RNAi treatment of cerebral ischemia

文档序号:1408836 发布日期:2020-03-06 浏览:22次 中文

阅读说明:本技术 大脑缺血的RNAi治疗 (RNAi treatment of cerebral ischemia ) 是由 J.温 于 2017-12-29 设计创作,主要内容包括:本发明提供治疗大脑缺血的新方法,其包括给予聚合物纳米胶囊或其组合物。所述纳米胶囊包括聚合物壳和RNAi(例如miRNA)分子,其中聚合物壳包括a)至少一种带正电荷的单体,b)至少一种可降解的交联剂,和c)至少一种中性单体;且所述RNAi分子为患有大脑缺血的受试者提供治疗益处。本发明还公开了所述聚合物纳米胶囊的组合物。(The present invention provides a novel method of treating cerebral ischemia comprising administering a polymeric nanocapsule or a composition thereof. The nanocapsule comprises a polymeric shell and an RNAi (e.g., miRNA) molecule, wherein the polymeric shell comprises a) at least one positively charged monomer, b) at least one degradable crosslinking agent, and c) at least one neutral monomer; and the RNAi molecules provide a therapeutic benefit to a subject suffering from cerebral ischemia. The invention also discloses a composition of the polymer nanocapsule.)

1. A polymeric nanocapsule comprising a polymeric shell and a microrna (mirna) molecule, wherein the polymeric shell comprises a) at least one positively charged monomer, b) at least one degradable crosslinking agent, and c) at least one neutral monomer.

2. The polymeric nanocapsule of claim 1 wherein the polymeric shell further comprises at least one non-degradable crosslinking agent.

3. The polymeric nanocapsule of claim 1 or 2, wherein the at least one degradable crosslinker comprises a crosslinker selected from the group consisting of Glycerol Dimethacrylate (GDMA), 1, 3-glycerol dimethacrylate, glycerol 1, 3-diglycerol alkyd diacrylate, N' -bis (acryloyl) cystamine, bis [2- (methacryloyloxy) ethyl ] phosphate, and a diacrylylated polypeptide.

4. The polymeric nanocapsule of any one of the preceding claims wherein the at least one degradable crosslinker comprises Glycerol Dimethacrylate (GDMA).

5. The polymeric nanocapsule of any one of the preceding claims wherein the at least one positively charged monomer is selected from the group consisting of N- (3-aminopropyl) methacrylamide (APM), N- (3-aminopropyl) methacrylamide hydrochloride, propen-spermine, dimethylaminoethyl methacrylate, (3-acrylamidopropyl) trimethylammonium hydrochloride, N- (3- ((4- ((3-aminopropyl) amino) butyl) amino) propyl) methacrylamide, N- (3- ((4-aminobutyl) amino) propyl) acrylamide, N- (3- ((4-aminobutyl) amino) propyl) methacrylamide, N- (2- ((2-aminoethyl) (meth) amino) ethyl) acrylamide N- (2- ((2-aminoethyl) (meth) amino) ethyl) methacrylamide, N- (piperazin-1-ylmethyl) acrylamide, N- (piperazin-1-ylmethyl) methacrylamide, N- (2- (bis (2-aminoethyl) amino) ethyl) acrylamide and N- (2- (bis (2-aminoethyl) amino) ethyl) methacrylamide.

6. The polymeric nanocapsule of any one of the preceding claims wherein the at least one positively charged monomer is N- (3-aminopropyl) methacrylamide (APM).

7. The polymeric nanocapsule of any one of the preceding claims wherein the at least one neutral monomer is selected from the group consisting of acrylamide (AAM) and poly (ethylene glycol) methyl ether acrylate (mPEG).

8. The polymeric nanocapsule of any one of the preceding claims which comprises at least two neutral monomers.

9. The polymer nanocapsule of claim 8, which comprises acrylamide (AAM) and poly (ethylene glycol) methyl ether acrylate (mPEG) as monomers.

10. The polymeric nanocapsule of claim 1, which comprises Glycerol Dimethacrylate (GDMA) as a degradable crosslinker, N- (3-aminopropyl) methacrylamide (APM) as a positively charged monomer, and acrylamide (AAM) and/or poly (ethylene glycol) methyl ether acrylate (mPEG) as neutral monomers.

11. The polymeric nanocapsule of any one of the preceding claims wherein the molar ratio of degradable crosslinker to total monomer is at least about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10 or higher, preferably at least about 1: 10.

12. The polymeric nanocapsule of any one of claims 1-10 wherein the molar ratio of degradable crosslinker to positively charged monomer is at least about 2:1, 1:2 or higher, preferably at least about 1: 1.

13. The polymeric nanocapsule of any one of the preceding claims wherein the molar ratio of positively charged monomer to neutral monomer is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, about 1:14, about 1:15, about 1:20 or higher, preferably at least about 1:10 or about 1: 11.

14. The polymer nanocapsule of claim 12 wherein the molar ratio of APM to mPEG is at least about 5:1, about 5:2, about 5:3, about 5:4, about 5:5 or higher, preferably at least about 5:1 or about 5: 2.

15. The polymeric nanocapsule of any one of the preceding claims wherein the molar ratio of total monomer and miRNA is at least about 1000:1, about 2000:1, about 3000:1, about 4000:1, about 5000:1, about 6000:1, about 7000:1, 8000:1, about 9000:1, about 10000:1 or higher, preferably at least about 4000:1 or about 5000: 1.

16. The polymeric nanocapsule of any one of the preceding claims wherein the miRNA is selected from miRNA that modulates a gene that induces reduced apoptosis, increased angiogenesis, increased neurogenesis, or increased neuroplasticity.

17. The polymeric nanocapsule of any one of the preceding claims wherein the miRNA is selected from mirnas that increase expression of at least one of VEGF, GFAP, CD31, CD34, BCL-2, NeuN, bromodeoxyuridine, nestin, PSA-NCAM, biscortin, Pax6, GAP-43, AKT, or HIF1- α.

18. The polymeric nanocapsule of any one of the preceding claims wherein the miRNA is selected from miRNA that reduces expression of caspase 3 or PTEN.

19. The polymeric nanocapsule of any one of claims 1-15 wherein the miRNA is miR-21.

20. The polymeric nanocapsule of any one of the preceding claims wherein the polymeric nanocapsule has a diameter of about 18nm to about 250nm, or a diameter of about 18nm to about 28 nm.

21. The polymeric nanocapsule of any one of the preceding claims having reduced non-specific protein adsorption as compared to a control nanocapsule without at least one neutral monomer, preferably mPEG.

22. The polymeric nanocapsule of any one of the preceding claims wherein the polymeric nanocapsule is stable at neutral pH and degradable at acidic pH, preferably at about pH 5.5.

23. The polymeric nanocapsule of any one of the preceding claims wherein the polymeric nanocapsule is degradable in late endosomes.

24. The polymeric nanocapsule of any one of the preceding claims, which is further conjugated to an agent, preferably a labeling agent and/or a targeting agent.

25. The polymeric nanocapsule of claim 24 wherein the labeling agent is selected from a fluorescent agent and/or a radioisotope.

26. The polymeric nanocapsule of claim 24 wherein the targeting agent is selected from TAT (transduction domain of human immunodeficiency virus type 1 (HIV-1)) peptide, diphtheria toxin, tetanus toxin, Tet1, capsid protein G23, Rabies Virus Glycoprotein (RVG) peptide, opioid peptide, glutathione, thiamine, leptin, angiopep, low density lipoprotein, insulin, melanotransferrin and transferrin.

27. The polymeric nanocapsule of claim 24 wherein the targeting agent delivers the polymeric nanocapsule to a cell type selected from the group consisting of endothelial cells, microglia, neurons, and astrocytes.

28. The polymeric nanocapsule of any one of the preceding claims further comprising a pharmaceutically acceptable carrier.

29. The polymeric nanocapsule of any one of the preceding claims having an internalization efficiency of at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least greater than 70%, or preferably at least about 60%.

30. The polymeric nanocapsule of any one of the preceding claims wherein administration of the polymeric nanocapsule induces a physiological process selected from the group consisting of reduced apoptosis, increased neurogenesis, increased neuroplasticity, or increased angiogenesis.

