1,128
Views
43
CrossRef citations to date
0
Altmetric
Review

Recent advances in polymeric materials for the delivery of RNA therapeutics

, , & ORCID Icon
Pages 1149-1167 | Received 08 Jul 2019, Accepted 02 Sep 2019, Published online: 12 Sep 2019

References

  • Gonçalves GAR, Paiva RD. Gene therapy: advances, challenges and perspectives. Einstein (São Paulo). 2017;15:369–375.
  • Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019. DOI:10.1038/s41573-019-0012-9
  • Eckstein F. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther. 2014;24:374–387.
  • Dirin M, Winkler J. Influence of diverse chemical modifications on the ADME characteristics and toxicology of antisense oligonucleotides. Expert Opin Biol Ther. 2013;13:875–888.
  • Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016;44:6518–6548.
  • Khvorova A, Watts JK. The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol. 2017;35:238–248.
  • Liang X-H, Shen W, Sun H, et al. Hsp90 protein interacts with phosphorothioate oligonucleotides containing hydrophobic 2′-modifications and enhances antisense activity. Nucleic Acids Res. 2016;44:3892–3907.
  • Castanotto D, Lin M, Kowolik C, et al. A cytoplasmic pathway for gapmer antisense oligonucleotide-mediated gene silencing in mammalian cells. Nucleic Acids Res. 2015;43:9350–9361.
  • Stein CA, Hansen JB, Lai J, et al. Efficient gene silencing by delivery of locked nucleic acid antisense oligonucleotides, unassisted by transfection reagents. Nucleic Acids Res. 2009;38:1–8.
  • Agrawal S, Kandimalla ER. Antisense and siRNA as agonists of Toll-like receptors. Nat Biotechnol. 2004;22:1533–1537.
  • Wilson PA, Kitson J, Turner G, et al. In silico and in vitro evaluation of exonic and intronic off-target effects form a critical element of therapeutic ASO gapmer optimization. Nucleic Acids Res. 2015;43:8638–8650.
  • Dowdy SF, Levy M. RNA therapeutics (almost) comes of age: targeting, delivery and endosomal escape. Nucleic Acid Ther. 2018;28:107–108.
  • Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet. 2003;4:346–358.
  • Wu Z, Yang H, Colosi P. Effect of genome size on AAV vector packaging. Mol Ther. 2009;18:80–86.
  • Chew WL, Tabebordbar M, Cheng JKW, et al. A multifunctional AAV–CRISPR–cas9 and its host response. Nat Methods. 2016;13:868–874.
  • Wisse E, Jacobs F, Topal B, et al. The size of endothelial fenestrae in human liver sinusoids: implications for hepatocyte-directed gene transfer. Gene Ther. 2008;15:1193–1199.
  • Huang Y, Hong J, Zheng S, et al. Elimination pathways of systemically delivered siRNA. Mol Ther. 2011;19:381–385.
  • Sabnis S, Kumarasinghe ES, Salerno T, et al. A novel amino lipid series for mRNA delivery: improved endosomal escape and sustained pharmacology and safety in non-human primates. Mol Ther. 2018;26:1509–1519.
  • Kleinman ME, Yamada K, Takeda A, et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature. 2008;452:591–597.
  • Coelho T, Adams D, Silva A, et al. Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med. 2013;369:819–829.
  • Kaczmarek JC, Kowalski PS, Anderson DG. Advances in the delivery of RNA therapeutics: from concept to clinical reality. Genome Med. 2017;9:60.
  • Jayaraman M, Ansell SM, Mui BL, et al. Maximizing the potency of siRNA lipid nanoparticles for hepatic gene silencing in vivo. Angew Chem Int Educ. 2012;51:8529–8533.
  • Andorko JI, Hess KL, Pineault KG, et al. Intrinsic immunogenicity of rapidly-degradable polymers evolves during degradation. Acta Biomater. 2016;32:24–34.
  • Kriegel C, Amiji M. Oral TNF-α gene silencing using a polymeric microsphere-based delivery system for the treatment of inflammatory bowel disease. J Control Release. 2011;150:77–86.
  • Knipe JM, Strong LE, Peppas NA. Enzyme- and pH-responsive microencapsulated nanogels for oral delivery of sirna to induce TNF-α knockdown in the intestine. Biomacromolecules. 2016;17:788–797.
  • Barba AA, Cascone S, Caccavo D, et al. Engineering approaches in siRNA delivery. Int J Pharm. 2017;525:343–358.
  • Godbey WT, Wu KK, Mikos AG. Size matters: molecular weight affects the efficiency of poly(ethylenimine) as a gene delivery vehicle. J Biomed Mater Res. 1999;45:268–275.
  • Boussif O, Lezoualc’h F, Zanta MA, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci U S A. 1995;92:7297–7301.
  • Demeneix B, Behr J-P. Polyethylenimine (PEI). Adv Genet. 2005;53PA:215–230.
  • Curtis KA, Miller D, Millard P, et al. Unusual salt and pH induced changes in polyethylenimine solutions. PLoS One. 2016;11:1–20.
  • Benjaminsen RV, Mattebjerg MA, Henriksen JR, et al. The possible “proton sponge “ effect of polyethylenimine (PEI) does not include change in lysosomal pH. Mol Ther. 2013;21:149–157.
  • Guo Z, Kong Z, Wei Y, et al. Effects of gene carrier polyethyleneimines on the structure and binding capability of bovine serum albumin. Spectrochim Acta - Part A Mol Biomol Spectrosc. 2017;173:783–791.
  • Lv H, Zhang S, Wang B, et al. Toxicity of cationic lipids and cationic polymers in gene delivery. J Control Release. 2006;114:100–109.
  • Boeckle S, von Gersdorff K, van der Piepen S, et al. Purification of polyethylenimine polyplexes highlights the role of free polycations in gene transfer. J Gene Med. 2004;6:1102–1111.
  • Kircheis R, Wightman L, Schreiber A, et al. Polyethylenimine/DNA complexes shielded by transferrin target gene expression to tumors after systemic application. Gene Ther. 2001;8:28–40.
  • Kafil V, Omidi Y. Cytotoxic impacts of linear and branched polyethylenimine nanostructures in A431 cells. BioImpacts. 2011;1:23–30.
  • Petersen H, Fechner PM, Martin AL, et al. Polyethylenimine-graft-poly(ethylene glycol) copolymers: influence of copolymer block structure on DNA complexation and biological activities as gene delivery system. Bioconjug Chem. 2002;13:845–854.
