CA2512144A1 - Solid surface for biomolecule delivery and high-throughput assay - Google Patents

Solid surface for biomolecule delivery and high-throughput assay Download PDF

Info

Publication number
CA2512144A1
CA2512144A1 CA002512144A CA2512144A CA2512144A1 CA 2512144 A1 CA2512144 A1 CA 2512144A1 CA 002512144 A CA002512144 A CA 002512144A CA 2512144 A CA2512144 A CA 2512144A CA 2512144 A1 CA2512144 A1 CA 2512144A1
Authority
CA
Canada
Prior art keywords
cells
transfection
lipid
biomolecule
cell
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.)
Granted
Application number
CA002512144A
Other languages
French (fr)
Other versions
CA2512144C (en
Inventor
Lei Yu
Kenji Matsumoto
Shouping Ji
Fusheng Du
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.)
Nitto Denko Corp
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2512144A1 publication Critical patent/CA2512144A1/en
Application granted granted Critical
Publication of CA2512144C publication Critical patent/CA2512144C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • 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
    • A61K47/644Transferrin, e.g. a lactoferrin or ovotransferrin
    • 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
    • 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
    • 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

Abstract

The present invention is related to a method for introducing biomolecules, such as nucleic acids, into cells by culturing cells on a solid surface which is coated with a transfection reagents and biomolecules.

Description

SOLID SURFACE FOR EIOMOLECULE DELIVERY AND
HIGH-THROUGHPUT ASSAY
Field of the invention The present invention is related to a method for introducing biomolecules, such as nucleic acids, into cells by culturing cells on a solid surface which is coated with a transfection reagents and biomolecules for routine transfection assays and high-throughput transfection assays. The present invention is also related to method of use and method of making the transfectable surface for such assays.
Baclcgrround of the Invention Gene transfection methods can be used to introduce nucleic acids into cells and are useful in studying gene regulation and function. High throughput assays that can be used to screen large sets of DNAs to identify those encoding products with properties of interest are particularly useful. Gene transfection is the delivery and introduction of biologically functional nucleic acids into a cell, such as a eukaryotic cell, in such a way that the nucleic acid retains its function within the cell. Gene transfection is widely applied in studies related to gene regulation, gene function, molecular therapy, signal transduction, drug screening, and gene therapy studies. As the cloning and cataloging of genes from higher organisms continues, researchers seelc to discover the function of the genes and to identify gene products with desired properties. This growing collection of gene sequences requires the development of systematic and high-throughput approaches to characterizing gene products and analyzing gene function, as well as other areas of research in cell and molecular biology.
Both viral and non-viral gene carriers have been used in gene delivery. Viral vectors have been shown to have higher transfection efficiency than non-viral carriers, but the safety of viral vectors hampers its applicability (Verma LM and Somia N.
Nature 389 (1997), pp. 239-242; Marhsall E. Scierace 286 (2000), pp. 2244-2245). Although non-viral transfection systems have not exhibited the efficiency of viral vectors, they have received significant attention, because of their theoretical safety when compared to viral vectors. In addition, viral vector preparation is a complicated and expensive process, which limits the application of viral vectors ira vitro. The preparation of non-viral carriers is simpler and more cost effective in comparison to preparation of viral carriers, malting synthetic gene carriers desirable as transfection reagents in irz vitYO studies.
Most non-viral vectors mimic important features of viral cell entry in order to overcome cellular barriers, which are meant to prevent infiltration by foreign genetic S material. Non-viral gene vectors, based on a gene carrier baclcbone, can be classified as a) lipoplexes, b) polyplexes, and c) lipopolyplexes. Lipoplexes are assemblies of nucleic acids with a lipidic component, which is usually cationic. Gene transfer by lipoplexes is called lipofection. Polyplexes are complexes of nucleic acids with cationic polymer.
Lipopolyplexes comprise both a lipid and a polymer component. Often such DNA
complexes are further modified to contain a cell targeting or an intracellular targeting moiety and/or a membrane-destabilizing component, for example, a viral protein or peptide or a membrane-disruptive synthetic peptide. Recently, bacteria and phages have also been described as shuttles for the transfer of nucleic acids into cells.
Most non-viral transfection reagents are synthetic cationic molecules and have been 1 S reported to "coat" the nucleic acid by 'interaction of the cationic sites on the canon and anionic sites on the nucleic acid. The positively-charged DNA-cationic molecule complex interacts with the negatively charged cell membrane to facilitate the passage of the DNA
through the cell membrane by non-specific endocytosis. (Schofield, Brit.
Microencapsulated. Bull, Sl(1):S6-71 (1995)). In most conventional gene transfection protocols, the cells are seeded on cell culture devices 16 to 24 hours before transfection.
The transfection reagent (such as a cationic polymer carrier) and DNA were usually prepared in separate tubes, and each respective solution was diluted in medium (containing no fetal bovine serum or antibiotics). The solutions were then mixed by carefully and slowing adding one solution to the other while continuously vortexing the mixture. The 2S mixture was incubated at room temperature for 1S-4S minutes to allow the transfection reagent-DNA complexes formation to remove the residues of serum and antibiotics. Prior to transfection, the cell culture medium was removed and the cells were washed with buffer. The solution containing DNA-transfection reagent complexes was added to the cells, and the cells were incubated for about 3-4 hours. The medium containing transfection reagent would then be replaced with fresh medium. The cells would finally be analyzed at one or more specific time point(s). This is obviously a time consuming procedure, particularly when the number of samples to be transfected is very large.
_2_ Several major problems exist in conventional transfectioy procedures. First, conventional procedures are time-consuming, particularly when there are many cell or gene samples to be used in transfection experiments. Also, the results derived from common transfection procedures are difficult to reproduce, due to the number of steps required. For instance, in producing the DNA-transfection reagent complex formation of the complex is influenced by concentration and volume of nucleic acid and reagents, pH, temperature, type of buffers) used, length and speed of vortexing, incubation time, and other factors.
Although the same reagents and procedure may be followed, different results may be obtained. Results derived from mufti-step procedures are often influenced by human or mechanical error or other variations at each step. In addition, refreshing the cell culture medium following transfection disturbs the cells and may cause them to detach from the surface on which they are cultured, thus leading to variation and unpredictability in the final results. Due to all the factors noted, conventional transfection methods require a highly skilled individual to perform the transfection experiment or assay.
Researchers require an easier and more cost effective method of transfecting cells, and a high-throughput method of transfecting cells is needed in order to transfect large sample numbers efficiently.
Summary of theTnvention A method for introducing biomolecules into eukaryotic cells is provided comprising (a) coating a solid surface with a biomolecule delivery reagent, (b) adding the biomolecules to be introduced into the eulcaiyotic cells onto the solid surface, (c) seeding cells on the solid surface at a sufficient density and under appropriate conditions for introduction of the biomolecules into the eukaryotic cells. According to embodiments of the invention the surface may be selected from the group consisting of flasks, dishes, mufti-well plates, glass slides, and implanted devices. The biomolecule delivery reagent or transfection reagent may be selected from the group consisting of polymers, lipids, lipid-polymers and/or their combinations and/or their derivatives containing a cell-targeting or an intracellular targeting moiety and/or a membrane-destabilizing component and one or more delivery eWancers.
According to embodiments of the invention the biomolecule delivery reagent can be affixed on the surface by evenly spreading the reagent on the surface or spotting said biomolecule delivery reagent in discrete areas of the surface. The solid surface coated with a biomolecule delivery reagent may further comprise a matrix reagent selected from the group consisting of proteins, peptides, polysaccharides, and polymers. The protein may be selected from gelatin, bovine serum albumin, and extracellular matrix components such as, but not limited to collagen, laminin, and fibronectin. The polymer may be selected from hydrogels, biodegradable polymers, and biocompatible materials.
According to embodiments of the invention a solid surface is coated with a biomolecule delivery reagent which further comprises a cell culture reagent selected from the group consisting of cytoreductive reagents, cell binding/attaching reagents, cell growing reagents, cell stimulating reagents, and cell inhibiting reagents.
Biomolecules may be selected from nucleotides, proteins, peptides, sugars, polysaccharides, and organic compounds. Preferably the biomolecules are selected from DNA, RNA, and DNA/RNA hybrids. The nucleotides may be circular (plasmid), linear, or single strand oligodeoxynucleotide. RNA may be single stranded (ribozyrne) or double stranded (siRNA).
Solid surfaces used according to methods described herein may be selected from, but not limited to, a slide or a multi-well plate.
Eul~aryotic cells used according to embodiments of the invention may be, but are not limited to, mammalian cells. The mammalian cells may be dividing cells or non-dividing cells. The mammalian cells may be transformed cells or primary cells.
The mammalian cells may be somatic cells or stem cells. The eul~aryotic cells may be plant, yeast, or insect cells.
A method of high throughput drug screening assay is provided comprising (a) affixing a delivery reagent to a solid surface, (b) affixing biomolecules to be introduced into eulcaryotic cells to said delivery reagent, (c) seeding cells on the surface bearing delivery reagent and biomolecules with sufficient density and under appropriate conditions for introduction of the biomolecules into the eulcaryotic cells, and (d) detecting eulcaryotic cells into which the biomolecule has been delivered.
The biomolecules may be selected from, but not limited to, nucleotides, proteins, peptides, sugars, polysaccharides, and organic compounds. The nucleotides may be selected from, but IlOt limited to, DNA, RNA, and DNA/RNA hybrid. The DNA may be circular (plasmid), linear, or single stranded oligodeoxynucleotide (ODN). The RNA may be single stranded (ribozyme) or double stranded (siRNA).

