AU2005316501B2 - Immobilized degradable cationic polymer for transfecting eukaryotic cells - Google Patents

Immobilized degradable cationic polymer for transfecting eukaryotic cells Download PDF

Info

Publication number
AU2005316501B2
AU2005316501B2 AU2005316501A AU2005316501A AU2005316501B2 AU 2005316501 B2 AU2005316501 B2 AU 2005316501B2 AU 2005316501 A AU2005316501 A AU 2005316501A AU 2005316501 A AU2005316501 A AU 2005316501A AU 2005316501 B2 AU2005316501 B2 AU 2005316501B2
Authority
AU
Australia
Prior art keywords
transfection
formula
cell
degradable
cells
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.)
Ceased
Application number
AU2005316501A
Other versions
AU2005316501A1 (en
Inventor
Chris P. Castello
Yasunobu Tanaka
Lei Yu
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
Nitto Denko Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nitto Denko Corp filed Critical Nitto Denko Corp
Publication of AU2005316501A1 publication Critical patent/AU2005316501A1/en
Application granted granted Critical
Publication of AU2005316501B2 publication Critical patent/AU2005316501B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0068General culture methods using substrates
    • 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
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/30Synthetic polymers

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Description

WO 2006/066001 PCT/US2005/045429 SOLID SURFACE WITH IMMOBILIZED DEGRADABLE CATIONIC POLYMER FOR TRANSFECTING EUKARYOTIC CELLS Related Applications This application claims priority to U.S. provisional application no. 60/637,344, filed 5 December 17, 2004, which is incorporated herein by reference. Background of the Invention Field of the Invention Embodiments of the invention relate to devices and methods for cell transfection. In particular, embodiments of the invention are directed to a cell transfection formula and to 10 a cell culture device that has been treated with the transfection formula. The treated cell culture device can be stored at room temperature and provides a transfection method that is simple and quick. Description of the Related Art Gene transfection methods can be used to introduce nucleic acids into cells and are 15 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 which are particularly useful. Gene transfection is the delivery and introduction of biologically functional nucleic acids into a cell, particularly a eukaryotic cell, in such a way that the nucleic acid retains its function within the cell. Gene transfection is widely applied 20 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 seek 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 25 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 applicability (Verma I.M and Somia N. Nature 389 30 (1997), pp. 239-242; Marhsall E. Science 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 -1- WO 2006/066001 PCT/US2005/045429 addition, viral vector preparation is a complicated and expensive process, which limits the application of viral vectors in vitro. The preparation of non-viral carriers is simpler and more cost effective in comparison to preparation of viral carriers, making synthetic gene carriers desirable as transfection reagents, particularly for in vitro studies. 5 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 material. Non-viral gene vectors, based on a gene carrier backbone, 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 10 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 15 described as shuttles for the transfer of nucleic acids into cells. Most non-viral transfection reagents are synthetic cationic molecules and have been reported to "coat" the nucleic acid by interaction of the cationic sites on the cation 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 20 through the cell membrane by non-specific endocytosis. (Schofield, Brit. Microencapsulated. Bull, 51(l):56-71 (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 are usually prepared in separate tubes, and each respective solution is diluted in medium (containing no fetal 25 bovine serum or antibiotics). The solutions are then mixed by carefully and slowing adding one solution to the other while continuously vortexing the mixture. The mixture is incubated at room temperature for 15-45 minutes to allow complex formation between the transfection reagent and the DNA and to remove residues of serum and antibiotics. Prior to transfection, the cell culture medium is removed and the cells are washed with buffer. The 30 solution containing the DNA-transfection reagent complexes is added to the cells, and the cells are incubated for about 3-4 hours. The medium containing the transfection reagent is then be replaced with fresh medium. The cells are finally analyzed at one or more specific -2- WO 2006/066001 PCT/US2005/045429 time point(s). This is obviously a time consuming procedure, particularly when the number of samples to be transfected is very large. Several major problems exist in conventional transfection procedures. First, conventional procedures are time-consuming, particularly when there are many cell or gene 5 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, the DNA-transfection reagent complex formation is influenced by concentration and volume of nucleic acid and reagents, pH, temperature, type of buffer(s) used, length and speed of vortexing, incubation time, and other factors. Although the same reagents and 10 procedure may be followed, different results may be obtained. Results derived from multi 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, 15 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. 20 Sabatini (U.S. 2002/0006664A1) describes a composition containing DNA which is deposited on a glass slide. However the system only allows transfection with the previously deposited DNA. This is a major disadvantage of this system. As it only provides for transfecting with previously deposited DNA, every researcher cannot use his or her desired nucleic acids. 25 U.S. Publication No. 2004/0138154A1, which is incorporated herein by reference, describes a cell culture/transfection device where the transfection is mediated by a lipid polymer. U.S. Publication No. 2005/0176132A1, also incorporated herein by reference, describes a calcium salt mediated transfectable cell culture device. US Publication No. 2003/0215395A1, incorporated herein by reference, describes 30 degradable polymers which can be used for gene delivery. As discussed above, conventional transfection is a lengthy and technically difficult procedure. Generally, three steps are required: 1) cells are seeded in a cell culture plate or -3- WO 2006/066001 PCT/US2005/045429 dish and incubated until sufficient confluence is achieved; 2) transfection reagent / nucleic acid complexes are prepared; and 3) nucleic acids of interest are added along with the transfection reagent and further incubation is carried out. Two incubation periods are needed and typically it takes more than two days to complete all the steps. In contrast, 5 embodiments of the present invention provide a simple procedure that involves only a single incubation step. A cell culture device, which has previously been coated with a transfection reagent, allows transfection by adding the nucleic acid of interest and the cell culture in succession. The transfected cells may then be cultured in the same device. Thus the cells may be transfected and cultured in the cell culture device without the need for 10 further manipulation of the cells immediately after the transfection step. Transfection efficiency is comparable to regular transfection, but the time required for the operation is reduced by more than one day. Embodiments of the invention include a transfectable cell culture device which greatly reduces the labor of transfection assays, and enables transfection with any nucleic acid of interest in an easy method with low cytotoxicity. 15 Also, the transfectable cell culture device of the invention is stable for long term storage at room temperature. Summary of the Invention Embodiments of the invention are directed to a device which includes a solid support coated with a transfection reagent mixture. Preferably, the transfection reagent in 20 the coating is not complexed with a biomolecule, such as a nucleic acid. Preferably, the solid support is polystyrene resin, epoxy resin or glass. Preferably, the coating is on the surface of the solid support. Preferably, the coating amount of the transfection reagent is from about 0.1 to about 100 Rg/cm2. Preferably, the transfection agent is a polymer. More preferably, the polymer is a cationic polymer. Preferably, the transfection agent comprises 25 a degradable cationic polymer. More preferably, the degradable cationic polymer is made by linking cationic compounds or oligomers with degradable linkers. The transfection agent may comprise both a degradable cationic polymer and a non-degradable cationic polymer. Preferably, the ratio of the non-degradable cationic polymer to the degradable cationic polymer is 1:0.5 to 1:20 (non-degradable : degradable) by weight. 30 In preferred embodiments, the transfection reagent includes a plurality of cationic molecules and at least one degradable linker molecule connecting said cationic molecules in a branched arrangement, wherein said cationic molecules are selected from: -4- WO 2006/066001 PCT/US2005/045429 (i) a cationic compound of formula (A) or (B) or a combination thereof: R N RR 6
-(NH-R,)R
8 R 3 - R 5 Formula B Formula A 5 wherein R 1 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; R is a straight chain alkylene group of the formula: -(CH2)a- wherein a is an integer number from 2 to 10; 10 R3 is a straight or branched chain alkylene group of the formula: -(CbH2b)- wherein b is an integer number from 2 to 10;
R