  • Malek A, Czubayko F, Aigner A. PEG grafting of polyethylenimine (PEI) exerts different effects on DNA transfection and siRNA-induced gene targeting efficacy. J Drug Target. 2008;16:124–139.
  • Urban-Klein B, Werth S, Abuharbeid S, et al. RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo. Gene Ther. 2005;12:461–466.
  • Rejman J, Tavernier G, Bavarsad N, et al. MRNA transfection of cervical carcinoma and mesenchymal stem cells mediated by cationic carriers. J Control Release. 2010;147:385–391.
  • Wang M, Wu B, Tucker JD, et al. Poly(ester amine) constructed from polyethylenimine and pluronic for gene delivery in vitro and in vivo. Drug Deliv. 2016;23:3224–3233.
  • Kim YH, Konák C, Ulbrich K. Polyethylenimine with acid-labile linkages as a biodegradable gene carrier. J Control Release. 2005;103:209–219.
  • Song J, Chen Y, Jiang S, et al. Efficient and non-toxic biological response carrier delivering tnf-α shrna for gene silencing in a murine model of rheumatoid arthritis. Front Immunol. 2016;7:1–11.
  • Song J, Li X, Li Y, et al. Biodegradable and biocompatible cationic polymer delivering microRNA-221/222 promotes nerve regeneration after sciatic nerve crush. Int J Nanomedicine. 2017;12:4195–4208.
  • Gosselin MA, Guo W, Lee RJ. Efficient gene transfer using reversibly cross-linked low molecular weight polyethylenimine. Bioconjug Chem. 2001;12:989–994.
  • Matar M, Slobodkin G, Rea-Ramsey A, et al. Synthesis and characterization of low molecular weight linear polyethylenimines for gene delivery. J Biomed Nanotechnol. 2006;2:53–61.
  • Neu M, Germershaus O, Mao S, et al. Crosslinked nanocarriers based upon poly(ethylene imine) for systemic plasmid delivery: in vitro characterization and in vivo studies in mice. J Control Release. 2007;118:370–380.
  • Mishra S, Webster P, Davis ME. PEGylation significantly affects cellular uptake and intracellular trafficking of non-viral gene delivery particles. Eur J Cell Biol. 2004;83:97–111.
  • Chiper M, Tounsi N, Kole R, et al. Self-aggregating 1.8 kDa polyethylenimines with dissolution switch at endosomal acidic pH are delivery carriers for plasmid DNA, mRNA, siRNA and exon-skipping oligonucleotides. J Control Release. 2017;246:60–70.
  • Chiper M, Tounsi N, Kole R, et al. Aromatic modification of low molecular weight PEI for enhanced gene delivery. Polymers (Basel). 2017;9:362.
  • Dahlman JE, Barnes C, Khan OF, et al. In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight. Nat Nanotechnol. 2014;9:648–655.
  • Xue W, Dahlman JE, Tammela T, et al. Small RNA combination therapy for lung cancer. Proc Natl Acad Sci. 2014;111:E3553–E3561.
  • Sager HB, Dutta P, Dahlman JE, et al. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction. 2016;8(342):342ra80–342ra80.
  • Sager HB, Hulsmans M, Lavine KJ, et al. Proliferation and recruitment contribute to myocardial macrophage expansion in chronic heart failure. Circ Res. 2016;119:853–864.
  • Yu D, Khan OF, Suvà ML, et al. Multiplexed RNAi therapy against brain tumor-initiating cells via lipopolymeric nanoparticle infusion delays glioblastoma progression. Proc Natl Acad Sci. 201701911. 2017. DOI:10.1073/pnas.1701911114
  • Wilson RA, Espinosa-Diez C, Kanner N, et al. MicroRNA regulation of endothelial TREX1 reprograms the tumour microenvironment. Nat Commun. 2016;7:1–10.
  • Love KT, Mahon KP, Levins CG, et al. Lipid-like materials for low-dose, in vivo gene silencing. Proc Natl Acad Sci. 2010;107:1864–1869.
  • Novobrantseva TI, Borodovsky A, Wong J, et al. Systemic RNAi-mediated gene silencing in nonhuman primate and rodent myeloid cells. Mol Ther Nucleic Acids. 2012;1:e4.
  • Whitehead KA, Dahlman JE, Langer RS, et al. Silencing or stimulation? siRNA delivery and the immune system. Annu Rev Chem Biomol Eng. 2011;2:77–96.
  • Chen Q, Osada K, Ishii T, et al. Homo-catiomer integration into PEGylated polyplex micelle from block-catiomer for systemic anti-angiogenic gene therapy for fibrotic pancreatic tumors. Biomaterials. 2012;33:4722–4730.
  • Khan OF, Kowalski PS, Doloff JC, et al. Endothelial siRNA delivery in nonhuman primates using ionizable low–molecular weight polymeric nanoparticles. Sci Adv. 2018;4:1–10.
  • Sago CD, Lokugamage MP, Paunovska K, et al. High-throughput in vivo screen of functional mRNA delivery identifies nanoparticles for endothelial cell gene editing. Proc Natl Acad Sci. 2018;115:E9944–E9952.
  • Yin H, Song C-Q, Dorkin JR, et al. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat Biotechnol. 2016;34:328–333.
  • Givens BE, Naguib YW, Geary SM, et al. Nanoparticle-based delivery of CRISPR/Cas9 genome-editing therapeutics. Aaps J. 2018;20:108.
  • Liu C, Zhang L, Liu H, et al. Delivery strategies of the CRISPR-Cas9 gene-editing system for therapeutic applications. J Control Release. 2017;266:17–26.
  • Suenaga T, Kohyama M, Hirayasu K, et al. Engineering large viral DNA genomes using the CRISPR-Cas9 system. Microbiol Immunol. 2014;58:513–522.
  • Ryu N, Kim M-A, Park D, et al. Effective PEI-mediated delivery of CRISPR-Cas9 complex for targeted gene therapy. Nanomed Nanotechnol Biol Med. 2018;14:2095–2102.
  • Sun W, Ji W, Hall JM, et al. Self-assembled DNA nanoclews for the efficient delivery of CRISPR-Cas9 for genome editing. Angew Chem Int Educ. 2015;54:12029–12033.
  • Sun W, Ji W, Hu Q, et al. Transformable DNA nanocarriers for plasma membrane targeted delivery of cytokine. Biomaterials. 2016;96:1–10.