31. The polymeric nanocapsule of any one of the preceding claims, wherein administration of the polymeric nanocapsule increases expression of at least one of VEGF, GFAP, CD31, CD34, BCL-2, NeuN, bromodeoxyuridine, nestin, PSA-NCAM, biscortin, Pax6, GAP-43, AKT, or HIF1- α.

32. The polymer nanocapsule of any one of the preceding claims, wherein administration of the polymer nanocapsule reduces expression of at least one of caspase 3 or PTEN.

33. The polymeric nanocapsule of any one of the preceding claims for administration to treat cerebral or cerebral ischemia/reperfusion injury.

34. A pharmaceutical composition comprising the polymeric nanocapsule of any one of the preceding claims.

35. The pharmaceutical composition of claim 34, which is administered at a dose of about 0.1 mg/kg, about 0.2 mg/kg, about 0.3mg/kg, about 0.4mg/kg, about 0.5mg/kg, about 0.6 mg/kg, about 0.7 mg/kg, about 0.8 mg/kg, about 0.9 mg/kg, about 1.0 mg/kg, about 1.5 mg/kg, about 2.0 mg/kg, a dose exceeding 2.0 mg/kg, or preferably a dose of about 0.5 mg/kg.

36. The pharmaceutical composition of any one of claims 34 or 35, wherein the composition is suitable for intravenous and/or intra-carotid administration.

37. A method of treating a disease characterized by a gene disorder comprising administering the polymeric nanocapsule of any one of claims 1-33 or the pharmaceutical composition of any one of claims 34-36, wherein the miRNA molecule knocks down or reduces expression of an overexpressed gene and/or increases expression of a gene whose expression is otherwise reduced or inhibited, thereby treating the disease.

38. A method of treating cerebral ischemia, or reperfusion injury comprising administering a polymeric nanocapsule, wherein the polymeric nanocapsule comprises a polymeric shell and an RNAi molecule; wherein the polymeric shell comprises a) at least one positively charged monomer, b) at least one degradable crosslinking agent, and c) at least one neutral monomer; and RNAi knockdown or decrease expression of an overexpressed gene and/or increase expression of a gene whose expression is otherwise reduced or suppressed, thereby treating cerebral ischemia, or reperfusion injury.

39. The method of claim 38, wherein the polymer shell further comprises at least one non-degradable crosslinking agent.

40. The method of claim 38 or 39, wherein the polymeric shell comprises at least one degradable crosslinker comprising a crosslinker selected from the group consisting of Glycerol Dimethacrylate (GDMA), 1, 3-glycerol dimethacrylate, glycerol 1, 3-diglycerol alkyd diacrylate, N' -bis (acryloyl) cystamine, bis [2- (methacryloyloxy) ethyl ] phosphate, and diacrylylated polypeptides.

41. The method of any one of claims 38-40, wherein the polymeric shell comprises at least one degradable crosslinker comprising Glycerol Dimethacrylate (GDMA).

42. The method of any of claims 38-41, wherein the polymeric shell comprises at least one positively charged monomer selected from the group consisting of N- (3-aminopropyl) methacrylamide (APM), N- (3-aminopropyl) methacrylamide hydrochloride, propylene-spermine, dimethylaminoethyl methacrylate, (3-acrylamidopropyl) trimethylammonium hydrochloride, N- (3- ((4- ((3-aminopropyl) amino) butyl) amino) propyl) methacrylamide, N- (3- ((4-aminobutyl) amino) propyl) acrylamide, N- (3- ((4-aminobutyl) amino) propyl) methacrylamide, N- (3- ((4-aminopropyl) amino) propyl) methacrylamide, N- (3-aminopropyl) amino) propyl) methacrylamide, N- (3-aminopropyl) amino) methyl acrylamide, N- (, N- (2- ((2-aminoethyl) (meth) amino) ethyl) acrylamide, N- (2- ((2-aminoethyl) (meth) amino) ethyl) methacrylamide, N- (piperazin-1-ylmethyl) acrylamide, N- (piperazin-1-ylmethyl) methacrylamide, N- (2- (bis (2-aminoethyl) amino) ethyl) acrylamide and N- (2- (bis (2-aminoethyl) amino) ethyl) methacrylamide.

43. The method of any one of claims 38-42, wherein the polymeric shell comprises at least one positively charged monomer that is N- (3-aminopropyl) methacrylamide (APM).

44. The method of any of claims 38-43, wherein the polymeric shell comprises at least one neutral monomer selected from the group consisting of acrylamide (AAM) and poly (ethylene glycol) methyl ether acrylate (mPEG).

45. The method of any of claims 38-44, wherein the polymeric shell comprises at least two neutral monomers.

46. The method of any of claims 38-45, wherein the polymeric shell comprises acrylamide (AAM) and poly (ethylene glycol) methyl ether acrylate (mPEG) as monomers.

47. The method of claim 38, wherein the polymer shell comprises Glycerol Dimethacrylate (GDMA) as a degradable crosslinker, N- (3-aminopropyl) methacrylamide (APM) as a positively charged monomer, and acrylamide (AAM) and/or poly (ethylene glycol) methyl ether acrylate (mPEG) as neutral monomers.

48. The method of any one of claims 38-47, wherein the molar ratio of degradable crosslinker to total monomer is at least about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, or higher, preferably at least about 1: 10.

49. The method of any one of claims 38-47, wherein the molar ratio of degradable crosslinker to positively charged monomer is at least about 2:1, 1:2, or higher, preferably at least about 1: 1.

50. The method of any one of claims 38-49, wherein the molar ratio of positively charged monomer to neutral monomer is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, about 1:14, about 1:15, about 1:20, or higher, preferably at least about 1:10 or about 1: 11.

51. The method of claim 49, wherein the molar ratio of APM to mPEG is at least about 5:1, about 5:2, about 5:3, about 5:4, about 5:5 or higher, preferably at least about 5:1 or about 5: 2.

52. The method of any one of claims 38-51, wherein the molar ratio of total monomers and miRNA is at least about 1000:1, about 2000:1, about 3000:1, about 4000:1, about 5000:1, about 6000:1, about 7000:1, 8000:1, about 9000:1, about 10000:1 or higher, preferably at least about 4000:1 or about 5000: 1.

53. The method of any one of claims 38-52, wherein RNAi is selected from RNAi modulating a gene that induces reduced apoptosis, increased angiogenesis, increased neurogenesis, or increased neuroplasticity.

54. The method of any one of claims 38-53, wherein the RNAi is selected from RNAi that increases expression of at least one of VEGF, GFAP, CD31, CD34, BCL-2, NeuN, bromodeoxyuridine, nestin, PSA-NCAM, biscortin, Pax6, GAP-43, AKT, or HIF1- α.

55. The method of any one of claims 38-54, wherein the RNAi is selected from a miRNA that decreases expression of caspase 3 or PTEN.

56. The method of any one of claims 38-55, wherein the RNAi molecules comprise at least one RNAi selected from the group consisting of microRNA (miRNA), antisense microRNA (AS-miRNA), small interfering RNA (siRNA), and small hairpin RNA (shRNA).

57. The method of claim 56, wherein the RNAi molecule comprises at least one miRNA or one AS-miRNA.

58. The method of any one of claims 38-53, wherein the miRNA is miR-21.

59. The method of any one of claims 38 to 58, wherein the polymeric nanocapsule has a diameter of about 18nm to about 250nm, or preferably a diameter of about 18nm to about 28 nm.

60. The method of any one of claims 38-59, wherein the polymer nanocapsule has reduced non-specific protein adsorption as compared to a control polymer nanocapsule without the at least one neutral monomer, preferably mPEG.

61. The method of any one of claims 38-60, wherein the polymeric nanocapsule is stable at neutral pH and degradable at acidic pH, preferably at about pH 5.5.

62. The method of any one of claims 38-61, wherein the polymeric nanocapsule is degradable in late endosomes.

63. The method of any one of claims 38-62, wherein the polymeric nanocapsule is further conjugated to an agent, preferably a labeling agent and/or a targeting agent.

64. The method of claim 63, wherein the labeling agent is a fluorescent agent and/or a radioisotope.

65. The method of claim 63, wherein the targeting agent is selected from the group consisting of TAT (transduction domain of human immunodeficiency virus type 1 (HIV-1)) peptides, diphtheria toxin, tetanus toxin, Tet1, G23, Rabies Virus Glycoprotein (RVG) peptide, opioid peptide, glutathione, thiamine, leptin, angiopep, low density lipoprotein, insulin, melanotransferrin, and transferrin.