4 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; R is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; Ri is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, Formula A, or another Formula B; 15 R7 is a straight or branched chain alkylene group of the formula: -(CcH 2 c)- in which c is an integer number from 2 to 10; and R8 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, Formula A, or another Formula B; (ii) a cationic dendritic or branched polyamidoamine (PAMAM) with terminated primary or 20 secondary amino groups; (iii) a cationic polyamino acid; or (iv) a cationic polycarbohydrate; and wherein said degradable linker molecule is represented by the formula: 25 A(Z)d wherein A is a spacer molecule having at least one degradable bond, Z is a reactive residue which reacts with amino group, and d is an integer equal to or more than two and wherein A and Z are bound covalently. -5- WO 2006/066001 PCT/US2005/045429 In preferred embodiments, the cationic compound or oligomer is poly(L-lysine) (PLL), polyethyleneimine (PEI), polypropyleneimine (PPI), pentaethyleneamine, NN' bis(2-aminoethyl)-1,3-propanediamine, N,N'-bis(2-aminopropyl)-ethylenediamine, spermine, spermidine, N-(2-aminoethyl)-1,3-propanediamine, N-(3-aminopropyl)-1,3 5 propanediamine, tri(2-aminoethyl)amine, 1,4-bis(3-aminopropyl)piperazine, N-(2 aminoethyl)piperazine, dendritic polyamidoamine (PAMAM), chitosan, or poly(2 dimethylamino)ethyl methacrylate (PDMAEMA). In preferred embodiments, the linker molecule is di- and multi-acrylates, di- and multi-acrylamides, di- and multi-isothiocyanates, di- and multi-isocyanates, di- and multi 10 epoxides, di- and multi-aldehydes, di-and multi-acyl chlorides, di- and multi-sulfonyl chlorides, di- and multi-halides, di- and multi-anhydrides, di- and multi-maleimides, di- and multi-N-hydroxysuccinimide esters, di- and multi-carboxylic acids, or di-and multi-a haloacetyl groups. In preferred embodiments, the linker molecule is 1,3-butanediol diacrylate, 1,4 15 butanediol diacrylate, 1,6-hexanediol diacrylate, 2,4-pentanediol diacrylate, 2-methyl-2,4 pentanediol diacrylate, 2,5-dimethyl-2,5-hexanediol diacrylate, poly(ethylene glycol) diacrylate, trimethylolpropane triacrylate, pentaerythritol tetraacrylate, di(trimethylolpropane) tetraacrylate, dipentaerythritol pentaacrylate, or a polyester with at least three acrylate or acrylamide side groups. 20 In preferred embodiments, the molecular weight of the polymer is from 500 da to 1,000,000 da. More preferably, the molecular weight of the polymer is from 2000 da to 200,000 da. In preferred embodiments, the molecular weight of the cationic compound or oligomer is from 50 da to 10,000 da. In preferred embodiments, the molecular weight of 25 the linker molecule is from 100 da to 40,000 da. Preferably, the solid support is a dish bottom, a multi-well plate, or a continuous surface. In some preferred embodiments, the transfection agent is covalently associated with a nucleic acid. In other preferred embodiments, the transfection agent is non-covalently 30 associated with a nucleic acid. In preferred embodiments, the device can be stored at room temperature for at least 5 months without significant loss of transfection activity. -6- WO 2006/066001 PCT/US2005/045429 Embodiments of the invention are directed to a method of cell transfection which includes the steps of adding a solution including a nucleic acid to be transfected to a device which includes a solid support coated with a transfection reagent mixture, adding eukaryotic cells to the solution; and incubating the cells and the nucleic acid solution to 5 allow cell transfection. Preferably, the incubation is for 5 min. to 3 hours. More preferably, the incubation is for 10 min. to 90 min. Preferably, the nucleic acid is DNA, RNA, DNA/RNA hybrid or chemically modified nucleic acid. More preferably, the DNA is circular (plasmid), linear, fragment or single strand oligonucleotide (ODN). More preferably, the RNA is single strand 10 (ribozyme) or double strand (siRNA). In some preferred embodiments, the cell is a mammalian cell. In some preferred embodiments, at least some of the cells undergo cell division. In some preferred embodiments, the cell is a transformed or primary cell. In some preferred embodiments, the cell is a somatic or stem cell. In some preferred embodiments, the cell is a plant cell. 15 Further aspects, features and advantages of this invention will become apparent from the detailed description of the preferred embodiments which follow. Brief Description of the Drawings These and other features of this invention will now be described with reference to 20 the drawings of preferred embodiments which are intended to illustrate and not to limit the invention. Figure 1 shows the cell shape of transfected 293 cells. The transfection agent treatments were linear polyethyleneimine (L-PEI) based polymer, lipid based polymer, degradable cationic polymer, and no treatment (intact 293 cells). 25 Figure 2 shows percentage of EGFP- positive cells. Figure 3 shows cell condition after transfection. Figure 4 shows the stability of a transfectable cell culture device in a mylar bag with 02 absorber. Figure 5 shows the stability of a transfectable cell culture device in a mylar bag with 30 CO 2 absorber. Figure 6 shows the stability of a transfectable cell culture device in a mylar bag with 02 and CO 2 absorber. -7- WO 2006/066001 PCT/US2005/045429 Figure 7 shows the stability of a transfectable cell culture device in a mylar bag. Detailed Description of the Preferred Embodiment While the described embodiment represents the preferred embodiment of the 5 present invention, it is to be understood that modifications will occur to those skilled in the art without departing from the spirit of the invention. The scope of the invention is therefore to be determined solely by the appended claims. Embodiments of the invention are directed to a transfection device and method which is simple, convenient, and efficient compared to conventional transfection assays. A 10 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 to be transfected and cells 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 key timing-consuming step, which is required 15 by conventional transfection procedures. Only approximately 40 minutes is required to complete the entire transfection process for 10 samples, compared to 2 to 5 hours or more required by current methods. This is particularly advantageous for high throughput transfection assays, in which hundreds of samples are tested at a time. As compared to conventional transfection, there are several advantages to the 20 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 5 min. to 3 hours, and it provides a high throughput method for transfection or drug delivery in 25 which large numbers of samples may be transfected at a time. In preferred embodiments, 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 30 addition of cells. An incubation period for a predetermined time allows the biomolecule and the transfection reagent(s) to form a complex for uptake by cells in the next step. Cells are then seeded on the surface of the cell culture device and incubated, without the -8- WO 2006/066001 PCT/US2005/045429 necessity of changing the medium, and the cells are analyzed. Changing medium during the transfection procedure is unnecessary. The methods described herein dramatically reduce the risk of error, by reducing the number of steps involved, thus increasing consistency and accuracy of the system. 5 The composition containing the transfection agent can be affixed to any suitable surface. 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 skin, endothelial tissue, bone, 10 cartilage), or 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 multi-well plate. For slides, such as a glass slide coated with poly-L-lysine (e.g., Sigma, Inc.), the transfection reagent is fixed on the surface and dried, and then a nucleic acid of interest or a 15 nucleic acid to be introduced into cells is introduced. Generally, the nucleic acid is spotted onto the glass slide in a microarray. The slide is incubated at room temperature for 30 minutes to form nucleic acid/transfection reagent complexes on the surface of the transfection device. The nucleic acid/transfection reagent complexes create a medium for use in high throughput microarrays, which can be used to study hundreds to thousands of 20 nucleic acids, or other biomolecules at the same time. In an alternative embodiment, the transfection reagents can be affixed on the surface of the transfection device in discrete, defined regions to form a microarray of transfection reagents. In this embodiment, biomolecules, such as nucleic acids, which are to be introduced into cells, are spread on the surface of the transfection device and incubated with the transfection reagent microarray. 