  • Zuckerman JE, Davis ME. Clinical experiences with systemically administered siRNA-based therapeutics in cancer. Nat Rev Drug Discov. 2015;14:843–856.
  • Chen X, Qiu YK, Owh C, et al. Supramolecular cyclodextrin nanocarriers for chemo- and gene therapy towards the effective treatment of drug resistant cancers. Nanoscale. 2016;8:18876–18881.
  • Ping Y, Hu Q, Tang G, et al. FGFR-targeted gene delivery mediated by supramolecular assembly between β-cyclodextrin-crosslinked PEI and redox-sensitive PEG. Biomaterials. 2013;34:6482–6494.
  • Davis ME, Snider GS, Kuekes PJ, et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464:1067–1070.
  • Zuckerman JE, Gritli I, Tolcher A, et al. Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA. Proc Natl Acad Sci. 2014;111:11449–11454.
  • Zhang Z, Wan T, Chen Y, et al. Cationic polymer-mediated CRISPR/Cas9 plasmid delivery for genome editing. Macromol Rapid Commun. 2018;1800068:1–8.
  • Truong NP, Gu W, Prasadam I, et al. An influenza virus-inspired polymer system for the timed release of siRNA. Nat Commun. 2013;4:1902–1907.
  • Truong NP, Jia Z, Burges M, et al. Self-catalyzed degradation of linear cationic poly(2-dimethylaminoethyl acrylate) in water. Biomacromolecules. 2011;12:1876–1882.
  • Cheng C, Convertine AJ, Stayton PS, et al. Multifunctional triblock copolymers for intracellular messenger RNA delivery. Biomaterials. 2012;33:6868–6876.
  • Cheng Y, Sellers DL, Tan JKY, et al. Development of switchable polymers to address the dilemma of stability and cargo release in polycationic nucleic acid carriers. Biomaterials. 2017;127:89–96.
  • Nelson CE, Kintzing JR, Hanna A, et al. Balancing cationic and hydrophobic content of PEGylated siRNA polyplexes enhances endosome escape, stability, blood circulation time, and bioactivity in vivo. ACS Nano. 2013;7:8870–8880.
  • Wang Y, Zhou K, Huang G, et al. A nanoparticle-based strategy for the imaging of a broad range of tumours by nonlinear amplification of microenvironment signals. Nat Mater. 2014;13:204–212.
  • Cheng Y, Wei H, Tan J-KY, et al. Nano-sized sunflower polycations as effective gene transfer vehicles. Small. 2016;12:2750–2758.
  • Cheng Y, Yumul RC, Pun SH. Virus-inspired polymer for efficient in vitro and in vivo gene delivery. Angew Chem. 2016;128:12192–12196.
  • Yen A, Cheng Y, Sylvestre M, et al. Serum nuclease susceptibility of mRNA cargo in condensed polyplexes. Mol Pharm. 2018;15:2268–2276.
  • Yan Y, Liu L, Xiong H, et al. Functional polyesters enable selective siRNA delivery to lung cancer over matched normal cells. Proc Natl Acad Sci. 2016;113:E5702–E5710.
  • Yan Y, Zhou K, Xiong H, et al. Aerosol delivery of stabilized polyester-siRNA nanoparticles to silence gene expression in orthotopic lung tumors. Biomaterials. 2017;118:84–93.
  • Harguindey A, Qu Y, Gupta TD, et al. Synthesis and assembly of click-nucleic-acid-containing PEG-PLGA nanoparticles for DNA delivery. Adv Mater. 2017;29:1700743.
  • Li Z, Liu X, Chen X, et al. Targeted delivery of Bcl-2 conversion gene by MPEG-PCL-PEI-FA cationic copolymer to combat therapeutic resistant cancer. Mater Sci Eng C. 2017;76:66–72.
  • Han S, Cheng Q, Wu Y, et al. Effects of hydrophobic core components in amphiphilic PDMAEMA nanoparticles on siRNA delivery. Biomaterials. 2015;48:45–55.
  • Vert M, Li SM, Spenlehauer G, et al. Bioresorbability and biocompatibility of aliphatic polyesters. J Mater Sci Mater Med. 1992;3:432–446.
  • Okamoto M, John B. Synthetic biopolymer nanocomposites for tissue engineering scaffolds. Prog Polym Sci. 2013;38:1487–1503.
  • Anderson JM, Shive MS. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Deliv Rev. 2012;64:72–82.
  • Sparks J, Scholz C. Evaluation of a cationic poly(beta-hydroxyalkanoate) as a plasmid DNA delivery system. Biomacromolecules. 2009;10:1715–1719.
  • Li Z, Loh XJ. Water soluble polyhydroxyalkanoates: future materials for therapeutic applications. Chem Soc Rev. 2015;44:2865–2879.
  • Yang XZ, Dou S, Sun T-M, et al. Systemic delivery of siRNA with cationic lipid assisted PEG-PLA nanoparticles for cancer therapy. J Control Release. 2011;156:203–211.
  • Lim YB, Kim SM, Suh H, et al. Biodegradable, endosome disruptive, and cationic network-type polymer as a highly efficient and nontoxic gene delivery carrier. Bioconjug Chem. 2002;13:952–957.
  • Li Z, Ning W, Wang J, et al. Controlled gene delivery system based on thermosensitive biodegradable hydrogel. Pharm Res. 2003;20:884–888.
  • Rivkin L, Gabai R, Rachmut IH, et al. Mutant KRAS is a druggable target for pancreatic cancer. Proc Natl Acad Sci. 2013;110:20723–20728.
  • Su W-P, Cheng F-Y, Shieh D-B, et al. PLGA nanoparticles codeliver paclitaxel and stat3 siRNA to overcome cellular resistance in lung cancer cells. Int J Nanomedicine. 2012;7:4269–4283.
  • Patil YB, Swaminathan SK, Sadhukha T, et al. The use of nanoparticle-mediated targeted gene silencing and drug delivery to overcome tumor drug resistance. Biomaterials. 2010;31:358–365.
  • Reul R, Nguyen J, Kissel T. Amine-modified hyperbranched polyesters as non-toxic, biodegradable gene delivery systems. Biomaterials. 2009;30:5815–5824.
  • Oster CG, Wittmar M, Unger F, et al. Design of amine-modified graft polyesters for effective gene delivery using DNA-loaded nanoparticles. Pharm Res. 2004;21:927–931.