66. The method of claim 63, wherein the targeting agent delivers the polymeric nanocapsule to a cell type selected from the group consisting of endothelial cells, microglia, neurons, and astrocytes.

67. The method of any one of claims 38-66, wherein the polymeric nanocapsule has an internalization efficiency of at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least greater than 70%, or preferably at least about 60%.

68. The method of any one of claims 38-67, wherein the polymeric nanocapsule further comprises a pharmaceutically acceptable carrier.

69. The method of any one of claims 38-68, wherein the polymeric nanocapsule is administered as a pharmaceutical composition.

70. The method of claim 69, wherein the pharmaceutical composition comprises a polymeric nanocapsule at a dose of about 0.1 mg/kg, about 0.2 mg/kg, about 0.3mg/kg, about 0.4mg/kg, about 0.5mg/kg, about 0.6 mg/kg, about 0.7 mg/kg, about 0.8 mg/kg, about 0.9 mg/kg, about 1.0 mg/kg, about 1.5 mg/kg, about 2.0 mg/kg, more than 2.0 mg/kg, or preferably at a dose of about 0.5 mg/kg.

71. The method of any one of claims 38-70, wherein administration of the polymeric nanocapsule induces a physiological process selected from the group consisting of reduced apoptosis, increased neurogenesis, increased neuroplasticity, and increased angiogenesis.

72. The method of any one of claims 38-71, wherein expression of at least one of VEGF, GFAP, CD31, CD34, BCL-2, NeuN, bromodeoxyuridine, nestin, PSA-NCAM, biscortin, Pax6, GAP-43, AKT, or HIF1- α is increased following administration.

73. The method of any one of claims 38-72, wherein expression of at least one of caspase 3 or PTEN is decreased following administration.

74. The method of any one of claims 38-73, wherein the polymeric nanocapsule or the pharmaceutical composition is administered parenterally, such as intravenously.

75. The method of any one of claims 38-74, wherein the method further comprises administering an additional treatment.

76. The method of claim 75, wherein the additional therapy is selected from the group consisting of anticoagulation therapy, antiplatelet therapy, and thrombolytic therapy.

77. A method of making the polymeric nanocapsule of any one of claims 1-33, comprising: a) dissolving the miRNA molecule in RNase-free water; b) dissolving at least one positively charged monomer, at least one neutral monomer, and at least one degradable crosslinker in deoxygenated and deionized water to create a monomer mixture; c) combining the solubilized miRNA molecules of step a) with the monomer mixture of step b); d) adding ammonium persulfate and N, N, N ', N' -tetramethylethylenediamine to the product of step c), and e) incubating the product of step d) in a serum-free medium.

Brief Description of Drawings

Figure 1 contains two subfigures, figure 1A and 1B, which depict schematic diagrams of miRNA nanocapsule synthesis and systemic delivery thereof for treatment of cerebral ischemia. The synthesis of miRNA nanocapsules comprises a step I for enriching monomers and cross-linking molecules around the miRNA molecule and a step I for forming n (miRNA) when growing a thin polymer shell around the miRNA molecule (fig. 1A). Systemic delivery of n (mirna) for ischemia therapy includes: (I) accumulation of n (mirna) in ischemic areas of brain tissue due to increased vascular permeability; (II) the cells take up n (miRNA) and release miRNA in the cytosol; and (III) the miRNA delivered promoted neuronal regeneration (fig. 1B).

Figure 2 contains 9 sub-figures, figures 2A-2I, which describe the structure, size, stability, protein adsorption and degradation kinetics of miR-21 nanocapsules. Agarose gel electrophoresis of n (miR-21) synthesized at different monomer/miRNA (M/R) molar ratios (FIG. 2A). FIG. 2B shows the stability of the synthesized Lipo/miR-21 complex and n (miR-21) at different M/R molar ratios after incubation with 10 mg/mL heparin. Shown here are Transmission Electron Microscope (TEM) images (fig. 2C), hydrodynamic size (fig. 2D), and zeta potential (fig. 2E) of n (miR-21) synthesized with an M/R ratio of 6000: 1. Hydrodynamic dimensions and zeta potential were measured with Dynamic Light Scattering (DLS) with a scattering angle of 173 °. Comparing the residues of native miR-21, Lipo/miR-21 and n (miR-21) after incubation with mouse whole serum or RNase for 2h at 37 ℃miR-21 levels (FIG. 2F). The residual miRNA was then extracted with chloroform/0.1% SDS-0.5M NaCl and measured with qRT-PCR. Data are shown as mean. + -. SD: (n=3)。**P<0.01、***P<0.001 andP<0.0001 (Turkey post hoc test followed by plain one-way ANOVA). Quantitative determination of n (miR-21) and Lipo/miR-21 adsorbed serum proteins after incubation with mouse whole serum is shown in FIG. 2G. Data are shown as mean ± SD (n = 3). **P<0.01 (Turkey post hoc test followed by plain one-way ANOVA). The relative scattered light intensity of n (miR-21) after incubation for 2h at 37 ℃ in 50mM sodium acetate buffer (pH 5.5) and 50mM HEPES buffer (pH 7.4) was compared (miR-21)I/I 0 X 100%) change, whereinI 0 Is the initial n (miR-21) scattered light intensity, andIis the scattered light intensity measured at different time points with a detection angle of 173 ° (fig. 2H). Data were normalized for direct comparison and expressed as mean ± SD (S) ((S))n= 5). After incubation in 50mM sodium acetate buffer (pH 5.5) and 50mM HEPES buffer (pH 7.4) for 2h at 37 deg.C, agarose gel electrophoresis of n (miR-21) is shown in FIG. 2I. The symbols "-" indicate "incubation without 1mg/mL heparin" and "+" indicate "incubation with 1mg/mL heparin", respectively.

FIG. 3 includes three sub-graphs, FIGS. 3A-3C, showing the characteristics of nanocapsules synthesized with different monomer to RNA (M: R) molar ratios, including synthesis parameters for n (miR-21) for a range of monomer to RNA molar ratios (FIG. 3A), size distribution (FIG. 3B), and zeta potential trend (FIG. 3C). Data in FIGS. 3B and 3C are presented as mean. + -. SEM ((C))n=3)。

FIG. 4 four panels, FIGS. 4A-4D, show the characteristics of nanocapsules synthesized with different mPEG/miRNA molar ratios at an M: R ratio of 6000:1, including synthesis parameters for n (miR-21) (FIG. 4A), agarose gel electrophoresis (FIG. 4B), size distribution and PDI (FIG. 4C) and zeta potential trends (FIG. 4D). Data in fig. 4C and 4D are presented as mean ± SEM (C: (D))n=3)。

FIG. 5 includes two subgraphs, FIGS. 5A and 5B, which depict fluorescence-assisted cell sorting analysis of C6 cells (FIG. 5A) and J774A.1 cells (FIG. 5B) treated with miR-21 and n (miR-21), which miR-21 and n (miR-21) are synthesized with different mPEG/miRNA molar ratios at a M: R ratio of 6000: 1. C6 and J774A.1 cells were treated with a sample of 50nM miRNA at 37 ℃ for 4 hr. MiR-21 was labeled with FITC. Trypan blue quenching analysis was performed before the measurement.

Figure 6 includes three subgraphs, figure 6A-6C, which depicts the properties of synthesized n (miR-21), including the stability of n (miR-21) synthesized at a monomer/miRNA molar ratio of 6000 in PBS (pH 7.4) at 4 ℃ (figure 6A), the temperature-dependent dimensional change of n (miR-21) in PBS buffer at different temperatures of 25, 37, and 60 ℃ (figure 6B), and the hydrodynamic size of n (miR-21) in the presence or absence of 10% serum in PBS buffer (figure 6C). All data in the three subgraphs are expressed as mean ± SEM (ii)n=3)。

FIG. 7 depicts the quantitative determination of the adsorbed serum proteins of n synthesized at M: R ratio 6000:1 (miR-21) and n synthesized at M: R ratio 6000:1 without mPEG (miR-21) (no-PEG n (miR-21)) after incubation with mouse whole serum. Data are shown as mean. + -. SD: (n=3)。*P<0.05 andP<0.01 (Turkey post hoc test followed by plain one-way ANOVA).