The eulcaryotic cells are preferably mammalian cells. The mammalian cells may be dividing cells or non-dividing cells and the cells may be transformed cells or primary cells.
The mammalian cells may be somatic cells or stem cells. The eulcaryotic cells may be selected from, but not limited to plant, bacterial, and insect cells.
Detecting cells into which the biomolecule has been delivered may be performed by detecting the biomolecule itself, its product, its target molecule, the products catalyzed or products regulated by the biomolecule.
Brief Description of the Drawings Figure 1 is a schematic of a transfection assay using transfectable cell culture devices or slides.
Figure 2 illustrates the effect of using a transfectable surface coated with various cationic polymers-gelatin transfection mixture on GFP reporter gene transfection in 923 cells in 96-well plate cell culture device system. The amount of Linear PEI, and NDT-CP-1 were showed in the figure. The amounts of Superfect were 15, 7.5 and 3.75~g/well, respectively. The ranges of GFP gene transfection efficiency are approximately 30-35%, and NDT-CP-1 showed the highest efficiency.
Figure 3 illustrates the effect of using a transfectable surface coated with various cationic polymers-gelatin transfection mixture on luciferase reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amounts of Linear PEI, NDT-CP-B-1 and NDT-CP-1 were 8 ,4, 2, and l~.g/ well, respectively. The amomts of Superfect were 15, 7.5 and 3.75~,g/well. The luciferase activities of all samples were higher than 5 x 10~ RLU/mg protein.
Figure 4 illustrates the effect of using a transfectable surface coated with various cationic lipid-polymer-gelatin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amount of lipid-polymer was shown as 8, 4, and2 ~,g/well. The GFP transfection efficiency could reach 20-25% in tested lipid-polymer transfection reagents.
Figure 5 illustrates the effect of using a transfectable surface coated with various cationic lipid-polymer-gelatin transfection mixture on luciferase reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The luciferase activity of NDT-LP-2 could reach higher than 10~ RLU/mg protein in a lipid-polymers-gelatin transfection mixture system.
Figure 6 illustrates the effect of using a transfectable surface coated with cationic lipid-gelatin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The transfection efficiency mediated by lipofectamine2000-gelatin transfection mixture in a 96-well-plate system could reach up to 30%.
Figure 7 illustrates the effect of using a transfectable surface coated with cationic lipid-gelatin transfection mixture on luciferase reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The luciferase activity of lipofectamine 2000 could reach up to near 10' RLU/mg proteins in lipid-gelatin transfection mixture system.
Figure 8 illustrates the effect of using a transfectable surface coated with cationic polymer-laminin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amounts of NDT-CP-1 and NDT-CP-B-1 are shown in the figure. The amounts of Superfect were 15 and 7.5 ~gl well respectively. The GFP transfection efficiency mediated by cationic polymer-laminin system could reach up to SO%.
Figure 9 illustrates the effect of using a transfectable surface coated with cationic polymer-laminin transfection mixture on luciferase reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amount of NDT-CP-1 and NDT-CP-B-1 are shown in the figure. The amounts of Superfect were 15 and 7.5 and 3.75 yg/
well respectively. The luciferase activity of cationic polymer-laminin system could reach up to 1 O8 RLU/mg protein.
Figure 10 illustrates the effect of using a trarisfectable surface coated with cationic lipid-polymer-laminin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amount of lipid-polymer was shown as 8, 4, and 2~.g/well. The GFP transfection efficiency mediated by lipid-polymer-laminin transfection mixture system could reach up to 45%.
Figure 11 illustrates the effect of using a transfectable surface coated with cationic lipid-polymer-laminin transfection mixture on luciferase reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amount of lipid-polyner is shown in the figure. The luciferase activity of lipid polymer-laminin transfection mixture system could reach up to 8 x 10' RLU/mg of protein.
Figure 12 illustrates the effect of using a transfectable surface coated with cationic lipid-laminin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The transfection efficiency mediated by lipofectamine-laminin transfection mixture system could reach up to 45%.
Figltre 13 illustrates the effect of using a transfectable surface coated with cationic lipid-lazninin transfection mixture on luciferase reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The luciferase activity of lipofectamine 2000-laminin tr ansfection mixture system could reach up to 1.5 x 10~ RLU/mg protein.
Figure 14 illustrates the effect of using a transfectable surface coated with cationic polymer-collagen transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amounts of NDT-CP-1 and NDT-CP-B-1 is shown in the figure. The amounts of Superfect were 15 and 7.5 ~g/well respectively. The GFP transfection efficiency mediated by cationic polymer-collagen transfection mixture system could reach up to 40%.
Figure 15 illustrates the effect of using a transfectable surface coated with cationic lipid-polymer-collagen transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amount of lipid-polymer is shown as 8, 4, and tug/ well. The transfection efficiency mediated by cationic lipid-polymer-collagen transfection mixture system could reach up to 35%.
Figure 16 illustrates the effect of using a transfectable surface coated with cationic lipid-collagen transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The transfection efficiency mediated by cationic lipid-collagen transfection mixture system could reach up to 35%, similar to those mediated by lipofectamine 2000-gelatin transfection mixture system.
Figure 17 illustrates the effect of using a transfectable surface coated with cationic polymer-gelatin-laminin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amounts of NDT-CP-1 and NDT-are shown in the figure. The amounts of Superfect were 15 and 7.5 ~,glwell respectively.
The GFP transfection efficiency mediated by the cationic polymer-gelatin-laminin transfection mixture system could reach up to 42%.
_7_ Figure 18 illustrates the effect of using a transfectable surface coated with cationic lipid-polymer-gelatin-laminin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The amount of lipid-polymer is shown as 8, 4, and 2ug/well. The GFP transfection efficiency mediated by the cationic lipid-polymer-gelatin-laminin transfection mixture system could reach up to 40%.
Figure 19 illustrates the effect of using a transfectable surface coated with cationic lipid-gelatin-laminin transfection mixture on GFP reporter gene transfection in 923 cells in a 96-well plate cell culture device system. The transfection efficiency mediated by cationic lipid-gelatin-laminin transfection mixture system could reach up to 30%.
Figure 20 illustrates a GFP reporter gene transfection assay in 923 cells with a transfectable glass slide system spotted with cationic polymer (NDT-CP-1)-gelatin or cationic lipid (lipofectamine2000)-gelatin transfection mixture. The transfectable slide was submerged in a GFP plasmid solution. Although the whole slide was covered with GFP
plasmid solution, only the cells that are on the spot where the transfection mixtures were applied exhibited a GFP signal. This indicates that the transfection reagents were well affixed on the glass slide without diffusion. These results indicated that the current technology would be useful in transfection array applications, which could screen thousands of the target genes or gene medicine in cell based transfection assays for genomic function studies or gene medicine development (antisense ODN or siRNA).
Figure 21 illustrates a GFP reporter gene transfection assay in 923 cells with a transfectable glass slide system coated with cationic polymer (NDT-CP-1)-gelatin or cationic lipid (lipofectamine2000)-gelatin transfection mixture. The transfectable slide was loaded with GFP plasmid DNA by spotting 1-4 ~.l GFP plasmid (20~.g/ml) solution and allowing the slide to air dry. The glass slide was then put on the bottom of a six-well plate, followed by seeding 293 cells. The GFP signal was analyzed by fluorescent microscope.
Only the areas spotted with GFP plasmid DNA showed green fluorescent signal, which indicated that the plasmid DNA was well affixed on the glass surface of spotted areas with transfection mixtures. The current technology is useful in transfection array applications, which could screen thousands of the target genes or gene medicine in cell based transfection assays for genomic function studies (cDNA library screening) or gene medicine development (antisense ODN or siRNA).
-g_ Figure 22 illustrates a GFP reporter gene transfection assay in 923 cells with a transfectabhe glass slide system spotted with cationic polymer (NDT-CP-1)-laminin or cationic lipid (lipofectamine2000)-laminin transfection mixture. The transfectable slide was submerged in GFP phasmid solution. Although the whole slide was covered with GFP
plasmid solution, only the cells that are on the spot where transfection mixtures were applied exhibited a GFP signal. Thus indicates that the transfection reagents were well affixed on the glass slide without diffusion. These results indicated that the current technology is useful in transfection array applications, which could screen thousands of the target genes or gene medicine in cell based transfection assays for genomic function studies or gene medicine development (antisense ODN or siRNA).
Figure 23 illustrates a GFP reporter gene transfection assay in 923 cells with a transfectable glass slide system coated with cationic polymer (NDT-CP-1)-laminin or cationic lipid (lipofectamine2000)-laminin transfection mixture. The transfectable slide was loaded with GFP plasmid DNA by spotting 1-4 ~.l GFP plasmid (20~,g/ml) solution and allowing the slide to air dry. The glass slide was then put on the bottom of a six-well plate, followed by seeding 293 cells. The GFP signal was analyzed by fluorescent microscope.
Only the areas spotted with GFP plasmid DNA showed green fluorescent signal, which indicates that the plasmid DNA was well affixed on the glass surface in the areas spotted with transfection mixtures. The current technology is useful in transfection array applications, which could screen thousands of the target genes or gene medicine in cell based transfection assays for genomic function studies (cDNA library screening) or gene medicine development (antisense ODN or siRNA).
Figure 24 illustrates the effect of using transfection reagent-laminin mixtures on antisense ODN transfection to Hela 705 Luc cells in a 96-well cell culture device. The results showed that transfectable surface composed of cationic polymer-haminin transfection mixture, lipid-polymer-laminin mixture, or lipid-laminin transfection mixture systems showed significant blocl~ing of the target RNA, which suggested that not only plasmid, but also oligonucleotide could be successfully delivered into mammalian cells by this strategy.
Figure 25 illustrates the effect of using transfection reagent-haminin mixtures on siRNA delivery to 293 cells in a 96-well cell culture device. In comparison to the non-siRNA control group, transfectable surfaces composed of cationic pohyner-laminin transfection mixture, lipid-polymer-laminin mixture, or lipid-laminin transfection mixture systems showed significant bloclcing of the target RNA, which suggested that not only plasmid, but also siRNA could be successfully delivered into mammalian cells by this strategy.
Figure 26 illustrates typical results of Tat peptide signal in HeLa cells. In Figure 26A, Tat was coated in solid surface with gelatin matrix. Figure 26B shows the non-Tat control. About 60-80% cells showed FITC signal in the Tat-coated group. This indicates that not only nucleic acid, but also peptides, can be delivered into cells according to embodiments of the present invention.
Figure 27 illustrates the effect of a targeting molecule (transferring) on cationic polymer (PLL)-laminin transfection mixture mediated gene transfer in HepG2 cells with transfectable surface technology in a 96-well cell culture device system. The results indicate that introduction of targeting molecules (transferring) in PLL-laminin based transfectable surface systems could significantly enhance transfection efficiency.
Figure 28 illustrates the effect of membrane disturbing peptide (VSVG peptide) on cationic polymer (PLL)-laminin transfection mixture mediated gene transfer in 293 cells with transfectable surface technology in a 96-well cell culture device system.
The results indicate that introduction of membrane disturbing peptide (VSVG peptide) in PLL-laminin based transfectable surface systems could significantly enhance transfection efficiency.
Figure 29 illustrate the effect of using transfection reagent-laminin mixtures in GFP
reporter gene delivery to HW-EC cells in a 96-well cell culture device. The GFP
transfection efficiency of NDT-CP-1 was about 20%, and the efficiency of Superfect was about 15%. The lipofectamine showed very low transfection efficiency (< 1.0%).
It indicated that not only transformed cell lines, such as 293, HeLa, HepG2 could be transfected, but also primary cells could be transfected by cationic polymer-laminin transfection mixture systems.
Figure 30 illustrates the effect of transfection reagent-laminin mixtures on luciferase reporter gene delivery to HUV-EC cells in a 96-well cell culture device. The transfection efficiency of NDT-CP-1 and Superfect transfection efficiency were 1.26x l0~and 8.26x 10~
RLU/mg protein respectively. Lipofectamine 2000 showed lower efficiency compared to the cationic polyner-laminin mixture system. This further confirmed that primary cells could be transfected with different gene according to embodiments of the present invention.