25 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 or more wells of a multi-well plate may be coated with one or more transfection reagent(s). Plates commonly used in transfection 30 are 96-well and 384-well plates. The transfection reagent can be evenly applied to the bottom of each well in the multi-well plate. Generally, the transfection reagent is applied to the bottom of plate in the range of about 0.1 to about l00pg/cm 2 . Further, the coating -9- WO 2006/066001 PCT/US2005/045429 amount of the transfection reagent may be varied depending on the type of well plate to be used. For example, for a 6-well plate, 12-well plate or 96-well plate, the coating concentration of the transfection reagent is preferably from about 0.5 to about 50pg/cm 2 , and more preferably from about 1 to 20ptg/cm 2 . In the case of a 384-well plate, the coating 5 concentration of the transfection reagent is preferably from about 0.5 to about 50pg/cm 2 , and more preferably from about 1 to 30pg/cm2. In another embodiment of the invention, a 10-cm cell culture dish is coated with a transfection reagent. The transfection reagent can be evenly applied to the bottom of dish. The transfection reagent may be applied to the bottom of dish in the range of about 0.1 to about 1 00pg/cm 2 , more preferably about 0.2 to 10 about 20 tg/cm 2 . Hundreds of nucleic acids 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 15 laboratories. The multi-well plate coated with transfection reagent can be widely used in most laboratories to study gene regulation, gene function, molecular therapy, and signal transduction. Also, if different kinds of transfection reagents are coated on the different wells of multi-well plates, the plates can be used to screen many transfection or delivery reagents efficiently. Recently, 1,536 and 3,456 well plates have been developed, which 20 may also be used according to the methods described herein. In preferred embodiments, the transfection device is stored in a material suitable for packaging which may be plastic (e.g., cellophane), an elastomeric material, thin metal, Mylar@, or other polyester film material. The storage may be with or without oxygen and/or carbon dioxide absorbers. The transfection plates prepared as described herein may 25 be stored for at least 5 months at room temperature with retention of significant cell transfecting activity. The transfection reagent is preferably a cationic compound which can introduce biomolecules, such as nucleic acids into cells. Preferred embodiments use cationic oligomers, such as low molecular weight polyethyleneimine (PEI). More preferably, the 30 transfection agent is a degradable cationic polymer. Optionally, the transfection agent includes a cell-targeting or an intracellular-targeting moiety and/or a membrane destabilizing component, as well as delivery enhancers. -10- WO 2006/066001 PCT/US2005/045429 In general, delivery enhancers fall into two categories. These are viral carrier systems and non-viral carrier systems. As human viruses have evolved ways to overcome the barriers to transport into the nucleus discussed above, viruses or viral components are useful in transport of nucleic acid into cells. Additionally, the degradable polymers may be 5 conjugated to or associated with a viral or non-viral protein to enhance transfection efficiency. For example, vesicular stomatitis virus G protein (VSVG) and other peptides or proteins which are known to those of skill in the art may be added to the polymers in order to improve transfection efficiency. Another example of a viral component useful as a delivery enhancer is the 10 hemagglutinin peptide (HA-peptide). This viral peptide facilitates transfer of biomolecules into cells by endosome disruption. At the acidic pH of the endosome, this protein causes release of the biomolecule and carrier into the cytosol. Non-viral delivery enhancers may be either polymer-based or lipid-based. They are generally polycations which act to balance the negative charge of the nucleic acid. 15 Polycationic polymers have shown significant promise as non-viral gene delivery enhancers due in part to their ability to condense DNA plasmids of unlimited size and to safety concerns with viral vectors. Examples include peptides with regions rich in basic amino acids such as oligo-lysine, oligo-arginine or a combination thereof and polyethylenimine (PEI). These polycationic polymers facilitate transport by condensation of DNA. Branched 20 chain versions of polycations such as PEI and Starburst dendrimers can mediate both DNA condensation and endosome release (Boussif, et al. (1995) Proc. Natl. Acad. Sci USA vol. 92: 7297-7301). PEI is a highly branched polymer with terminal amines that are ionizable at pH 6.9 and internal amines that are ionizable at pH 3.9 and because of this organization, can generate a change in vesicle pH that leads to vesicle swelling and eventually, release 25 from endosome entrapment. Another means to enhance delivery is to design a ligand on the transfection reagent. The ligand must have a receptor on the cell that has been targeted. Biomolecule delivery into the cell is then initiated by receptor recognition. When the ligand binds to its specific cell receptor, endocytosis is stimulated. Examples of ligands which have been used with 30 various cell types to enhance biomolecule transport are galactose, transferrin, the glycoprotein asialoorosomucoid, adenovirus fiber, malaria circumsporozite protein, epidermal growth factor, human papilloma virus capsid, fibroblast growth factor and folic -11- WO 2006/066001 PCT/US2005/045429 acid. In the case of the folate receptor, the bound ligand is internalized through a process tended potocytosis, where the receptor binds the ligand, the surrounding membrane closes off from the cell surface, and the internalized material then passes through the vesicular membrane into the cytoplasm (Gottschalk, et al. (1994) Gene Ther 1:185-191). 5 Various agents have been used for endosome disruption. Besides the HA-protein described above, defective-virus particles have also been used as endosomolytic agents (Cotten, et al. (July 1992) Proc. Natl. Acad. Sci. USA vol. 89: pages 6094-6098). Non viral agents are either amphiphillic or lipid-based. The release of biomolecules such as DNA into the cytoplasm of the cell can be 10 enhanced by agents that either mediate endosome disruption, decrease degradation, or bypass this process all together. Chloroquine, which raises the endosomal pH, has been used to decrease the degradation of endocytosed material by inhibiting lysosomal hydrolytic enzymes (Wagner, et al. (1990) Proc Natl Acad Sci USA vol. 87: 3410-3414). Branched chain polycations such as PEI and starburst dendrimers also promote endosome release as 15 discussed above. To completely bypass endosomal degradation, subunits of toxins such as Diptheria toxin and Pseudomonas exotoxin have been utilized as components of chimeric proteins that can be incorporated into a gene/gene carrier complex (Uherek, et al.(1998) J Biol. Chem. vol. 273: 8835-8841). These components promote shuttling of the nucleic acid 20 through the endosomal membrane and back through the endoplasmic reticulum. Once in the cytoplasm, the nucleic acid must find its way to the nucleus. Localization to the nucleus may be enhanced by inclusion of a nuclear localization signal on the nucleic acid-carrier. A specific amino acid sequence that functions as a nuclear localization signal (NLS) is used. The NLS on a cargo-carrier complex interacts with a 25 specific nuclear transport receptor protein located in the cytosol. Once the cargo-carrier complex is assembled, the receptor protein in the complex is thought to make multiple contacts with nucleoporins, thereby transporting the complex through a nuclear pore. After a cargo-carrier complex reaches its destination, it dissociates, freeing the cargo and other components. 30 Subsequences from the SV40 large T-antigen has been used for transport into nuclei. This short sequence from SV40 large T-antigen acts as a signal that causes the transport of associated macromolecules into the nucleus. -12- WO 2006/066001 PCT/US2005/045429 Biodegradable cationic polymers typically exhibit low cytotoxicity, but also low transfection efficiency due to rapid degradation, making them less competitive against other carriers for gene transfer and other applications. These degradable cationic polymers improve transfection efficiency by linking low molecular weight cationic compounds or 5 oligomers together with degradable linkers. The linker molecules may contain biologically, physically or chemically cleavable bonds, such as hydrolysable bonds, reducible bonds, a peptide sequence with enzyme specific cleavage sites, pH sensitive, or sonic sensitive bonds. The degradation of these polymers may be achieved by methods including, but not limited to hydrolysis, enzyme digestion, and physical degradation methods, such as optical 10 cleavage (photolysis). One of the advantages of the degradable cationic polymers described herein is that degradation of the polymers is controllable in terms of rate and site of polymer degradation, based on the type and structures of the linkers. In preferred embodiments, the transfection reagent includes a plurality of cationic 15 molecules and at least one degradable linker molecule connecting said cationic molecules in a branched arrangement. Cationic oligomers, such as low molecular weight polyethyleneimine (PEI), low molecular weight poly(L-lysine) (PLL), low molecular weight chitosan, and low molecular weight PAMAM dendrimers, can be used to make the polymers described herein. 20 Furthermore, any molecule containing amines with more than three reactive sites can be used. Cationic oligomers may be selected from, but are not limited to: (i) a cationic compound of formula (A) or (B) or a combination thereof: 25 7R2 -R4 R -N\ R 6 -(-NH-R-)Ra R I 5 Formula B Formula A wherein R 1 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; 30 R 2 is a straight chain alkylene group of the formula: -(CH 2 )a- wherein a is an integer number from 2 to 10; -13- WO 2006/066001 PCT/US2005/045429
R
3 is a straight chain alkylene group of the formula: -(CbH2b)- wherein b is an integer number from 2 to 10;
R
4 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B;
R
5 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; 5 R 6 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, Formula A, or another Formula B;
R
7 a straight or branched chain alkylene group of the formula: -(CeH 2 c)- in which c is an integer number from 2 to 10; and R8 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, Formula A, or another Formula B; 10 (ii) a cationic dendritic or branched polyamidoamine (PAMAM) with terminated primary or secondary amino groups; (iii) a cationic polyamino acid; and (iv) a cationic polycarbohydrate. Examples of such cationic molecules include, but are not limited to, the cationic 15 molecules shown in Table 1. Table 1: Structures of cationic compounds and oligomers according to preferred embodiments of the invention Symbol Name Structure Pentaethylenehexamine H C1