  • Yan Y, Siegwart DJ. Scalable synthesis and derivation of functional polyesters bearing ene and epoxide side chains. Polym Chem. 2014;5:1362–1371.
  • Yan Y, Xiong H, Zhang X, et al. Systemic mRNA delivery to the lungs by functional polyester-based carriers. Biomacromolecules. 2017;18:4307–4315.
  • Jensen DK, Jensen LB, Koocheki S, et al. Design of an inhalable dry powder formulation of DOTAP-modified PLGA nanoparticles loaded with siRNA. J Control Release. 2012;157:141–148.
  • Colombo S, Cun D, Remaut K, et al. Mechanistic profiling of the siRNA delivery dynamics of lipid-polymer hybrid nanoparticles. J Control Release. 2015;201:22–31.
  • Zuo ZQ, Chen K-G, Yu X-Y, et al. Promoting tumor penetration of nanoparticles for cancer stem cell therapy by TGF-β signaling pathway inhibition. Biomaterials. 2016;82:48–59.
  • Siegwart DJ, Whitehead KA, Nuhn L, et al. Combinatorial synthesis of chemically diverse core-shell nanoparticles for intracellular delivery. Proc Natl Acad Sci. 2011;108:12996–13001.
  • Zhu X, Xu Y, Solis LM, et al. Long-circulating siRNA nanoparticles for validating prohibitin1-targeted non-small cell lung cancer treatment. Proc Natl Acad Sci. 2015;112:7779–7784.
  • Zhou P, Pavlova ER, Bagrov DV, et al. Delivery siRNA with a novel gene vector for glioma therapy by targeting Gli1. Int J Nanomedicine. 2018;13:4781–4793.
  • Liu Y, Sun Y, Rai K, et al. Systemic delivery of CRISPR/Cas9 with PEG-PLGA nanoparticles for chronic myeloid leukemia targeted therapy. Biomater Sci. 2018;6:1592–1603.
  • Zhao W, Zhang C, Li B, et al. Lipid polymer hybrid nanomaterials for mRNA delivery. Cell Mol Bioeng. 2018;11:397–406.
  • Green JJ, Zugates G, Tedford N, et al. Combinatorial modification of degradable polymers enables transfection of human cells comparable to adenovirus. Adv Mater. 2007;19:2836–2842.
  • Dosta P, Segovia N, Cascante A, et al. Surface charge tunability as a powerful strategy to control electrostatic interaction for high efficiency silencing, using tailored oligopeptide-modified poly(beta-amino ester)s (PBAEs). Acta Biomater. 2015;20:82–93.
  • Dosta P, Ramos V, Borrós S. Stable and efficient generation of poly(β-amino ester)s for RNAi delivery. Mol Syst Des Eng. 2018;3:677–689.
  • Lynn DM, Langer R. Degradable Poly(β-amino esters): synthesis, characterization, and self-assembly with plasmid DNA. J Am Chem Soc. 2000;122:10761–10768.
  • Anderson DG, Peng W, Akinc A, et al. A polymer library approach to suicide gene therapy for cancer. Proc Natl Acad Sci. 2004;101:16028–16033.
  • Akinc A, Anderson DG, Lynn DM, et al. Synthesis of poly(β-amino ester)s optimized for highly effective gene delivery. Bioconjug Chem. 2003;14:979–988.
  • Anderson DG, Levenberg S, Langer R. Nanoliter-scale synthesis of arrayed biomaterials and application to human embryonic stem cells. Nat Biotechnol. 2004;22:863–866.
  • Murphy JE, Uno T, Hamer JD, et al. A combinatorial approach to the discovery of efficient cationic peptoid reagents for gene delivery. Proc Natl Acad Sci U S A. 1998;95:1517–1522.
  • Capasso Palmiero U, Kaczmarek JC, Fenton OS, et al. Poly(β-amino ester)-co-poly(caprolactone) terpolymers as nonviral vectors for mRNA delivery in vitro and in vivo. Adv Healthc Mater. 2018;7:e1800249.
  • Kaczmarek JC, Patel AK, Kauffman KJ, et al. polymer-lipid nanoparticles for systemic delivery of mRNA to the lungs. Angew Chem Int Educ. 2016;55:13808–13812.
  • Kaczmarek JC, Pan L, Ma Z, et al. Optimization of a degradable polymer–lipid nanoparticle for potent systemic delivery of mRNA to the lung endothelium and immune cells. Nano Lett. 2018;18:6449–6454.
  • Zhang X, Tang W, Yang Z, et al. PEGylated poly(amine-co-ester) micelles as biodegradable non-viral gene vectors with enhanced stability, reduced toxicity and higher in vivo transfection efficacy. J Mater Chem B. 2014;2:4034.
  • Kim J, Kang Y, Tzeng SY, et al. Synthesis and application of poly(ethylene glycol)-co-poly(β-amino ester) copolymers for small cell lung cancer gene therapy. Acta Biomater. 2016;41:293–301.
  • Deng X, Zheng N, Song Z, et al. Trigger-responsive, fast-degradable poly(β-amino ester)s for enhanced DNA unpackaging and reduced toxicity. Biomaterials. 2014;35:5006–5015.
  • Liu Y, Chen J, Tang Y, et al. Synthesis and characterization of quaternized poly(β-amino ester) for highly efficient delivery of small interfering RNA. Mol Pharm. 2018;15:4558–4567.
  • Wang D, Zhao T, Zhu X, et al. Bioapplications of hyperbranched polymers. Chem Soc Rev. 2015;44:4023–4071.
  • Huang JY, Gao Y, Cutlar L, et al. Tailoring highly branched poly(β-amino ester)s: a synthetic platform for epidermal gene therapy. Chem Commun. 2015;51:8473–8476.
  • Cutlar L, Brandt S, Esch S, et al. Highly branched poly(β-amino esters): synthesis and application in gene delivery. Biomacromolecules. 2015;16:2609–2617.
  • Gao Y, Falatach R, McGlone C, et al. Highly branched poly(β-amino esters) for non-viral gene delivery: high transfection efficiency and low toxicity achieved by increasing molecular weight. Biomacromolecules. 2016;17:3640–3647.
  • Wang WW, Waters MR, Perrotti A, et al. The transition from linear to highly branched poly(β-amino ester)s: branching matters for gene delivery. Sci Adv. 2016;2:1–14.
  • Eltoukhy AA, Siegwart DJ, Alabi CA, et al. Effect of molecular weight of amine end-modified poly(β-amino ester)s on gene delivery efficiency and toxicity. Biomaterials. 2012;33:3594–3603.