Fig. 8 includes 8 subgraphs, fig. 8A-8H, depicting cytotoxicity, cellular internalization efficiency, non-specific phagocytosis, and intracellular activity of miRNA nanocapsules. Confocal micrographs of intracellular distribution of miR-21, n (miR-21) and Lipo/miR-21 in C6 cells after 4 hours of incubation at 37 ℃ are shown in FIG. 8A. Scale bar: 20 μm. miR-21 is labeled with FITC. Cells were counterstained with DAPI (for the nucleus), DiI (for the cell membrane). Scale bar: 20 μm. Fluorescence-assisted cell sorting (FACS) analysis of C6 cells after 4 hours incubation with miR-21 and n (miR-21) at 37 ℃ is shown in FIG. 8B. miR-21 is labeled with FITC. Trypan blue quenching analysis was performed before the measurement. Cell viability assay for n (miR-NC) in C6 cells after 4 hours incubation at 37 ℃ is shown in FIG. 8C. Data are shown as mean ± SD (n = 5). Fluorescence images of J774A.1 mouse macrophages after incubation with n (miR-21) and Lipo/miR-21 for 1 hour at 37 ℃ are shown in FIG. 8D. MiR-21 was labeled with FITC. Cells were stained with Hoechst 33342 for nuclear imaging. Scale bar: 100 μm. For the incubation with n (miR-21) and Lipo/miR-21Mean fluorescence intensities obtained from FACS analysis of macrophages, compare histograms (fig. 8E). Data are shown as mean. + -. SD: (n=3)。***P<0.001 (Turkey post hoc test followed by plain one-way ANOVA). Real-time quantitative PCR analysis was performed to compare miR-21 levels in C6 cells treated with different samples in serum-free medium or 50% human serum medium (fig. 8F). Data are shown as mean. + -. SD: (n=3)。*P<0.05 andP<0.01 (Turkey post hoc test followed by ordinary two-way ANOVA). Western blot analysis of protein expression in C6 cells 48 hours after sample administration is shown in fig. 8G. Quantification of protein expression levels was expressed as normalized grey values obtained from figure 8G by ImageJ software (figure 8H).

FIG. 9 includes 3 sub-graphs, FIGS. 9A-9C, which depict the characteristics of J774A.1 mouse macrophages after n (miR-21) treatment. FIG. 9A compares fluorescence images of J774A.1 mouse macrophages 1 hour after incubation with n (miR-21) and non-PEG n (miR-21). Cells were stained with Hoechst 33342 for nuclear imaging. MiR-21 was labeled with FITC. Scale bar: 100 μm. FACS analysis of J774A.1 macrophages was performed after incubation with n (miR-21), non-PEG n (miR-21) and Lipo/miR-21 (FIG. 9B). Histograms were performed to compare the mean fluorescence intensity obtained from FACS analysis of macrophages after incubation with n (miR-21) and non-PEG n (miR-21) (fig. 9C). Data are shown as mean. + -. SD: (n=3)。*P<0.05 (Turkey post hoc test followed by plain one-way ANOVA).

FIG. 10 includes 3 sub-graphs, FIGS. 10A-10C, which depict quantitative real-time PCR analysis of miR-21 levels in C6 cells, C6 cells were treated with various samples in serum-free medium or 50% human serum medium. Data are shown as mean. + -. SD: (n=3)。**P<0.01 (Turkey post hoc test followed by ordinary two-way ANOVA).

Fig. 11 includes 2 subgraphs, fig. 11A and 11B, which depict western blot analysis of protein expression from C6 cells 48 hours after sample administration (fig. 11A) and normalized protein expression levels from ImageJ software quantitative analysis (fig. 11B).

FIG. 12 includes 9 sub-diagrams, FIGS. 12A-12I, which depictThe biodistribution, pharmacokinetics and administration route comparison of the miRNA nanocapsule are described. FIG. 12A shows in vivo fluorescence images (top) of ischemic (transient focal cerebral ischemia) model rats or non-ischemic rats, and intravenously: (B)i.vEx vivo fluorescence images of brain tissue collected 24 hours after administration of Cy5.5-labeled miR-21 nanocapsules (n (Cy5.5-miR-21)) at a dose of 0.5mg/kg miR-21 (bottom), and an equal volume of PBS was administered as a control. ROI analysis of fluorescence signals of collected brain tissue (fig. 12B) and mature miR-21 levels (fig. 12C) were performed. Data are shown as mean. + -. SD: (n=5)。*P<0.05 andP<0.001 (Turkey post hoc test followed by ordinary two-way ANOVA). Ex vivo fluorescence images (FIG. 12D) and ROI analysis of fluorescence signals (FIG. 12E) show major organs collected 24 hours after injection of Lipo/Cy5.5-miR-21 and n (Cy5.5-miR-21) (dose 0.5mg miR-21/kg rat) and an equal volume of PBS was given as a control. Data are shown as mean. + -. SD: (n=5)。*P<0.05 (two-tailed unpaired Student t-test). FIG. 12F compares the blood circulation profiles of n (Cy5.5-miR-21) in rats after intravenous injection of Lipo/Cy5.5-miR-21 and n (Cy5.5-miR-21) (dose 0.5mg/kg miR-21). Illustration is shown: first 2 hours of enlarged blood circulation profile, which is plotted on the logarithmic y-axis. Data are shown as mean. + -. SEM: (n= 5). FIG. 12G compares in vivo fluorescence images of ischemic model rats (top), and in carotid artery: (C)i.cEither) ori.vEx vivo images of brain collected 24 hours after injection of different doses of n (cy5.5-miR-21) (below). ROI analysis of fluorescence signal (fig. 12H) and mature miR-21 levels (fig. 12I) were measured on brain tissue collected in fig. 12G. Data are shown as mean. + -. SEM: (n= 5). Level of significance isP<0.01 andP<0.001 (Turkey post hoc test followed by plain one-way ANOVA).

FIG. 13 depicts representative H & E stain images of brain tissue obtained from sacrificed MCAO/R or normal rats.

Figure 14 includes 6 subgraphs, 14A-14F, which depict the efficacy of miRNA nanocapsules as a therapeutic for transient focal cerebral ischemia. FIG. 14A compares the neurological work of transient focal cerebral ischemia model rats after administration of samplesCan be used. Arrows indicate time points of sample administration. Neurological deficit scores were tested every other day prior to each injection. Data are shown as mean. + -. SEM: (n=10)。*P<0.05 andP<0.01 (Bonferroni multiple comparison test followed by two-way RMANOVA). TTC staining of brain tissue obtained from day 7 sacrificed rats is shown in fig. 14B. The quantified cerebral infarct volume was measured from fig. 14B (fig. 14C). Data are shown as mean. + -. SEM: (n=5)。*P<0.05 andP<0.01. (Turkey post hoc test followed by ordinary one-way ANOVA). Representative HE staining images of the infarct area are shown in fig. 14D. Scale bar: 400 μm. A representative fluorescent in situ hybridization assay for miR-21 in the infarct zone was determined (FIG. 14E). Blue for DAPI and red for miR-21. Scale bar: 100 μm. A representative immunohistochemistry image of the infarct area is shown in fig. 14F. Scale bar: 400 μm (up) and scale: 150 μm (lower).

Fig. 15 includes 2 sub-graphs, fig. 15A and 15B, depicting representative fluorescence images of TUNEL analysis of the infarct region of brain tissue (fig. 15A) (blue for DAPI and green for FITC) and the apoptosis index of the infarct region of brain tissue obtained from quantitative analysis of the TUNEL analysis (fig. 165). Data are shown as mean. + -. SEM: (n=3)。***P<0.001 (Turkey post hoc test followed by plain one-way ANOVA).

Figure 16 depicts representative H & E stained images of major organs after a 7 day treatment period.

Detailed Description

Definition of

For convenience, the meanings of certain terms and phrases used in the specification, examples, and appended claims are provided below.

As used herein, the term "target" refers to a strand of DNA or RNA that is double-stranded and is complementary to a strand of DNA or RNA (including all transcribed regions) that serves as a transcription matrix. The target gene, usually the sense strand, is a gene whose expression is selectively inhibited or silenced by RNA interference and/or microrna.