H
2 N N NH 2 C2 Linear polyethylenimine (H Mw=423) N} Branched '5 C3 polyethylenimine NH H 2 N (Mw=600) H 2 N/r-N Branched ,-N C4 polyethylenimine '\NH NH (Mw-1200)
H
2 N
H
2 N N,N'-Bis(2-aminopropyl)- H C5 ethylenediamine
H
2 N -N N NH 2 H Spermine H C6
H
2 N ' N _-- N , NH 2 H C7 N-(2-aminoethyl)-1,3-
H
2 N N -,NH 2 propanediamine H -14- WO 2006/066001 PCT/US2005/045429 Symbol Name Structure C8 N-(3-aminopropyl)-1,3~ H2N N NH 2 propanediamine H N,N'-Bis(2-aminoethyl)- H H C9 1,3-propanediamine
H
2 N N N NH 2 C10 Poly(amidoamine) PAMAM Dendrimer C11 Poly(propyleneimine) DAB-Am-16 dendrimer C12 Spermidine H2NHNH2 1,4-Bis(3-aminopropyl) C13 piperazine C14 12- HN N NH 2 Aminoethyl)piperazine H 2
H
2 N N NH 2 C15 Tri(2-aminoethyl)amine
NH
2 C16 Poly(L-lysine) Cationic polymers used herein may include primary or secondary amino groups, which can be conjugated with active ligands, such as sugars, peptides, proteins, and other molecules. In a preferred embodiment, lactobionic acid is conjugated to the cationic 5 polymers. The galactosyl unit provides a useful targeting molecule towards hepatocyte cells due to the presence of galactose receptors on the surface of the cells. In a further embodiment, lactose is conjugated to the degradable cationic polymers in order to introduce galactosyl units onto the polymer. Degradable linking molecules include, but are not limited to, di- and multi 10 acrylates, di- and multi-methacrylates, di- and multi-acrylamides, di- and multi isothiocyanates, di- and multi-isocyanates, di- and multi-epoxides, di- and multi-aldehydes, di- and multi-acyl chlorides, di- and multi-sulfonyl chlorides, di- and multi-halides, di- and multi-anhydrides, di- and multi-malemides, di- and multi-carboxylic acids, di- and multi-a haloacetyl groups, and di- and multi-N-hydroxysuccinimide esters, which contain at least 15 one biodegradable spacer. The following formula describes a linker which may be used according to preferred embodiments: A(Z)d -15- WO 2006/066001 PCT/US2005/045429 wherein A is a spacer molecule having at least one degradable bond, Z is a reactive residue which reacts with amino group, and d is an integer equal to or more than two and wherein A and Z are bound covalently. 5 Several embodiments of reactive residues of the linker molecules have been illustrated in Table 2, however these examples are not limiting to the scope of the invention. Reactive residues may be selected from, but are not limited to, acryloyl, maleimide, halide, carboxyl acylhalide, isocyanate, isothiocyanate, epoxide, aldehyde, sulfonyl chloride, and N-hydroxysuccinimide ester groups or combinations thereof. 10 Table 2: Structures of biodegradable linker molecules used in preferred embodiments of the invention Symbol Name Structure 1,3-Butanediol 0 0 Li diacrylate H 2 C=HC-C-0-CHCH 2
CH
2 -0-C-CH=CH 2 CH, 2-Methyl-2,4- 0 CH3 0 L2 pentanediol diacrylat H 2 C=HC-C-0-CHCH 2
C--OC-CH=CH
2
CH
3
CH
3 Trimethylolpropane 0 L3 triacrylate
CH
3
CH
2 C0OCH 2
~C-CH=CH
2
)
3 2,4-Pentanediol 0 0 L4 diacrylate H 2 C=HC-C-0-CHCH 2 CH-0-C-CH=CH 2
CH
3
OH
3 Pentaerythritol 0 L5 tetraacrylate C CH 2 0-C--CH=CH 2
)
4 Dipentaerythritol 0 pentaacrylate 2
(H
2 C=HC-c-OH 2 C H 2
H
2 11 L6
HOH
2 pna a c-C-0-C-C(CH 2 0-C-CH=CH 2
)
3 Di(trimethylolprop 0 ane) tetraacrylate H 2 C=HC-C0-OH 2 Cx H 2
/CH
2 0-0-CH=CH 2 L7 C 2
H
5
~C-CH
2
-OC-C-C
2
H
5
H
2
C=HC-C-OH
2 C CH 2 0-C-CH=CH 2 o 0 1,4-Butanediol 0I diacrylate H 2 C=HC-C-0-CH 2
CH
2
CH
2
CH
2 -0-C-CH=CH 2 1,6-Hexanediol diacrylate H 2 C=HC-C-0-CH 2
(CH
2
)
4
CH
2
-O-C-CH=CH
2 2,5-Dimethyl-2,5- 0 OH 3
CH
3 0 Li0 hexanediol H 2 C=HC-C-0-CCH 2
CH
2
C-O-C-CH=CH
2
CH
3
CH
3 -16- WO 2006/066001 PCT/US2005/045429 Symbol Name Structure diacrylate The degradation rates of the polymers can be controlled by changing the polymer composition, feed ratio, and the molecular weight of the polymers. For example, when linkers with bulkier alkyl groups are used, the polymers will degrade slower. Also, 5 increasing molecular weight will cause a decrease in the degradation rate in some cases. Degradation rates of the polymers may be controlled by adjusting the ratio of cationic polymer to linker or by changing the various degradable linker molecules. Acrylate linkers are much cheaper than disulfide-containing linkers, because synthesis of the disulfide-containing linkers is more difficult. Acrylate linkers can be 10 hydrolysable in any water solution. Therefore a polymer containing acrylate linkers can be degraded in various environments as long as it contains water. Thus, polymers containing acrylate linkers have broad applications compared to disulfide-linker-containing polymers. In addition, the degradation rate of polymers with disulfide-linkers are usually the same, but the degradation rate of polymers synthesized with acrylate linkers can vary depending on 15 the different acrylate linkers used. In some embodiments, the transfection 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 20 and biocompatible materials. The polysaccharide can be any compound that can form a membrane and coat the delivery reagent, such as 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 25 reagent. The transfection agent may comprise both a degradable cationic polymer and a non-degradable cationic polymer. The ratio of the non-degradable cationic polymer to the degradable cationic polymer is preferably from 1:0.5 to 1:20 (non-degradable : degradable) by weight, and more preferably from 1:2 to 1:10 by weight. According to another embodiment, a gelatin-transfection reagent mixture, 30 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 -17- WO 2006/066001 PCT/US2005/045429 double demonized water, may be affixed to the transfection device. In a further embodiment a cell culture reagent (e.g., fibronectin, collagen, salts, sugars, protein, or peptides) may also be present in the gelatin-transfection reagent mixture. The mixture is evenly spread onto a surface, such as a slide or multi-well plate, thus producing a transfection surface bearing the 5 gelatin-transfection reagent mixture. In alternative embodiments, different transfection reagent-gelatin mixtures may also be spotted in 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 can be dried at specific 10 temperatures or humidity or in a vacuum-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 keeping cytotoxicity at an acceptable level, the concentration of 15 transfection reagent is at the optimal level. The concentration of transfection reagent will generally be in the range of about 1.0 ptg/ml to about 1000 ig/ml. In preferred embodiments, the concentration is from about 10 Rg /ml to about 600 pg/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% (w/v) of 20 the transfection reagent solution. In preferred embodiments, the molecules to be introduced into cells are nucleic acids. The nucleic acid can be DNA, RNA, DNA/RNA hybrid, peptide nucleic acid (PNA), etc. If the DNA used is present in a vector, the vector can be of any type, such as a plasmid (e.g., plasmid carrying green fluorescence protein (GFP) gene and/or luciferase (luc) gene) 25 or viral-based vector (e.g. pLXSN). The DNA can also be linear fragment with a promoter sequence (such as 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 a single strand oligodeoxynucleotide (ODN), for example antisense ODN, it can be introduced into cells to 30 regulate target gene expression. In embodiments using RNA the nucleic acid may be single stranded (antisense RNA and ribozyme) or double stranded (RNA interference, SiRNA). In most cases, the RNA is modified in order to increase the stability of RNA and improve its -18- WO 2006/066001 PCT/US2005/045429 function in down regulation of gene expression. In peptide nucleic acid (PNA), the nucleic acid backbone is replaced by peptide, which makes the molecule more stable. The methods described herein can be used to introduce nucleic acids into cells for various purposes, for example molecular therapy, protein function studies, or molecule mechanism studies. 5 Under appropriate conditions, a nucleic acid solution is added into the transfection device, which has been coated with the transfection reagent, to form a nucleic acid transfection reagent complex. The nucleic acids are preferably dissolved in cell culture medium without fetal bovine serum and antibiotics, for example Dulbecco's Modified Eagles Medium (DMEM). However, any appropriate cell culture media may be used 10 including, but not limited to, Minimum Essential Eagle, F-12 Kaighn's Modification medium, or RPMI 1640 medium. If the transfection reagent is evenly affixed on the slide, the nucleic acid solution can be spotted onto discrete locations on the slide. Alternatively, transfection reagents may be spotted on discreet locations on the slide, and the nucleic acid solution can simply be added to cover the whole surface of the transfection device. If the 15 transfection reagent is affixed on the bottom of multi-well plates, the nucleic acid solution is simply added into different wells by multi-channel pipette, automated device, or other delivery methods which are well known in the art. The resulting product (transfection device coated with transfection reagent and desired nucleic acid) is incubated for approximately 5 min. to 60 min., more preferably, from 25-30 minutes at room 20 temperature to form the nucleic acid/transfection reagent complex. In some embodiments, for example, if different nucleic acid samples are spotted on discrete locations of the slide, the DNA solution will be removed to produce a surface bearing the nucleic acid samples in complex with the transfection reagent. In other alternate embodiments, the nucleic acid solution can be kept on the surface. Secondly, cells in an appropriate medium, such as 25 DMEM, 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 nucleic acids of interest into the plated cells. In alternate embodiments, the cells are mixed with the biomolecule or nucleic acid. The cell/biomolecule mixture is then added to the transfection device and incubated at room temperature. 