  • Patel AK, Kaczmarek JC, Bose S, et al. Inhaled nanoformulated mRNA polyplexes for protein production in lung epithelium. Adv Mater. 2019;31:1805116.
  • Zeng M, Kang Z, Tian Z, et al. Highly branched poly(5-amino-1-pentanol-co-1, 4butanediol diacrylate) for high performance gene transfection. Polymers. 2017;9:161.
  • Mastrobattista E, Hennink WE. Polymers for gene delivery: charged for success. Nat Mater. 2012;11:10–12.
  • Richardson SCW, Pattrick NG, Stella Man YK, et al. Poly(amidoamine)s as potential nonviral vectors: ability to form interpolyelectrolyte complexes and to mediate transfection in vitro. Biomacromolecules. 2001;2:1023–1028.
  • Rajasekaran D, Srivastava J, Ebeid K, et al. Combination of nanoparticle-delivered siRNA for astrocyte elevated gene-1 (AEG-1) and all-trans retinoic acid (ATRA): an effective therapeutic strategy for hepatocellular carcinoma (HCC). Bioconjug Chem. 2015;26:1651–1661.
  • Zhang X-Q, Intra J, Salem AK. Conjugation of polyamidoamine dendrimers on biodegradable microparticles for nonviral gene delivery. Bioconjug Chem. 2007;18:2068–2076.
  • Kim HJ, Gupta MK, Page JM, et al. siRNA delivery from triblock copolymer micelles with spatially-ordered compartments of PEG shell, siRNA-loaded intermediate layer, and hydrophobic core. Biomaterials. 2014;35:4548–4556.
  • Liu H, Wang H, Yang W, et al. Disulfide cross-linked low generation dendrimers with high gene transfection efficacy, low cytotoxicity, and low cost. J Am Chem Soc. 2012;134:17680–17687.
  • Lin C, Zhong Z, Lok MC, et al. Novel bioreducible poly(amido amine)s for highly efficient gene delivery. Bioconjug Chem. 2007;18:138–145.
  • Parmar RG, Busuek M, Walsh ES, et al. Endosomolytic bioreducible poly(amido amine disulfide) polymer conjugates for the in vivo systemic delivery of siRNA therapeutics. Bioconjug Chem. 2013;24:640–647.
  • Parmar RG, Poslusney M, Busuek M, et al. Novel endosomolytic poly(amido amine) polymer conjugates for systemic delivery of siRNA to hepatocytes in rodents and nonhuman primates. Bioconjug Chem. 2014;25:896–906.
  • Kretzmann JA, Zhang X, Zhu Y, et al. Synthetically controlling dendrimer flexibility improves delivery of large plasmid DNA. Chem Sci. 2017;8:2923–2930.
  • Xiong SD, Chen Y-J, Chen H-L, et al. Cationic fluorine-containing amphiphilic graft copolymers as DNA carriers. Biomaterials. 2010;31:2673–2685.
  • Kasuya MCZ, Nakano S, Katayama R, et al. Evaluation of the hydrophobicity of perfluoroalkyl chains in amphiphilic compounds that are incorporated into cell membrane. J Fluor Chem. 2011;132:202–206.
  • Percec V, Imam MR, Peterca M, et al. Self-organizable vesicular columns assembled from polymers dendronized with semifluorinated janus dendrimers act as reverse thermal actuators. J Am Chem Soc. 2012;134:4408–4420.
  • Wang M, Liu H, Li L, et al. A fluorinated dendrimer achieves excellent gene transfection efficacy at extremely low nitrogen to phosphorus ratios. Nat Commun. 2014;5:1–8.
  • Uchida H, Miyata K, Oba M, et al. Odd–even Effect of repeating aminoethylene units in the side chain of N-substituted polyaspartamides on gene Transfection profiles. J Am Chem Soc. 2011;133:15524–15532.
  • Miyata K, Oba M, Nakanishi M, et al. Polyplexes from poly(aspartamide) bearing 1,2-diaminoethane side chains induce pH-selective, endosomal membrane destabilization with amplified transfection and negligible cytotoxicity. J Am Chem Soc. 2008;130:16287–16294.
  • Akagi D, Oba M, Koyama H, et al. Biocompatible micellar nanovectors achieve efficient gene transfer to vascular lesions without cytotoxicity and thrombus formation. Gene Ther. 2007;14:1029–1038.
  • Itaka K, Ohba S, Miyata K, et al. Bone regeneration by regulated in vivo gene transfer using biocompatible polyplex nanomicelles. Mol Ther. 2007;15:1655–1662.
  • Harada-Shiba M, Takamisawa I, Miyata K, et al. Intratracheal gene transfer of adrenomedullin using polyplex nanomicelles attenuates monocrotaline-induced pulmonary hypertension in rats. Mol Ther. 2009;17:1180–1186.
  • Cavallaro G, Licciardi M, Scirè S, et al. New pegylated polyaspartamide-based polyplexes as gene delivery vectors. Nanomedicine. 2010;5:243–258.
  • Licciardi M, Cavallaro G, Amato G, et al. New copolymers graft of α,β-poly(N-2-hydroxyethyl)-d,l- aspartamide obtained from atom transfer radical polymerization as vector for gene delivery. React Funct Polym. 2012;72:268–278.
  • Suma T, Miyata K, Ishii T, et al. Enhanced stability and gene silencing ability of siRNA-loaded polyion complexes formulated from polyaspartamide derivatives with a repetitive array of amino groups in the side chain. Biomaterials. 2012;33:2770–2779.
  • Uchida H, Itaka K, Nomoto T, et al. Modulated protonation of side chain aminoethylene repeats in N-substituted polyaspartamides promotes mRNA transfection. J Am Chem Soc. 2014;136:12396–12405.
  • Takemoto H, Chen Y-J, Chen H-L, et al. Polyion complex stability and gene silencing efficiency with a siRNA-grafted polymer delivery system. Biomaterials. 2010;31:8097–8105.
  • Cavallaro G, Vatanara A, Najafabadi AR, et al. Synthesis and characterization of polyaspartamide copolymers obtained by ATRP for nucleic acid delivery. Int J Pharm. 2014;466:246–257.
  • Cavallaro G, Farra R, Craparo EF, et al. Galactosylated polyaspartamide copolymers for siRNA targeted delivery to hepatocellular carcinoma cells. Int J Pharm. 2017;525:397–406.