As used herein, the term "polymeric nanocapsule" refers to a composition comprising a "polymeric shell" and an "RNAi molecule" such as a "microrna molecule".

As used herein, the term "polymeric shell" refers to the polymeric portion of the microrna polymeric nanocapsule that comprises one or more positively charged polymeric monomers, one or more crosslinkers, and optionally one or more neutral polymeric monomers. Examples of positively charged monomers, crosslinkers, and neutral monomers are provided in table 1, table 2, and table 3 of U.S. patent application publication No. US 2015/0071999, which is incorporated herein by reference in its entirety.

As used herein, the term "microrna molecule" refers to any microrna (abbreviated "miRNA") that is a small, non-coding RNA molecule (about 22 nucleotides) that is either naturally found (e.g., in plants, animals, and certain viruses), or artificially synthesized, that plays a role in RNA silencing and post-transcriptional regulation of gene expression (see Ambros (2004)Nature. 431:350-355; Bartel (2004)Cell116:281-297). While naturally occurring mirnas are typically located intracellularly, some mirnas, commonly referred to as circulating mirnas or extracellular mirnas, are also found in the extracellular environment (including various biological fluids and cell culture media). The miRNA is complementary to a portion of one or more messenger rnas (mrnas). Animal mirnas are generally complementary to sites in the 3' UTR, while plant mirnas are generally complementary to coding regions of mrnas. Perfect or near perfect base pairing with the target RNA promotes cleavage of the RNA. This is a typical pattern of plant mirnas. In animals, pairing is often imperfect. mirnas also sometimes cause DNA methylation and histone modification at promoter sites, which affect the expression of target genes. The 9 miRNA mechanisms of action are described and assembled in a unified mathematical model: Cap-40S onset inhibition; 60S ribosome unit ligation inhibition; inhibition of elongation; ribosome shedding (premature termination); co-translational nascent protein degradation; isolation in the P-body; mRNA decay (destabilization); mRNA cleavage; and transcriptional repression by microRNA-mediated chromatin recombination followed by gene silencing (Morozova)et al.(2012)RNA18:1635-1655)。

As used herein, the terms "degradable polymer" and "non-degradable polymer" refer to the ability of a polymer described herein to degrade into smaller fragments. In certain embodiments of the invention, the degradable polymer will decompose at physiological pH. In certain embodiments, the degradable polymer may decompose at about pH 7.4. In certain embodiments, a mixture of degradable and non-degradable polymers may result in a degradable polymer mixture. In certain embodiments, a mixture of degradable and non-degradable polymers will decompose at physiological pH. In certain embodiments, the mixture of degradable and non-degradable polymers may decompose at about pH 7.4.

As used herein, the terms "mutant gene" and "target mutant gene" refer to a gene that comprises at least one point mutation relative to the corresponding normal, non-mutated cellular gene (referred to herein as the "corresponding wild-type gene"). The terms mutant gene and target mutant gene specifically include any variant of a normal cellular gene or gene fragment (e.g., an oncogene) whose expression is associated with a disease or disorder.

As used herein, the term "conjugate" or "conjugating agent" or "surface-conjugated targeting agent" or "polymer nanocapsule conjugate" refers to any moiety, such as a protein or an effective portion thereof, that is conjugated to a polymer nanocapsule and provides specific targeting of the polymer nanocapsule to the surface of a particular cell type, thereby allowing targeted delivery of RNAi molecules (e.g., micrornas) to a particular cell type. For example, the conjugating agent may bind to a cell-specific cell surface receptor, thereby bringing the polymeric nanocapsule in direct proximity to the target cell. In certain embodiments, conjugates for achieving specific targeting of polymer nanocapsules include CD4, CD8, CD45, CD133, aHLA, and transferrin. In other embodiments, the conjugate may be a cell-specific antibody or fragment thereof. Other examples of conjugates for targeting specific cell types are described in the detailed description below.

The term "complementary RNA strand" (also referred to herein as "antisense strand") refers to a dsRNA strand that is complementary to an mRNA transcript formed during expression of a target gene or a processed product thereof. As used herein, the term "complementary nucleotide sequence" refers to a region on a complementary RNA strand that is complementary to a region of an mRNA transcript of a target mutant gene (i.e., the "corresponding nucleotide sequence" of the target gene). "dsRNA" refers to a ribonucleic acid molecule having a duplex structure comprising two complementary and antiparallel nucleic acid strands. Not all dsRNA nucleotides necessarily exhibit Watson-crick base pairs. The maximum number of base pairs is the number of nucleotides in the shortest strand of the dsRNA. The RNA strands may have the same or different number of nucleotides. The length of the complementary nucleotide region of the complementary RNA strand is less than 25, preferably 19-24, more preferably 20-24, even more preferably 21-23, and most preferably 22 or 23 nucleotides. The length of the complementary RNA strand is less than 30, preferably less than 25, more preferably 21-24, and most preferably 23 nucleotides. Dsrnas comprising complementary or antisense strands of this length (referred to as "short interfering RNAs" or "sirnas") are particularly effective in inhibiting expression of target mutant genes. "introduced" refers to uptake or absorption in a cell, as understood by one of skill in the art. The uptake or uptake of nucleic acids, e.g., micrornas, can occur by cellular processes or by auxiliary agents or devices. For example, in vivo delivery, micrornas can be injected into a tissue site or administered systemically. In vitro delivery includes methods known in the art, such as electroporation and lipofection.

As used herein, "selective inhibition of expression" means that the dsRNA and/or miRNA has a greater inhibitory effect on the target mutant gene than the expression of the corresponding wild-type gene. Preferably, the expression level of the target mutant gene is less than 98%, less than 95%, less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20% or less than 10% of the expression level of the corresponding wild-type gene.

As used herein and known in the art, the term "identity" is a relationship between two or more polynucleotide sequences, as determined by comparing the sequences. Identity also means the degree of sequence relatedness between polynucleotide sequences, as determined by the match between strings of such sequences. Identity can be readily calculated (see, e.g., computing Molecular Biology, Lesk, A. M., eds., Oxford University Press, New York (1998), and Biocomputing: information and Genome Projects, Smith, D.W., ed., Academic Press, New York (1993), both of which are incorporated herein by reference). Although there are many methods to measure identity between two polynucleotide sequences, this term is well known to those skilled in the art (see, e.g., Sequence Analysis in Molecular Biology, von Heinje, g., Academic Press (1987); and Sequence Analysis Primer, gribskov, m. and deveux, j., eds., m.stockton Press, New York (1991)). Methods commonly used to determine identity between sequences include, for example, the methods disclosed in Carillo, h, and Lipman, d., SIAM j. Applied math, (1988) 48: 1073. As used herein, "substantially identical" means that there is a high degree of homology (preferably 100% sequence identity) between the sense strand of the dsRNA and/or miRNA and the corresponding portion of the target gene. However, dsrnas and/or mirnas having greater than 90% or 95% sequence identity may be used in the present invention, and therefore sequence variations that can be expected due to gene mutations, strain polymorphisms, or evolutionary divergence can be tolerated. Although 100% identity is preferred, the dsRNA and/or miRNA may comprise a single or multiple base pair random mismatch between the RNA and the target gene.

As used herein, the term "treatment" refers to the application or administration of a therapeutic agent to a patient, or to an isolated tissue or cell line from a patient, who has a disorder, such as a disease or condition, a disease symptom, or a susceptibility to a disease, with the purpose of treating, curing, alleviating, altering, treating, ameliorating, improving, or affecting the disease, the disease symptom, or the susceptibility to the disease.

As used herein, a therapeutic agent that "prevents" a disorder, or disease refers to a therapeutic agent, such as a polymeric nanocapsule (or a composition comprising the same) described herein, that reduces the occurrence of the disorder or condition in a treated sample relative to an untreated control sample in a statistical sample, or delays the occurrence or reduces the severity of one or more symptoms of the disorder or condition relative to an untreated control sample.

As used herein, a "pharmaceutical composition" includes a pharmacologically effective amount of a therapeutic agent, such as a miRNA, and a pharmaceutically acceptable carrier. As used herein, "pharmacologically effective amount," "therapeutically effective amount," or simply "effective amount" refers to an amount of RNA effective to produce a predetermined pharmacological, therapeutic, or prophylactic result. For example, if a given clinical treatment is deemed effective when at least a 25% reduction in a measurable parameter associated with a disease or disorder, then a therapeutically effective amount of the drug for treating the disease or disorder is the amount necessary to reduce the parameter by at least 25%.