Figure 31 illustrates the cell survival fraction of 293 cells after transfection in a 96 well plate coated with transfection reagent-gelatin. All samples showed a high survival fraction (>65%). It indicates that the cytotoxicity of transfection reagents used according to methods described herein are acceptable.
Figure 32 illustrates the effect of introduction of cytoreductive reagents (glutamine) in a transfection reagents-laminin based transfectable surface on cytotoxicity improvement in 293 cells. The "Non" indicates samples without glutamine, and "G" indicates samples with glutamine. The results showed that glutamine (cytoreductive reagent) significantly improved transfection cytotoxicity compared to that without glutamine group, which indicated that cytoreductive reagents are excellent candidates for reducing cytotoxicity caused by transfection reagents and transfection procedures.
Figure 33 illustrates the effect of introduction of cell attaching reagents and cell stimulating reagent (laminin) in a transfection reagents-gelatin based transfectable surface on cytotoxicity improvement to 293 cells. "L" indicates samples with laminin and "Non"
indicates samples without laminin. The results indicate that the cell attaching reagent, such as laminin, significantly improved cytotoxicity caused by transfection reagents and transfection procedures in a transfectable surface technology system.
Figure 34 illustrates an evaluation of shelf life of transfectable surfaces in stability studies. The results showed that there are no significant differences in transfection performances in flash-made transfectable sl~rfaces or that with the treatment at 37°C for 9 days, which indicated that the shelf life of transfectable surface could be 1.5 years when stored at 4°C.
Figure 35 is a table illustrating the structures of NDT synthesized polymers and lipid-polymers.
Detailed Description of the Preferred Embodiment A novel transfection device and method are described herein which is simple, convenient, and efficient compared to conventional transfection assays. A
transfection device is made according to methods described herein by affixing a transfection reagent on the solid surface of a cell culture device. By using this device, researchers need only add a nucleic acid or other biomolecule carrier system to the surface of the cell culture device.
There is no need to pre-mix the DNA or biomolecule with a transfection reagent. This removes a l~ey timing-consuming step, which is required by conventional transfection procedures. Scientists only require approximately 40 minutes to complete the entire transfection process for 10 samples, compared to 2 to 5 hours or more required by current methods. This is particularly favorable for high throughput transfection assays, in which hundreds of samples will be tested at a time.
As compared to conventional transfection, there are several advantages to the new method described herein. It provides a transfection device that is very easy to store, and it provides a simple method for biomolecule delivery or gene transfection in which no biomaterial/transfection reagent mixing step is required. The transfection procedure described herein can be finished in a short period of time, for instance approximately 40 minutes, and it provides a high throughput method for transfection or drug delivery in which large numbers of samples may be transfected at a time.
A novel method and device for gene delivery are described herein, which overcome the common problems encountered in conventional transfection assays described above.
Transfection reagents are simply coated onto the surface of a cell culture device, which can be easily commercialized and mass-produced. Customers, researchers for instance, need only add a biomolecule, such as a nucleic acid of interest, directly to the surface of a cell culture device in order to prepare the device prior to transfection. Cells are then seeded on the surface of the cell culture device and incubated, without changing the medium, and the cells are analyzed. Changing medium during transfection procedure is unnecessary. The methods described herein dramatically reduce the rislc of error, by reducing the number of steps involved, thus increasing consistency and accuracy of the system.
According to the methods described herein, transfection reagents were affixed on the surface of a slide, multi-well plate, or other surface to form a transfection device. By using this device, people need only add DNA or other biomolecule to the surface and allow the transfection reagent to form a complex with the DNA or biomolecule. This reaction occurs in approximately 30 minutes, then cells are seeded on the surface and incubated in suitable condition for introduction of the biomoloecule(s) into the cells.
Any suitable surface that can be used to affix the nucleic acid/biomolecule-containing mixture to its surface can be used. For example, the surface can be glass, plastics (such as polytetrafluoroethylene, polyvinylidenedifluoride, polystyrene, polycarbonate, polypropylene), silicon, metal, (such as gold), membranes (such as nitrocellulose, methylcellulose, PTFE or cellulose), paper, biomaterials (such as protein, gelatin, agar), tissues (such as slcin, endothelial tissue, bone, cartilage), minerals (such as hydroxylapatite, graphite). According to preferred embodiments the surfaces may be slides (glass or poly-L-lysine coated slides) or wells of a mufti-well plate.
For slides, such as a glass slide coated with poly-L-lysine (e.g., Sigma, Inc.), the transfection reagents are fixed on the surface and dried, and then a nucleic acid of interest or a nucleic acid to be introduced into cells, a protein, peptide, or small molecule drug is introduced. The slide is incubated at room temperature for 30 minutes to form biomolecule/transfection reagent complexes on the surface of the transfection device. The biomolecule/transfection reagent complexes create a medium for use in high throughput microarrays, which can be used to study hundreds to thousands of nucleic acids, proteins, peptides and other small molecular drugs at same time. In an alternative embodiment, the transfection reagents or drug delivery reagents can be affixed on the surface of the transfection device in discrete, defined regions to form a microarray of transfection reagents or drug delivery reagents. In this embodiment, molecules, such as nucleic acids, which are to be introduced into cells, are spread on the surface of the transfection device along with a transfection or delivery reagent. This method can be used in screening transfection reagents or other delivery reagents from thousands of compounds. The results of such a screening method can be examined through computer analysis.
In another embodiment of the invention one of more well of a mufti-well plate may be coated with a transfection or drug delivery reagent. Plates commonly used in transfection and drug screening are 96-well and 384-well plates. The transfection or biomolecule delivery reagent can be evenly applied to the bottom of plate.
Hundreds of nucleic acids, proteins, peptides or other biomolecules are then added into the well~(s) by, for instance, a multichannel pipette or automated machine. Results of transfection are then determined by using a microplate reader. This is a very convenient method of analyzing the transfected cells, because microplate readers are commonly used in most biomedical laboratories. The mufti-well plate coated with transfection or biomolecule delivery reagent can be widely used in most laboratories to study gene regulation, gene function, molecular therapy, and signal transduction, as well as drug screening. Also, if different lcinds of transfection reagents are coated on the different wells of mufti-well plates, the plates can be used to screen many transfection or delivery reagents relatively efficiently.
Recently, 1,536 and 3,456 well plates have been developed, which may also be used according to the methods described herein.
The transfection reagent or delivery reagent are preferably cationic compounds which can introduce biomolecules, such as nucleic acids, proteins, peptides, sugars, polysaccharides, organic compounds, and other biomolecules into cells.
Preferred embodiments use cationic oligomers, such as low molecular weight polyethyleneimine (PEI), low molecular weight poly(L-lysine) (PLL), low molecular weight chitosan, or low molecular weight dendrimers. According to their modular composition, reagents can be classified as: lipids, polymers, lipid-polymers and/or their combinations and/or their derivatives, which contain a cell-targeting or an intracellular-targeting moiety and/or a membrane-destabilizing component, as well as delivery enhancers.
According to an one embodiment, the delivery reagent can be mixed with a matrix, such as proteins, peptides, polysaccharides, or other polymers. The protein can be gelatin, collagen, bovine serum albumin or any other protein that can be used in affixing proteins to a surface. The polymers can be hydrogels, copolymers, non-degradable or biodegradable polymers and biocompatible materials. The polysaccharide can be any compound that can form a membrane and coat the delivery reagent, such chitosan. Other reagents, such as cytotoxicity reductive reagents, cell binding reagents, cell growing reagents, cell stimulating reagents or cell inhibiting reagents and the compounds for culturing specific cells, can be also affixed to the transfection device along with the transfection or delivery reagent.
According to another embodiment, a gelatin-transfection reagent mixture, comprising transfection reagent (e.g., lipid, polymer, lipid-polymer or membrane destabilizing peptide) and gelatin that is present in an appropriate solvent, such as water or double deionized water, may be affixed to the transfection device. hi a further embodiment a cell culture reagent may also be present in the gelatin-transfection reagent mixture. The mixture is evenly spread onto a surface, such as a slide and multi-well plate, thus producing a transfection surface bearing the gelatin-transfection reagent mixture. In alternative embodiments, different transfection reagent-gelatin mixtures may also be spotted on discrete regions on the surface of the transfection device. The resulting product is allowed to dry completely under suitable conditions such that the gelatin-transfection reagent mixture is affixed at the site of application of the mixture. For example, the resulting product to can be dried at specific temperatures or humidity or in a vacutun-dessicator.
The concentration of transfection reagent present in the mixture depends on the transfection efficiency and cytotoxicity of the reagent. Typically there is a balance between transfection efficiency and cytotoxicity. At concentrations in which a transfection reagent is most efficient, while lceeping cytotoxicity at an acceptable level, the concentration of transfection reagent is at the optimal level. The concentration of transfection reagent will generally be in the range of about 1.,0 ~ghnl to about 1000 ~.g/ml. In preferred embodiments, the concentration is from about 40 ~g /ml to about 600 p,g/ml.
Similarly, the concentration of gelatin or another matrix depends on the experiment or assay to be performed, but the concentration will generally be in the range of 0.01% to 0.5% of the transfection reagent. According to embodiments shown in the examples the gelatin concentration is about 0.2% of the transfection reagent.
The molecules to be introduced into cells can be nucleic acids, proteins, peptides, peptide nucleic acid (PNA) and other biomolecules. The nucleic acid can be DNA, RNA
and DNA/hybrid, etc. If the DNA used is present in a vector, the vector can be of any type, such as a plasmid (e.g. example, pCMV-GFP, pCMV-luc) or viral-based vector(e.g.
pLXSN). The DNA can also be linear fragment with a promoter sequence (such CMV
promoter) at the 5' end of the cDNA to be expressed and a poly A site at the 3' end. These gene expression elements allow the cDNA of interest to be transiently expressed in mammalian cells. If the DNA is the single strand oligodeoxynucleotide (ODN), for example antisense ODN, it can be introduced into cells to regulate target gene expression.
In embodiments using RNA the nucleic acid may be single stranded (antisense RNA and ribozyrne) or double stranded (RNA interference, SiRNA). In most cases, the RNA is ~ modified in order to increase the stability of RNA and improve its function in down regulation of gene expression. W peptide nucleic acid (PNA), the nucleic acid backbone is replaced by peptide, which malces the molecule more stable. In particular embodiments the methods described herein can be used to introduce proteins, peptides and other molecules into cells for various proposes, for example molecular therapy, protein function studies, or molecule mechanism studies.
Under appropriate conditions, the biomolecules are added into the transfction device, which is coated with transfection or delivery reagent(s), to form biomolecule/delivery reagent complexes. The biomolecules are preferably dissolved in cell culture medium without fetal bovine serum and antibiotics, for example Dulbecco's Modified Eagles Medium (DMEM). If the transfection or delivery reagent is evenly affixed on the slide, the biomolecules can be spotted onto discrete locations on the slide.
Alternatively, transfection or delivery reagents may be spotted on descrete locations on the slide, and the biomolecules can simply be added to cover the whole sL~rface of the transfection device. If the transfection reagent or delivery reagent are affixed on the bottom of multi-well plates, the biomolecules are simply added into different wells by multi-channel pipette, automated device, or other method. The resulting product (transfection device coated with transfection or delivery reagent and biomolecules) is incubated for approximately 25 minutes room temperature to form the bio-molecule/transfection reagent (or delivery reagent) complexes. In some cases, for example, the different kind of biomolecules are spotted on discrete location of slide, the DNA solution will be remove to produce a surface bearing biomolecules in complex with transfection reagent.
In other case, the biomolecules solution can be kept on the surface. Sequently, cells in an appropriate medium and appropriate density are plated onto the surface. The resulting product (a surface bearing biomolecules and plated cells) is maintained under conditions that result in entry of the biomolecules into plated cells.
Suitable cells for use according to the methods described herein include prokaryotes, yeast, or higher eukaryotic cells, including plant and animal cells, especially mammalian cells. Eukaryotic cells, such as mammalian cells (e.g., human, monkey, canine, feline, bovine, or murine cells), bacterial, insect or plant cells, are plated onto the transfection device, which is coated with transfection or delivery reagent and biomolecules, in sufficient density and under appropriate conditions for introduction/entry of the biomolecule into the eulcaryotic cells and either expression of the DNA or interaction of the biomolecule with cellular components. In particular embodiments the cells maybe selected from hematopoietic cells, neuronal cells, pancreatic cells, hepatic cells, chondrocytes, osteocytes, or myocytes. The cells can be fully differentiated cells or progenitor/stem cells.
W preferred embodiments, eulcaryotic cells are grown in Dulbecco's Modified Eagles Medium (DMEM) containing 10% heat-inactivated fetal bovine serum (FBS) with L-glutamine and penicilliustreptomycin (pen/strep). It will be appreciated by those of skill in the art that certain cells should be cultured in a special medium, because some cells need special nutrition, such as growth factors and amino acids. The optimal density of cells depends on the cell types and the purpose of experiment. For example, a population of 70-80% confluent cells is preferred for gene transfection, but for oligonucleotide delivery the optimal condition is 30-50% confluent cells. In an example embodiment, if Sx104 293 cells/well were seeded onto a 96 well plate, the cells would reach 90%
confluency at 18-24 hours after cell seeding. For HeLa 705 cells, only 1 x 104 cells/well are needed to reach a similar confluent percentage in a 96 well plate.
After the cells are seeded on the surface containing biomolecules/delivery reagent, the cells are incubated under optimal conditions for the cell type (e.g.
37°C, 5-10% COZ).
The culture time is dependent on the purpose of experiment. Typically, the cells are incubated for 24 to 48 hours for cells to express the target gene in the case of gene transfection experiments. In the analysis of intracellular trafficlcing of biomolecules in cells, minutes to several hours of incubation may be required and the cells can be observed at defined time points.
The results of biomolecule delivery can be analyzed by different methods. In the case of gene transfection and antisense nucleic acid delivery, the target gene expression level can be detected by reporter genes, such as green fluorescent protein (GFP) gene, luciferase gene, or (3-galactosidase gene expression. The signal of GFP can be directly observed under microscope, the activity of luciferase can be detected by a luminometer, and the blue product catalyzed by (3-galactosidase can be observed under microscope or determined by a microplate reader. One of skill in the art is familiar with how these reporters function and how they may be introduced into a gene delivery system.
The nucleic acid and its product, the protein, peptide, or other biomolecules delivered according to methods described herein and the target modulated by these biomolecules can be determined by various methods, such as detecting immunofluorescence or enzyme imtnunocytochemistiy, autoradiography, or in situ hybridization. If immunofluorescence is used to detect expression of am encoded protein, a fluorescently labeled antibody that binds the target protein is used (e.g., added to the slide under conditions suitable for binding of the antibody to the protein). Cells containing the protein are then identified by detecting a fluorescent signal. If the delivered molecules can modulate gene expression, the target gene expression level can also be determined by methods such as autoradiography, in situ hybridization, and in situ PCR. However, the identification method depends on the properties of the delivered biomolecules, their expression product, the target modulated by it, and/or the final product resulting from delivery of the biomolecules.
EXAMPLES
Transfection reagent Branched PEIaso (polyethyleneimine of Mw 25KDa) and Linear PEIZSO (Mw 25KDa) were purchased from Polysciences Inc. (Warrington, PA, USA).
Superfect~TM~
(Qiagen, Valencia, CA) solutions were used as provided by the manufacturers.
Transfection Reagent LipofectAMINETM was purchased from Life Technologies (Gaithersburg, MD) and was used as provided by the manufacturers. NDT-CP-B-1 (degradable) and NDT-CP-1 (non-degradable) were polymeric transfection reagent synthesized by PEI~oo (Mw 600 Da) with different linkers. NDT-LP-1 was lipid-polymer containing polymer and lipid structure on the same molecule. The structures of NDT
polymeric transfection reagents are shown in Figure 35.
Transfectable surface preparation Transfection surface prepared by gelatin based tr~ansfection mixture 0.2% Gelatin Preparation Gelatin powder Type B: 225 Bloom (Sigma, catalog #G-9391) was dissolved in sterile MilliQ water by gently swirling the solution for 15 minutes in a 60°C water bath.
The 0.2% gelatin solution was then cooled at room temperature, and while still warm (~37 40°C), the solution was filtered through a 0.45 ~.m cellular acetate membrane (CA). One hundred ml of solution was prepared and stored in 50 ml aliquots of the filtered gelatin solution at 4°C.
Preparation of transfection mixture with gelatin All transfection reagents were diluted in the 0.2% gelatin solution. The concentration of linear PEIZS~~ and specially synthesized polymer samples ranged from 320.0 ~.g/ml to 40.O~.g/ml, and the concentration of branched PEIZS~~ ranged from 160.O~,g/ml to 20.O~.g/ml. The concentration of Superfect ranged from 600.0 to 75.0 ~g/ml, and lipofectamine concentration ranged from 200.0 ~,ghnl to 25.0 ~.g/ml. The concentration of polymers and lipid-polymer synthesized (NDT) ranged from 320.0 ~.ghnl to 40.O~,g/ml.
Malting transfectable surface with 96 well plate and transfection reagent-gelatin mixture The 25 ~.l transfection/gelatin solution was added to each well of a 96 well plate.
The plate was shaken for several seconds to male sure the entire bottom surfaces were covered by transfection/gelatin solution. Then the plate was allowed to air-dry in a tissue culture hood for several hours (approximately 5 -6 hours). The dried plate was stored at 4°C and ready for use (Figure 2-7).
Making transfectable surface of glass slide by spotting transfection reagent -gelatin mixture The 1-4 ~.l transfection/gelatin solution was spotted on a PLL coated slide.
The slide was put on a clean hood for approximately 1 hour until completely dried. The dried slides were stored at 4°C and ready for use (Figure 20, 21).
T~ahsfection surface p~epaYed by lay~ain.in based tf~ansfection mixtuf°e Laminin (Sigma, catalog # L2020) was diluted in PBS to a final concentration of 40.0 ~,g/ml and stored at 4°C.
Preparation of transfection mixture with laminin All transfection reagents were diluted in the 40.O~,g/ml laminin solution. The concentration of linear PEIZS~~ and specially synthesized polymer samples ranged from 320.0 ~,g/ml to 40.O~g/ml, and the concentration of branch PEI25~~ ranged from 160.O~.g/ml to 20.O~,g/ml. The concentration of Superfect ranged from 600.0 to 75.0 l.~g/ml, and lipofectamine concentration ranged from 200.0 ~.g/ml to 25.0 ~,g/ml. The concentration of polymers and lipid-polyner synthesized (NDT) ranged from 320.0 ~.g/ml to 40.O~.g/ml.
Malting transfectable surface with 96 well plate and transfection reagent-laminin mixture The 25 ~,l transfection/laminin solution was added to each well of a 96 well plate.
The plate was shalten for several seconds to matte sure the entire bottom surfaces were cover by transfectiougelatin solution. Then the plate was allowed to air dry in a tissue culture hood for several hours (approximately 5 -6 hours). The dried plate was stored at 4°C and ready for use (Figure 8-13).
Malting transfectable surface of glass slide by spotting transfection reagent -laminin mixture The 1-4 ~,1 transfection/gelatin solution was spotted on a PLL coated slide.
The slide was placed on a clean hood for about 1 hour and allowed to dry completely. The dried slide was stored at 4°C and ready for use (Figure 22-23).