  • Lu H, Wang J, Bai Y, et al. Ionic polypeptides with unusual helical stability. Nat Commun. 2011;2:206–209.
  • Deming TJ. Facile synthesis of block copolypeptides of defined architecture. Nature. 1997;390:386–389.
  • Ulkoski D, Scholz C. Impact of cationic charge density and pegylated poly(amino acid) tercopolymer architecture on their use as gene delivery vehicles. Part 2: DNA protection, stability, cytotoxicity, and transfection efficiency. Macromol Biosci. 2018;18:e1800109.
  • Martin ME, Rice KG. Peptide-guided gene delivery. Aaps J. 2007;9:E18–E29.
  • Pati D, Shaikh AY, Hotha S, et al. Synthesis of glycopolypeptides by the ring opening polymerization of O-glycosylated-α-amino acid N-carboxyanhydride (NCA). Polym Chem. 2011;2:805.
  • Kramer JR, Deming TJ. Glycopolypeptides with a redox-triggered helix-to-coil transition. J Am Chem Soc. 2012;134:4112–4115.
  • Yu M, Nowak AP, Deming TJ, et al. Methylated mono- and diethyleneglycol functionalized polylysines: nonionic, α-helical, water-soluble polypeptides. J Am Chem Soc. 1999;121:12210–12211.
  • Chen C, Wang Z, Li Z. Thermoresponsive polypeptides from pegylated poly-L-glutamates. Biomacromolecules. 2011;12:2859–2863.
  • Yakovlev I, Deming TJ. Analogues of poly(l-phosphoserine) via living polymerization of phosphonate-containing N-carboxyanhydride monomers. ACS Macro Lett. 2014;3:378–381.
  • Miyata K, Nishiyama N, Kataoka K. Rational design of smart supramolecular assemblies for gene delivery: chemical challenges in the creation of artificial viruses. Chem Soc Rev. 2012;41:2562–2574.
  • Cheever MA. `Mac’. Twelve immunotherapy drugs that could cure cancers. Immunol Rev. 2008;222:357–368.
  • Diaz-San Segundo F, Dias CC, Moraes MP, et al. Poly ICLC increases the potency of a replication-defective human adenovirus vectored foot-and-mouth disease vaccine. Virology. 2014;468:283–292.
  • Pollack IF, Jakacki RI, Butterfield LH, et al. Peptide vaccine therapy for childhood gliomas. Neurosurgery. 2013;60:113–119.
  • Dhodapkar MV, Sznol M, Zhao B, et al. Induction of antigen-specific immunity with a vaccine targeting NY-ESO-1 to the dendritic cell receptor DEC-205. Sci Transl Med. 2014;6.
  • Sabbatini P, Tsuji T, Ferran L, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res. 2012;18:6497–6508.
  • Morse MA, Chapman R, Powderly J, et al. Phase I study utilizing a novel antigen-presenting cell-targeted vaccine with toll-like receptor stimulation to induce immunity to self-antigens in cancer patients. Clin Cancer Res. 2011;17:4844–4853.
  • Wada H, Nagase H, Kurose K, et al. NY-ESO-1 protein cancer vaccine with poly-ICLC and OK-432. J Immunother. 2017;40:140–147.
  • Caskey M, Robert I, Heyer V, et al. Synthetic double-stranded RNA induces innate immune responses similar to a live viral vaccine in humans. J Exp Med. 2011;208:2357–2366.
  • van Rossenberg SMW, van Keulen A, Drijfhout J-W, et al. Stable polyplexes based on arginine-containing oligopeptides for in vivo gene delivery. Gene Ther. 2004;11:457–464.
  • Morris VB, Labhasetwar V. Arginine-rich polyplexes for gene delivery to neuronal cells. Biomaterials. 2015;60:151–160.
  • Putnam D, Zelikin AN, Izumrudov VA, et al. Polyhistidine-PEG:DNA nanocomposites for gene delivery. Biomaterials. 2003;24:4425–4433.
  • Lallana E, Rios de la Rosa JM, Tirella A, et al. Chitosan/hyaluronic acid nanoparticles: rational design revisited for RNA delivery. Mol Pharm. 2017;14:2422–2436.
  • Liang L, Zhang L, Peng G, et al. Molecular dynamics study on the mechanism of polynucleotide encapsulation by chitosan. Sci Rep. 2017;7:1–9.
  • Ahmed M, Narain R. The effect of molecular weight, compositions and lectin type on the properties of hyperbranched glycopolymers as non-viral gene delivery systems. Biomaterials. 2012;33:3990–4001.
  • Germershaus O, Mao S, Sitterberg J, et al. Gene delivery using chitosan, trimethyl chitosan or polyethylenglycol-graft-trimethyl chitosan block copolymers: establishment of structure–activity relationships in vitro. J Control Release. 2008;125:145–154.
  • Liu X, Howard KA, Dong M, et al. The influence of polymeric properties on chitosan/siRNA nanoparticle formulation and gene silencing. Biomaterials. 2007;28:1280–1288.
  • Nafee N, Taetz S, Schneider M, et al. Chitosan-coated PLGA nanoparticles for DNA/RNA delivery: effect of the formulation parameters on complexation and transfection of antisense oligonucleotides Nanomedicine Nanotechnology. Biol Med. 2007;3:173–183.
  • Taetz S, Nafee N, Beisner J, et al. The influence of chitosan content in cationic chitosan/PLGA nanoparticles on the delivery efficiency of antisense 2′-O-methyl-RNA directed against telomerase in lung cancer cells. Eur J Pharm Biopharm. 2009;72:358–369.
  • Mahiny AJ, Dewerth A, Mays LE, et al. In vivo genome editing using nuclease-encoding mRNA corrects SP-B deficiency. Nat Biotechnol. 2015;33:2–6.
  • Bousbaa H, Singh A, Bousbaa H, et al. Mad2 checkpoint gene silencing using epidermal growth factor receptor-targeted chitosan nanoparticles in non-small cell lung cancer model. Mol Pharm. 2014;11:3515–3527.
  • Nascimento AV, Ten Hoopen HW, Storm G, et al. Biodistribution and pharmacokinetics of Mad2 siRNA-loaded EGFR-targeted chitosan nanoparticles in cisplatin sensitive and resistant lung cancer models. Nanomedicine. 2016;11:767–781.