The term "pharmaceutically acceptable carrier" refers to a carrier used for administration of a therapeutic agent. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The term specifically excludes cell culture media. For orally administered drugs, pharmaceutically acceptable carriers include, but are not limited to, pharmaceutically acceptable excipients such as inert diluents, disintegrants, binders, lubricants, sweeteners, flavoring agents, coloring agents, and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binders may include starch and gelatin, while lubricants, if present, are typically magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract.

The term "cerebral ischemia" refers to cerebral ischemia and/or cerebrovascular ischemia, a condition in which cerebral blood flow is insufficient to meet metabolic demand. This leads to poor oxygen supply or hypoxia of the brain, which in turn leads to death of brain tissue or cerebral infarction/ischemic stroke. It is a subtype of stroke that is associated with subarachnoid and intracerebral hemorrhage. Ischemia leads to altered brain metabolism, reduced metabolic rates, and energy crisis. The broad term "stroke" can be divided into 3 categories: cerebral ischemia, subarachnoid hemorrhage, and intracerebral hemorrhage. Cerebral ischemia can be further subdivided into thrombotic, embolic, and hypoperfusion depending on the cause. Thrombotic and embolic are generally focal or multifocal in nature, while low perfusion affects the whole brain. Focal cerebral ischemia occurs when blood clots occlude the cerebral blood vessels. Focal cerebral ischemia reduces blood flow to a specific area of the brain, increasing the risk of cell death in that specific area. It may be caused by thrombosis or embolism. Global cerebral ischemia occurs when blood flow to the brain is stopped or sharply reduced. This is usually caused by cardiac arrest. Symptoms may be transient if sufficient circulation is restored within a short period of time. However, brain damage can be permanent if a significant amount of time passes before recovery. Although reperfusion is necessary to protect as much brain tissue as possible, it can also lead to reperfusion injury. Reperfusion injury is divided into the subsequent injury after the ischemic tissue has returned to blood supply. In the present application, the scope of cerebral ischemia is broad enough to encompass any thrombotic, embolic, and hypoperfusion ischemia, plus any other ischemia that is associated with and/or affects brain function. The main symptoms of cerebral ischemia include impairment of vision, physical movement and speech. The causes of cerebral ischemia are diverse, ranging from sickle cell anemia to congenital heart defects. Symptoms of cerebral ischemia may include confusion, blindness, coordination problems, and physical weakness. Other effects that may be caused by cerebral ischemia are stroke, cardiopulmonary arrest and irreversible brain damage. Interruption of cerebral blood flow for more than 10 seconds leads to unconsciousness, while interruption of blood flow for more than a few minutes often leads to irreversible brain damage.

The present invention provides a new strategy for encapsulating microrna molecules in cross-linked polymer nanocapsules by self-assembly and in situ polymerization techniques. In certain embodiments, the polymer nanocapsules have a diameter of about 20-100 nm. The small diameter of the polymer nanocapsules protects the RNA molecules to the maximum extent from external RNase attack and serum neutralization.

In certain embodiments, the RNA is selected from an RNA that modulates a gene that induces decreased apoptosis, increased angiogenesis, increased neurogenesis, or increased neuroplasticity. In certain embodiments, the miRNA is selected from a miRNA that modulates a gene that induces decreased apoptosis, increased angiogenesis, increased neurogenesis, or increased neuroplasticity.

In certain embodiments, the polymeric nanocapsule is 10 nm to 20 nm, 20 to 25 nm, 25 nm to 30nm, 30nm to 35 nm, 35 nm to 40 nm, 40 nm to 45 nm, 45 nm to 50nm, 50nm to 55 nm, 55 nm to 60 nm, 60 nm to 65nm, 70 to 75 nm, 75 nm to 80 nm, 80 nm to 85 nm, 85 nm to 90 nm, 90 nm to 95 nm, 95 nm to 100nm, or 100nm to 110 nm. In certain embodiments, the diameter of the polymeric nanocapsule is about 10 nm, 11 nm, 12 nm, 13 nm, 14nm, 15 nm, 16 nm, 17 nm, 18nm, 19 nm, 20 nm, 21 nm, 22 nm, 23 nm, 24 nm, 25 nm, 26nm, 27 nm, 28 nm, 29 nm, 30nm, 31 nm, 32 nm, 33 nm, 34 nm, 35 nm, 36 nm, 37 nm, 38 nm, 39nm, 40 nm, 41 nm, 42 nm, 43 nm, 44 nm, 45 nm, 46 nm, 47 nm, 48 nm, 49 nm, 50nm, 51 nm, 52nm, 53 nm, 54 nm, 55 nm, 56 nm, 57 nm, 58 nm, 59 nm, 60 nm, 61 nm, 62 nm, 63 nm, 64 nm, 65nm, 66 nm, 67 nm, 68 nm, 69 nm, 65nm, 50nm, or more, 70nm, 71 nm, 72 nm, 73 nm, 74 nm, 75 nm, 76 nm, 77 nm, 78nm, 79 nm, 80 nm, 81 nm, 82 nm, 83 nm, 84 nm, 85 nm, 86 nm, 87 nm, 88 nm, 89 nm, 90 nm, 91nm, 92 nm, 93 nm, 94 nm, 95 nm, 96 nm, 97 nm, 98 nm, 99 nm, 100nm, 101 nm, 102 nm, 103nm, 104 nm, 105 nm, 106 nm, 107 nm, 108 nm, 109 nm, 110 nm, 111 nm, 112 nm, 113 nm, 114nm, 115 nm, 116 nm, 117 nm, 118 nm, 119 nm, or 120 nm. In certain embodiments, the diameter of the polymeric nanocapsule is 120 nm to 130 nm, 130 nm to 140 nm, 140 nm to 150 nm, 150 nm to 160 nm, 160 nm to 170nm, 170nm to 180 nm, 180 nm to 190 nm, 190 nm to 200 nm, 200 nm to 210 nm, 220 nm to 230nm, 230nm to 240 nm, 240 nm to 250nm, or greater than 250 nm.

In certain embodiments, the polymeric nanocapsules disclosed herein include non-targeting and targeting capabilities, greater efficiency, and/or lower adverse immune responses. For example, higher efficiency may be the result of increased uptake and more targeted delivery.

In addition, highly stable microrna molecules within the protective nanocapsule can be completely released when the nanostructured polymer shell degrades in endosomes and lysosomes. In certain embodiments, non-targeting polymer-encapsulated microrna molecules can be transduced into primary cells, such as PBMCs, in vitro with higher efficiency and lower cytotoxicity compared to the low efficiency and high toxicity of liposomal transduction. In certain embodiments.

Importantly, by selecting and designing the appropriate polymer charge, the method of delivering microrna molecules for a particular purpose can be modulated (e.g., targeted by conjugating moieties to the polymer nanocapsules described herein).

In certain embodiments, the ratio of degradable to non-degradable crosslinker is 1:1, 1:2, 2:1, 1:3, 2:3, 3:1, 3:2, 4:1, 1:4, 4:3, 3:4, 5:1, 1:5, 2:5, 5:2, 5:3, 3:5, 4:5, 5:4, 6:1, 1:6, 1:7, 7:1, 2:7, 7:2, 3:7, 7:3, 4:7, 7:4, 5:7, 7:5, 6:7, 7:6, 8:1, 1:8, 3:8, 8:3, 5:8, 8:5, 7:8, 8:7, 9:1, 1:9, 2:9, 9:2, 4:9, 9:4, 5:9, 9:5, 7:9, 9: 8:9, 9: 4:9, 9:4, 5:9, 9: 9, 9: 5:9, 9 7:10, 10:7, 9:10, 10:9, or any other ratio known to those skilled in the art to be useful. In certain embodiments, the molar ratio of degradable crosslinker to positively charged monomer is at least about 2:1, 1:2 or higher, preferably at least about 1: 1. In certain embodiments, the molar ratio of positively charged monomer to neutral monomer is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, about 1:14, about 1:15, about 1:20, or higher, preferably at least about 1:10 or about 1: 11.