Ti~arZSfectiora surface pr~epar~ed by collagen based tr~ansfection mixture Collagen (Sig~.na, catalog # C8919) was diluted in PBS to a final concentration of 120.0 ~,g/ml and stored at 4°C.
Preparation of transfection mixture with laminin All transfection reagents were diluted in the 120.0 ~.g/ml of collagen solution. The concentration of linear PEIZSn and specially synthesized polymer samples ranged from 320.0 ~.g/ml to 40.O~,g/ml, and the concentration of branched PEI25~~ ranged from 160.O~,g/ml to 20.O~,g/ml. The concentration of Superfect ranged from 600.0 to 75.0 ~g/ml, and lipofectamine concentration ranged from 200.0 ~,glml to 25.0 ~ghnl. The concentration of polymers and lipid-polymer synthesized (NDT) ranged from 320.0 ~g/ml to 40.O~,g/ml.
Malting transfectable surface with 96 well plate and transfection reagent-collagen mixture The 25 ~l transfection/collagen solution was added to each well of a 96 well plate.
The plate was shalten for several seconds to male sure the entire bottom surfaces were covered by transfection/gelatin solution. Then the plate was allowed to dry in a tissue culture hood for several hours (approximately 5 -6 hours). The dried plate was stored at 4°C and ready for use (Figure 14-16).
Ti~ansfection surface pr~epar~ed by gelatirrllaminin based tr~ansfection mixture Laminin (Sigma, catalog # L2020) was diluted in 0.2% gelatin to a final concentration of 40.0 ~,g/ml and stored at 4°C.
Preparation of transfection mixture with gelatin and laminin All transfection reagents were diluted in the 40.0 yg/ml laminin0.2% gelatin solution. The concentration of linear PEI25~~ and specially synthesized polymer samples ranged from 320.0 ~,g/ml to 40.Op,g/ml, and the concentration of branch PEIZS~< ranged from 160.O~g/ml to 20.O~,g/ml. The concentration of Superfect ranged from 600.0 to 75.0 yg/ml, and lipofectamine concentration ranged from 200.0 p,g/ml to 25.0 ~.g/ml. The concentration of polyners and lipid-polymer synthesized (NDT) ranged from 320.0 ~,g/ml to 40.O~,g/ml.
Malting transfectable surface with 96 well plate and transfection reagent-gelatin-laminin mixture The 25 ~,1 transfection/gelatin/laminin solution was added to each well of a 96 well plate. The plate was shalten for several second to malte sure the entire bottom surfaces were covered by transfection/gelatin solution. Then the plate was allowed to dry in a tissue culture hood for several hours (approximately 5 -6 hours). The dried plate was stored at 4°C and ready for use (Figure 17-19).
Plasmid DNA pre aration The plasmids pCMV-GFP and pCMV-luc were constructed according to standard' DNA recombinant protocols. The expression of green fluorescent protein (GFP) and firefly luciferase gene cDNA was controlled by the human cytomegalovirus (CMV) promoter, and the transcripts were stabilized by a gene expression enhancer, chicl~en (3-globulin intron.
The plasmids were amplified in DHSa E. coli and purified with Qiagen Plasmid Max Preparation Kit according to the manufacturer's instruction. The quantity and quality of the purified plasmid DNA was assessed by spectrophotometric analysis at 2f0 and 280 mn, as well as by electrophoresis in 0.8% agarose gel. Purified plasmid DNA was dissolved in sterile ddHaO and stored at -20°C.
Preparation of DNA solution with DMEM
pCMV-GFP or pCMV-luc plasmid was diluted in DMEM to a final concentration of 10~.g/ml. Thirty (30) ~,1 DNA solution was 'added to the transfectable surface of 96-well plate or glass slide, and incubated at room temperature for 20-30 min.
Antisense oligonucleotide preparation Luciferase 705 reporter gene system was developed by Dr. Kole in University of Northern Carolina (Kang SH et al. Biochemistry 1998;37(18):6235-9). In this system, human (3-globin with mutation at 705 was inserted into the sequence between luciferase cDNA. This plasmid was introduced into HeLa cell for stable gene expression;
the cell line was termed as HeLa 1uc705. Usually the cells exhibit low luciferase activity, because the gene products (luciferases) with wrong splicing exhibit no activities.
However, the antisense oligonucleotide binding to 705 sequence will bloclc the wrong splicing site and produce luciferase protein with biological activity. Luciferase 705 is used as functional model for evaluation of the efficiency of antisense oligonucleotide delivery.
Higher luciferase activity indicates higher efficiency of antisense delivery.
In present studies, the l8nt 2'-O-methyl-phosphorothioate oligonucleotide binding to 1uc705 sequence was synthesized. The sequence is CCUCUUACCUCAGUUACA. The antisense oligo was diluted in optimal MEM, final concentration was 0.6 ynol/L. 30.1 of antisense oligo was add to the transfectable surface of each well described previously and incubated at room temperature for 25 min (Figure 24).