30 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. In preferred embodiments, eukaryotic cells, such as mammalian cells -19- WO 2006/066001 PCT/US2005/045429 (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 reagent and nucleic acids of interest, in sufficient density and under appropriate conditions for introduction/entry of the nucleic acids of interest into the eukaryotic cells and either 5 expression of the DNA or interaction of the biomolecule with cellular components. In particular embodiments the cells may be 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. In preferred embodiments, eukaryotic cells are grown in Dulbecco's Modified 10 Eagles Medium (DMEM) containing 10% heat-inactivated fetal bovine serum (FBS) with L-glutamine and penicillin/streptomycin (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. Appropriate media for culture of particular cell types are known to those of skill in the art. The optimal density of cells 15 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. For example, if 5x 104 293 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 lx104 cells/well are needed to reach a similar confluent 20 percentage in a 96 well plate. After the cells are seeded on the surface containing the nucleic acid samples/transfection reagent, the cells are incubated under optimal conditions for the cell type (e.g. 37*C, 5-10% C0 2 ). 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 25 the case of gene transfection experiments. In the analysis of intracellular trafficking 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 30 level can be detected by reporter genes, such as green fluorescent protein (GFP) gene, luciferase gene, or P-galactosidase gene expression. The signal of GFP can be directly observed under a microscope, the activity of luciferase can be detected by a luminometer, -20- WO 2006/066001 PCT/US2005/045429 and the blue product catalyzed by p-galactosidase can be observed under a 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, or other biomolecules delivered according to methods 5 described herein and the target modulated by these biomolecules can be determined by various methods, such as detecting immunofluorescence or enzyme immunocytochemistry, autoradiography, or in situ hybridization. If immunofluorescence is used to detect expression of an 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 10 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 15 resulting from delivery of the biomolecules. Example 1 Preparation of Degradable Cationic Polymer The synthesis of a polymer which is derived from polyethylenimine oligomer with molecular weight of 600 (PEI-600) and 2,4-pentandiol diacrylate (PDODA) is provided as a 20 general procedure for preparation of a degradable cationic polymer. To a vial, 4.32 g of PEI-600 in 25 ml of methylene chloride were added by using pipette or syringe. 2.09 g of PDODA was quickly added to the above PEI-600 solution with stirring. The reaction mixture was stirred for 4 hours at room temperature (20 C). Then, the reaction mixture was neutralized by adding 50 ml of 2M HCl. The white precipitate was centrifuged, washed 25 with methylene chloride, and dried at room temperature under reduced pressure. Example 2 Preparation of Transfectable Cell Culture Device with Degradable Cationic Polymer Degradable cationic polymer was prepared as indicated in Example 1. Linear polyethyleneimine (L-PEI) based polymer and lipid based polymers were used for 30 transfecting plasmid DNA into mammalian cells in vitro to evaluate the transfection efficiency. For L-PEI based polymer, jet PEI (Qbiogene) transfection reagent was used. Lipofectamine2000 (Invitrogen) and N-[1-(2,3-dioleoyloxy)propyl]-N,N,N -21- WO 2006/066001 PCT/US2005/045429 trimethylammonium salts (DOTAP; Sigma-Aldrich) were employed as lipid based polymers. Degradable cationic polymer and DOTAP were dissolved in methanol, and jet PEI and Lipofectamine2000 were diluted by deionized water. Flat bottom 96-well cell culture plates (bottom surface: 0.32 cm2 per each well; BD Biosciences) were treated with 5 these polymer solutions. The actual amounts affixed on the bottom were as follows: (a) Degradable cationic polymer; 3 pg per well, thus 9.4 pig/cm 2 , (b) jet PEI; 1 pl per well, (c) Lipofectamine2000; 0.375 gg per well, (d) DOTAP; 2 and 4 pmole per well. These plates were dried at room temperature under reduced pressure and sealed in a vacuum pack until use. 10 Example 3 Transfection with Transfectable Cell Culture Device for 293 Cells 25 or 50 ng of pEGFP-N1 plasmid (purchased from Clontech) in 25 pl of opti MEM I (Invitrogen) was added in each well and kept at room temperature for 25 minutes. 15 Then, 5 X 10 4 of 293 cells in 100 pl of Dulbecco's modified Eagle Medium (DMEM) (Invitrogen) with 10% calf serum (Invitrogen) were added and incubated at 37'C in 7.5% of
CO
2 . After 24 to 36 hrs. incubation, transfection efficiency was estimated by observing EGFP fluorescence by using epifluorescent microscope (1X70, Olympus). Transfection efficiencies are shown in Table 3. Degradable cationic polymer and jet 20 PEI, i.e. L-PEI based polymer showed high transfection efficiency. Table 3 Polymer EGFP-positive cells Degradable cationic polymer 60 -70% Jet PEI 60% Upofectamine2000 -Less than 10% DOTAP 4 pmole/well 0% DOTAP 2 pmole/well 0% Example 4 Evaluation of Cytotoxicity 25 Cytotoxicity of the described method was evaluated. Cell shape of 293 cells, transfected as indicated in Example 3, were compared by microscopic observation (Figure 1). Cells transfected by using degradable polymer showed normal shape, which was similar -22- WO 2006/066001 PCT/US2005/045429 to intact 293 cells. However, those transfected by using L-PEI based polymer (jet PEI) and lipid based polymer (Lipofectamine2000) were rounded. We concluded that the degradable cationic polymer can deliver genes without damaging cells. 5 Example 5 Optimization of Degradable Cationic Polymer Amount Various amounts of degradable cationic polymer were affixed on the cell culture devices, and transfection efficiency was evaluated. 96-well cell culture plates were coated 10 with degradable cationic polymer by the same protocol as shown in Example 2. The actual amount of polymer was as follows: 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 10 and 20 pg per well. Then, transfection was carried out as described in Example 3 and transfected cells were incubated at 37'C in 7.5% of CO 2 . Amount of plasmid DNA added before seeding cells was 0.13, 0.25, 0.50 or 1.0 pg per well. After 40 hours incubation, percentage of 15 fluorescing cells and cell condition were estimated by epifluorescent microscopy. Figure 2 shows percentages of EGFP-positive cells after transfection. High transfection efficiencies were allocated between 2.5 to 5.0 pg per well (thus, 7.8 to 16 pg/cm 2 ) of degradable cationic polymer with 0.25 and 0.5 pg per well (thus, 0.78 to 1.6 pg/cm 2 ) of plasmid DNA. Cell condition in these experiments is shown in Figure 3. Cells transfected in the 20 plates with L-PEI and lipid based polymers had rounded shape and had aggregated. The morphology was due to cytotoxicity. Cell condition was acceptable when the amount of degradable cationic polymer affixed on the bottom of the plate was from 2.5 to 5.0 pg per well. Also, all the plasmid DNA conditions that we tested gave us good cell condition with degradable cationic polymer if the amount was from 2.5 to 5.0 pg per well. 25 Example 6 Stability Stndy There are products in the market, in which there is a coating on the surface of cell culture devices for a special purpose, for example, to assist cell growth. Normally, the 30 coating material is a kind of protein, like collagen or fibronectin. As they are temperature sensitive, these cell culture devices require refrigerated storage which is a disadvantage, especially if they are bulky. For this reason, stability at room temperature is an important feature. -23- WO 2006/066001 PCT/US2005/045429 The cell culture/transfection devices of this invention were tested to study their stability after long-term storage. The cell transfection devices were prepared as described in Example 2, and vacuum-sealed in Mylar Bags (Dupont Corp.), which is a film with an oxygen barrier material and aluminum foil with or without oxygen and carbon dioxide 5 absorbers. Storage was at 25*C. Then, transfection efficiency with plasmid DNA carrying luciferase gene (pCMV-LUC) was tested periodically. The procedure for transfectable cell culture devices was as described in Example 3 except the plasmid DNA was different. Luciferase activity of cells were determined by using a Dynex MLX Microtiter @ plate luminometer and Luciferase Assay System (Promega Corp. Madison, WI USA) to 10 determine transfection efficiency. Figures 4, 5 and 6 show change of transfection efficiencies after storage at 25 C with 02 and/or CO 2 absorbing materials in Mylar Bags. There was no obvious decrease of transfection efficiency after 5 month storage. Moreover, even when cell culture devices were kept at 250 C in Mylar Bags without 02 and/or CO 2 absorbing materials, transfection 15 efficiency was stable after 5 month and still quite high (Figure 7). The cell culture devices of this invention are quite stable at room