In certain embodiments, the miRNA is selected from mirnas that increase expression of at least one of VEGF, GFAP, CD31, CD34, BCL-2, NeuN, bromodeoxyuridine, nestin, PSA-NCAM, biscortin, Pax6, GAP-43, AKT, or HIF1- α.

Some exemplary degradable and non-degradable crosslinkers are listed in table 2 of US 2015/0071999.

In certain embodiments, the polymer nanocapsule is designed to degrade within 1 hour, or 2 hours, or 3 hours, or 4 hours, or 5 hours, or 6 hours, or 7 hours, or 8 hours, or 9 hours, or 10 hours, or 11 hours, or 12 hours, or 13 hours, or 14 hours, or 15 hours, or 16 hours, or 17 hours, or 18 hours, or 19 hours, or 20 hours, or 21 hours, or 22 hours, or 23 hours, or 1 day, or 2 days, or 3 days, or 4 days, or 5 days, or 6 days, or 1 week, or 2 weeks, or 3 weeks, or 1 month, or any combination thereof. In certain embodiments, the polymeric nanocapsules are designed to degrade at any of the above rates at physiological pH. In certain embodiments, the polymeric nanocapsule is designed to degrade at any of the above rates following administration to a subject in need thereof.

In certain embodiments, microrna molecules can be efficiently delivered to specific sites in the body. In certain embodiments, the targeting agent (i.e., conjugate or conjugating agent) is conjugated to the polymeric nanocapsule. In certain embodiments, conjugation prevents dissociation of the targeting agent from the polymer particle. In certain embodiments, cyclodextrins and/or adamantanes may be used for targeting agent conjugation.

In certain embodiments, the invention is practiced using non-targeted and targeted polymer nanocapsule microrna molecule delivery with high efficiency and low toxicity for in vitro testing and targeting to specific tissues and organs in vivo by intravenous injection.

The increased stability of the RNAi/miRNA molecule encapsulated by the cross-linked polymer also ensures its long-term circulation in vivo before reaching the targeted site. In summary, delivery of microrna molecules as described herein can provide significant efficiency, enhanced stability, and minimal toxicity.

Monomers and crosslinking agents

Different monomers and cross-linkers can be used to encapsulate the RNAi molecules (e.g., micrornas) by in situ polymerization, such as those taught in the specification and others well known in the art.

In certain embodiments, the molar ratio of total monomers and miRNA is at least about 1000:1, about 2000:1, about 3000:1, about 4000:1, about 5000:1, about 6000:1, about 7000:1, 8000:1, about 9000:1, about 10000:1 or higher, preferably at least about 4000:1, or about 5000: 1.

Polymer nanocapsule conjugates

In certain embodiments, the microrna molecules are targeted into cells using surface-conjugated targeting agents on optimized nanocapsules. Of particular interest, the polymer nanocapsule conjugates can be used to target immune cells, lung cells, eye cells, liver cells, kidney cells, brain cells, central nervous system cells, peripheral nervous system cells, heart cells, cancer cells, proliferating cells, virally or retroviral infected cells, stem cells, skin cells, intestinal cells, and/or auditory cells. In certain embodiments, the targeting agent delivers the polymeric nanocapsule to a cell type selected from the group consisting of endothelial cells, microglia, neurons, and astrocytes.

In certain embodiments, conjugates for achieving specific targeting of polymer nanocapsules include CD4, CD8, CD45, CD133, aHLA, and transferrin. In certain embodiments, the conjugates used to achieve specific targeting of the polymeric nanocapsule include any one or more cluster of differentiation or designated Cluster (CD) markers. For example, CD markers include CDX, where X can be any of 1-340. As described herein, the term "CD1", for example, means all CD1 variants and subtypes. This applies to all CD markers described herein.

In certain embodiments, conjugates for achieving specific targeting of polymeric nanocapsules include any one or more of AFP, β -catenin, BMI-1, BMP-4, c-kit, CXCL12, SDF-1, CXCR4, decorin, E-cadherin, cadherin 1, EGFR, ErbB1, Endoglin, EpCAM, TROP-1, Fepsilon RI A, FCER1A, L1CAM, LMO2, Nodal, Notch-1, PDGFRB, Podoplanin, PTEN, Sonic Hedgehog, STAT3, Syndec-1, transferrin receptor, and vimentin.

In certain embodiments, conjugates for achieving specific targeting of polymeric nanocapsules include any one or more of ALK, AFP, B2M, β -hCG, BCR-ABL, BRAF, CA15-3, CA19-9, CA-125, calcitonin, CEA (carcinoembryonic antigen), CD20, chromogranin a, cytokeratin or fragment thereof, EGFR, estrogen receptor, progesterone receptor, fibrin, fibrinogen, HE4, HER2/neu, IgG variants, KIT, lactate dehydrogenase, nuclear matrix protein 22, PSA, thyroglobulin, uPA, PAI-1, and ova.

In certain embodiments, the targeting agent is selected from TAT (transduction domain of human immunodeficiency virus type 1 (HIV-1)) peptides, diphtheria toxin, tetanus toxin, Tet1, G23, Rabies Virus Glycoprotein (RVG) peptide, opioid peptide, glutathione, thiamine, leptin, angiopep, low density lipoprotein, insulin, melanotransferrin, and transferrin.

Any of the labels described herein or acceptable to those of skill in the art may be used alone or in combination with one or more additional labels to achieve the desired targeting of a particular cell.

All other cell and/or tissue specific markers known to those skilled in the art are incorporated herein by reference.

Methods of treating diseases caused by expression of target genes

In one embodiment, the invention relates to a method of treating a subject having or at risk of developing a disease caused by expression of a target mutant gene. In certain embodiments, the polymeric nanocapsules described herein can be used to treat or prevent one or more cell proliferation and/or differentiation disorders. In certain embodiments, the polymeric nanocapsules may be used to treat or prevent one or more immune or immunodeficiency disorders. In certain embodiments, the polymeric nanocapsules are useful for treating or preventing viral replication or viral infection. In certain embodiments, the polymeric nanocapsules can be used to treat one or more neurological or neurodegenerative disorders. In certain embodiments, the polymeric nanocapsules are useful for treating or preventing cancer.

In certain embodiments, the polymeric nanocapsules are useful for treating or preventing stroke or an ischemic disease or disorder, particularly cerebral ischemia.

In certain embodiments, the polymeric nanocapsules are useful for treating or preventing advanced cancer, congenital pachymenia, age-related macular degeneration, choroidal neovascularization, metastatic melanoma without CNS metastasis, chronic myeloid leukemia, solid tumors, advanced solid tumors, optic atrophy, non-arterial anterior ischemic optic neuropathy, pancreatic cancer, pancreatic ductal adenocarcinoma, diabetic macular edema, hypercholesterolemia, colorectal cancer with liver metastasis, pancreatic cancer with liver metastasis, gastric cancer with liver metastasis, breast cancer with liver metastasis, ovarian cancer with liver metastasis, preeclampsia, neuroblastoma, ocular hypertension, open angle glaucoma, ocular pain, dry eye syndrome, kidney injury, acute renal failure, delayed graft function, renal graft complications, TBX3 overexpression, and diabetic retinopathy.

In certain embodiments, the polymeric nanocapsules are useful for treating or preventing viral infections. In certain embodiments, the polymeric nanocapsules are useful for treating or preventing retroviral infections. In certain embodiments, the polymeric nanocapsules are useful for treating or preventing HIV or AIDS infection. In particular embodiments, the polymeric nanocapsules are useful for inhibiting retroviral infection. In certain embodiments, the polymeric nanocapsules can be used to prevent, prevent or down-regulate retrovirus or viral replication. In certain embodiments, administration of the polymeric nanocapsule induces a physiological process selected from the group consisting of anti-apoptosis, neurogenesis, and angiogenesis.

In certain embodiments, the polymeric nanocapsules described herein may be administered by intravitreal injection, parenterally, intravenously, by injection into the callus at the base of one foot, by oral, subcutaneous or by any other mode of drug administration.

In certain embodiments, the method comprises administering to the subject a pharmaceutical composition of the invention such that expression of the target mutant gene is silenced or down-regulated. In certain embodiments, the subject is a mammal, such as a human. Due to the high specificity of the polymer nanocapsules of the present invention, they can specifically target mutant genes of affected cells and tissues, as described herein.