siRNA preparation siRNA is a double stranded RNA fragment with 21 to 25 bp, which can bind and destroy target mRNA and lead to down regulation of gene expression levels. In this experiment, the luciferase plasmid and siRNA synthetic cassette which targets luciferase gene were prepared in opti MEM and were added into the transfectable plate described previously and incubated for 25 min. The amounts of luciferase plasmids were 0.5 ~.glwell and siRNA synthetic cassettes were about 0.5 p,g/ well (Figure 25).
Tat Peptide delivery Biotin labeled Tat peptide of various concentrations (50, 25, 12.5 and 6.3 p,g/ well) were prepared with 0.2% gelatin solution and coated on a 96 well plate, respectively. The plate was dried by sitting in a cell culture hood for several hours. 1.5 x 104 HeLa cells/well were seeded on the plate coated with peptide and incubated at 37°C for 4h. The cells were fixed with 0.2% glutaraldehyde /PBS for 5 min followed by treatment with 10%
methanol.
After being bloclced with 10% serum at 37°C for 30min, the cells were incubated with streptavidin-FITC at 37°C for 30min. The cells were washed with PBS and the fluorescent signal was observed under a fluorescent microscope. If the peptide were successfully delivered into cells, the biotin-conjugated peptide can specifically bind to strreptavidin-FITC and lead to peptides be able to produce fluorescent signals. The increased FITC signal in cells indicates that more peptides were transported into the cells (Figure 26).
The effect of tar~etin moiety in transfection mixture on transfectable surface system mediated gene transfer Transferrin conjugated poly-L-lysine preparation Transferrin can be absorbed by liver cells in a transferrin receptor mediated endocytosis pathway. Transferrin has been successfully reported as cell targeting molecular that could improve gene delivery efficiency in liver cells (Wagner E, Ogris M, Zauner W.
Adv Drug Deliv Rev 1998; 30(1-3):97-113).
Effect of tar eg t~ molecule (Transferrin~on cationic polymeric transfection reagent mediated gene transfer in laminin-based transfection mixture system In this experiment, 25p,1 of poly-L-lysine conjugated with transferrin (PLL-T) was coated on a 96 well plate with 40.Op.g/ml laminin. Poly-L-lysine (PLL) was used as a control. The ranges of concentration of PLL-T and PLL are from 320.O~,g/ml to 40.O~g/ml.
After air drying, 25p,1 luciferase plasmid solution (20.Op,g/ml, with the polymer/DNA ratio of 16:1 to 2:1.) was added into the plate and incubated at room temperature for 25 min.
The dried plate was ready to use in related experiments (Figure 27).
Effect of membrane-destabilizing component in transfection mixture on transfectable surface system mediated gene transfer VSVG is a viral envelope protein with membrane-destabilizing properties resulting in membrane fusion and disruption of the membranes of the cell. VSVG has been used as a gene transfection enhancer which can dramatically increase the cationic polymer (PLL) mediated gene transfection. A peptide from the cell fusion domain of VSVG
protein was synthesized. The sequence of this peptide is RRRQGTWLNPGFPPQSCGYATVTDARRR, with the amino acid arginine at the end of the C and N terminal respectively in order to improve the solubility of the peptide.
VSVG peptide and poly-L-lysine were diluted in 40.0 ~.g/ml laminin. The concentration of VSVG peptide was 1.0 mg/ml, aild the concentration of PLL
ranged from 640 ~.g/ml to 160.0 ~.g/ml. In the control group only PLL was diluted in laminin solution at same concentration. 25 ~1 of the PLL or PLL+VSVG peptide solution was added into 96 well plate and let it air dry. The amount of PLL was 16.0, 8.0 and 4.O~.g/well respectively (Figure 2~).

Effect of cytoreductive reagent in transfection mixture on transfectable surface system mediated gene transfer Glutamine is a cytoreductive reagent which can protect cells against ammonia-induced cytotoxicity (Nalcamura E and Hagen S J. Am J Physiol Gastointest liver Physiol 283 61264-1275, (2002)). Since almost all cationic polymers or cationic lipids contain amine groups, the addition of glutamine into the transfection mixture for transfectable surface preparation plays a role in protection against the cytotoxicity of transfection.
Glutamine (100mmo1/L) and different transfection reagents (NDT-CP-B-1, NDT-CP-1, Superfect and lipofectamine 2000) were diluted in 0.2% gelatin solution, and 25 ~.1 of the solution containing Glutamine/transfection reagent/gelatin was added into a 96 well plate and allowed to air dry. Then 251 GFP plasmid solution in opti MEM (20.Oyg/ml) was added into each well and incubated at room temperature for 25 min. The dried plate was ready for use (Figure 32).
The effect of cell attaching_reagent in transfection mixture on transfectable surface system mediated gene transfer Laminin is a matrix for support cell growth and differentiation. Laminin is a commonly used cell attaching reagent. In this experiment, 0.2% gelatin or 0.2 % gelatin and 40.0 ~.g/ml laminin mixture was used for diluting transfection reagents NDT-CP-B-1, NDT-CP-1 , NDT-LP-1 NDT-LP-2, Superfect and lipofectamine 2000, and 251 solution was added into 96 well plate. ,The amounts of NDT polymers used in each well were 8, 4 or 2~,g, respectively, the amounts of Superfect were 15, 7.5 and 3.8~.g/well respectively, and the amounts of lipofectamine 2000 were 2.5, 1.25 and 0.63~,g/well respectively. After air drying, 25 ~,1 of GFP plasmid in Opti MEM (20~,g/ml) was added into each well and incubated at room temperature for 25 min, then S~e 104 293 cells/well were seeded in the plate. The cells were further incubated at 37°C for 24 hours. The transfection efficiency and cytotoxicity were analyzed by fluorescent microscope and MTT assay (Figure 33).
Cell culture HEK 293T cells was maintained in DMEM (Gibco) containing 10% fetal bovine serum, 100 units/ml penicillin and 100 ~.g/ml streptomycin. In this media the cells had a doubling time of about 20 hours, and the cells were split every 3-4 days to avoid over confluency.

HeLa 705 cell line was from Human cervical carcinoma HeLa cells after introducing firefly luciferase gene with a mutant (3-globin intron (a mutation at 705 position) which results in a mutated luciferease protein due to the incorrect splicing.
However, the mutated intron can be corrected by a specific antisense oligo nucleotide after it blocl~s the wrong splicing site (Fang SH et al. Biochemistry 1998;37(18):6235-9). The cell line was maintained in DMEM (Gibco) containing 10% fetal bovine serum, units/ml penicillin and 100 ~ghnl streptomycin. 200~g /ml hygromycin was added into medium to maintain the luc-705 plasmid. In this media the cells had a doubling time of about 20 hours, and the cells were split every 3-4 days to avoid over confluency.
Human liver tumor cell line HepG2 was maintained in a,-MEM medium (Gibco) containing 10% fetal bovine serum, 100 unitshnl penicillin and 100 ~.g/ml streptomycin. In this media the cells had a doubling time of about 20 hours, and the cells were split every 3-4 days to avoid over confluency.
Human primary endothelial cell HW-EC cell line was grew and maintained in EBM medium (Cambrex Corp.) containing 10% fetal bovine serum, 100 units/ml penicillin and 100 ~g/ml streptomycin, and different types of growth factors according to the manufacturers instructions (Figure 29-30).
Cell preparation and seeding onto the transfectable surface Immediately prior to transfection, in a tissue culture hood, cells were harvested from a 10 cm dish as follows:
a. Media was removed, cells were rinsed with 2 ml of PBS, the solution was allowed to spread over the plate and then the solution was removed immediately.
b. 0.5 ml trypsin-EDTA was added to the cells and evenly spread over plate and the trypsin-EDTA was then immediately removed.
c. The cells were allowed to sit in the hood for 3-5 minutes. The plate was then agitated to dislodge cells from the surface of the plate.
d. Six (6) ml of 37°C full medium was added to the plate of cells, and the solution was pipetted up and down 12-15 times with a 10 ml pipette until a single cell suspension was obtained, while avoiding the creation of too much froth. Another 14m1 of medium was added and the cells were completely suspended in the solution.
e. Cells were quantified in a hemocytometer.