temperature. The device can be stored without special storage conditions. Example 7 20 Preparation of Non-degradable Cationic Polymer Non-degradable polymer was prepared as follows: Approximately 5 g of polyethlenimine (Aldrich, product number: 408727) was dissolved in 50 ml of dichloromethane, then 100 ml of 2.OM hydrogen chloride in diethyl ether (Aldrich, product number: 455180) was added and mixed well to form polymer hydrochloride. Then, the 25 polymer hydrochloride was collected by centrifuge, and rinsed with 150 ml of diethyl ether. This rinse with diethyl ether was carried out twice. The resultant precipitation after the rinse was dried under vacuum condition at room temperature for 3 hours. Then, the dried powder was stored at 4 "C with desiccant until use. -24- WO 2006/066001 PCT/US2005/045429 Example 8 Preparation of 96-well Transfectable Cell Culture Device with Degradable Cationic Polymer and Non-degradable Cationic Polymer Degradable cationic polymer was prepared as indicated in Example 1. Non 5 degradable cationic polymer was obtained as described in Example 7. Both polymers were dissolved in methanol and mixed together to make a coating solution. The final concentration of each polymer was: Degradable cationic polymer; 40 gg/ml, and Non degradable cationic polymer; 10 pg/ml. Then, flat bottom 96-well cell culture plates (bottom surface: 0.32 cm 2 per each well; BD Biosciences) were treated with the coating 10 solution. Actually, 25 pl of the coating solution was dispensed in each well, and dried under vacuum condition to remove methanol. Under these coating conditions, 1 pg of degradable cationic polymer was affixed on each well of a 96-well plate; therefore the density of the degradable cationic polymer was 3.1 [g/cm 2 . Also, 0.25 ig of non degradable cationic polymer was affixed on each well of the 96-well plate so that the 15 density of the non-degradable cationic polymer was 0.78 pLg/cm 2 . In total, 1.25pig of polymer was affixed on each well of the 96-well plate; therefore the density of polymer was 3.9 pig/cm 2 . The cell culture devices prepared in this example were vacuum sealed in Mylar Bags with desiccant, and stored at room temperature until further use. 20 Example 9 Preparation of 12-well Transfectable Cell Culture Device with Degradable Cationic Polymer and Non-degradable Cationic Polymer Degradable cationic polymer was prepared as indicated in Example 1. Non degradable cationic polymer was obtained as described in Example 7. Both polymers were 25 dissolved in methanol and mixed together to make a coating solution. The final concentration of each polymer was: Degradable cationic polymer; 80 pg/ml, and Non degradable cationic polymer; 10 pLg/ml. Then, flat bottom 12-well cell culture plates (bottom surface: 3.8 cm 2 per each well; BD Biosciences) were treated with these polymer solutions. 100 pl of the coating solution was dispensed in each well, and dried under 30 vacuum condition to remove methanol. Under these coating conditions, 8.0 pg of degradable cationic polymer was affixed on each well of a 12-well plate so that the density of the degradable cationic polymer was 2.1 ptg/cm 2 and 1.0 gg of non-degradable cationic -25- WO 2006/066001 PCT/US2005/045429 polymer was affixed on each well of the 12-well plate so that the density of the non degradable cationic polymer was 0.26 jig/cm 2 . In total, 9.0 Rg of polymer was affixed on each well of the 12-well plate; therefore the density of polymer was 2.4 pig/cm 2 . The cell culture devices prepared in this example were vacuum sealed in Mylar Bags with desiccant, 5 and stored at room temperature until further use. Example 10 Preparation of 6-well Transfectable Cell Culture Device with Degradable Cationic Polymer and Non-degradable Cationic Polymer 10 Degradable cationic polymer was prepared as indicated in Example 1. Non degradable cationic polymer was obtained as described in Example 7. Both polymers were dissolved in methanol and mixed together to make a coating solution. The final concentration of each polymer was: Degradable cationic polymer; 80 pg/ml, and Non degradable cationic polymer; 10 pg/ml. Then, flat bottom 6-well cell culture plates (bottom 15 surface: 9.6 cm 2 per each well; BD Biosciences) were treated with the coating solution. 200 jil of the coating solution was dispensed in each well, and dried under vacuum condition to remove methanol. Under these coating conditions, 16 pig of degradable cationic polymer was affixed on each well of a 6-well plate so that the density of the degradable cationic polymer was 1.7 jg/cm 2 and, 2.0 jg of non-degradable cationic polymer was affixed on 20 each well of the 6-well plate so that the density of the non-degradable cationic polymer was 0.21 jig/cm 2 . In total 18 jig of polymer was affixed on each well of the 6-well plate; therefore the density of polymer was 1.9 pg/cm 2 . The cell culture devices prepared in this example were vacuum sealed in Mylar Bags with desiccant, and stored at room temperature until further use. 25 Example 11 Transfection with 96-well Transfectable Cell Culture Devices Prepared with Degradable and Non-degradable Cationic Polymers Mammalian cells were incubated in 10-cm cell culture dishes, rinsed with 30 phosphate-buffered saline, and treated with trypsin solution. Then, the trypsinized cells were diluted in appropriate cell culture medium with serum to prepare a cell suspension. The cell density used in this example is shown in Table 4. -26- WO 2006/066001 PCT/US2005/045429 pEGFP-Nl plasmid was diluted in opti-MEM, and the final concentration was adjusted to 10 pg/ml. Then, 25 I of the plasmid solution was added in each well of the 96 well transfectable cell culture device prepared as indicated in Example 8, and kept at room temperature for 25 minutes. Then, 100 l of the cell suspension was added in the well, and 5 incubated at 37'C in 7.5% of CO 2 . After 36 to 48-hour incubation, transfection efficiency was estimated by observing EGFP fluorescence by using epifluorescent microscope (IX70, Olympus). Table 4 indicates the percentage of the cells with EGFP fluorescence in various mammalian cell lines. The 96-well transfectable cell culture device in this invention 10 transfected various mammalian cell lines with high efficiency. Example 12 Transfection with 12-well Transfectable Cell Culture Devices Prepared with Degradable and Non-degradable Cationic Polymers 15 Manmmalian cells were incubated in 10-cm cell culture dishes, rinsed with phosphate-buffered saline, and treated with trypsin solution. Then, the trypsinized cells were diluted in appropriate cell culture medium with serum to prepare cell suspension. The cell density used in this example is shown in Table 4. pEGFP-N1 plasmid was diluted in opti-MEM, and the final concentration was 20 adjusted to 5 gg/ml. Then, 200 [L1 of the plasmid solution was added in each well of the 12 well transfectable cell culture device prepared as indicated in Example 9, and kept at room temperature for 25 minutes. Then, 1 ml of the cell suspension was added in the well, and incubated at 37'C in 7.5% of CO 2 . After 36 to 48-hour incubation, transfection efficiency was estimated by observing EGFP fluorescence by using epifluorescent microscope (IX70, 25 Olympus). Table 4 indicates the percentage of the cells with EGFP fluorescence in various mammalian cell lines. The 12-well transfectable cell culture device in this inventiontransfected various mammalian cell lines with high efficiency. -27- WO 2006/066001 PCT/US2005/045429 Example 13 Transfection with 6-well Transfectable Cell Culture Devices Prepared with Degradable and Non-degradable Cationic Polymers Mammalian cells were incubated in 10-cm cell culture dishes, rinsed with 5 phosphate-buffered saline, and treated with trypsin solution. Then, the trypsinized cells were diluted in appropriate cell culture medium with serum to prepare cell suspension. The cell density used in this example is shown in Table 4. pEGFP-Nl plasmid was diluted in opti-MEM, and the final concentration was adjusted to 5 ig/ml. Then, 400 pl of the plasmid solution was added in each well of the 6 10 well transfectable cell culture device prepared as indicated in Example 10, and kept at room temperature for 25 minutes. Then, 2 ml of the cell suspension was added in the well, and incubated at 37*C in 7.5% of CO 2 . After 36 to 48-hour incubation, transfection efficiency was estimated by observing EGFP fluorescence by using epifluorescent microscope (IX70, Olympus). 15 Table 4 indicates the percentage of the cells with EGFP fluorescence in various mammalian cell lines. The 6-well transfectable cell culture device in this invention transfected various mammalian cell lines with high efficiency. Table 4 Percentage of cells with fluorescence, and initial cell density 20 % EGFP Initial Cell Density (cells/ml) Cell Line 6-well 12-well 96-well 6-well 12-well 96-well 293 80 80 80 2.5x10 5 2.5x10 5 2.5x105 705 80 80 80 1.5x1 05 1.5x1 05 1.5x1 05 COS-7 70 70 70-80 1.5-2.Ox105 1.5x10 5 1.5x105 HT-1080 70-80 70 70 0.5-1.0x10 5 0.5x10 5 1.0x10 5 HeLa 70 80 70 1.0-2.0x10 5 1.0x10 5 0.5x10 5 25 MDCK 50 60 1.0x1 05 1.5x1 05 CHO-K1 30-40 50 50 1.5x10 5 2.0x10 5 2 .0x105 DU145 30-40 40 30-40 1.5-2.0x10 5 1.5x105 1.5x105 A549 20-30 20-30 30-40 2.0x105 2.0x105 2.0x105 CV-1 20-30 30 20-30 1.0x10 5 1.5x10 5 1.5x105 HepG2 20 30 10-20 1.0-2.0x10 5 1.5x105 1.5x105 30 It will be understood by those of skill in the art that numerous and various modifications can be made without departing from the spirit of the present invention. -28- Therefore, it should be clearly understood that the forms of the present invention are illustrative only and are not intended to limit the scope of the present invention. Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps. The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates. -29-

