling.
  • the inventors proposed testing this approach by generating a phage library displaying Her variants and screen the library for internalizing Her species lacking proliferative signaling by isolating internalized phage from quiescent cells, and re-panning on non-proliferating human breast cancer cells.
  • a library of Her sequences containing different mutations distributed across the coding sequence will be produced by error-prone PCR and staggered extension, which entails repeated cycles of denaturation and brief annealing/extension after initial priming of template sequences (Zhao et al., 1998).
  • the resulting library will be inserted into the appropriate vector arms for transfer into T7Select bacteriophage (which is developed to display whole proteins), and recombinant phage produced following manufacturer's instructions (Novagen, Gibbstown, NJ, USA).
  • an initial titer of 10 A 12 phage will be added to MDA-MB-435 cells maintained in media containing mitotic inhibitor such as taxol, and at about 30-45 min later (the time required for binding and internalization; Medina-Kauwe et al., 2000) the cells will be harvested by trypsin/EDTA (to remove non-internalized phage) and membrane-extracted to isolate the vesicle fraction of crude virus (Qproteome Plasma Membrane Protein Kit, Qiagen Inc., Valencia, CA, USA), which can then be isolated by CsCl banding. Three to four rounds of selection (i.e.
  • isolated virus will be characterized in the following ways. First, fresh cells receiving enriched virus will be fixed and processed for immunofluorescence using an anti-phage antibody (Sigma- Aldrich, St. Louis, MO, USA) to confirm that the isolated phage still internalize. Separate cells treated in parallel will be assessed for proliferation rate in comparison to mock and untreated cells, by metabolic assay.
  • an anti-phage antibody Sigma- Aldrich, St. Louis, MO, USA
  • the Her sequence from isolated phage will be excised and inserted into a bacterial expression vector for recombinant protein production (Medina-Kauwe et al., 2001a), then mutant Her tested for internalization and proliferative activity in MDA-MB- 435 human breast cancer cells as described earlier, in comparison to parental Her as well as mock and untreated cells.
  • the mutant clones will be sequenced to identify mutated regions, and inserted back into the HerPBKlO expression cassette, replacing parental 'Her'.

Abstract

The invention relates to methods for isolating traffic-enhancing mutants of drug delivery proteins. In one embodiment, the invention provides a carrier for delivering a therapeutic agent to an organelle, comprising a polypeptide encoded by a mutant penton base gene. In another embodiment, the invention provides a method of enhancing trafficking to a cell by administering a composition comprising a penton base (PB) protein with one or more mutations that enhance cellular entry.