f. The HEK 293T, HepG2 cells were diluted to a final concentration of 4-Sx105ce11/well in a sterile basin, and 100.1 (4-Sx104cells) were be seeded for each well in a 96-well plate. The optimal concentration of HeLa 705 and HUV-EC cells were 1-2x 105ce11/ml.
GFP Reporter Gene Transfection assay Green fluorescent protein (GFP) gene was used in an initial screening. After transfection, the GFP signal in cells was observed under fluorescent microscope (Olympus, filter 520nm). Cells were photographed using a lOX objective. The percentage of cells with GFP signal in transfected cultures was determined from cotmts of three fields for optimal cationic polymer amounts.
Luciferase assay Measurement of luciferase activity was performed using a chemiluminescent assay following the manufacturer's instructions (Luciferase Assay System; Promega, Madison, WI, USA). Briefly, thirty hours after gene transfer, the cells were rinsed twice with PBS
and then were lysed with lysis buffer (1% Triton X-100, 100 mM K3P04, 2 mM
dithiothreitol, 10% glycerol, and 2 mM EDTA pH 7.8) for 15 min at room temperature. A
10-~,l aliquot of cell lysate was then mixed with 50-~,l of luciferase assay reagent with an injector at room temperature in the luminometer. Light emission was measured in triplicate over 10 seconds and expressed as RLUs (relative light units). Relative light units (RLU) were normalized to the protein content of each sample, determined by BSA
protein assay (Pierce, Rocl~ford, Illinois). All the experiments were conducted in triplicate.
Cytotoxici~ assay-MTT assay The cytotoxicities of transfection reagents on mammalian cells were evaluated using 3-[4,5 dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) method.
Foray (40) hours after transfection, 101 of MTT solution (5.0 mg/ml, Sigma) was added to each well, and incubated for 3 hrs. The medium was then removed and 200-~,l DMSO was added to dissolve the formazan crystals. The absorbance of the solution was measured at 570 mn.
Cell viabilities was calculated using the equation: Viability (%) = f AbsS~o (sample) /Abss~o ~°°"t,~°p) x 100 (Figure 31).
Stabilit,~tudy LPEI, BPEI, NDT-CP-B-1, NDT-CP-1, Superfect and lipofectamine in 0.2% gelatin were coated in 96-well plate as described previously. After air-drying, the plate was incubated at 37°C for 9 days. The plate was then ready for use. 25 ~,1/well luciferase plasmid (20.0 p,g/ml in optimal MEM) was added to the plates (before and after 37°C
incubation) and incubated for 25 min. 5x104 293 cells were seed in the plate and incubated at 37°C for 48 hours. The gene transfection efficiency of the transfectable surface of the flash plate or of 37°C incubation for 9 days was compared (Figure 34).
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention. All references cited herein are incorporated herein by reference.

Claims (26)

1. A method for introducing biomolecules into eukaryotic cells in vitro comprising: (a) providing a solid surface at least partially coated with a cationic polymer or a lipid, (b) adding the biomolecules to be introduced into the eukaryotic cells onto the solid surface, (c) seeding cells on the solid surface at a sufficient density and under appropriate conditions for introduction of the biomolecules into the eukaryotic cells.
2. The method of claim 1, wherein the surface is selected from the group consisting of flasks, dishes, multi-well plates, glass slides, and implanted devices.
3. The method of claim 1, wherein said solid surface is coated with a lipid-polymer.
4. The method of claim 1, wherein said polymer or lipid contain an additional reagent selected from the group consisting of a cell-targeting moiety, an intracellular targeting moiety, a membrane-destabilizing component, and one or more transfection enhancers.
5. The method of claim 1, wherein said cationic polymer or lipid is affixed on the surface by evenly spreading said cationic polymer or lipid on the surface or spotting said cationic polymer or lipid in discrete areas of the surface.
6. The method of claim 1, wherein the cationic polymer or lipid contains a matrix reagent selected from the group consisting of proteins, glycoproteins, peptides, polysaccharides, and polymers or mixtures thereof.
7. The method of claim 6, wherein said protein is selected from gelatin, collagen, laminin, fibronectin, and bovine serum albumin or a mixture thereof.
8. The method of claim 6, wherein said polymer is selected from hydrogels, biodegradable polymers, and biocompatible materials.
9. The method of claim 1, wherein said cationic polymer or lipid contains a cell culture reagent selected from the group consisting of cytoreductive reagents, cell binding reagents, cell growing reagents, cell stimulating reagents, and cell inhibiting reagents.
10. The method of claim 1, wherein the solid surface is selected from a slide or a multi-well plate.
11. The method of claim 1, wherein the eukaryotic cells are mammalian cells.
12. The method of claim 11, wherein the mammalian cells are dividing cells or non-dividing cells.
13. The method of claim 11, wherein the mammalian cells are transformed cells or primary cells.
14. The method of claim 11, wherein the mammalian cells are somatic cells or stem cells.
15. The method of claim 1, wherein the eukaryotic cells are plant cells.
16. The method of claim 1, wherein the cationic polymer or lipid is affixed on the surface by evenly spreading said cationic polymer or lipid on the solid surface or spotting said cationic polymer or lipid on the solid surface manually or by an automated mechanism.
17. The method of claim 1, wherein the biomolecule is selected from the group consisting of DNA, RNA, and DNA/RNA hybrid.
18. The method of claim 1, wherein the biomolecule is selected from the group consisting of a linear molecule, a plasmid, and single stranded oligodeoxynucleotide (ODN).
19. The method of claim 1, wherein the biomolecule is selected from the group consisting of single stranded RNA (ribozyme) or double stranded RNA (siRNA).
20. The method of claim 1, wherein the eukaryotic cells are insect cells.
21. A method of determining whether a biomolecule can enter a cell comprising (a) introducing the biomolecule into said cell by the method of any one of the foregoing claims, and (b) detecting whether the biomolecule has been delivered to the cells.
22. The method of claim 21 wherein the biomolecules are selected from the group consisting of nucleotides, proteins, peptides, sugars, polysaccharides, and organic compounds.
23. The method of claim 21, wherein the cells are selected from the group consisting of plant, insect, and bacterial cells.
24. The method of claim 1, wherein the cationic polymer or lipid is affixed on the surface manually or by an automated mechanism.
25. The method of claim 21, wherein said detecting is performed by detecting said biomolecule, a product of the biomolecule, activity of a biomoloecule, activity of the product of the biomolecule, a target molecule, a product catalyzed by the biomolecule or a product regulated by the biomolecule.
26. The method of claim 1 wherein the biomolecules are selected from the group consisting of nucleotides, proteins, peptides, sugars, polysaccharides, and organic compounds.
CA2512144A 2003-01-13 2003-12-04 Solid surface for biomolecule delivery and high-throughput assay Expired - Fee Related CA2512144C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10/341,059 2003-01-13
US10/341,059 US20040138154A1 (en) 2003-01-13 2003-01-13 Solid surface for biomolecule delivery and high-throughput assay
PCT/US2003/038498 WO2004065636A1 (en) 2003-01-13 2003-12-04 Solid surface for biomolecule delivery and high-throughput assay

Publications (2)

Publication Number Publication Date
CA2512144A1 true CA2512144A1 (en) 2004-08-05
CA2512144C CA2512144C (en) 2013-07-02

Family

ID=32711435

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2512144A Expired - Fee Related CA2512144C (en) 2003-01-13 2003-12-04 Solid surface for biomolecule delivery and high-throughput assay

Country Status (12)

Country Link
US (3) US20040138154A1 (en)
EP (2) EP1587954B1 (en)
JP (1) JP4580764B2 (en)
KR (1) KR100863774B1 (en)
CN (1) CN1738911B (en)
AT (1) ATE477336T1 (en)
AU (1) AU2003298863B2 (en)
CA (1) CA2512144C (en)
DE (1) DE60333793D1 (en)
DK (1) DK2253718T3 (en)
HK (1) HK1088637A1 (en)
WO (1) WO2004065636A1 (en)