In the prevention of disease, the target gene may be a gene essential for the initiation or maintenance of the disease, or a gene that has been identified as being associated with a higher risk of contracting the disease. In the treatment of disease, the polymeric nanocapsule may be contacted with a cell or tissue exhibiting the disease. As one non-limiting example, the polymeric nanocapsule can deliver RNA or miRNA that is substantially identical to all or part of the cancer-associated mutant gene, or RNA or miRNA expressed at high levels in the tumor cell can be contacted with or introduced into the cancer cell or tumor gene. As another non-limiting example, the polymeric nanocapsule can deliver RNA or miRNA that is substantially identical to all or a portion of a viral or retroviral disease-associated mutant gene. In particular, a non-limiting example of a retroviral disease that can be treated with the polymeric nanocapsules described herein is HIV.

Non-limiting examples of cell proliferation and/or differentiation disorders include cancers, such as carcinomas, sarcomas, metastatic disorders, or hematopoietic tumor disorders, such as leukemias. Metastases may be caused by a variety of primary tumor types, including but not limited to tumors of prostate, colon, lung, breast and liver origin. As used herein, the terms "cancer," "hyperproliferative," and "tumor" refer to cells that have the ability to grow autonomously, i.e., an abnormal disease state characterized by rapid proliferative cell growth. These terms are intended to include all types of cancer growth or carcinogenic processes, metastatic tissues, or malignantly transformed cells, tissues, or organs, regardless of the type or stage of invasive histopathology. Proliferative disorders also include hematopoietic neoplastic disorders, including diseases associated with proliferating/neoplastic cells of hematopoietic origin, such as diseases caused by myeloid, lymphoid or erythroid lineages or precursors thereof.

Mutations in cellular genes that directly or indirectly control cell growth and differentiation are thought to be the major cause of cancer. There are approximately 30 gene families called oncogenes, which have been implicated in the formation of human tumors. One such family, the members of the RAS gene family, is carried in a wide range of eukaryotes and is often found mutated in human tumors. The polymer nanocapsules of the present invention can be used to target such oncogenes to knock down or prevent their expression.

In addition to oncogenes, the methods and compositions of the present invention may also be applied to other disease-associated target genes having point mutations. Gene mutations have been reported in more than 1000 different human genes. Data on these mutations and their associated phenotypes have been collated and can be obtained online from two major databases: one line Mendelian affinity inMan in Baltimore and Human Gene Mutation Database in Cardiff. For example, the frequency of CG to TG or CA mutations in the human genome is high due to deamination of 5' methyl-cytosine. Short deletions or insertions of less than 20 nucleotides are also very common mutations in humans. See, e.g., Antonarakis, s. e., eur. Pediatr. (2000) 159(3): 5173-8.

In addition, Sacidanandmam et al describe a map of human genomic sequence variation that contains 142 ten thousand single nucleotide polymorphisms that is helpful for identifying genes that are biomedically important for diagnosis and treatment (Sacidanandmam, R., et al, Nature (2001) 409(6822):821-2 and Nature (2001) 409(6822): 822-3). This map integrates all publicly available SNPs and other genomic features of the described genes. It is estimated that 60,000 SNPs fall within exons (coding and untranslated regions), and that 85% of the exons are within 5 kb of the most recent SNP. Clifford et al provide expression-based genetic/physical maps of single nucleotide polymorphisms identified by cancer genomic dissection (Clifford, r., et al., Genome Res (2000) 10(8): 1259-65). In addition to SNP profiles, Sachidanandam et al also provide profiles of SNPs containing genes expressed in breast, colon, kidney, liver, lung or prostate tissue.

Thus, the microrna molecule polymer nanocapsules of the present invention can be used to target such mutant genes to knock down or prevent their expression.

In certain embodiments, the method of treatment comprises administering additional treatment. In certain embodiments, the additional therapy is selected from anticoagulation therapy, antiplatelet therapy, or thrombolytic therapy.

Method for inhibiting expression of mutant gene

In yet another aspect, the invention relates to a method of inhibiting mutant gene expression in a subject. The method comprises administering to the subject a composition of the invention such that expression of the mutant gene is silenced as compared to a corresponding wild-type gene. In certain embodiments, the subject is a mammal. In certain embodiments, the mammal is a human.

Due to their high specificity, the miRNA nanocapsules of the present invention can specifically target RNA (primary or processed) of the target mutant gene at surprisingly low doses. Compositions and methods for inhibiting expression of a target gene using a polymer nanocapsule can be performed as described herein.

In certain embodiments, the invention encompasses administering a composition comprising a polymeric nanocapsule, wherein the polymeric nanocapsule comprises a nucleotide sequence, such as a miRNA targeted to a mutant gene. When the subject to be treated is a mammal, such as a human, the compositions can be administered by any method known in the art, including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal, and topical (including buccal and sublingual) administration. In certain preferred embodiments, the compositions are administered by intravenous or intraperitoneal (intraperitoneal) infusion or injection.

The method of inhibiting the expression of a target gene can be applied to any mutant gene whose silencing is desired, thereby selectively inhibiting its expression. Non-limiting examples of human genes that can be used for targeted silencing include oncogenes cytokinin genes, idiotypic protein genes, prion genes, genes expressing molecules that induce angiogenesis, genes encoding adhesion molecules, genes encoding cell surface receptors, genes encoding proteins involved in metastasis and/or invasion processes, proteases and molecules that regulate apoptosis and cell cycle, genes expressing EGF receptors, genes encoding multidrug resistance 1 gene (MDR1 gene), genes that allow viral uptake and replication, genes that cause neurodegenerative disorders, genes that cause protein aggregation and/or accumulation, genes that cause hormone up-or down-regulation, genes that cause neurological disorders, genes that cause heart disorders, and genes that cause psychological disorders. Those skilled in the art will understand which genes are contained in the broad category of exemplary genes described above.

Preparation method

In certain embodiments, the polymer nanocapsules described herein are prepared in order to achieve a particular size, target a particular site for gene downregulation, and downregulate a particular gene. The size of the polymer nanocapsules described herein can be determined according to the polymer to crosslinker ratio as described herein. For example, targeted delivery can be achieved using a conjugation agent attached (i.e., conjugated) to the exterior of the polymeric nanocapsules described herein. In addition, specific binding of the RNAi molecules of the polymer nanocapsules described herein to a particular gene (thereby reducing expression of a particular gene) can be achieved by designing the RNAi molecules using methods known to those skilled in the art. See, e.g., Birmingham et al, "A Protocols for designing siRNAs with high functionality and specificity," Nature Protocols, 2007; 2(9): 2068-78. In addition, by adjusting the ratio of degradable to non-degradable polymers as described herein, the ability of the polymeric nanocapsule to deliver RNAi molecules to a target site can be optimized and determined.

Administration method

The pharmaceutical compositions encompassed by the present invention may be administered by any method known in the art, including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration. In certain preferred embodiments, the pharmaceutical composition is administered by intravenous or intraperitoneal (intraperitoneal) infusion or injection.

The term "subject" contemplated for administration includes, but is not limited to, humans (i.e., male or female of any age group, such as pediatric subjects (e.g., infants, children, adolescents) or adult subjects (e.g., young adults, middle-aged adults, or elderly adults) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys), mammals including commercially relevant mammals such as cows, pigs, horses, sheep, goats, cats, and/or dogs, and/or birds including commercially relevant birds such as chickens, ducks, geese, quail, and/or turkeys.

In certain embodiments, the disclosed compositions are administered nasally in a range from once daily, to once weekly, to once every two weeks, to once monthly. In a preferred embodiment, the microparticles and compositions are administered nasally. As used herein, the term "nasal" or "nasally administered" refers to the delivery of microparticles to the nasal mucosa of a subject, with the contents of the microparticles being absorbed directly into the nasal tissue.

In certain embodiments, the polymeric nanocapsules, microparticles and compositions comprising them are for systemic administration.

In certain embodiments, administration of the polymeric nanocapsule increases expression of at least one of VEGF, GFAP, CD31, CD34, BCL-2, NeuN, biscortin, nestin, PSA-NCAM, biscortin, Pax6, GAP-43, AKT, or HIF1- α.

Examples

47页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用锆-89标记的超小纳米颗粒及其方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类