  • Corbet C, Ragelle H, Pourcelle V, et al. Delivery of siRNA targeting tumor metabolism using non-covalent PEGylated chitosan nanoparticles: identification of an optimal combination of ligand structure, linker and grafting method. J Control Release. 2016;223:53–63.
  • Gao Z, Huang W, Jin M, et al. Chitosan-based nanoparticles for survivin targeted siRNA delivery in breast tumor therapy and preventing its metastasis. Int J Nanomedicine. 2016;11:4931–4945.
  • Siahmansouri H, Somi MH, Babaloo Z, et al. Effects of HMGA2 siRNA and doxorubicin dual delivery by chitosan nanoparticles on cytotoxicity and gene expression of HT-29 colorectal cancer cell line. J Pharm Pharmacol. 2016;68:1119–1130.
  • Liu Y, Reineke TM. Degradation of poly(glycoamidoamine) DNA delivery vehicles: polyamide hydrolysis at physiological conditions promotes DNA release. Biomacromolecules. 2010;11:316–325.
  • McLendon PM, Fichter KM, Reineke TM. Poly(glycoamidoamine) vehicles promote pDNA uptake through multiple routes and efficient gene expression via caveolae-mediated endocytosis. Mol Pharm. 2010;7:738–750.
  • Tranter M, Liu Y, He S, et al. In vivo delivery of nucleic acids via glycopolymer vehicles affords therapeutic infarct size reduction in vivo. Mol Ther. 2012;20:601–608.
  • Dong Y, Dorkin JR, Wang W, et al. Poly(glycoamidoamine) brushes formulated nanomaterials for systemic siRNA and mRNA delivery in vivo. Nano Lett. 2016;16:842–848.
  • Ahmed M, Jawanda M, Ishihara K, et al. Impact of the nature, size and chain topologies of carbohydrate-phosphorylcholine polymeric gene delivery systems. Biomaterials. 2012;33:7858–7870.
  • Xue L, Ingle NP, Reineke TM. Highlighting the role of polymer length, carbohydrate size, and nucleic acid type in potency of glycopolycation agents for pDNA and siRNA delivery. Biomacromolecules. 2013;14:3903–3915.
  • Diaz-Dussan D, Nakagawa Y, Peng -Y-Y, et al. Effective and specific gene silencing of epidermal growth factor receptors mediated by conjugated oxaborole and galactose-based polymers. ACS Macro Lett. 2017;6:768–774.
  • Singhsa P, Diaz-Dussan D, Manuspiya H, et al. Well-defined cationic N-[3-(Dimethylamino)propyl]methacrylamide hydrochloride-based (Co)polymers for siRNA delivery. Biomacromolecules. 2018;19:209–221.
  • Luo X, Wang W, Dorkin JR, et al. Poly(glycoamidoamine) brush nanomaterials for systemic siRNA delivery in vivo. Biomater Sci. 2017;5:38–40.
  • Mathew AP, Cho KH, Uthaman S, et al. Stimuli-regulated smart polymeric systems for gene therapy. Polymers. 2017;9:152.
  • Schaffer DV, Fidelman NA, Dan N, et al. Vector unpacking as a potential barrier for receptor-mediated polyplex gene delivery. Biotechnol Bioeng. 2000;67:598–606.
  • Estrela JM, Ortega A, Obrador E. Glutathione in cancer biology and therapy. Crit Rev Clin Lab Sci. 2006;43:143–181.
  • Gamcsik MP, Kasibhatla MS, Teeter SD, et al. Glutathione levels in human tumors. Biomarkers. 2012;17:671–691.
  • Ping Y, Li Y, Tian C, et al. Redox-responsive hyperbranched poly(amido amine)s with tertiary amino cores for gene delivery. Biomacromolecules. 2013;14:2083–2094.
  • van der Aa LJ, Vader P, Storm G, et al. Optimization of poly(amido amine)s as vectors for siRNA delivery. J Control Release. 2011;150:177–186.
  • Nam JP, Kim S, Kim SW. Design of PEI-conjugated bio-reducible polymer for efficient gene delivery. Int J Pharm. 2018;545:295–305.
  • Breunig M, Hozsa C, Lungwitz U, et al. Mechanistic investigation of poly(ethylene imine)-based siRNA delivery: disulfide bonds boost intracellular release of the cargo. J Control Release. 2008;130:57–63.
  • Zhu C, Zheng M, Meng F, et al. Reversibly Shielded DNA polyplexes based on bioreducible PDMAEMA-SS-PEG-SS-PDMAEMA triblock copolymers mediate markedly enhanced nonviral gene transfection. Biomacromolecules. 2012;13:769–778.
  • Kim Y, Uthaman S, Nurunnabi M, et al. Synthesis and characterization of bioreducible cationic biarm polymer for efficient gene delivery. Int J Biol Macromol. 2018;110:366–374.
  • Matsumoto S, Christie RJ, Nishiyama N, et al. Environment-responsive block copolymer micelles with a disulfide cross-linked core for enhanced siRNA delivery. Biomacromolecules. 2009;10:119–127.
  • Kwon YJ. Before and after endosomal escape: roles of stimuli-converting siRNA/polymer interactions in determining gene silencing efficiency. Acc Chem Res. 2012;45:1077–1088.
  • Yang XC, Chai MY, Zhu Y, et al. Facilitation of gene transfection with well-defined degradable comb-shaped Poly(glycidyl methacrylate) derivative vectors. Bioconjug Chem. 2012;23:618–626.
  • Nie Y, Günther M, Gu Z, et al. Pyridylhydrazone-based PEGylation for pH-reversible lipopolyplex shielding. Biomaterials. 2011;32:858–869.
  • Manganiello MJ, Cheng C, Convertine AJ, et al. Diblock copolymers with tunable pH transitions for gene delivery. Biomaterials. 2012;33:2301–2309.
  • Convertine AJ, Diab C, Prieve M, et al. pH-responsive polymeric micelle carriers for siRNA drugs. Biomacromolecules. 2010;11:2904–2911.
  • Convertine AJ, Benoit DSW, Duvall CL, et al. Development of a novel endosomolytic diblock copolymer for siRNA delivery. J Control Release. 2009;133:221–229.
  • Convertine AJ, Benoit DSWW, Duvall CL, et al. Development of a novel endosomolytic diblock copolymer for siRNA delivery. J Control Release. 2009;133:221–229.
  • Cheng Y, Yumul RC, Pun SH. Virus-inspired polymer for efficient in vitro and in vivo gene delivery. Angew Chemie Int Ed. 2016;55:12013–12017.
  • McKinlay CJ, Vargas JR, Blake TR, et al. Charge-altering releasable transporters (CARTs) for the delivery and release of mRNA in living animals. Proc Natl Acad Sci. 2017;114:E448–E456.
  • Haabeth OAW, Blake TR, McKinlay CJ, et al. mRNA vaccination with charge-altering releasable transporters elicits human T cell responses and cures established tumors in mice. Proc Natl Acad Sci. 2018;115:E9153–E9161.
  • Benner NL, Near KE, Bachmann MH, et al. Functional dna delivery enabled by lipid-modified charge-altering releasable transporters (CARTs). Biomacromolecules. 2018;19:2812–2824.
  • Haabeth OAW, Blake TR, McKinlay CJ, et al. local delivery of Ox40l, Cd80, and Cd86 mRNA kindles global anticancer immunity. Cancer Res. 2019;79:1624–1634.
  • McKinlay CJ, Benner NL, Haabeth OA, et al. Enhanced mRNA delivery into lymphocytes enabled by lipid-varied libraries of charge-altering releasable transporters. Proc Natl Acad Sci. 2018;115:E5859–E5866.
  • Benner NL, McClellan RL, Turlington CR, et al. Oligo(serine ester) charge-altering releasable transporters: organocatalytic ring-opening polymerization and their use for in vitro and in vivo mrna delivery. J Am Chem Soc. 2019;141:8416–8421.
  • Gribble FM, Loussouarn G, Tucker SJ, et al. A novel method for measurement of submembrane ATP concentration. J Biol Chem. 2000;275:30046–30049.
  • Cambre JN, Sumerlin BS. Biomedical applications of boronic acid polymers. Polymer (Guildf). 2011;52:4631–4643.
  • Matsumoto A, Yoshida R, Kataoka K. Glucose-responsive polymer gel bearing phenylborate derivative as a glucose-sensing moiety operating at the physiological pH. Biomacromolecules. 2004;5:1038–1045.
  • Naito M, Ishii T, Matsumoto A, et al. A phenylboronate-functionalized polyion complex micelle for ATP-triggered release of siRNA. Angew Chem Int Educ. 2012;51:10751–10755.
  • Naito M, Yoshinaga N, Ishii T, et al. enhanced intracellular delivery of sirna by controlling ATP-responsivity of phenylboronic acid-functionalized polyion complex micelles. Macromol Biosci. 2018;18:1700357.
  • Kim J, Lee YM, Kim H, et al. Phenylboronic acid-sugar grafted polymer architecture as a dual stimuli-responsive gene carrier for targeted anti-angiogenic tumor therapy. Biomaterials. 2016;75:102–111.
  • Yoshinaga N, Ishii T, Naito M, et al. polyplex micelles with phenylboronate/gluconamide cross-linking in the core exerting promoted gene transfection through spatiotemporal responsivity to intracellular pH and ATP concentration. J Am Chem Soc. 2017;139:18567–18575.
  • Kim Y, Binauld S, Stenzel MH. zwitterionic guanidine-based oligomers mimicking cell-penetrating peptides as a nontoxic alternative to cationic polymers to enhance the cellular uptake of micelles. Biomacromolecules. 2012;13:3418–3426.
  • You J-O, Almeda D, Ye GJ, et al. Bioresponsive matrices in drug delivery. J Biol Eng. 2010;4:15.
  • Choi JL, Tan J-KY, Sellers DL, et al. Guanidinylated block copolymers for gene transfer: A comparison with amine-based materials for in vitro and in vivo gene transfer efficiency. Biomaterials. 2015;54:87–96.
  • Xu X, Wu J, Liu Y, et al. Ultra-pH-responsive and tumor-penetrating nanoplatform for targeted siRNA delivery with robust anti-cancer efficacy. Angew Chem Int Educ. 2016;55:7091–7094.
  • Zhou J, Wu Y, Wang C, et al. pH-sensitive nanomicelles for high-efficiency sirna delivery in vitro and in vivo: an insight into the design of polycations with robust cytosolic release. Nano Lett. 2016;16:6916–6923.
  • Elzoghby AO, Samy WM, Elgindy NA. Protein-based nanocarriers as promising drug and gene delivery systems. J Control Release. 2012;161:38–49.
  • Monfort DA, Koria P. Recombinant elastin-based nanoparticles for targeted gene therapy. Gene Ther. 2017;24:610–620.
  • Elangovan S, Khorsand B, Do A-V, et al. Chemically modified RNA activated matrices enhance bone regeneration. J Control Release. 2015;218:22–28.
  • Badieyan ZS, Berezhanskyy T, Utzinger M, et al. Transcript-activated collagen matrix as sustained mRNA delivery system for bone regeneration. J Control Release. 2016;239:137–148.
  • Chen THH, Bae Y, Furgeson DY, et al. Biodegradable hybrid recombinant block copolymers for non-viral gene transfection. Int J Pharm. 2012;427:105–112.
  • Elsner MB, Herold HM, Müller-Herrmann S, et al. Enhanced cellular uptake of engineered spider silk particles. Biomater Sci. 2015;3:543–551.
  • Kozlowska AK, Florczak A, Smialek M, et al. Functionalized bioengineered spider silk spheres improve nuclease resistance and activity of oligonucleotide therapeutics providing a strategy for cancer treatment. Acta Biomater. 2017;59:221–233.
  • Urello MA, Kiick KL, Sullivan MO. A CMP-based method for tunable, cell-mediated gene delivery from collagen scaffolds. J Mater Chem B. 2014;2:8174–8185.
  • Urello MA, Kiick KL, Sullivan MO. Integration of growth factor gene delivery with collagen-triggered wound repair cascades using collagen-mimetic peptides. Bioeng Transl Med. 2016;1:207–219.
  • Luo T, Kiick KL. Collagen-like peptide bioconjugates. Bioconjug Chem. 2017;28:816–827.
  • Numata K, Kaplan DL. Silk-based gene carriers with cell membrane destabilizing peptides. Biomacromolecules. 2010;11:3189–3195.
  • Numata K, Mieszawska-Czajkowska AJ, Kvenvold LA, et al. Silk-based nanocomplexes with tumor-homing peptides for tumor-specific gene delivery. Macromol Biosci. 2012;12:75–82.
  • Numata K, Reagan MR, Goldstein RH, et al. Spider silk-based gene carriers for tumor cell-specific delivery. Bioconjug Chem. 2011;22:1605–1610.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.