Claims (30)

1. A device for cell transfection comprising a solid support coated only with a composition consisting of a transfection reagent which is not complexed to a biomolecule, 5 wherein the transfection reagent comprises a plurality of cationic molecules and at least one degradable linker molecule connecting said cationic molecules in a branched arrangement, wherein said cationic molecules are selected from the group consisting of: (i) a cationic compound of formula (A) or (B) or a combination thereof: R- -N\ R 6 -(NH-R 7 -)-R 8 R 3 j-R 5 Formula B 10 Formula A Formula A wherein R' is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; R 2 is a straight chain alkylene group of the formula: -(CH2)a- wherein a is an integer 15 number from 2 to 10; R 3 is a straight chain alkylene group of the formula: -(CbH2b)- wherein b is an integer number from 2 to 10; R 4 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; 20 R 5 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, another Formula A, or Formula B; R6 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, Formula A, or another Formula B; R 7 is a straight chain alkylene group of the formula: -(CeH 2 c)- in which c is an integer 25 number from 2 to 10; and -30- R 8 is a hydrogen atom, an alkyl of 2 to 10 carbon atoms, Formula A, or another Formula B; (ii) a cationic dendritic or branched polyamidoamine (PAMAM) with terminated primary or secondary amino groups; 5 (iii) a cationic polyamino acid; and (iv) a cationic polycarbohydrate; and wherein said degradable linker molecule is represented by the formula: A(Z)d wherein A is a spacer molecule having at least one degradable bond, Z is a reactive 10 residue which reacts with amino group, and d is an integer equal to or more than two and wherein A and Z are bound covalently, and wherein the device is capable of storage at room temperature for at least 5 months without significant loss of transfection activity. 15
2. The device of claim 1, wherein the solid support is selected from the group consisting of polystyrene resin, epoxy resin and glass.
3. The device of claim 1, wherein the coating is on the surface of the solid support. 20
4. The device of claim 3, wherein the coating amount of the transfection reagent is from about 0.1 to about 100 pg/cm 2 .
5. The device of claim 1, wherein the transfection agent is a cationic polymer.
6. The device of claim 1, wherein the transfection agent comprises a degradable 25 cationic polymer.
7. The device of claim 6, wherein the degradable cationic polymer comprises cationic compounds or oligomers linked together by one or more degradable linkers. 30
8. The device of claim 6, wherein the transfection agent further comprises a non degradable cationic polymer. -31 -
9. The device of claim 8, wherein the ratio of the non-degradable cationic polymer to the degradable cationic polymer is from 1:0.5 to 1:20 by weight. 5
10. The device of claim 7, wherein the cationic compound or oligomer is selected from the group consisting of poly(L-lysine) (PLL), polyethyleneimine (PEI), polypropyleneim ine (PPI), pentaethyleneamine, N,N'-bis(2-aminoethyl)-1,3- propanediamine, N,N'-bis(2-aminopropyl)-ethylenediamine, spermine, spermidine, N-(2- aminoethyl)-1,3 propanediamine, N-(3-aminopropyl)-l,3-propanediamine, tri(2- aminoethyl)amine, 1,4-bis(3 10 aminopropyl)piperazine, N-(2-aminoethyl)piperazine, dendritic polyamidoamine (PAMAM), chitosan, and poly(2-dimethylamino)ethyl methacrylate (PDMAEMA).
11. The device of claim 7, wherein the linker molecule is selected from the group consisting of di- and multi-acrylates, di- and multi-acrylamides, di- and multi- isothiocyanates, 15 di- and multi-isocyanates, di- and multi-epoxides, di- and multi-aldehydes, di-and multi-acyl chlorides, di- and multi-sulfonyl chlorides, di- and multi-halides, di- and multi-anhydrides, di- and multi-maleimides, di- and multi-N-hydroxysuccinimide esters, di- and multi-carboxylic acids, and di-and multi-a-haloacetyl groups. 20
12. The device of claim 7, wherein the linker molecule is selected from the group consisting of 1,3-butanediol diacrylate, 1,4-butanediol diacrylate, 1,6-hexanediol diacrylate, 2,4-pentanediol diacrylate, 2-methyl-2,4-pentanediol diacrylate, 2,5-dimethyl-2,5- hexanediol diacrylate, poly(ethylene glycol) diacrylate, trimethylolpropane triacrylate, pentaerythritol tetraacrylate, di(trimethylolpropane) tetraacrylate, dipentaerythritol pentaacrylate, and a 25 polyester with at least three acrylate or acrylamide side groups.
13. The device of claim 7, wherein the molecular weight of the polymer is from 500 da to 1,000,000 da. 30
14. The device of claim 7, wherein the molecular weight of the polymer is from 2000 da to 200,000 da. - 32 -
15. The device of claim 7, wherein the molecular weight of the cationic compound or oligomer is from 50 da to 10,000 da. 5
16. The device of claim 7, wherein the molecular weight of the linker molecule is 100 da to 40,000 da.
17. The device of claim 1, wherein the solid support is a dish bottom, a multi-well plate, or a continuous surface. 10
18. A method of cell transfection comprising: adding a solution comprising a nucleic acid to be transfected to the device of claim 1; adding eukaryotic cells to the device; and incubating the cells and the nucleic acid solution to allow cell transfection. 15
19. The method of claim 18, wherein the incubation is for 5 min. to 3 hours.
20. The method of claim 18, wherein the incubation is for 10 min. to 90 min. 20
21. The method of claim 18, wherein the nucleic acid is selected from the group consisting of DNA, RNA, DNA/RNA hybrid and chemically-modified nucleic acid.
22. The method of claim 21, wherein the DNA is circular plasmid, linear, fragment or single strand oligodeoxynucleotide . 25
23. The method of claim 21, wherein the RNA is single strand ribozyme or double strand siRNA.
24. The method of claim 18, wherein the cell is a mammalian cell. 30
25. The method of claim 18, wherein at least some of the cells undergo cell division. -33 -
26. The method of claim 18, wherein the cell is a transformed or primary cell.
27. The method of claim 18, wherein the cell is a somatic or stem cell. 5
28. The method of claim 18, wherein the cell is a plant cell.
29. The method of claim 18, wherein the solution is added to the device after storage of the device for at least 5 months. 10
30. A device according to any one of claims 1-17, or a method according to any one of claims 18-29, substantially as hereinbefore described with reference to the figures and/or examples. -34 -
AU2005316501A 2004-12-17 2005-12-14 Immobilized degradable cationic polymer for transfecting eukaryotic cells Ceased AU2005316501B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US63734404P 2004-12-17 2004-12-17
US60/637,344 2004-12-17
PCT/US2005/045429 WO2006066001A2 (en) 2004-12-17 2005-12-14 Immobilized degradable cationic polymer for transfecting eukaryotic cells