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070269891A9 (en) * 2003-01-13 2007-11-22 Yasunobu Tanaka Solid surface with immobilized degradable cationic polymer for transfecting eukaryotic cells
US20040138154A1 (en) * 2003-01-13 2004-07-15 Lei Yu Solid surface for biomolecule delivery and high-throughput assay
US7358223B2 (en) * 2004-10-04 2008-04-15 Nitto Denko Corporation Biodegradable cationic polymers
KR20130010036A (en) * 2004-12-17 2013-01-24 닛토덴코 가부시키가이샤 Immobilized degradable cationic polymer for transfecting eukaryotic cells
US20120269886A1 (en) 2004-12-22 2012-10-25 Nitto Denko Corporation Therapeutic agent for pulmonary fibrosis
PT2727583T (en) 2004-12-22 2021-12-27 Nitto Denko Corp Drug carrier and drug carrier kit for inhibiting fibrosis
US9572886B2 (en) 2005-12-22 2017-02-21 Nitto Denko Corporation Agent for treating myelofibrosis
US7700541B2 (en) * 2006-04-06 2010-04-20 Nitto Denko Corporation Biodegradable cationic polymers
CA2706124A1 (en) * 2006-07-07 2008-01-10 Aarhus Universitet Nanoparticles for nucleic acid delivery
TWI407971B (en) 2007-03-30 2013-09-11 Nitto Denko Corp Cancer cells and tumor-related fibroblasts
US20080312174A1 (en) * 2007-06-05 2008-12-18 Nitto Denko Corporation Water soluble crosslinked polymers
WO2009036368A2 (en) * 2007-09-14 2009-03-19 Nitto Denko Corporation Drug carriers
TW201004646A (en) * 2008-07-30 2010-02-01 Nitto Denko Corp Drug carriers
CN102010508B (en) * 2010-10-19 2012-10-03 北京大学 Cationic polymer and preparation method and application thereof
US9637775B2 (en) 2012-02-13 2017-05-02 Neumodx Molecular, Inc. System and method for processing biological samples
US11931740B2 (en) 2012-02-13 2024-03-19 Neumodx Molecular, Inc. System and method for processing and detecting nucleic acids
US11485968B2 (en) 2012-02-13 2022-11-01 Neumodx Molecular, Inc. Microfluidic cartridge for processing and detecting nucleic acids
WO2013123035A1 (en) * 2012-02-13 2013-08-22 Molecular Systems Corporation System and method for processing and detecting nucleic acids
US9604213B2 (en) 2012-02-13 2017-03-28 Neumodx Molecular, Inc. System and method for processing and detecting nucleic acids
ES2741749T3 (en) 2012-10-25 2020-02-12 Neumodx Molecular Inc Method and materials for the isolation of nucleic acid materials
US11293021B1 (en) 2016-06-23 2022-04-05 Inscripta, Inc. Automated cell processing methods, modules, instruments, and systems
US9982279B1 (en) 2017-06-23 2018-05-29 Inscripta, Inc. Nucleic acid-guided nucleases
US10011849B1 (en) 2017-06-23 2018-07-03 Inscripta, Inc. Nucleic acid-guided nucleases
KR20220104847A (en) 2017-06-30 2022-07-26 인스크립타 인코포레이티드 Automated cell processing methods, modules, instruments, and systems
US10858761B2 (en) 2018-04-24 2020-12-08 Inscripta, Inc. Nucleic acid-guided editing of exogenous polynucleotides in heterologous cells
US10557216B2 (en) 2018-04-24 2020-02-11 Inscripta, Inc. Automated instrumentation for production of T-cell receptor peptide libraries
CN114854720A (en) 2018-06-30 2022-08-05 因思科瑞普特公司 Apparatus, modules and methods for improved detection of editing sequences in living cells
US11142740B2 (en) 2018-08-14 2021-10-12 Inscripta, Inc. Detection of nuclease edited sequences in automated modules and instruments
US11214781B2 (en) 2018-10-22 2022-01-04 Inscripta, Inc. Engineered enzyme
CA3134168A1 (en) 2019-03-25 2020-10-01 Inscripta, Inc. Simultaneous multiplex genome editing in yeast
US11001831B2 (en) 2019-03-25 2021-05-11 Inscripta, Inc. Simultaneous multiplex genome editing in yeast
CA3139122C (en) 2019-06-06 2023-04-25 Inscripta, Inc. Curing for recursive nucleic acid-guided cell editing
US11203762B2 (en) 2019-11-19 2021-12-21 Inscripta, Inc. Methods for increasing observed editing in bacteria
AU2020407048A1 (en) 2019-12-18 2022-06-09 Inscripta, Inc. Cascade/dCas3 complementation assays for in vivo detection of nucleic acid-guided nuclease edited cells
EP4081196A1 (en) * 2019-12-23 2022-11-02 Universidad del País Vasco/Euskal Herriko Unibertsitatea Golden lipid nanoparticles for gene therapy
WO2021154706A1 (en) 2020-01-27 2021-08-05 Inscripta, Inc. Electroporation modules and instrumentation
US20210332388A1 (en) * 2020-04-24 2021-10-28 Inscripta, Inc. Compositions, methods, modules and instruments for automated nucleic acid-guided nuclease editing in mammalian cells
EP4214314A1 (en) 2020-09-15 2023-07-26 Inscripta, Inc. Crispr editing to embed nucleic acid landing pads into genomes of live cells
US11512297B2 (en) 2020-11-09 2022-11-29 Inscripta, Inc. Affinity tag for recombination protein recruitment
US11306298B1 (en) 2021-01-04 2022-04-19 Inscripta, Inc. Mad nucleases
EP4274890A1 (en) 2021-01-07 2023-11-15 Inscripta, Inc. Mad nucleases
US11884924B2 (en) 2021-02-16 2024-01-30 Inscripta, Inc. Dual strand nucleic acid-guided nickase editing

Family Cites Families (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5264618A (en) * 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US6509032B1 (en) * 1991-08-28 2003-01-21 Mcmaster University Cationic amphiphiles
US5283185A (en) * 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
US5919455A (en) * 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
EP0769063A1 (en) * 1994-06-27 1997-04-23 The Johns Hopkins University Targeted gene delivery system
US5767168A (en) * 1995-03-30 1998-06-16 The Proctor & Gamble Company Biodegradable and/or compostable polymers made from conjugated dienes such as isoprene and 2,3-dimethyl-1, 3-butadiene
US6051429A (en) 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
JP3851369B2 (en) * 1995-08-24 2006-11-29 ミレニウム・バイオロジクス・インコーポレイテッド Multiwell bone cell culture device
EP0904031A4 (en) 1996-05-29 2002-05-02 Cell Genesys Inc Cationic polymer/lipid nucleic acid delivery vehicles
JP2000514440A (en) * 1996-07-09 2000-10-31 ザ ジョーンズ ホプキンス ユニバーシティー Gene transfer system
DE19743135A1 (en) 1997-09-30 1999-04-01 Hoechst Marion Roussel De Gmbh Biologically compatible low molecular weight polyethyleneimines
US6072101A (en) * 1997-11-19 2000-06-06 Amcol International Corporation Multicomponent superabsorbent gel particles
US6475994B2 (en) * 1998-01-07 2002-11-05 Donald A. Tomalia Method and articles for transfection of genetic material
ATE346156T1 (en) * 1999-09-17 2006-12-15 Whitehead Biomedical Inst REVERSE TRANSFECTION PROCESS
US20020006664A1 (en) * 1999-09-17 2002-01-17 Sabatini David M. Arrayed transfection method and uses related thereto
US8143195B2 (en) * 2000-01-24 2012-03-27 Yingjian Wang Arrays for bringing two or more reagents in contact with one or more biological targets and methods for making and using the arrays
US6319715B1 (en) 2000-04-21 2001-11-20 Cornell Research Foundation, Inc. Method of enhancing the delivery of nucleic acids using silica nanoparticles
US20020009807A1 (en) 2000-05-18 2002-01-24 Lance Kam Lipid bilayer array method and devices
US20040120979A1 (en) * 2000-06-02 2004-06-24 Roessler Blake J. Delivery systems comprising biocompatible and bioerodable membranes
US6696038B1 (en) 2000-09-14 2004-02-24 Expression Genetics, Inc. Cationic lipopolymer as biocompatible gene delivery agent
US6897067B2 (en) * 2000-11-03 2005-05-24 Regents Of The University Of Michigan Surface transfection and expression procedure
CA2427916C (en) * 2000-11-03 2012-01-17 The Regents Of The University Of Michigan Surface transfection and expression procedure
WO2002100435A1 (en) * 2001-06-11 2002-12-19 Centre Hospitalier Universitaire De Montreal Compositions and methods for enhancing nucleic acid transfer into cells
US6586524B2 (en) * 2001-07-19 2003-07-01 Expression Genetics, Inc. Cellular targeting poly(ethylene glycol)-grafted polymeric gene carrier
US20030215395A1 (en) * 2002-05-14 2003-11-20 Lei Yu Controllably degradable polymeric biomolecule or drug carrier and method of synthesizing said carrier
US20040048260A1 (en) * 2002-09-10 2004-03-11 Fu-Hsiung Chang Transfection of nucleic acid
US20040138154A1 (en) * 2003-01-13 2004-07-15 Lei Yu Solid surface for biomolecule delivery and high-throughput assay
US7125709B2 (en) * 2004-02-10 2006-10-24 Nitto Denko Corporation Culture device and method for eukaryotic cell transfection

Also Published As

Publication number Publication date
EP1587954B1 (en) 2010-08-11
KR100863774B1 (en) 2008-10-16
HK1088637A1 (en) 2006-11-10
ATE477336T1 (en) 2010-08-15
AU2003298863A1 (en) 2004-08-13
EP1587954A1 (en) 2005-10-26
DK2253718T3 (en) 2012-10-08
CN1738911A (en) 2006-02-22
JP4580764B2 (en) 2010-11-17
EP2253718A3 (en) 2010-12-15
DE60333793D1 (en) 2010-09-23
US20070072171A1 (en) 2007-03-29
US20040138154A1 (en) 2004-07-15
KR20050096130A (en) 2005-10-05
CN1738911B (en) 2012-09-05
CA2512144C (en) 2013-07-02
EP1587954A4 (en) 2006-04-12
AU2003298863B2 (en) 2008-04-24
US20070020761A1 (en) 2007-01-25
JP2006513703A (en) 2006-04-27
EP2253718B1 (en) 2012-07-04
US8192989B2 (en) 2012-06-05
WO2004065636A1 (en) 2004-08-05
EP2253718A2 (en) 2010-11-24

Similar Documents

Publication Publication Date Title
US8192989B2 (en) Solid surface for biomolecule delivery and high-throughput assay
JP4739352B2 (en) Solid surface with immobilized degradable cationic polymer for transfecting eukaryotic cells
US20060134790A1 (en) Solid surface with immobilized degradable cationic polymer for transfecting eukaryotic cells
CA2581632C (en) Biodegradable cationic polymers
KR20100017956A (en) Peg-pei copolymers for nucleic acid delivery
Park et al. Visualization of transfection of hepatocytes by galactosylated chitosan-graft-poly (ethylene glycol)/DNA complexes by confocal laser scanning microscopy
Geisse et al. Large-scale transient expression of therapeutic proteins in mammalian cells
US7446099B2 (en) Compositions and methods for biodegradable polymer-peptide mediated transfection
US7125709B2 (en) Culture device and method for eukaryotic cell transfection
EP1723240B1 (en) Culture device and method for eukaryotic cell transfection
Schlaeger et al. Transient transfection in mammalian cells: a basic study for an efficient and cost-effective scale up process

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20141204