Description

ISOLATING TRAFFIC-ENHANCING MUTANTS OF DRUG DELIVERY PROTEIN
GOVERNMENT RIGHTS The invention was made with government support under Grant Nos. CA129822 and CA140995 awarded by the National Institutes of Health. The government has certain rights in the invention.
FIELD OF INVENTION
The invention provides methods and compositions for cell penetration function to improve drug delivery to specific organelles.
BACKGROUND OF THE INVENTION
Directed evolution has been used to generate pseudotyped adeno-associated virus (AAV) capsids with novel tropism, reduced non-specific delivery, and immune evasion (Kwon and Schaffer, 2008; Maheshri et al., 2006). Biopanning of phage display and whole viral libraries in vitro and in vivo with different selective pressures has generated proteins with desired properties such as improved ligand binding and uptake, immune interactions, or enzyme activities (Yuan et al., 2005). Thus, this methodology can be a powerful and more effective alternative to rational mutation for creating new protein variants with improved features. The most recently developed approach to this process employs error-prone PCR and staggered extension to create a library of variants that can be screened against different cell types to select out cell-specific targeted vectors (Zhao et al, 1998). To date, a process has not yet been developed to isolate protein variants with improved intracellular trafficking functions. This invention introduces a new procedure and new protein molecules with improved cell penetration functions for drug delivery resulting from the procedure. SUMMARY OF THE INVENTION
Various embodiments include a method of enhancing trafficking to a cell, comprising providing a composition comprising a penton base (PB) protein with one or more mutations that enhance cellular entry and administering an effective dosage of the composition to the cell. In one embodiment, the one or more mutations is 111C and/or 333F. In another embodiment, the composition targets the cytoplasm and/or nucleus of the cell. In another embodiment, the composition further comprises a therapeutic drug. In another embodiment, the cell is a tumor cell. In another embodiment, the one or more mutations is SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7,SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, and/or SEQ ID NO: 20. Other embodiments include a method for producing a drug delivery molecule that targets an organelle. The method includes the following steps: a) obtaining a polynucleotide encoding the penton base (PB) gene and generating mutants of the polynucleotide, b) cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors, c) transforming cells with the phage library, d) fractioning the transformed cells and harvesting the organelle from the transformed cells, e) amplifying the phages from the harvested organelles, f) transforming cells with the amplified phages from the harvested organelle, g) repeating steps (d), (e), (f), and (g), h) titering the phages from the harvested organelles from each round, i) selecting the phages with the highest titer and obtaining the sequences of the mutant polynucleotide from the phage, and j) producing polypeptides encoded by the sequences, where the polypeptides are the drug delivery molecules that target organelles. In another embodiment, the organelle is selecting from the group consisting of mitochondrion, Golgi apparatus, endoplasmic reticulum, nucleus, ribosomes, plasma membrane and cytosol. In another embodiment, the cells are mammalian or non-mammalian cells. In another embodiment, the mutants are generated using any one or more of PCR- based methods, chemical mutagenesis, ultraviolet-induced mutagenesis or a combination thereof. In another embodiment, the drug delivery molecule comprises a targeting domain, an endosomolytic ligand domain and a positively charged domain.
Other embodiments include a carrier for delivering a therapeutic agent to an organelle, comprising a polypeptide encoded by one or more penton base (PB) mutations that enhance cellular entry. In another embodiment, the one or more penton base (PB) mutations include 111C and/or 333F. In another embodiment, the carrier further comprises a polylysine motif. In another embodiment, the carrier further comprises a targeting domain of heregulin. In another embodiment, the one or more PB mutations comprises a C-terminal deletion. Other embodiments include a therapeutic agent comprising a carrier for delivering a therapeutic agent to an organelle comprising a polypeptide encoded by one or more penton base (PB) mutations that enhance cellular entry and a therapeutic drug. In another embodiment, the therapeutic drug is a chemotherapeutic agent.
Various other embodiments include a method of producing a carrier without proliferative activity, comprising a) obtaining a polynucleotide encoding the receptor binding domain of heregulin (Her) and generating mutants in the polynucleotide, b) cloning the mutant Her polynucleotides into phage vectors and generating a phage library comprising the phage vectors, c) transforming MDA-MB-435 cells with the phage library in the presence of mitotic inhibitors, d) fractioning the transformed cell and extracting the membrane fraction of the MDA-MB-435 cells, e) harvesting membrane phages from the membrane fraction, f) transforming the membrane phages into MDA-MB-435 cells in the presence of mitotic inhibitors, g) repeating steps (d), (e), and (f), h) monitoring MDA-MB-435 cell proliferation in each round and selecting the membrane phages with the lowest MDA-MB-435 cell proliferation, i) obtaining the sequences of the Her polynucleotide mutants in the selected membrane phages, and j) producing polypeptides encoded by the Her sequences and penton base gene, where the polypeptides are the carrier without proliferative activity.
Other embodiments include a carrier for delivering therapeutics to the nucleus, comprising a polypeptide encoded by mutant Her sequences. In another embodiment, the carrier further comprises a polypeptide encoding penton base (PB) protein and a polylysine motif. In another embodiment, the PB protein is a mutant penton base protein.
Other embodiments include a therapeutic agent comprising a carrier and a therapeutic drug.
BRIEF DESCRIPTION OF FIGURES
Figures 1 depicts, in accordance with an embodiment herein, the biopanning strategy.
Figure 2 depicts, in accordance with an embodiment herein, a PCR-based random mutagenesis of the penton base gene. The PCR product (just above the 1600bp band) contains lOOng DNA, based on densitometry analysis, giving us a total yield of ~4800ng. As the initial target DNA was 97ng, we have a yield/initial ratio of ~50 and a duplication number of ~5.6, which when extrapolated against the standard curve provided in the manufacturer's protocol (Genemorph II, Strategene, La Jolla, CA, USA), corresponds to a mutation rate of ~8/kb.
Figure 3 depicts, in accordance with an embodiment herein, isolation of phage displaying penton base variants with enhanced partitioning in nuclear compartment. A T7 phage library displaying randomly mutagenized PB was added to lxl 0A6 HeLa cells at lxl 0A8 pfu following the conditions described in the text for cell binding and uptake. Cells were harvested by trypsinization to remove any surface-bound phage, then fractionated using a commercial fractionation kit (Qproteome Cell Compartment Kit; Qiagen Inc., Valencia, CA, USA). Phage was PEG-precipitated overnight from each fraction following standard procedures, then resuspended in TB bacterial media and added to BLT5403 bacteria to amplify the isolated phage. Amplified phage was titered by plaque assay, then re-panned on HeLa using the same titer and conditions as described earlier. The phage obtained from cytosolic fractions underwent 3 rounds of cytosolic biopanning, whereas that obtained from nuclear fractions underwent 2 rounds. Figure 4 depicts, in accordance with an embodiment herein, the alignment of trafficking variants isolated from biopanning.
Figure 5 depicts, in accordance with an embodiment herein, full-length mutant, 111C, exhibits enhanced trafficking to cytoplasm and nucleus. Figure 5(A) depicts protein was precipitated from each fraction by acetone precipitation and pellets were resuspended in 40uL desalting buffer, followed by analysis via SDS-PAGE and Western blotting. Figure 5(B) depicts analysis of band densities using Image J show an increase of 111C in both cytosolic and nuclear fractions compared to the wild-type protein. Trafficking and Cell fractionation assay: Adherent HeLa cells growing in flasks were detached with 5mL lXPBS+2mM EDTA at 37C incubation with agitation for 50 min and transferred to 15 mL conical tubes. The density of the cells were measured, and cells were distributed into separate tubes at 5x106 cells per tube. Cells were washed with IX PBS + Ca2+ + Mg2+ three times to remove the EDTA, and cell pellets were resuspended in 0.7 ml Buffer A (20mM HEPES, pH 7.4; 2mM MgC12; 3% BSA in DMEM). Wild-type (WT) or mutant (111C, 333F) penton base protein (450 ug) was added to separate cell aliquots and mixtures were incubated at 4C for 2hrs with agitation to promote receptor binding, followed by incubation at 37C for 2h with agiatation to promote internalization of receptor-bound protein. Control treatments received no protein (NP). Cells were then collected and processed for subcellular fractionation using the Qproteome Cell Compartment assay kit (Qiagen).
Figure 6 depicts, in accordance with an embodiment herein, PB mutants, 111C and 333F, exhibit enhanced nuclear entry compared to wild-type PB. Figure 6(A) depicts intracellular trafficking and immunocytochemistry. Procedure followed the established protocols detailed in Gene Therapy (2006) 13, 821-836. Anti-penton base antibody (Ad5 antibody) was used at 1 :500 dilution. Alexa-Fluor 488 Goat Anti-Rabbit was used at 1 :400 dilution (2nd antibody). Phalloidin was used at 1 : 100 dilution, and DAPI used at 300nM. Green, wild-type (WT) or mutant (111C, 333F) PB; Red, actin; Blue, nucleus. Figure 6(B) depicts quantification of protein trafficking. Sixteen cells from each treatment represented in the left panel was selected for quantification. The counts were based on the histogram of each image in Adobe Photoshop. Bars represent green pixel counts within the 80-255 window in the green channel.
Figure 7 depicts, in accordance with an embodiment herein, an updated Alignment of PB Mutants. New peptide lengths are based on amino acid sequences translated from nucleic acid sequences of mutant clones. Sequences are provided in Figures 8-17 herein.
Figure 8 depicts, in accordance with an embodiment herein, nucleic Acid and peptide sequence of mutated PB from Fraction 111 Clone A (111 A).
Figure 9 depicts, in accordance with an embodiment herein, nucleic acid and predicted amino acid sequence of mutated PB from Fraction 111 Clone C (111C).
Figure 10 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 111 Clone G (111G).
Figure 11 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 331 Clone E (33 IE). Figure 12 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 331 Clone I (3311).
Figure 13 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 331 Clone J (331 J).
Figure 14 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 333 Clone A (333A). Figure 15 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 333 Clone D (333D).
Figure 16 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Fraction 333 Clone E (333E).
Figure 17 depicts, in accordance with an embodiment herein, nucleic acid and peptide sequence of mutated PB from Franction 333, clones F, G, and H (333F, 333G, 333H).
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the abbreviation "PB" means penton base.
A variety of reagents have been developed to deliver therapeutic genes and drugs to diseased cells, and include liposomes, synthetic polymers, peptides, proteins, viruses, and viral nanoparticles (Medina-Kauwe, 2006; MedinaKauwe et al., 2005). Typically, the particles formed by these reagents require modifications to facilitate delivery of gene or drug payloads. Such modifications include appending targeting ligands or antibodies, membrane penetrating agents, and/or intracellular targeting (such as nuclear targeting) agents. These modifications are usually introduced through rational design and thus each new variant generated by such modification requires empirical testing. This can be time consuming and labor intensive, and runs the risk of yielding suboptimal activity.
Past and present attempts to improve cell membrane penetration and/or intracellular trafficking have used rational design to conjugate cell penetration or intracellular targeting peptides to drug carriers. Such an approach requires empirical testing of each molecule. Numerous types of cell penetration peptides have been tested for enhanced gene and drug delivery, and include AntP, TAT, GALA, honey bee melittin, and similar peptides (Medina- Kauwe, 2006; Medina-Kauwe et al, 2005). Likewise, nuclear targeting activity has been added to gene delivery agents via appendage of different poly-basic domains such as the SV40 NLS, HMG-1, protamine, and similar peptides or proteins (MedinaKauwe, 2006; Medina- Kauwe et al, 2005). While each of these peptides possess the capacity to penetrate cell membranes or target intracellular compartments, including the nucleus, their activities are altered when covalently coupled to another molecule or expressed as a fusion protein to another molecule. Moreover, empirical testing of each different variant of these peptides is time consuming and labor intensive. Therefore, while rational design and empirical testing is the existing solution to this problem, this approach, has its limitations.
The invention described herein circumvents the time and effort required for rational design and empirical testing of cell penetration/intracellular trafficking proteins/peptides by using selective pressure to isolate protein mutants that have acquired improved cell penetration, intracellular trafficking, and/or subcellular targeting. The protein mutants derived from this process have unique advantages over the parent proteins or existing gene/drug delivery proteins currently in use because of the improved features acquired through artificial evolution/selective pressure. Finally, the specific proteins isolated by the process described below will provide an advantage over existing cell penetrating peptides because of their improved membrane lysis and trafficking features. Therefore, they can be used to augment gene and drug delivery, and thus enhance therapeutic efficacy of particles used in nanomedicine. The invention provides a method for producing a drug delivery molecule that targets an organelle. The method includes the steps of (a) obtaining a polynucleotide encoding the penton base gene and generating mutants of the polynucleotide; (b) cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors; (c) transforming cells with the phage library; (d) fractioning the transformed cells and harvesting the organell from the transformed cells; (e) amplifying the phages from the harvested organelles; (f) transforming cells with the amplified phages from the harvested organelle; (g) repeating steps (d), (e), (f), and (g); (h) titering the phages from the harvested organelles from each round; (i) selecting the phages with the highest titer and obtaining the sequences of the mutant polynucleotide from the phage; and (j) producing polypeptides encoded by the sequences, wherein the polypeptides are the drug delivery molecules that target organelles.
In some embodiments, the organelle is selecting from the group consisting of mitochondrion, Golgi apparatus, endoplasmic reticulum, nucleus, ribosomes, plasma membrane and cytosol. The cells may be mammalian or non-mammalian cells. Mutants may be generated using any random mutagenesis methods or targeted mutagenesis methods. Examples of methods that may be used to generate mutants include but are not limited to any one or more of PCR-based methods, chemical mutagenesis, ultraviolet-induced mutagenesis or a combination thereof. Mutations in the penton base gene may be any one or more of insertions, deletions, substitutions or a combination thereof.
In an embodiment of the invention, the drug delivery molecule comprises a targeting domain, an endosomolytic ligand domain and a positively charged domain.
The invention also provides a carrier for delivering a therapeutic agent to an organelle, comprising a polypeptide encoded by a mutant penton base gene. The mutation in the penton base gene may be isolated by a) obtaining a polynucleotide encoding the penton base gene and generating mutants of the polynucleotide; (b) cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors; (c) transforming cells with the phage library; (d) fractioning the transformed cells and harvesting the organell from the transformed cells; (e) amplifying the phages from the harvested organelles; (f) transforming cells with the amplified phages from the harvested organelle; (g) repeating steps (d), (e), (f), and (g); (h) titering the phages from the harvested organelles from each round; (i) selecting the phages with the highest titer and obtaining the sequences of the mutant polynucleotide from the phage; and (j) producing polypeptides encoded by the sequences, wherein the polypeptides are the drug delivery molecules that target organelles. The carrier further comprises a polylysine motif and a targeting domain, for example the targeting domain of heregulin.
The invention further provides a therapeutic agent comprising the carrier described above and a therapeutic drug. A therapeutic drug may be any drug that, for example, treats, inhibits, prevents, mitigates the effects of, reduce the severity of, reduce the likelihood of developing, slow the progression of and/or cure, a disease. Diseases targeted by the therapeutic agents include but are not limited to carcinomas, sarcomas, lymphomas, leukemia, germ cell tumors, blastomas, antigens expressed on various immune cells, and antigens expressed on cells associated with various hematologic diseases, autoimmune diseases, and/or inflammatory diseases. Therapeutic agents may be a chemotherapeutic agent.
The invention also provides a method of producing a carrier without proliferative activity. The method comprises (a) obtaining a polynucleotide encoding the receptor binding domain of heregulin (Her) and generating mutants in the polynucleotide; (b) cloning the mutant Her polynucleotides into phage vectors and generating a phage library comprising the phage vectors; (c) transforming MDA-MB-435 cells with the phage library in the presence of mitotic inhibitors; (for example taxol); (d)fractioning the transformed cell and extracting the membrane fraction of the MDA-MB-435 cells; (e) harvesting membrane phages from the membrane fraction; (f) transforming the membrane phages into MDA-MB-435 cells in the presence of mitotic inhibitors; (g) repeating steps (d), (e), and (f); (h) monitoring MDA-MB- 435 cell proliferation in each round and selecting the membrane phages with the lowest MDA-MB-435 cell proliferation; (i) obtaining the sequences of the Her polynucleotide mutants in the selected membrane phages; and (j) producing polypeptides encoded by the Her sequences and the penton base gene, wherein the polypeptides are the carrier without proliferative activity. Mutations in the Her gene may be any one or more of insertions, deletions, substitutions or a combination thereof.
The invention further provides a carrier for delivering therapeutics to the nucleus, comprising a polypeptide encoded by the mutant Her sequences wherein the mutant Her is obtained according to the method described above. The carrier further comprises a polypeptide encoding penton base protein and a polylysine motif. The penton base protein may be a mutant protein.
EXAMPLES Example 1
Isolation of traffic-enhancing mutants.
Previously, a recombinant adenovirus penton base protein was developed to target and deliver a variety of therapeutic molecules to tumor cells in vitro and in vivo (Agadjanian et al., 2012; Agadjanian et al., 2009; Agadjanian et al., 2006; Medina-Kauwe et al., 2001a; Medina-Kauwe et al, 2001b; Rentsendorj et al, 2008). Currently, the penton base recombinant protein (the same one that comprises the 'PB' domain of HerPBKlO, used to target therapeutics to HER2+ tumor cells; (Agadjanian et al., 2012; Agadjanian et al., 2009; Agadjanian et al, 2006; MedinaKauwe et al, 2001b; Rentsendorj et al, 2008) proceeds through multiple cell entry routes after cell binding, some but not all of which support membrane penetration and entry into the cytosol (Rentsendorj et al., 2006). To improve upon this function and enhance the delivery and penetration of therapeutics into cell targets, a directed evolution approach was used to isolate penton base variants with enhanced cell penetration activity by using nuclear accumulation as a readout, as endosomal escape enables entry into the nucleus (Rentsendorj et al, 2006).
Procedures: This two-step process involves: 1. Creation of a mutant library through random mutagenesis, and 2. Introduction of a selective pressure to isolate variants with improved function, depending on the screening process. Here, isolation of phage that survive entry into the cytosol and/or nucleus will serve as the selective pressure, which is expected to yield variants with enhanced cell penetration activity (Summarized in Fig. 1). Accordingly, the inventors have randomly mutagenized the penton base gene and generated a library of mutants cloned into a . T7 phage vector. Based on previously established directed evolution studies (Cherry et al, 1999; Shafikhani et al, 1997; Wan et al., 1998; You and Arnold, 1996), the inventors aimed for a mutation frequency of 1-4 amino acid changes (or 2-7 nucleotide changes) per gene. Based on the product yield using a specialized error-prone polymerase chain reaction (PCR) method (GeneMorphll Random Mutagenesis Kit; Stratagene, La Jolla, CA, USA), the inventors achieved an estimated mutation frequency of ~8 nucleotides/kb or 13.6 nucleotides per penton base gene (Fig. 2). The inventors inserted this product into a T7- Select phage vector and packaged recombinant phage to produce an amplified library titer of 5xl010 pfu/mL. The library was panned onto HeLa cells (which express integrin receptors for binding and uptake of the PB protein). Phage were incubated on the cells at 4°C for lh to promote receptor binding but not uptake, then cells were washed and incubated for 2h at 37°C to promoted internalization. After uptake, harvested cells underwent fractionation to isolate cytosolic and nuclear fractions. Phage amplified from each fraction then underwent repeated biopanning, and corresponding sequential fractions were extracted from cell harvests (i.e. nuclear phage isolated from round 1 were amplified and added back to cells, followed by repeat isolation of nuclear fractions to re-obtain nuclear phage). After either two or three rounds of biopanning, phage isolated from each repeat fraction was titered to determine the relative enrichment of nuclear/cytosolic phage from the mutagenized library compared to phage displaying wild-type penton base.
Results: The non-mutagenized parent phage, T7-PB, yielded a nuclear/cytosolic phage titer ratio of less than 1 , indicating that the proportion of phage arriving at the nucleus by 2h was less than the proportion of phage that remained in the cytoplasm (Fig. 3). In contrast, after 2 rounds of biopanning and isolation of nuclear phage (3 -3 a), a shift was observed toward higher nuclear accumulation compared to cytoplasmic retention, with a significant increase compared to T7-PB (P=0.05) (Fig. 3). Even phage isolated from 3 rounds of cytosolic fraction panning (1-1-1) showed a relative, though not highly significant (P=0.07), increase in nuclear partitioning compared to T7-PB (Fig. 3).
After three rounds of biopanning and isolation of cytosolic and nuclear mutants, the inventors sequenced clones picked randomly from each enriched population and found that the majority of clones isolated from both cytosolic and nuclear fractions encoded carboxy- [C-] terminal truncated protein (Fig. 4). The 287LDV and 340RGD integrin binding motifs located near the middle of the wild-type penton base (wt PB) linear sequence were not retained in most of the truncated clones. One of the truncated mutants contains the LDV but not RGD motif, whereas the remaining truncations lack both LDV and RGD motifs. The full-length clones isolated from the biopanning retain both LDV and RGD motifs but also contain several point mutations that introduce potential function-altering amino acid changes (Fig. 4). Among these are a Leu60Trp replacement in cytosolic fraction clone 111C; and Lys375Glu, Val449Met, and Pro469Ser amino acid changes in nuclear fraction clone 333F. To test the ability of each isolated variant to impart enhanced cytosolic and/or nuclear penetration, the intracellular trafficking of each will be compared to the parent protein by immunofluorescence and confocal microscopy, and confirmed by subcellular fractionation. Specifically, as the full-length mutants (11 1C and 333F) and mutant 331J retain the integrin binding motifs, these variants are tested in comparison to wt PB, as they are predicted to enter cells via integrin binding and uptake. Meanwhile, as the remaining truncated variants lack any receptor-binding motifs, these will be inserted into HerPBKlO to replace the PB domain, and tested in comparison to parental HerPBKlO, which enters cells via human epidermal growth factor receptor (HER) binding and uptake.
Example 2
Isolation of receptor-binding and endocytosis mutants with blunted signaling.
Rationale: The tumor-targeted cell penetration protein, HerPBKlO, is specifically directed to the human epidermal growth factor receptor (HER) via inclusion of the receptor binding domain of heregulin, designated here as the 'Her' domain of HerPBKlO (Medina-Kauwe et al., 2001b). However, ligation of heregulin receptors can induce signaling that may result in tumor cell proliferation, differentiation, and in some cases, apoptosis, depending on several factors including receptor heterodimer ratio, ligand subtype, cell type, and presence of certain intracellular molecules (Aguilar et al, 1999; Lewis et al, 1996; Weinstein et al, 1998). The possibility of inducing adverse effects such as tumor progression leads to examining whether the selective pressure introduced by mitotic inhibitors can select receptor binding and endocytosis mutants that lack proliferative signaling. The inventors proposed testing this approach by generating a phage library displaying Her variants and screen the library for internalizing Her species lacking proliferative signaling by isolating internalized phage from quiescent cells, and re-panning on non-proliferating human breast cancer cells.
Procedure: A library of Her sequences containing different mutations distributed across the coding sequence will be produced by error-prone PCR and staggered extension, which entails repeated cycles of denaturation and brief annealing/extension after initial priming of template sequences (Zhao et al., 1998). The resulting library will be inserted into the appropriate vector arms for transfer into T7Select bacteriophage (which is developed to display whole proteins), and recombinant phage produced following manufacturer's instructions (Novagen, Gibbstown, NJ, USA). Based on biopanning of evolved AAV capsids, an initial titer of 10A12 phage will be added to MDA-MB-435 cells maintained in media containing mitotic inhibitor such as taxol, and at about 30-45 min later (the time required for binding and internalization; Medina-Kauwe et al., 2000) the cells will be harvested by trypsin/EDTA (to remove non-internalized phage) and membrane-extracted to isolate the vesicle fraction of crude virus (Qproteome Plasma Membrane Protein Kit, Qiagen Inc., Valencia, CA, USA), which can then be isolated by CsCl banding. Three to four rounds of selection (i.e. adding isolated virus to fresh cells and repeating membrane extraction) will be performed and isolated virus will be characterized in the following ways. First, fresh cells receiving enriched virus will be fixed and processed for immunofluorescence using an anti-phage antibody (Sigma- Aldrich, St. Louis, MO, USA) to confirm that the isolated phage still internalize. Separate cells treated in parallel will be assessed for proliferation rate in comparison to mock and untreated cells, by metabolic assay. Second, the Her sequence from isolated phage will be excised and inserted into a bacterial expression vector for recombinant protein production (Medina-Kauwe et al., 2001a), then mutant Her tested for internalization and proliferative activity in MDA-MB- 435 human breast cancer cells as described earlier, in comparison to parental Her as well as mock and untreated cells. The mutant clones will be sequenced to identify mutated regions, and inserted back into the HerPBKlO expression cassette, replacing parental 'Her'. The various methods and techniques described above provide a number of ways to carry out the application. Of course, it is to be understood that not necessarily all objectives or advantages described can be achieved in accordance with any particular embodiment described herein. Thus, for example, those skilled in the art will recognize that the methods can be performed in a manner that achieves or optimizes one advantage or group of advantages as taught herein without necessarily achieving other objectives or advantages as taught or suggested herein. A variety of alternatives are mentioned herein. It is to be understood that some preferred embodiments specifically include one, another, or several features, while others specifically exclude one, another, or several features, while still others mitigate a particular feature by inclusion of one, another, or several advantageous features.
Furthermore, the skilled artisan will recognize the applicability of various features from different embodiments. Similarly, the various elements, features and steps discussed above, as well as other known equivalents for each such element, feature or step, can be employed in various combinations by one of ordinary skill in this art to perform methods in accordance with the principles described herein. Among the various elements, features, and steps some will be specifically included and others specifically excluded in diverse embodiments.
Although the application has been disclosed in the context of certain embodiments and examples, it will be understood by those skilled in the art that the embodiments of the application extend beyond the specifically disclosed embodiments to other alternative embodiments and/or uses and modifications and equivalents thereof.
In some embodiments, the terms "a" and "an" and "the" and similar references used in the context of describing a particular embodiment of the application (especially in the context of certain of the following claims) can be construed to cover both the singular and the plural. The recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (for example, "such as") provided with respect to certain embodiments herein is intended merely to better illuminate the application and does not pose a limitation on the scope of the application otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the application.
Preferred embodiments of this application are described herein, including the best mode known to the inventors for carrying out the application. Variations on those preferred embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. It is contemplated that skilled artisans can employ such variations as appropriate, and the application can be practiced otherwise than specifically described herein. Accordingly, many embodiments of this application include all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the application unless otherwise indicated herein or otherwise clearly contradicted by context.
All patents, patent applications, publications of patent applications, and other material, such as articles, books, specifications, publications, documents, things, and/or the like, referenced herein are hereby incorporated herein by this reference in their entirety for all purposes, excepting any prosecution file history associated with same, any of same that is inconsistent with or in conflict with the present document, or any of same that may have a limiting affect as to the broadest scope of the claims now or later associated with the present document. By way of example, should there be any inconsistency or conflict between the description, definition, and/or the use of a term associated with any of the incorporated material and that associated with the present document, the description, definition, and/or the use of the term in the present document shall prevail.
In closing, it is to be understood that the embodiments of the application disclosed herein are illustrative of the principles of the embodiments of the application. Other modifications that can be employed can be within the scope of the application. Thus, by way of example, but not of limitation, alternative configurations of the embodiments of the application can be utilized in accordance with the teachings herein. Accordingly, embodiments of the present application are not limited to that precisely as shown and described.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of enhancing trafficking to a cell, comprising:
providing a composition comprising a penton base (PB) protein with one or more mutations that enhance cellular entry; and
administering an effective dosage of the composition to the cell.
2. The method of claim 1, wherein the one or more mutations is 111C and/or 333F.
3. The method of claim 1, wherein the composition targets the cytoplasm and/or nucleus of the cell.
4. The method of claim 1, wherein the composition further comprises a therapeutic drug.
5. The method of claim 1, wherein the cell is a tumor cell.
6. The method of claim 1, wherein the one or more mutations is SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7,SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, and/or SEQ ID NO: 20.
7. A method for producing a drug delivery molecule that targets an organelle comprising: a. obtaining a polynucleotide encoding the penton base (PB) gene and generating mutants of the polynucleotide;
b. cloning the mutant polynucleotide into a phage vector and generating a phage library comprising the phage vectors;
c. transforming cells with the phage library;
d. fractioning the transformed cells and harvesting the organelle from the transformed cells;
e. amplifying the phages from the harvested organelles; f. transforming cells with the amplified phages from the harvested organelle;
g. repeating steps (d), (e), (f), and (g);
h. titering the phages from the harvested organelles from each round;
i. selecting the phages with the highest titer and obtaining the sequences of the mutant polynucleotide from the phage; and
j. producing polypeptides encoded by the sequences,
wherein the polypeptides are the drug delivery molecules that target organelles.
8. The method of claim 7, wherein the organelle is selecting from the group consisting of mitochondrion, Golgi apparatus, endoplasmic reticulum, nucleus, ribosomes, plasma membrane and cytosol.
9. The method of claim 7, wherein the cells are mammalian or non-mammalian cells.
10. The method of claim 7, wherein mutants are generated using any one or more of PCR- based methods, chemical mutagenesis, ultraviolet-induced mutagenesis or a combination thereof.
11. The method of claim 7, wherein the drug delivery molecule comprises a targeting domain, an endosomolytic ligand domain and a positively charged domain.
12. A carrier for delivering a therapeutic agent to an organelle, comprising a polypeptide encoded by one or more penton base (PB) mutations that enhance cellular entry.
13. The carrier of claim 12, wherein the one or more penton base (PB) mutations include 1 l lC and/or 333F.
14. The carrier of claim 12, further comprising a polylysine motif.
15. The carrier of claim 12, further comprising a targeting domain of heregulin.
16. The carrier of claim 12, wherein the one or more PB mutations comprises a C-terminal deletion.
17. A therapeutic agent comprising the carrier of claim 12 and a therapeutic drug.
18. The therapeutic agent of claim 17, wherein the therapeutic drug is a chemotherapeutic agent.
19. A method of producing a carrier without proliferative activity, comprising:
(a) obtaining a polynucleotide encoding the receptor binding domain of heregulin (Her) and generating mutants in the polynucleotide;
(b) cloning the mutant Her polynucleotides into phage vectors and generating a phage library comprising the phage vectors;
(c) transforming MDA-MB-435 cells with the phage library in the presence of mitotic inhibitors;
(d) fractioning the transformed cell and extracting the membrane fraction of the MDA-MB-435 cells;
(e) harvesting membrane phages from the membrane fraction;
(f) transforming the membrane phages into MDA-MB-435 cells in the presence of mitotic inhibitors;
(g) repeating steps (d), (e), and (f);
(h) monitoring MDA-MB-435 cell proliferation in each round and selecting the membrane phages with the lowest MDA-MB-435 cell proliferation;
(i) obtaining the sequences of the Her polynucleotide mutants in the selected membrane phages; and
(j) producing polypeptides encoded by the Her sequences and penton base gene, wherein the polypeptides are the carrier without proliferative activity.
20. A carrier for delivering therapeutics to the nucleus, comprising a polypeptide encoded by the mutant Her sequences of claim 19.
21. The carrier of claim 20, further comprising a polypeptide encoding penton base (PB) protein and a polylysine motif.
22. The carrier of claim 20, wherein the PB protein is a mutant penton base protein.
23. A therapeutic agent comprising the carrier of claim 19 and a therapeutic drug.
24. The therapeutic agent of claim 23, wherein the therapeutic drug is a chemotherapeutic agent.
PCT/US2013/053493 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein WO2014022811A1 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
MX2015001366A MX366347B (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein.
RU2015105052A RU2015105052A (en) 2012-08-03 2013-08-02 Isolation of drug delivery protein mutants enhancing directed migration
CN201380041215.1A CN104619839A (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
EP13826083.1A EP2880160B1 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
CA2881582A CA2881582A1 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
PL13826083T PL2880160T3 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
US14/419,233 US10036009B2 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
AU2013296218A AU2013296218B2 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
KR1020157005438A KR102115630B1 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
ES13826083T ES2703052T3 (en) 2012-08-03 2013-08-02 Isolation of protein-enhancing drug delivery mutants
BR112015002004A BR112015002004A2 (en) 2012-08-03 2013-08-02 isolation of drug-transporting protein-enhancing traffic mutants
JP2015525636A JP6618360B2 (en) 2012-08-03 2013-08-02 Isolation of transport-enhancing variants of drug delivery proteins
DK13826083.1T DK2880160T3 (en) 2012-08-03 2013-08-02 ISOLATION OF TRANSPORT INCREASING MUTANTS OF PHARMACEUTICAL PROCESSING PROTEIN
IL236955A IL236955A0 (en) 2012-08-03 2015-01-28 Isolating traffic-enhancing mutants of drug delivery protein
ZA2015/00739A ZA201500739B (en) 2012-08-03 2015-02-02 Isolating traffic-enhancing mutants of drug delivery protein
IN1196DEN2015 IN2015DN01196A (en) 2012-08-03 2015-02-13
US16/017,866 US10752893B2 (en) 2012-08-03 2018-06-25 Isolating traffic-enhancing mutants of drug delivery protein
AU2019204815A AU2019204815A1 (en) 2012-08-03 2019-07-04 Isolating Traffic-Enhancing Mutants Of Drug Delivery Protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261679306P 2012-08-03 2012-08-03
US61/679,306 2012-08-03

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/419,233 A-371-Of-International US10036009B2 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein
US16/017,866 Continuation US10752893B2 (en) 2012-08-03 2018-06-25 Isolating traffic-enhancing mutants of drug delivery protein

Publications (1)

Publication Number Publication Date
WO2014022811A1 true WO2014022811A1 (en) 2014-02-06

Family

ID=50028570

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/053493 WO2014022811A1 (en) 2012-08-03 2013-08-02 Isolating traffic-enhancing mutants of drug delivery protein

Country Status (18)

Country Link
US (2) US10036009B2 (en)
EP (1) EP2880160B1 (en)
JP (2) JP6618360B2 (en)
KR (1) KR102115630B1 (en)
CN (1) CN104619839A (en)
AU (2) AU2013296218B2 (en)
BR (1) BR112015002004A2 (en)
CA (1) CA2881582A1 (en)
DK (1) DK2880160T3 (en)
ES (1) ES2703052T3 (en)
IL (1) IL236955A0 (en)
IN (1) IN2015DN01196A (en)
MX (1) MX366347B (en)
PL (1) PL2880160T3 (en)
PT (1) PT2880160T (en)
RU (1) RU2015105052A (en)
WO (1) WO2014022811A1 (en)
ZA (1) ZA201500739B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9850293B2 (en) 2014-04-04 2017-12-26 Cedars-Sinai Medical Center Targeting trastuzumab-resistant HER2+ breast cancer with a HER3-targeting nanoparticle
US10183078B2 (en) 2014-01-17 2019-01-22 Cedars-Sinai Medical Center Receptor targeting constructs and uses thereof
US11795454B2 (en) 2009-01-23 2023-10-24 Cedars-Sinai Medical Center Targeted delivery system

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9078927B2 (en) 2007-07-06 2015-07-14 Cedars-Sinai Medical Center Self-assembling complex for targeting chemical agents to cells
BR112015002004A2 (en) * 2012-08-03 2017-07-04 Cedars Sinai Medical Center isolation of drug-transporting protein-enhancing traffic mutants
AU2014262653B2 (en) 2013-05-08 2018-07-26 Cedars-Sinai Medical Center Targeting corroles for tumor toxicity and MRI

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
WO1998035036A1 (en) 1997-02-10 1998-08-13 Genentech, Inc. Heregulin variants
WO2002024934A1 (en) * 2000-09-22 2002-03-28 Consorzio Interuniversitario Per Le Biotecnologie Chimeric vectors and their use for heterologous genes transfer
EP1266963A1 (en) * 2001-06-15 2002-12-18 Crucell Holland B.V. Chimaeric phages
WO2009009441A2 (en) 2007-07-06 2009-01-15 Cedars-Sinai Medical Center Self-assembling complex for targeting chemical agents to cells
US20120004181A1 (en) 2009-01-23 2012-01-05 Cedars-Sinai Medical Center Targeted delivery system

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5439829A (en) 1991-01-30 1995-08-08 Eli Lilly And Company Immobilization of biologically active molecules by changing the Oxidation state of a chelated transition metal ion
US6287792B1 (en) 1991-06-17 2001-09-11 The Regents Of The University Of California Drug delivery of antisense oligonucleotides and peptides to tissues in vivo and to cells using avidin-biotin technology
GB9223084D0 (en) 1992-11-04 1992-12-16 Imp Cancer Res Tech Compounds to target cells
FI943242A (en) 1993-07-15 1995-01-16 Jcr Pharmaceutical Co Ltd A remedy for kidney and liver diseases
US6465253B1 (en) * 1994-09-08 2002-10-15 Genvec, Inc. Vectors and methods for gene transfer to cells
US6066322A (en) 1995-03-03 2000-05-23 Millennium Pharmaceuticals, Inc. Methods for the treatment of immune disorders
EA199800475A1 (en) * 1995-11-28 1998-12-24 Дженвек, Инк. VECTORS AND METHODS FOR TRANSFERING GENES IN CELLS
GB9709421D0 (en) 1997-05-10 1997-07-02 Zeneca Ltd Chemical compounds
IL140855A0 (en) 1998-08-18 2002-02-10 Univ California Preventing airway mucus production by administration of egf-r antagonists
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
CA2391534A1 (en) 1999-11-15 2001-05-25 Drug Innovation & Design, Inc. Selective cellular targeting: multifunctional delivery vehicles
CA2415319A1 (en) 2000-06-16 2001-12-20 Martek Biosciences Corporation Recombinant phycobiliprotein and phycobiliprotein linker fusion proteins and uses therefore
WO2002062823A2 (en) 2001-02-02 2002-08-15 Yale University Peptides for facilitating composite receptor expression and translocation of macromolecules
SE0201863D0 (en) 2002-06-18 2002-06-18 Cepep Ab Cell penetrating peptides
US7329638B2 (en) 2003-04-30 2008-02-12 The Regents Of The University Of Michigan Drug delivery compositions
US7205387B2 (en) 2003-08-28 2007-04-17 Agency For Science, Technology And Research Recombinant polypeptide useful for neurotrophin receptor mediated gene delivery and as neurotrophin agonist
US20060014712A1 (en) 2004-05-30 2006-01-19 Cemines, Inc. Controlled delivery of therapeutic compounds
US8057821B2 (en) 2004-11-03 2011-11-15 Egen, Inc. Biodegradable cross-linked cationic multi-block copolymers for gene delivery and methods of making thereof
US20110052697A1 (en) 2006-05-17 2011-03-03 Gwangju Institute Of Science & Technology Aptamer-Directed Drug Delivery
JP4479755B2 (en) 2007-07-03 2010-06-09 ソニー株式会社 ORGANIC ELECTROLUMINESCENT ELEMENT AND ORGANIC ELECTROLUMINESCENT DISPLAY DEVICE
PT2220241T (en) * 2007-11-28 2016-12-26 Univ Pennsylvania Adenovirus comprising a simian e adenovirus sadv-39 capsid hexon protein and uses thereof
WO2013052859A2 (en) 2011-10-05 2013-04-11 Genvec, Inc. Adenoviral vector-based respiratory syncytial virus (rsv) vaccine
BR112015002004A2 (en) 2012-08-03 2017-07-04 Cedars Sinai Medical Center isolation of drug-transporting protein-enhancing traffic mutants
AU2014262653B2 (en) 2013-05-08 2018-07-26 Cedars-Sinai Medical Center Targeting corroles for tumor toxicity and MRI
KR102434541B1 (en) 2014-01-17 2022-08-19 세다르스-신나이 메디칼 센터 Receptor targeting constructs and uses thereof
AU2015240460A1 (en) 2014-04-04 2016-09-15 Cedars-Sinai Medical Center Targeting trastuzumab-resistant HER2+breast cancer with a HER3-targeting nanoparticle
BR112018074304A2 (en) * 2016-05-27 2019-10-01 Cedars Sinai Medical Center nanoparticle compositions, kit and method for treating an individual with drug resistant cancer

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5559099A (en) * 1994-09-08 1996-09-24 Genvec, Inc. Penton base protein and methods of using same
WO1998035036A1 (en) 1997-02-10 1998-08-13 Genentech, Inc. Heregulin variants
WO2002024934A1 (en) * 2000-09-22 2002-03-28 Consorzio Interuniversitario Per Le Biotecnologie Chimeric vectors and their use for heterologous genes transfer
EP1266963A1 (en) * 2001-06-15 2002-12-18 Crucell Holland B.V. Chimaeric phages
WO2009009441A2 (en) 2007-07-06 2009-01-15 Cedars-Sinai Medical Center Self-assembling complex for targeting chemical agents to cells
US20120004181A1 (en) 2009-01-23 2012-01-05 Cedars-Sinai Medical Center Targeted delivery system

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
GENE THERAPY, vol. 13, 2006, pages 821 - 836
See also references of EP2880160A4

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11795454B2 (en) 2009-01-23 2023-10-24 Cedars-Sinai Medical Center Targeted delivery system
US10183078B2 (en) 2014-01-17 2019-01-22 Cedars-Sinai Medical Center Receptor targeting constructs and uses thereof
US11369687B2 (en) 2014-01-17 2022-06-28 Cedars-Sinai Medical Center Receptor targeting constructs and uses thereof
US9850293B2 (en) 2014-04-04 2017-12-26 Cedars-Sinai Medical Center Targeting trastuzumab-resistant HER2+ breast cancer with a HER3-targeting nanoparticle

Also Published As

Publication number Publication date
US20180298376A1 (en) 2018-10-18
EP2880160A1 (en) 2015-06-10
ES2703052T3 (en) 2019-03-06
US10752893B2 (en) 2020-08-25
JP2015527343A (en) 2015-09-17
JP6618360B2 (en) 2019-12-11
BR112015002004A2 (en) 2017-07-04
EP2880160A4 (en) 2016-04-20
AU2013296218B2 (en) 2019-07-18
RU2015105052A (en) 2016-09-27
DK2880160T3 (en) 2019-01-14
PT2880160T (en) 2018-12-17
IL236955A0 (en) 2015-03-31
CN104619839A (en) 2015-05-13
EP2880160B1 (en) 2018-09-19
KR20150038543A (en) 2015-04-08
IN2015DN01196A (en) 2015-06-26
US10036009B2 (en) 2018-07-31
PL2880160T3 (en) 2019-03-29
ZA201500739B (en) 2018-07-25
KR102115630B1 (en) 2020-05-27
MX366347B (en) 2019-07-05
CA2881582A1 (en) 2014-02-06
AU2013296218A1 (en) 2015-02-19
JP2020002138A (en) 2020-01-09
US20150240231A1 (en) 2015-08-27
MX2015001366A (en) 2015-08-13
AU2019204815A1 (en) 2019-07-25

Similar Documents

Publication Publication Date Title
US10752893B2 (en) Isolating traffic-enhancing mutants of drug delivery protein
WO2017215619A1 (en) Fusion protein producing point mutation in cell, and preparation and use thereof
US20150010475A1 (en) Crlf-2 binding peptides, protocells and viral-like particles useful in the treatment of cancer, including acute lymphoblastic leukemia (all)
CN113347993A (en) Mutant vaccinia virus and uses thereof
US8729038B2 (en) Down regulation of the gene expression by means of nucleic acid-loaded virus-like particles
US9879254B2 (en) Targeting RNAs to microvesicles
JPWO2013018778A1 (en) Immunotherapeutic cells comprising a modified nucleic acid construct encoding a Wilms tumor gene product or fragment thereof, a method for producing the cell, and the nucleic acid construct
US20240084330A1 (en) Compositions and methods for delivering cargo to a target cell
KR20180092989A (en) Transposon systems, kits containing them, and uses thereof
KR20200006111A (en) Compositions and methods for loading chemical and biological agents / molecules into extracellular vesicles
CN109563490A (en) Herpesviral with modified Glycoprotein B
Legendre et al. Construction and exploitation in model experiments of functional selection of a landscape library expressed from a phagemid
Liu et al. Systematic evolution of ligands by exosome enrichment: A proof‐of‐concept study for exosome‐based targeting peptide screening
TWI515203B (en) Nuclear localization signal peptides derived from vp2 protein of chicken anemia virus and uses of said peptides
JP2024503728A (en) How to identify peptide therapeutics to treat various conditions
WO2023158486A2 (en) Cell-type specific targeting contractile injection system
US8502015B1 (en) Method of inducing cancer
WO2024006988A2 (en) Engineered delivery vesicles and uses thereof
CA2463960A1 (en) Transient immortalization
CN113913459A (en) Method for preparing high-purity chimeric antigen receptor modified T cell by non-viral method and application thereof
Kritz Peptides from phage display libraries for targeted gene delivery via the p75NTR neurotrophic receptor

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13826083

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 236955

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/001366

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2015525636

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14419233

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2881582

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2013296218

Country of ref document: AU

Date of ref document: 20130802

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2013826083

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157005438

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015105052

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015002004

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112015002004

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150128