Publications (2)

Publication Number Publication Date
AU2005316501A1 AU2005316501A1 (en) 2006-06-22
AU2005316501B2 true AU2005316501B2 (en) 2010-10-21

Family

ID=36353906

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005316501A Ceased AU2005316501B2 (en) 2004-12-17 2005-12-14 Immobilized degradable cationic polymer for transfecting eukaryotic cells

Country Status (7)

Country Link
EP (1) EP1836293A2 (en)
JP (1) JP4739352B2 (en)
KR (2) KR20130010036A (en)
CN (1) CN101124316A (en)
AU (1) AU2005316501B2 (en)
CA (1) CA2590820A1 (en)
WO (1) WO2006066001A2 (en)

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6692700B2 (en) 2001-02-14 2004-02-17 Handylab, Inc. Heat-reduction methods and systems related to microfluidic devices
US7829025B2 (en) 2001-03-28 2010-11-09 Venture Lending & Leasing Iv, Inc. Systems and methods for thermal actuation of microfluidic devices
US8895311B1 (en) 2001-03-28 2014-11-25 Handylab, Inc. Methods and systems for control of general purpose microfluidic devices
US6852287B2 (en) 2001-09-12 2005-02-08 Handylab, Inc. Microfluidic devices having a reduced number of input and output connections
US7010391B2 (en) 2001-03-28 2006-03-07 Handylab, Inc. Methods and systems for control of microfluidic devices
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
EP2407243B1 (en) 2003-07-31 2020-04-22 Handylab, Inc. Multilayered microfluidic device
US8852862B2 (en) 2004-05-03 2014-10-07 Handylab, Inc. Method for processing polynucleotide-containing samples
AU2005269527B2 (en) 2004-07-26 2011-12-01 Pfenex Inc. Process for improved protein expression by strain engineering
US7358223B2 (en) 2004-10-04 2008-04-15 Nitto Denko Corporation Biodegradable cationic polymers
US9572886B2 (en) 2005-12-22 2017-02-21 Nitto Denko Corporation Agent for treating myelofibrosis
ES2692380T3 (en) 2006-03-24 2018-12-03 Handylab, Inc. Method to perform PCR with a cartridge with several tracks
US7998708B2 (en) 2006-03-24 2011-08-16 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US11806718B2 (en) 2006-03-24 2023-11-07 Handylab, Inc. Fluorescence detector for microfluidic diagnostic system
US10900066B2 (en) 2006-03-24 2021-01-26 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
US8765076B2 (en) 2006-11-14 2014-07-01 Handylab, Inc. Microfluidic valve and method of making same
WO2008060604A2 (en) 2006-11-14 2008-05-22 Handylab, Inc. Microfluidic system for amplifying and detecting polynucleotides in parallel
JP5444553B2 (en) * 2007-04-27 2014-03-19 フェネックス インコーポレイテッド Methods for rapidly screening microbial hosts to identify specific strains with improved yield and / or quality of heterologous protein expression
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US8105783B2 (en) 2007-07-13 2012-01-31 Handylab, Inc. Microfluidic cartridge
US9618139B2 (en) 2007-07-13 2017-04-11 Handylab, Inc. Integrated heater and magnetic separator
US8133671B2 (en) 2007-07-13 2012-03-13 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US8182763B2 (en) 2007-07-13 2012-05-22 Handylab, Inc. Rack for sample tubes and reagent holders
US8324372B2 (en) * 2007-07-13 2012-12-04 Handylab, Inc. Polynucleotide capture materials, and methods of using same
US9186677B2 (en) 2007-07-13 2015-11-17 Handylab, Inc. Integrated apparatus for performing nucleic acid extraction and diagnostic testing on multiple biological samples
US8287820B2 (en) 2007-07-13 2012-10-16 Handylab, Inc. Automated pipetting apparatus having a combined liquid pump and pipette head system
USD787087S1 (en) 2008-07-14 2017-05-16 Handylab, Inc. Housing
EP2697657B8 (en) 2011-04-15 2017-08-16 Becton, Dickinson and Company Scanning real-time microfluidic thermocycler and methods for synchronized thermocycling and scanning optical detection
USD692162S1 (en) 2011-09-30 2013-10-22 Becton, Dickinson And Company Single piece reagent holder
EP3273253B1 (en) 2011-09-30 2020-08-26 Becton, Dickinson and Company Unitized reagent strip
CN104040238B (en) 2011-11-04 2017-06-27 汉迪拉布公司 Polynucleotides sample preparation apparatus
JP6262152B2 (en) 2012-02-03 2018-01-17 ベクトン・ディキンソン・アンド・カンパニーBecton, Dickinson And Company External file for distribution of molecular diagnostic tests and determination of compatibility between tests
CN102827756B (en) * 2012-06-25 2014-04-09 华南农业大学 Agrobacterium local invasion method applied in soybean hypocotyl explant transformation system, and device thereof
CN105579584B (en) * 2013-06-28 2020-08-28 埃泽瑞斯公司 Compositions for introducing RNA into cells
US20180214572A1 (en) * 2014-11-10 2018-08-02 Ethris Gmbh Induction of osteogenesis by delivering bmp encoding rna
UA127540C2 (en) * 2016-10-05 2023-10-04 Сінгента Партісіпейшнс Аг Improvements in or relating to gene silencing
CN109735573A (en) * 2019-02-19 2019-05-10 武汉普诺赛生命科技有限公司 A kind of transient transfection reagent and its application method
EP3798250A1 (en) * 2019-09-25 2021-03-31 University College Dublin Hyperbranched cationic polymers useful as nucleic acid delivery vectors for transfecting
CN112195193A (en) * 2020-09-30 2021-01-08 上海交通大学 Application of single-stranded DNA in gene transfection
CN114904003B (en) * 2021-02-09 2023-09-29 广州立得生物医药科技有限公司 Use of ionizable cationic lipid analog materials as nucleic acid drug delivery vehicles or transfection reagents

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002042447A2 (en) * 2000-11-03 2002-05-30 Regents Of The University Of Michigan Surface transfection and expression procedure
WO2002077264A2 (en) * 2001-03-22 2002-10-03 Whitehead Institute For Biomedical Research Arrayed transfection method and uses related thereto

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2383423A1 (en) * 1999-09-17 2001-03-22 Whitehead Institute For Biomedical Research Reverse transfection method
US20030215395A1 (en) * 2002-05-14 2003-11-20 Lei Yu Controllably degradable polymeric biomolecule or drug carrier and method of synthesizing said carrier
WO2003106636A2 (en) * 2002-06-14 2003-12-24 Mirus Corporation Novel methods for the delivery of polynucleotides to cells
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
US6878374B2 (en) * 2003-02-25 2005-04-12 Nitto Denko Corporation Biodegradable polyacetals
GB0320627D0 (en) * 2003-09-03 2003-10-01 Imp College Innovations Ltd Methods
US7125709B2 (en) * 2004-02-10 2006-10-24 Nitto Denko Corporation Culture device and method for eukaryotic cell transfection
ATE477325T1 (en) * 2004-02-18 2010-08-15 Nitto Denko Corp CULTURE DEVICE AND METHOD FOR TRANSFECTION OF EUKARYONTIC CELLS

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002042447A2 (en) * 2000-11-03 2002-05-30 Regents Of The University Of Michigan Surface transfection and expression procedure
WO2002077264A2 (en) * 2001-03-22 2002-10-03 Whitehead Institute For Biomedical Research Arrayed transfection method and uses related thereto

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BIOCONJUGATE CHEM, vol 13, 2002, pages 812-821 *

Also Published As

Publication number Publication date
KR101263173B1 (en) 2013-05-13
WO2006066001A3 (en) 2006-09-08
CN101124316A (en) 2008-02-13
KR20130010036A (en) 2013-01-24
KR20070101859A (en) 2007-10-17
AU2005316501A1 (en) 2006-06-22
CA2590820A1 (en) 2006-06-22
JP2008523810A (en) 2008-07-10
WO2006066001A2 (en) 2006-06-22
JP4739352B2 (en) 2011-08-03
EP1836293A2 (en) 2007-09-26

Similar Documents

Publication Publication Date Title
AU2005316501B2 (en) Immobilized degradable cationic polymer for transfecting eukaryotic cells
US20070269891A9 (en) Solid surface with immobilized degradable cationic polymer for transfecting eukaryotic cells
US8192989B2 (en) Solid surface for biomolecule delivery and high-throughput assay
AU2005294699B2 (en) Biodegradable cationic polymers
US20080312174A1 (en) Water soluble crosslinked polymers
US20060147376A1 (en) Controllably degradable polymeric biomolecule or drug carrier and method of synthesizing said carrier
US7125709B2 (en) Culture device and method for eukaryotic cell transfection
EP1723240B1 (en) Culture device and method for eukaryotic cell transfection
AU2012200435A1 (en) "Biodegradable cationic polymers"

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired