Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jun 23.
Published in final edited form as: Adv Drug Deliv Rev. 2020 Jun 23;156:119–132. doi: 10.1016/j.addr.2020.06.014

Polymeric Vehicles for Nucleic Acid Delivery

Alexandra S Piotrowski-Daspit 1, Amy C Kauffman 1,2, Laura G Bracaglia 1, W Mark Saltzman 1,3,4,5,*
PMCID: PMC7736472  NIHMSID: NIHMS1609335  PMID: 32585159

Abstract

Polymeric vehicles are versatile tools for therapeutic gene delivery. Many polymers—when assembled with nucleic acids into nanoparticles or polyplexes—can protect the cargo from degradation and clearance in vivo, and facilitate its transport into intracellular compartments. Design options in polymer synthesis yield a comprehensive range of molecules and resulting vehicle formulations. These properties can be manipulated to achieve stronger association with nucleic acid cargo and cells, improved endosomal escape, or sustained delivery depending on the application. Here, we describe current approaches for polymer use and related strategies for gene delivery in preclinical and clinical applications. Polymer vehicles delivering genetic material have already achieved significant therapeutic endpoints in vitro and in animal models. From our perspective, with preclincal assays that better mimic the in vivo environment, improved strategies for target specificity, and scalable techniques for polymer synthesis, the impact of this therapeutic approach will continue to expand.

Keywords: gene delivery, gene editing, polymeric vehicle, nanocarriers, polyplex, nanoparticle, biodegradable, biocompatible, sustained release, in vivo

Graphical abstract

graphic file with name nihms-1609335-f0001.jpg

1. Introduction

1.1. Introduction to nucleic acid delivery

Nucleic acids are powerful tools to exogenously regulate cellular processes that can have an impact on the whole organism. Strategic delivery of these molecules can make changes to the expression of genes in an organism in either permanent or transient ways. For therapeutic purposes, nucleic acids can be used to correct a disease-related inherited gene by adding, removing, or replacing genetic material, or to make cells and tissues behave in a certain way by activating, suppressing, or supplementing gene expression [1, 2]. Applications of this therapeutic strategy are growing as our understanding of human gene regulation and the links between genetic defects and the molecular basis of disease expands.

Nucleic acid-based therapeutics must act intracellularly, but most nucleic acids are rapidly cleared and degraded after systemic administration and do not readily cross the plasma membrane. Therefore, vectors that facilitate their transport inside cells can enhance the efficacy of these therapeutic strategies [3]. Successful intracellular delivery of nucleic acids has been achieved using a wide variety of tools and techniques, including physical methods, viral vectors, and non-viral vehicles [4]. Within the category of non-viral vehicles are polymeric delivery systems (Figure 1), which facilitate the protected transport of nucleic acids through extracellular space and through the plasma membrane. There are several types of delivery vehicles that can be formulated, including polyplexes, nanoconjugates, micelles, nanocapsules, dendrimers, and nanoparticles (NPs). Polyplexes are common formulations used for the delivery of nucleic acids, particularly with cationic polymers. These vehicles are spontaneously formed by entropically driven electrostatic interactions between positively charged polymers, often containing ionizable amine (N) groups, and negatively charged nucleic acids containing phosphate (P) groups. For gene delivery purposes, an excess of polymer is typically used (N/P ratio >1) to condense nucleic acid cargo, generating vehicles with a positive surface charge [5].

Figure 1. Polymeric vehicles for nucleic acid delivery: cargo and formulations.

Figure 1.

Polymeric nucleic acid delivery vehicle formulation menu including example choices of cargo, polymers, modifications, and end vehicle design. Vehicle designs include polyplexes – short-lifetime electrostatic complexes that require an excess of polymer to nucleic acids to be formed, nanoconjugates - very short-lifetime linear polycations with limited structure and random organization, micelles - MW-dependent lifetime core-shell complex composed of dynamic amphiphilic polymers, nanocapsules - natural polymer shell complex that encapsulates cargo, dendrimer - branched polymer complex with dendritic encapsulation of nucleic acids that can have peripheral functionalized for improve delivery, and nanoparticles - solid particles with homogeneous loading of nucleic acids that require degradation to assist cargo release.

There are various design criteria that must be met to ensure that polymeric vehicles reach their final destination and deliver their cargo effectively. These criteria depend on target cell populations, the duration over which the genetic material needs to be present/expressed, and the properties (ex. size/molecular weight, sensitivity to degradation, etc.) of the nucleic acid material to be delivered. The major advantages of this class of vehicles stem from versatile polymer chemistries and physical characteristics which impart an element of control to the delivery strategy; polymers can be manipulated to provide controlled release of cargo, support transfection, and affect biodistribution. This review provides an overview of polymer materials used to deliver nucleic acids along with the current status of clinical trials. We also outline the major challenges faced by polymeric vehicles for gene delivery as well as strategies to overcome them.

1.2. Types of nucleic acid cargo

Therapeutic nucleic acids encompass a wide range of sizes, stabilities, as well as various modes and intracellular sites of activity. The diversity of these molecules is important to consider when designing delivery strategies (Figure 1), as their properties will affect encapsulation efficiency as well as delivery vehicle characteristics.

There are several varieties of therapeutic RNA molecules, most of which are small and intended to act in the cytosol without entry into the nucleus. Since they are not integrated into the genome and are eventually degraded, their activity is temporary. They are often chemically modified in order to improve their stability and bioavailability [6]. Such modifications can include 2’ hydroxyl modifications that introduce fluorine or methoxy groups or replacement of the phosphodiester backbone to phosphorothioates, phosphorodiamidates, or polyamides to reduce susceptibility to nucleases [7].

The smallest RNA molecules are small interfering RNAs (siRNA) and microRNAs (miRNAs), which are both about 20 base pairs (bp) in length. siRNAs are short duplex, non-coding RNA molecules ~20 bp in length. They act to silence specific genes after they are expressed through the RNA interference (RNAi) pathway. Specifically, siRNA molecules silence a single targeted messenger RNA (mRNA) sequence [8]. Similarly, miRNAs are non-coding RNAs that regulate translation, but act through partial recognition of mRNA targets. Transfer RNAs (tRNAs, ~70–90 nucleotides) interact with ribosomes to incorporate a desired amino acid based on the native mRNA sequence. The largest RNA molecules are single-stranded mRNAs, which direct the synthesis of an encoded protein in the cytoplasm. The size of these molecules is dependent on the length of the gene encoded, which can be up to thousands of nucleotides.

With sizes in the kilobase range, the largest therapeutic nucleic acids by far are DNA molecules. The most extensively studied form of DNA is plasmid DNA (pDNA). pDNAs are circular double-stranded molecules that are capable of replicating autonomously in a cellular host. They consist of an anionic phosphodiester backbone and a stable deoxyribose structure. These large nucleic acids are capable of strong electrostatic interactions and condensation, particularly with cationic carriers [9]. As with all DNA molecules, successful transfection of pDNA requires transport to the nucleus to be effective. These effects can be long-lasting, especially if components of the plasmid are integrated into the host genome.

Therapeutic nucleic acids also include synthetic DNA or RNA analogs such as peptide nucleic acids (PNAs). PNAs have a charge-neutral peptide-like polyamide backbone with nucleobases capable of hybridization with DNA or RNA with high affinity. They are resistant to degradation by nucleases and have increased serum stability compared to other nucleic acids [10], but are also hydrophobic and not easily taken up by cells. However, these properties can be advantageous for loading into hydrophobic polymer NPs [11]. PNA monomers are composed of N-(2-aminoethyl)-glycine units with nucleobases attached via methylene carbonyl linkages [12]. Therapeutically, PNAs can be utilized in multiple ways to manipulate gene expression, including antisense, anti-miR, and gene editing [11]. These nucleic acids range in size, with larger therapeutic molecules up to ~40 nucleotides in length. As PNAs have a different backbone compared to their natural nucleic acid counterparts, they cannot be incorporated into the host genome.

Gene editing agents can also be delivered in polymeric vehicles for genetic manipulation. In addition to synthetic PNAs mentioned above, there are several families of nuclease-based gene editing agents including meganucleases, zinc finger nucleases (ZFNs), TALE nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated proteins (Cas). Each of these nucleases can be programmed to introduce single or double-strand breaks in the genome [13, 14]. The CRISPR/Cas9 system has been the most widely used gene editing approach since its discovery in 2013, in part due to the relative ease of design for various applications [15]. CRISPR/Cas9 gene editing platforms use a single guide RNA (sgRNA) to direct the bacterial Cas9 enzyme to a target locus in the genome [16, 17]. In nucleic acid form, Cas9 can be delivered as a plasmid (~8–10 kilobases) or mRNA (~4000 nucleotides) alongside a sgRNA (~100 nucleotides) which can also be delivered in plasmid form; a donor DNA molecule (up to ~100 nucleotides) is also delivered to serve as a template if a site specific genome modification is desired [18]. Protein and mRNA forms of Cas9 are often preferred as they limit the duration nuclease activity, reducing the potential for off-target modification of the genome. Incorporating these gene editing components together results in large and complex nucleic acid payloads within any delivery vehicle.

1.3. Extracellular and intracellular barriers to nucleic acid delivery in vivo

Upon in vivo administration, polymeric delivery vehicles encounter a complex set of extracellular and intracellular barriers which remain a key challenge in clinical translation (Figure 2). For each application, accessibility of target tissues and cell types needs to be carefully considered when choosing a route of administration. If administered intravenously, delivery vehicles can interact with molecules present in the blood, such as serum proteins which can accumulate on carrier surfaces. The formation of a “protein corona” around polymeric carriers can change the surface properties and hinder the effectiveness of the delivery vehicle by altering the carrier’s interactions with cells and tissues [19, 20]. Most intravenously injected carriers, particularly those that are larger than 100 nm, rapidly accumulate in the liver due to phagocytosis by Kupffer cells. Avoiding this rapid liver accumulation is essential to achieve the long carrier circulation times that are needed to increase accumulation in other tissues. The ability of non-viral vectors to evade liver clearance and accumulate in target tissues depends on their size and surface properties. Small vectors (<100 nm) and vectors with certain coatings that confer stealth properties by reducing protein corona formation remain in circulation longer and may be better able to accumulate in organs other than the liver [21]. Of course, the carrier must be large enough (>10 nm) to avoid efficient filtration by the kidney. Aside from elimination from the blood, once the polymer vehicle reaches the organ of interest, there may be additional physical barriers that prevent access to certain cell types including vascular impermeability and hydrostatic pressure preventing extravasation, poor local distribution, as well difficultly traversing through local mucus or matrix barriers. If the target tissue of interest is accessible by local administration, such as direct injection into the eye, many of the challenges associated with systemic delivery can be avoided.

Figure 2. Extracellular and intracellular barriers to nucleic acid delivery in vivo.

Figure 2.

A schematic illustrating physiological barriers relevant to polymeric nucleic acid carriers at the organism level, organ/tissue level, and cell level. Systemically administered vehicles must adequately circulate in the blood, accumulate in and penetrate target tissues, be internalized by target cells, and achieve intracellular cargo release.

Once delivery vehicles reach their target cells, they face additional barriers [22]. The first challenge is cellular association and entry, which can occur in a non-targeted or targeted manner via specific receptors on the cell surface. Cell association and uptake of delivery vehicles will depend on the interactions between the delivery vehicle and cell membrane, and generally occurs by endocytosis [23]. Endosomal escape of the carrier is essential to avoid lysosomal degradation of the cargo [24]. Delivery vehicles must facilitate transport of cargo to the cytosol or the nucleus for the genetic manipulation to take place, after which remnants of the delivery vehicle must be eliminated without disrupting normal cellular processes.

In sum, to be successful as nucleic acid carriers, polymeric vehicles must perform multiple functions. They must efficiently load and protect nucleic acid cargo, evade the immune system and premature clearance mechanisms, achieve cellular uptake and endosomal escape, and disappear without toxicity. Designing vehicles to fulfill all of these criteria has been challenging, but recent approaches aim to modify or combine traditional polymers into materials that both capitalize on proven strengths and balance pitfalls to create well-rounded vehicles for various applications.

2. Polymeric vehicles for nucleic acid delivery

Polymer physicochemical properties (ex. composition, molecular weight, and polydispersity) can be modified to achieve specialized formulations for nucleic acid delivery. One common feature in most polymers designed for nucleic acid delivery is the incorporation of cationic groups, with two purposes: first, to aid with the loading of negatively charged nucleic acid cargo, and second, to faciliate the interaction with negatively charged glycoproteins on the cell membrane [25]. Intended applications may dictate stability, size, and cargo requirements, which can be achieved with various formulations. Some commonly used formulations are illustrated in Figure 1. Both naturally derived and synthetic polymers have been utilized for nonviral delivery systems, although an advantage of synthetic polymers is the ability to incorporate versatile chemistries in a controlled manner, providing more options for the final formulation.

2.1. Early linear polycations

One of the first linear polycations investigated for nucleic acid delivery was diethylaminomethyl (DEAE) dextran. This polycationic derivative of the carbohydrate dextran was shown to enhance poliovirus RNA transfection in 1965 and simian virus 40 viral DNA three years later [26, 27]. The advantages of DEAE-dextran include its chemical simplicity, reproducibility, and low cost; disadvantages consisting of low transfection efficiency, cytotoxicity in primary cells, and inhibition of cell growth in vitro have limited its use in vivo [28].

Polylysines and polyamines have also been used as gene carriers. Polyamines and nucleic acids spontaneously condense into compact structures that closely resemble viruses [29, 30]. Polylysines were the first polymers used in targeted gene transfection strategies by incorporating specific ligands for cell surface receptors [31, 32]. Vehicles formulated from these polycations can be toxic (with low molecular weight polymers tending to be more toxic), and endosomal escape can be inefficient due to complete protonation at neutral pH, which limits their endosomal buffering ability. To overcome these limitations, copolymers which incorporate these low molecular weight polycations with other polymer blocks, such as polyesters, can be synthesized [33].

2.2. Proton sponges and amphiphilic polycations

Proton sponges and amphiphilic polymers are more efficient at endosomal escape than early linear polycations. Often, proton sponges are composed of multiple blocked linear and/or non-linear polycations that enable a larger buffering capacity and enhanced physicochemical properties (such as increased molecular weight for stability). These materials generally contain many proton-accepting groups including primary, secondary, and tertiary amines, and can achieve endosomal escape by causing an influx of chloride and water into the endosome upon increased protonation in an acidified environment. This influx leads to subsequent bursting of the endosome from the osmotic pressure and membrane destabilization [34]. One example is polyamidoamine (PAMAM), which was developed in the early 1990s and demonstrated effective pDNA transfer [35]. Dendrimers, like PAMAM, inherently contain hundreds of coupling sites with primary amine termini due to their multivalent structure, and as such have been extensively explored as drug and gene carriers [36]. Indeed, the abundance of charged coupling sites enables efficient nucleic acid loading of even large structures. On the other hand, the high cation density leads to non-specific cellular uptake and systemic toxicity in vivo [3740]. A strategy to improve specificity is to utilize the amine groups to chemically attach cell-specific ligands that enhance binding to delivery targets. Using this technique, PAMAM-based formulations have been used to deliver gene therapy to injured microglia [41] and tumor cells [42]; another similar dendrimer (polypropylenimine) was used to treat prostate cancer cells [43]. These strategies report no secondary effects to healthy tissues. Conjugation of poly(ethylene glycol) (PEG) to PAMAM also reduces toxicity and improves efficacy [44].

Another well-known proton sponge is polyethylenimine (PEI), which has become a “gold standard“ for gene delivery. The PEI polymer family consists of linear and branched polymers with varying molecular weights and structures, adding to their versatility. These materials have been used to deliver a wide variety of nucleic acids including pDNA, miRNA, and siRNA both in vitro and in vivo [4548]. While transfection efficiency is high, the mechanism of action for PEI and similar polymers remains unclear [34, 4953]. Beyond the osomotic effect of a proton sponge, it is possible that the higher charge density of PEI molecules results in direct interaction with the endosomal membrane and subsequent permeabilization. Another possible mechanism for endosomal lysis by PEI is thermomechanical disruption, driven by swelling of PEI vehicles to more than four times their original size with a temperature shift [54, 55]. While membrane disruptions are helpful for transfection, the high charge density of this polymer can cause extensive damage to cellular membranes, leading to apoptosis, necrosis, and inhibition of ATP synthesis. Additionally, polyplexes with rapid release of nucleic acid cargo may raise concerns over the stability of the vehicle for delivery. Strategies that address this “polyplex dilemma” are described in detail in a recent review by Wagner et al. [56]. Other limitations of PEI are that it is not degradable, it exhibits molecular weight-dependent cytotoxicity, and can stimulate a systemic immune response [5760]. To address some of these limitations, PEI has been used in combination with other, more biocompatible polymers. For example, blending or grafting PEI with PLGA produces stable, cationic carriers [61] that have been complexed simultaneously with large nucleic acids (pDNA) as well as peptides and have shown improved transfection over branched PEI alone [62, 63]. Along these lines, other groups have developed derivatives of PEI, by forming hybrids of PEI with other polymers or lipids [64]. Another way to reduce the cytotoxicity of polymers with high charge density is through rational architectural changes. For example, one hypothesis suggests that increased branch density of dendrimers reduces toxicity [65]. This was demonstrated in studies of linear branched and cyclic branched poly(2-dimethylaminoethyl methacrylate) (pDMAEMA) [66]. Alternatively, the high surface charge of PEI was shielded when heparin was conjugated to the surface, increasing hemocompatibility and transfection [67].

Some amphiphilic polymers, such as poly-alkyl-carboxylic acids and polyvinyl ethers, appear to enhance endosomal escape due to their hydrophobic domains. This process can be futher enhanced by incorporating redox-sensitive and pH-dependent chemical groups into the polymer. For example, pyridyl disulfide (PDSA) [68] is pH-sensitive and, under acidic conditions, can enhance endosomal membrane disruption due to increased hydrophobicity [6973]. Many amphiphilic polymers lack cationic groups and therefore exhibit reduced nucleic acid binding, though cationic groups can sometimes be incorporated into the polymer design. For example, cationic polyvinyl ethers contain amine groups and achieve both efficient nucleic acid loading and transfection activity [74, 75].

2.3. Biodegradable polymers

Many non-biodegradable polymers have no established way to leave cells and tissues after administration. The prolonged exposure to foreign materials and the potential accumulation of materials can lead to toxicity. Biocompatibility is enhanced with the use of biodegradable polymers as carriers, particularly if the polymer degrades to non-toxic and natural metabolites. Both naturally-derived and synthetic polymers can be biodegradable, making them potentially safer options for systemic gene delivery.

2.3.1. Natural polymers

Among naturally-derived polymers, structural proteins and polysaccharides, such as cationic collagen derivatives and chitosan, have been investigated as gene delivery vehicles. Cationic collagenous proteins have been used for nucleic acid delivery to articular cartilage and bone for regenerative medicine and metastatic tumor treatment [7681]. Chitosan, a linear cationic polysaccharide, is produced by the deactylation of chitin (poly-d-glucosamine). It is non-toxic, even at high concentrations [82, 83], and can be formulated into polyplexes for effective protection and transfection of small and large nucleic acid cargos [84, 85]. For example, chitosan has been recently used to deliver miR-124 to rat microglia ex vivo [86], osteopathic tumors [87], tumors in multiple myeloma [88], and for in vivo bone regeneration [89]. An advantage of chitosan compared to other cationic polymers with higher charge densities (e.g. PEI) is the ready dissociation between nucleic acid cargo and polymer upon internalization, leading to more efficient gene transfection [90]. However, incorporating chitosan and PEI into a combined vehicle can take advantage of the stronger nucleic acid assoication with PEI while reducing negative consequences from charge density [9195].

Cyclodextrins (CDs) are another family of naturally-derived carbohydrate-based polymers with favorable physiochemical properties for gene delivery. CDs in α-, β-, or γ- forms can be used either as stand-alone vectors or in combination with other cationic polymers (including PEI) [96104]. β-CD can form host-guest interactions with adamantane which has supported the formulation of β-CD, adamantane-PEG, and PEI polyplex blends [105]. These formulations acheived high transfection efficiency in the liver, metastatic tumors, and via oral administration [106109].

2.3.2. Synthetic polymers

Synthetic biodegradable polymers are generated by assembling low molecular weight monomers into polymers via bioreversible linkages such as sulfide or ester bonds. Examples of widely used biodegradable synthetic polymers are poly(lactic acid) (PLA), poly(glycolic acid) (PGA), and random or block copolymers such as poly(lactic-co-glycolic acid) (PLGA) [110]. These materials degrade slowly via bulk hydrolysis, a process which can facilitate sustained release of cargo. PLGA-based delivery systems have been used for a wide variety of therapeutic agents [111], including nucleic acids such as siRNA, miRNA, and PNAs [112114]. Importantly, PLGA has been approved by the FDA for certain delivery applications. Practically, these materials have been difficult to use for nucleic acid delivery, as formulations usually have low nucleic acid loading, reducing their effectiveness [115, 116]. Precomplexation of nucleic acids with spermidine improves loading of siRNA [112]. Alternatively, PLGA can be blended or copolymerized with cationic polymers such as PEI [63], poly(beta amino) esters (PBAEs) [117], poly-L-arginine [118], and chitosan [119] for enhanced gene transfection. Interestingly, a number of these studies capitalized on the chemical properties of both polymers to deliver both nucleic acids and peptides [63].

PBAEs are a family of degradable cationic polymers with the ability to effectively encapsulate nucleic acids, promote cellular uptake, and act as a proton sponge to achieve endosomal escape [120]. Like polylysine, PEI, and PAMAM, PBAEs contain amine groups which interact with negatively charged nucleic acids, but they also have hydrolyzable moieties which render them degradable. PBAEs are relatively easy to synthesize compared to other cationic degradable polyesters and they are amendable to custom modifications [121], enabling the generation of large libraries of polymers, which can be screened to identify structures that are optimized in different applications [122, 123]. Strategic modifications to the polymer structure have been reported to optimize delivery to pediatric brain cancer cells [124], hepatocellular carcinoma [125], and retinal pigment epithelial cells [126]. While PBAEs are biodegradable and exhibit high transfection efficiencies, the high density of amine groups in these polymers makes them susceptible to aggregation with anionic serum molecules and increases toxicity [127, 128]. In blended formulations of PBAE and PLGA, cytotoxicity was reduced, which decreased as the ratio of PBAE to PLGA decreased [129].

Poly(amine-co-esters) (PACEs) are another class of biodegradable cationic polymers designed to have lower charge densities to reduce toxicity. These polymers are synthesized via copolymerization of hydrophobic monomers and amine-containing monomers that promote nucleic acid association and endosomal escape [130, 131]. PACE synthesis is often accomplished with an enzyme (lipase) as the catalyst, making the polymer easy to purify and free from potentially toxic metal catalysts used to synthesize many other degradable polymers. The physical properties of PACE can be controlled by composition and molecular weight, and these materials can be formulated into various delivery vehicles including polyplexes and solid NPs [132]. The lower charge density of PACE, compared to PEI and PBAE, improves biocompatibility, while maintaining enough charge to promote transfection. Like other cationic polymers, PACE polyplexes have the tendency to aggregate in serum, although this can be mediated through the use of coatings with synthetic peptides, apolipids, or surface modifications [132134].

Various synthetic strategies now exist to control polymerization reactions and, therefore, the properties of the resulting polymer. Until recently, the evolution of sequence-defined synthetic polymers in the laboratory was limited to analogs of nucleic acids and polypeptides [135137]. With new knowledge and technical capabilities, sequence-defined synthetic polymers are possible. This synthesis approach provides high chemical precision and flexible design of the resulting polymer, and is therefore a powerful tool to systematically evaluate the effect of minor chemical changes on nucleic acid delivery. Examples include controlled free-radical polymerizations such as reversible addition-fragmentation chain transfer (RAFT) polymerization and atom transfer radical polymerization (ATRP) [138]. RAFT polymerization, in particular, can result in high end-group fidelity and has been used to synthesize amine-based polymers for nucleic acid delivery [139]. Another example is highly functionalized nucleic acid polymer (HFNAP) libraries which are the first in-lab evolution of sequence-defined synthetic polymers that are not limited to the constraints of polymerases or ribosomes [140]. A ligase-based polymerization method provides great flexibility for this polymer system to produce libraries of different side-chain compositions for parallel experimentation to gain structure-function relationships between the resulting polymers and the genetic code to find the best candidates for the application of interest. Sequence control can also be achieved with approaches that are similar to peptide synthesis: in sequence-defined cationic oligo(aminoamide) (OAA) polymers, for example, monomer and functional group placement in the polymer chain is controlled by using stepwise solid phase synthesis [141]. By generating libraries of polymers built with minor chemical changes, OAAs with different architectures and structural motifs were identified for improved stability as a cationic carrier for formation of siRNA [142], for Cas9/sgRNA lipo-OAA complexes [143] and for combinations of pDNA and siRNA [144]. While techniques to control polymer sequence and structure are time-consuming, they enable control over the properties of the polymer, including hydrophilicity/hydrophobicity, charge density, functional domains, and architecture.

2.4. Vehicle characteristics and surface modifications

Strategic modifications to polymer vehicles can manipulate their behavior in vivo. Chemical or physical changes to the vehicle profile can capitalize on natural transport phenomena (transport-based) or can include molecules to aid in specific binding (molecular binding-based). Perhaps most simply, the size of polymer vehicle can significantly alter transport capabilities in the blood. A recent study comparing PAMAM dendrimers with minor size differences (4.3nm vs 6.5nm) found profound changes to circulation time and enhanced brain accumulation with the larger polymers [145]; another study found that the size of PLGA NPs determined the fraction that were rapidly accumulating in the liver, and the fraction that were able to target the bone marrow and lung [146]. Similarly, reducing the size of PEI/pDNA complexes resulted in the evasion of normal clearance mechanisms for these polyplexes [147]. In addition to evading clearance, it has long been appreciated that the size of the carrier is an important consideration in capitalizing on enhanced permeability and retention (EPR) effect seen in tumors and other instances of leaky vasculature [148]. However, this strategy does not apply to many tissue types and can result in accumulation in off-target regions [149, 150].

Another tool to accomplish increased circulation time is through the incorporation of stealth coatings, such as PEG, as described above. Originating in the 1960s [151], PEGylation is a strategy to reduce aggregation, improve stability, reduce clearance, and increase systemic circulation time in vivo [152]. Hydrophilic PEG moieties sterically hinder interactions with the neighboring NPs or serum proteins and blood components by creating a hydrated cloud around the NP [153, 154]. Unfortunately, the use of PEG to enhance circulation time has other consequences. The steric hindrance and stealth properties can prevent intended cellular uptake and reduce transfection efficiency [155]. Further, there is still a fraction of serum proteins that can adhere to the vehicle even after PEGylation [154]. In addition, there have been reports of several adverse events including anti-PEG antibody production [156] and complement activation [157]. Despite these potential limitations, the addition of PEG to polymer vehicles remains a popular technique, and is included in many of in vivo applications [123, 158].

An alternative to PEGylation is the addition of other hydrophilic moieties, such as hyperbranched polyglycerol (HPG). HPG has a dendrimer-like structure which increases the availability of surface-accessible hydroxyl groups [159]. The structure of HPG can be beneficial for further chemical modifications. For example, the vicinal diols can be chemically modified to aldehyde groups, conferring bioadhesive properties [160]. Like PEG modifications, HPG also increases blood circulation time when added to NP surface, and in some cases may outperform PEG in extended circulation half-life and reduced liver accumulation [161]. Advantages of HPG are low toxicity and reduced immunogenicity in vivo [162]. Other molecules added to the surface can also affect biodistribution of injected NPs based on natural transport mechanisms. Different types of cholesterol, for example, can direct NPs and oligonucleotides to accumulate in either the liver exclusively (low density lipoprotein) or spread throughout the liver, gut, kidney, and steroidogenic organs (high density lipoprotein) [163, 164].

To enhance vehicle accumulation within a certain cell type or tissue, molecules can be incorporated into the vehicle that will increase the likelihood of vehicle binding. Although this strategy will not help the polymer vehicle “home” to a particular site, in many cases it can increase the concentration delivered to a target site if the vehicle encounters this target naturally [165]. Using this strategy to deliver nucleic acids to tumors, many recent reports describe the use of sugars, proteins or drugs attached to the surface of the vehicle that will bind to uniquely upregulated receptors on cells [42, 43, 85, 118, 166]. This effort is summarized in Figure 3, which illustrates in vivo gene delivery applications, which have primarily been targeted to tumors. Enhanced accumulation at the tumor site was observed when hyaluronic acid (which binds to the CD44 receptor) [42, 85, 118], lactoferrin [43], folate [167169], or epidermal growth factor [170] were used. Similarly, lactose, which binds to hepatocytes through asialoglycoprotein receptors, improved transfection efficiency of poly(lysine)-PEG vehicles in hepatocellular carcinomas [166]. More details on nanoparticle designs made to enhance delivery in the tumor microenvironment can be found in the recent review by Mukalel et al. [171]. Another study demonstrated enhanced pDNA delivery using the drug eprosartan to bind to upregulated ATR1 in myocardial ischemic regions [172].

Figure 3. Nucleic acid delivery in polymeric vehicles grouped by target organ.

Figure 3.

Heat maps in this figure demonstrate the relative number of publications that have described polymer delivery vehicles for targeted gene delivery per organ or tissue type. The rodent representation is compiled from approximately 100 studies between 2015 and 2020. The non-human primate is compiled from approximately 8 studies between 2005 and 2020, and the human representation is compiled from 11 clinical trials presented in Table 1. These studies were identified using key words such as “gene delivery”, “in vivo” and “polymer” in the Web of Science database. In order to be included, investigations had to report activity-based assays demonstrating effective gene delivery.

In addition to the molecules described above, a number of smaller peptides have also resulted in enhanced tumor accumulation. Certain peptides such as Arg-Gly-Asp (RGD) [173, 174] and RGERPPR [175] are frequently used. New peptides, discovered through phage display libraries, can enhance binding to specific, hard-to-access sites. Some examples of novel peptides identified in this way include the tartrate-resistant acid phosphatase (TRAP)-targeted peptide for bone regeneration [176] and an epithelial targeted peptide for lung delivery [177]. Although not yet applied to polymer vehicles, a recent study using peptides discovered with phage display increased nanoligand siRNA carriers in the brain from ~2% (random peptide) to 6% using the targeted peptide [178]. Traditional binding molecules such as antibodies and aptamers can be used as well for specific binding if highly expressed antigens are identified [71, 179182]. Furthermore, coating NPs with a complete cell membrane is a biomimetic strategy that can prolong circulation and can be used for immune cell targeting [183]. Ultimately, any of these strategies can be used to enhance the accumulation of polymer vehicles at a target site, which can increase the therapeutic efficacy from a single administered dose.

2.5. Clinical trials

Worldwide clinical trials for nucleic acid therapy encompass different approaches with approximately 70% using viral vectors and 30% using non-viral vectors. While there have not yet been any polymeric NPs for gene delivery approved by the FDA, there are several recent and ongoing clinical trials (Table 1). The development of safe, efficient, and controllable delivery vehicles remains a bottleneck to clinical translation, highlighting the need for further vehicle optimization.

Table 1.

Clinical trials evaluating polymeric vehicles to deliver therapeutic nucleic acids.

Product Name Delivery System Cargo Disease/Target Organ Administration Route Phase Status Identifier
CALAA-01 CD NP with PEG siRNA
(RRM2)
solid tumor systemic (IV) I terminated NCT00689065
BC-819/PEI PEI NP pDNA
(diphtheria toxin A)
superficial bladder cancer local II completed NCT00595088
BC-819/PEI PEI NP pDNA
(diphtheria toxin A)
superficial bladder cancer local I completed NCT01878188
BC-819 PEI NP pDNA
(diphtheria toxin A)
ovarian cancer systemic (IP) I/II completed NCT00826150
BC-819 PEI NP pDNA
(diphtheria toxin A)
pancreatic cancer local II terminated NCT01413087
BC-819 PEI NP pDNA
(diphtheria toxin A)
non-muscle invasive bladder cancer local II active NCT03719300
DTA-H19 PEI NP pDNA
(diphtheria toxin A)
superficial bladder cancer local I/II completed NCT00393809
DTA-H19 PEI NP pDNA
(diphtheria toxin A)
Pancreatic neoplasms local I/II completed NCT00711997
CYL-02 JetPEI pDNA
(SSTR2 + DCK::UMK)
pancreatic cancer local I completed NCT01274455
CYL-02 JetPEI pDNA
(SSTR2 + DCK::UMK)
pancreatic cancer local II recruiting NCT02806687
MK 4261/JetPEI JetPEI RNA
(DDX58 activator)
solid tumor local I active NCT03739138
BO-112 JetPEI dsRNA
(TLR-3, MDA5, RIG-I agonist)
solid tumor local I active NCT02828098
EGEN-001 PEG-PEI-
cholesterol
pDNA
(IL-12)
ovarian cancer systemic (IP) I terminated NCT00137865
EGEN-001 PEG-PEI-
cholesterol
pDNA
(IL-12)
ovarian cancer systemic (IP) I completed NCT00473954
EGEN-001 PEG-PEI-
cholesterol
pDNA
(IL-12)
fallopian tube/ peritoneal/ovarian cancer systemic (IP) II completed NCT01118052
EGEN-001 PEG-PEI-
cholesterol
pDNA
(IL-12)
fallopian tube/ peritoneal/ ovarian cancer systemic (IP) I completed NCT01489371
GEN-1 PEG-PEI-
cholesterol
pDNA
(IL-12)
fallopian tube/ peritoneal/ ovarian cancer systemic (IP) I active NCT02480374
siG12D LODER PLGA NP siRNA
(KRASG12D)
pancreatic cancer local I completed NCT01188785
siG12D LODER PLGA NPs siRNA
(KRASG12D)
pancreatic cancer local II recruiting NCT01676259
SNS01-T PEI carrier siRNA
(eIF5A)
pDNA
(eIF5AK50R)
myeloma/ lymphoma/ leukemia systemic (IV) I/II unknown NCT01435720

In 2010, Davis et al. reported on the first polymeric NP-based gene delivery system (CALAA-01) tested in a Phase I trial for solid cancers [184]. CALAA-01 is a NP system in which an siRNA against the M2 subunit of ribonucleotide reductase (RRM2) is encapsulated. The nucleic acid is loaded into a NP composed of a CD-based polymer and PEG with a human transferrin protein (TF) targeting ligand on the surface. Results from the clinical trial indicated that these NPs could deliver siRNA to melanoma cells after systemic administration and that an antiproliferative response was achieved. Though apparently successful in delivery of the siRNA to several types of cancer cells, this trial was terminated after Phase I when patients experienced dose-limiting toxicity. Preclinical studies indicated that primary toxicities were in the liver and kidneys as elevations in liver enzymes, creatinine, and blood urea nitrogen were observed [185]. However, in humans, most adverse events consisted of hypersensitivity and acute immune responses. Subsequently, several cationic PEI-based systems (ex. BC-819/PEI, BC-819, DTA-H19) were developed for clinical trials for the treatment of various cancer types after local administration [186]. However, the substantial cytotoxicity of PEI polymers at the preclinical level has limited their clinical application and consequently several modifications have been investigated. For example, glycosylated JetPEI was used in CYL-02 to deliver SSTR2 and DCK::UMK in pDNA form to inhibit tumor cell proliferation and increase chemosensitivity of tumor cells to gemcitabine, respectively [187]. In this case, treatment-related toxicities were mild. Further, EGEN-001, a PEG-PEI-cholesterol polymer-lipid hybrid vehicle was developed for the delivery of an IL-12-expressing plasmid for immunotherapy of epithelial ovarian cancer. This clinical trial progressed to Phase II but resulted in patients exhibiting adverse side effects, including fever, chills, nausea, vomiting, anemia, thrombocytopenia, and leukopenia [188]. Another trial using PLGA-based NPs, a polymer system that has been used safely in many FDA-approved drug delivery and medical device applications, delivering siRNA targeting the oncogene KRAS for pancreatic cancer treatment is currently recruiting for Phase II. In summary, polymeric vehicle-based gene therapy at the clinical level is still in its infancy, and the polymeric NPs that have progressed to clinical trials with published results have mostly failed to meet their rigorous endpoints. Further, the adverse events reported thus far, though mild in some cases, suggest that the therapeutics that have been investigated at the clinical level exhibit off-target activity. However, it is difficult to determine whether these adverse effects are due to the polymeric vehicles themselves, their nucleic acid cargo, or administered combination therapies where applicable. Nonetheless, it is clear that therapeutics involving polymeric carriers must still overcome challenges associated with safety and efficacy before translation to humans. In particular, advances that reduce cytotoxicity while increasing transfection efficiency and target specificity are needed. Strategies to improve vehicle design are outlined below. However, it is important to note that increasing the complexity of polymeric carriers increases the difficulty of maintaining the high quality standards needed for clinical trials.

3. Strategies to mitigate current challenges facing polymeric nucleic acid delivery

While innovation to produce novel polymeric materials and delivery vehicles continues at a rapid pace, significant challenges remain. The clinical viability of any nucleic acid delivery system requires proper dissemination without interfering with normal bodily functions, eliciting an immune response, or producing adverse effects in non-target organs [189]. We have discussed strategies and examples for balancing biocompatibility and transfection efficiency in our review of polymeric delivery systems above. In broad terms, new innovations that focus on in vivo efficacy and target specificity as well as translation to commercial production will lead to increased clinical usage of this class of therapeutics. Here, we describe these challenges and identify forward-looking strategies to address the issues.

3.1. In vivo efficacy and target specificity

Many nucleic acid delivery systems have been successful in vitro but are not effective in vivo. This discrepancy is likely due, at least in part, to the lack of physiologically relevant screening tools and difficulty controlling tissue tropism. It is well understood that traditional 2D monolayer culture systems, which are widely used for initial screens, are not a realistic representation of cell behavior in the human body. Fluid flow, 3D cell and tissue organization, and vehicle clearance are critical aspects of gene delivery that are not adequately addressed using conventional in vitro experimental designs [190]. Further, animal models of disease can be inadequate, costly, and not truly predicative of human results [191, 192]. As shown in Figure 3, a number of studies deliver genetic material to specific human cancer cell lines (hepatocellular, prostate, lung, breast) which are grown in rodents as tumors. The physical constraints of this type of delivery may not mimic organ-specific cellular organization and limit the predictive results of these experiments.

Strategy: improve preclinical screening models to more accurately predict in vivo activity

Advancements in technologies that create platforms to better mimic human disease and physiological barriers can enhance or replace typical in vitro and in vivo preclinical studies. 3D cell culture systems have emerged over the past few decades as physiologically relevant cellular environments. Using technologies such as 3D printing and microfluidic devices, cell culture environments can mimic the tortuous and complex tissue environments found in tumors, the liver, and other tissue types [193195]. In particular, micropatterning of cell culture devices allows researchers to incorporate sophisticated gradients and patterns of signaling molecules, growth factors, or environmental confinement [193, 194]. Evaluating polymeric vehicle efficiency in these systems can help to elucidate the effect that physical constraints have on vehicle stability and cell association.

In addition to replicating the physical environment, cells with genetically accurate human disease can be produced from human induced pluripotent stem cells (iPSCs) with the advent of CRISPR/Cas9 and other gene editing technologies. The ability to generate iPSCs from somatic cells provides tremendous promise for gene delivery screening as it is possible to produce many different cell types from patient cells [196]. Candidate therapies can be tested directly in cells modified to include defects responsible for hematological, neuronal, muscular, and other disorders [197]. iPSCs are also more physiologically relevant cells with which to conduct preclinical safety studies than immortalized cell lines.

Lastly, new assays are being developed to rapidly screen many gene delivery vehicles. In one example, Yonamine and colleagues modified an ELISA (enzyme-linked immunosorbent assay) assay into a high throughput screening (HTS) method to select NPs with high affinity to target proteins [198]. The screen identified polymeric NPs with distinctive functional group compositions that exhibited high affinity to either histone or fibrinogen which allowed for identification of synthetic polymers with affinity for the target. These types of assays are often used to screen only one aspect of the delivery process, but they can be a powerful addition to the more complex models described above. In vivo, other factors may be relevant and vehicle specific, such as time in circulation and biodistribution. For high-throughput in vivo screening, we recently described an accessible approach to measure the circulation half-life of fluorescently labeled agents after intravenous injection [199]. Further, Dahlman and colleagues have designed a way to directly compare the biodistribution of many formulations of nanoparticles at once by labeling the encapsulated DNA with unique barcodes [200]. Evaluating the activity of many polymer vehicles side-by-side in the intended setting can provide valuable insights to behavior in the physiological environment.

Strategy: strategic administration and new vehicle designs for improved target specificity

As described, using molecular targeting strategies for systemic delivery can enhance accumulation at a specific site, but often will still result in unwanted accumulation elsewhere [173, 201, 202] and cannot provide access to every organ [41]. One strategy to overcome this challenge is site-specific administration, either by using an implant in response to an injury that anchors polymer delivery vehicles at the site [203, 204] or through an injection into an accessible organ or tissue (e.g. eye [126], spinal cord [62], brain [124]). Similarly, localized genetic manipulation has been achieved by pre-treating cells with polymeric vehicles ex vivo, and then transplanting the treated cells back into the patient [63, 113]. This strategy takes advantage of sustained delivery available with some polymer vehicles to maintain the nucleic acid activity throughout cellular development or differentiation. Finally, in instances where organs or tissues are being transplanted out of the body, ex vivo normothermic organ perfusion (EVNP) could provide a period of privileged access to deliver therapeutics without off-target possibilities. EVNP is being investigated for many organ transplant practices, and both polymeric NPs and naked siRNA have been successfully delivered during this process [205, 206]. Ex vivo treatment or direct injections can be invasive and are not suited for all applications. Another strategy for systemic delivery is to temporarily blockade the reticuloendothelial system with decoy materials, allowing the critical cargo to circulate without liver interference [207209]. Recent work in the field of lipid NP-mediated gene delivery has also shown that modulating the internal charge of delivery vehicles can enable tuning of biodistribution and tissue-specific gene delivery [210].

Alternatively, stimuli responsive polymers offer a tool to constrain nucleic acid availability to only intended regions [211]. In this strategy, polymers react to environmental or applied stimuli, and a conformational or chemical change triggers the release of the nucleic acid cargo. Externally applied stimuli such as a magnetic field, an electric field, ultrasonic waves, or light can be spatially applied for treatment of tumors or organ specific pathologies [212214]. For example, near-infrared light was used to trigger PEI-derived NP decomposition-driven release of siRNA to tumors in vivo [215217]. Environmental factors that are unique to the cell, tissue, or pathology (inflammation, cancer, infection) can also be used, such as pH [218223], temperature [224226], and the presence of ATP (reviewed in [64]). Polymers incorporating disulfide bonds can be transformed into thiols via reduction in the presence of glutathione (GSH) or reactive oxygen species (ROS), triggering a release of nucleic acid cargo [227229]. ROS-sensitive polymers are particularly useful tools for cancer cells that exhibit higher levels of ROS radicals and GSH [230].

3.2. Translation to commercial production

Polymeric materials must be inexpensive to produce, easy to synthesize, facile to purify and characterized completely to accomplish commercial-scale production. Control over polymerization mechanisms and chain polydispersity are key factors that can lead to variable performance [231, 232]. Since polymers are generally not a specified molecular entity, their characterization for manufacturing can prove difficult with regard to achieving Good Manufacturing Practice (GMP) certification. In terms of vehicle production, the size, morphology, and functionality of polymeric carriers need to be reproducible and scalable. For example, on a research scale, formulation processes such as nanoprecipitation and emulsion evaporation begin with milligrams of a dissolved polymer and end with the formation of precipitates that require the removal of solvents. On an industrial scale, grams to kilograms of materials would be required, with accompanying changes to techniques which may change formulation properties [233]. Many formulations, especially polyplexes, are unstable and cannot be stored long term, and as such, stability and shelf-life need to be considered as well.

Strategy: improved synthesis, purification, and formulation methods

Precise and inexpensive synthesis of polymers is a challenge, especially due to the cost of reactor systems, materials (especially catalysts and initiators), and purification equipment. Enzymatic catalysts can provide a cost-effective and environmentally friendly solution for biodegradable polymers compared to conventional metal or salt catalysts [234]. For example, bacterial-derived lipase can perform essential synthesis steps while maintaining high selectivity, high efficiency, the ability to operate under mild conditions, and potential catalyst recyclability [235241]. In a non-aqueous environment, more efficient catalysis can occur due to the increased solubility of the substrates in solvents, a reduction in the number of side-reactions, and a shift in the reaction equilibrium to favor synthesis over hydrolysis [242, 243]. Enzymes can also be separated from the reaction mixture as they are often immobilized to an inert bead which can be collected via filtration [244, 245]. Lastly, their thermal stability can be enhanced, allowing for synthesis reactions to occur more efficiently at higher temperatures [246].

As cationic charge in polymers has continued to be essential for nucleic acid encapsulation and delivery, appropriate synthesis schemes incorporating specific amine and other end groups are crucial. For example, specified crosslinking with 1,1′-carbonyldiimidazole (CDI) is a useful technique to obtain user-defined end groups. Activation and cross-linking with CDI results in a variety of gene delivery polymers with precise end group chemistries for efficient delivery of nucleic acid cargos [247250]. Subtle differences in the structure and properties of polymeric materials used for gene delivery can lead to dramatic differences in transfection efficiency and CDI chemistry is an efficient way to develop a library of materials [133].

As described above, it is also desirable to have control over the entire polymer sequence and not just the end group composition. Multistep flow synthesis and iterative exponential growth (Flow-IEG) is a technique has been utilized for macromolecules such as PNAs and synthetic RNAs, and has been adapted for polymers with semi-automatic processes with molecular weight distribution control [251, 252]. The development of the next generation of polymeric materials could utilize an iterative approach to find the appropriate chain length, charge density, and end group chemistry to form the most efficient non-viral vectors in a scalable manner [251, 253257].

Alongside advanced apparatus systems and novel catalysts, new polymer purification equipment and techniques have been employed with the same scalability. Enzyme catalyst reactions often require solvents for appropriate viscosity for mixing during synthesis [239, 241, 243]. This could be seen as a disadvantage as purification requires the complete removal of the solvent, but it can be advantageous as removal of the polymer and separation of the catalyst can be completed in a series of wash steps that utilize solvent miscibility principles [132]. Other methods of advanced purification, including the use of supercritical fluids and innovative membrane filtration, have aided in large scale polymer syntheses [258262].

Scale-up of formulation methods requires added consideration of the fragility of nucleic acid cargo, where techniques such as membrane extrusion and supercritical fluid technology are not applicable. Nonetheless, a few methods have been developed in order to produce NPs at larger scales with desired characteristics, such as microfluidic mixing technologies for nanoprecipitation processes [263]. Such processes still require the removal of organic solvent, achievable with tangential fluid flow systems. However, these methods are not amenable to the production of surface functionalized or targeted polymeric carriers, which may require additional processing steps.

4. Conclusions

The future of nucleic acid-based therapies will depend on innovations in delivery systems. For polymeric vehicles, this will involve the development of materials that excel at nucleic acid association, are less toxic/immunogenic, and are efficient at nucleic acid delivery both in vitro and in vivo. It will also be essential to define the mechanisms by which these materials achieve delivery to maximize effectiveness and minimize unexpected side effects. Studies aimed at understanding the structure-function relationships of polymeric vectors and the mechanisms by which they interact with cells in physiological environments are likely to produce promising candidates. Determining the requirements for optimal timing and longevity of genetic manipulation for each application will also help to guide vehicle design. Given the rapid progress in the field, nanoscale polymeric delivery systems will likely continue to serve an important role in the clinical translation of gene therapies.

Acknowledgements

This work was supported by grants from the National Institutes of Health (UG3 HL147352 and R01 HL125892). ASP was supported by two NIH National Research Service Awards (NRSA): a training grant (T32 GM86287) and an individual postdoctoral fellowship (F32 HL142144). LGB and ACK were supported by an NIH NRSA training grant (T32 DK101019), and LGB was later supported by a second NIH NRSA training grant (T32 DK007276).

Abbreviations

ATRP

atom transfer radical polymerization

bp

base pairs

Cas

CRISPR-associated protein

CD

cyclodextrin

CDI

1,1′-carbonyldiimidazole

CRISPR

clustered regularly interspaced short palindromic repeats

DEAE

diethylaminomethyl

DMSO

dimethyl sulfoxide

ELISA

enzyme-linked immunosorbent assay

EPR

enhanced permeability and retention

EVNP

ex vivo normothermic organ perfusion

Flow-IEG

flow synthesis and iterative exponential growth

GMP

Good Manufacturing Practice

GSH

glutathione

HCA

high content analysis

HFNAP

highly functionalized nucleic aid polymer

HPG

hyperbranched polyglycerol

HTS

high-throughput screening

iPSC

induced pluripotent stem cell

mRNA

messenger RNA

miRNA

micro RNA

N

amine group

NIR

near-infrared

NP

nanoparticle

OAA

oligo(aminoamide)

P

phosphate group

PACE

poly(amine-co-ester)

PAMAM

polyamidoamine

PBAE

poly(beta amino) ester

pDMAEMA

poly(2-dimethylaminoethyl methacrylate

pDNA

plasmid DNA

PDSA

pyridyl disulfide

PEG

poly(ethylene glycol)

PEI

polyethylenimine

PGA

poly(glycolic acid)

PLA

poly(lactic acid)

PLGA

poly(lactic-co-glycolic acid)

PNA

peptide nucleic acid

RAFT

reversible addition-fragmentation chain transfer

RGD

Arg-Gly-Asp

RNAi

RNA interference

ROS

reactive oxygen species

RRM2

M2 subunit of ribonucleotide reductase

sgRNA

guide RNA

siRNA

small interfering RNA

TALEN

TALE nuclease

TF

human transferrin protein

TRAP

tartrate-resistant acid phosphatase

tRNA

transfer RNA

ZFN

zinc finger nuclease

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of Interest

Amy C. Kauffman was a Postdoctoral Fellow at Yale University and transitioned to an employee of Corning Life Sciences during the time this review was completed. During the time of writing this review, ASP and WMS were consultants to Trucode Gene Repair Inc.

References

  • [1].Dunbar CE, High KA, Joung JK, Kohn DB, Ozawa K, Sadelain M, Gene therapy comes of age, Science, 359 (2018). [DOI] [PubMed] [Google Scholar]
  • [2].Orkin SH, Reilly P, Paying for future success in gene therapy, Science, 352 (2016) 1059. [DOI] [PubMed] [Google Scholar]
  • [3].Zhao Y, Huang L, Lipid nanoparticles for gene delivery, Advances in genetics, 88 (2014) 13–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Luo D, Saltzman WM, Synthetic DNA delivery systems, Nature biotechnology, 18 (2000) 33–37. [DOI] [PubMed] [Google Scholar]
  • [5].Pack DW, Hoffman AS, Pun S, Stayton PS, Design and development of polymers for gene delivery, Nat Rev Drug Discov, 4 (2005) 581–593. [DOI] [PubMed] [Google Scholar]
  • [6].Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR, Anderson DG, Non-viral vectors for gene-based therapy, Nature Reviews Genetics, 15 (2014) 541. [DOI] [PubMed] [Google Scholar]
  • [7].Pun SH, Hoffman AS, B.8 - Nucleic Acid Delivery, in: Ratner BD, Hoffman AS, Schoen FJ, Lemons JE (Eds.) Biomaterials Science (Third Edition), Academic Press; 2013, pp. 1047–1054. [Google Scholar]
  • [8].Gavrilov K, Saltzman WM, Therapeutic siRNA: principles, challenges, and strategies, Yale J Biol Med, 85 (2012) 187–200. [PMC free article] [PubMed] [Google Scholar]
  • [9].Hardee CL, Arévalo-Soliz LM, Hornstein BD, Zechiedrich L, Advances in Non-Viral DNA Vectors for Gene Therapy, Genes (Basel), 8 (2017) 65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Uhlmann E, Peyman A, Breipohl G, Will DW, PNA: Synthetic Polyamide Nucleic Acids with Unusual Binding Properties, Angew Chem Int Ed Engl, 37 (1998) 2796–2823. [DOI] [PubMed] [Google Scholar]
  • [11].Quijano E, Bahal R, Ricciardi A, Saltzman WM, Glazer PM, Therapeutic Peptide Nucleic Acids: Principles, Limitations, and Opportunities, The Yale journal of biology and medicine, 90 (2017) 583–598. [PMC free article] [PubMed] [Google Scholar]
  • [12].Nielsen PE, Egholm M, Buchardt O, Peptide nucleic acid (PNA). A DNA mimic with a peptide backbone, Bioconjug Chem, 5 (1994) 3–7. [DOI] [PubMed] [Google Scholar]
  • [13].Fernandez A, Josa S, Montoliu L, A history of genome editing in mammals, Mamm Genome, 28 (2017) 237–246. [DOI] [PubMed] [Google Scholar]
  • [14].Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, Liu DR, Search-and-replace genome editing without double-strand breaks or donor DNA, Nature, 576 (2019) 149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Knott GJ, Doudna JA, CRISPR-Cas guides the future of genetic engineering, Science, 361 (2018) 866–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Wang M, Glass ZA, Xu Q, Non-viral delivery of genome-editing nucleases for gene therapy, Gene Therapy, 24 (2017) 144–150. [DOI] [PubMed] [Google Scholar]
  • [17].Liu C, Zhang L, Liu H, Cheng K, Delivery strategies of the CRISPR-Cas9 gene-editing system for therapeutic applications, Journal of Controlled Release, 266 (2017) 17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Piotrowski-Daspit AS, Glazer PM, Saltzman WM, Debugging the genetic code: Non-viral in vivo delivery of therapeutic genome editing technologies, Current Opinion in Biomedical Engineering, 7 (2018) 24–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Nguyen VH, Lee B-J, Protein corona: a new approach for nanomedicine design, International journal of nanomedicine, 12 (2017) 3137–3151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Bertrand N, Grenier P, Mahmoudi M, Lima EM, Appel EA, Dormont F, Lim J-M, Karnik R, Langer R, Farokhzad OC, Mechanistic understanding of in vivo protein corona formation on polymeric nanoparticles and impact on pharmacokinetics, Nature Communications, 8 (2017) 777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Hoshyar N, Gray S, Han H, Bao G, The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction, Nanomedicine, 11 (2016) 673–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Peeler DJ, Sellers DL, Pun SH, pH-Sensitive Polymers as Dynamic Mediators of Barriers to Nucleic Acid Delivery, Bioconjugate Chemistry, 30 (2019) 350–365. [DOI] [PubMed] [Google Scholar]
  • [23].Lonn P, Kacsinta AD, Cui XS, Hamil AS, Kaulich M, Gogoi K, Dowdy SF, Enhancing Endosomal Escape for Intracellular Delivery of Macromolecular Biologic Therapeutics, Sci Rep, 6 (2016) 32301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Selby LI, Cortez-Jugo CM, Such GK, Johnston APR, Nanoescapology: progress toward understanding the endosomal escape of polymeric nanoparticles, Wiley interdisciplinary reviews. Nanomedicine and nanobiotechnology, 9 (2017). [DOI] [PubMed] [Google Scholar]
  • [25].Ni R, Feng R, Chau Y, Synthetic Approaches for Nucleic Acid Delivery: Choosing the Right Carriers, Life (Basel), 9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Vaheri A, Pagano JS, Infectious poliovirus RNA: a sensitive method of assay, Virology, 27 (1965) 434–436. [DOI] [PubMed] [Google Scholar]
  • [27].McCutchan JH, Pagano JS, Enhancement of the Infectivity of Simian Virus 40 Deoxyribonucleic Acid With Diethylaminoethyl-Dextran2, JNCI: Journal of the National Cancer Institute, 41 (1968) 351–357. [PubMed] [Google Scholar]
  • [28].Gulick T, Transfection using DEAE-dextran, Current protocols in cell biology, Chapter 20 (2003) Unit 20.24. [DOI] [PubMed] [Google Scholar]
  • [29].Laemmli UK, Characterization of DNA condensates induced by poly(ethylene oxide) and polylysine, Proceedings of the National Academy of Sciences of the United States of America, 72 (1975) 4288–4292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Chattoraj DK, Gosule LC, Schellman A, DNA condensation with polyamines. II. Electron microscopic studies, Journal of molecular biology, 121 (1978) 327–337. [DOI] [PubMed] [Google Scholar]
  • [31].Wu GY, Wu CH, Receptor-mediated in vitro gene transformation by a soluble DNA carrier system, The Journal of biological chemistry, 262 (1987) 4429–4432. [PubMed] [Google Scholar]
  • [32].Chowdhury NR, Wu CH, Wu GY, Yerneni PC, Bommineni VR, Chowdhury JR, Fate of DNA targeted to the liver by asialoglycoprotein receptor-mediated endocytosis in vivo. Prolonged persistence in cytoplasmic vesicles after partial hepatectomy, The Journal of biological chemistry, 268 (1993) 11265–11271. [PubMed] [Google Scholar]
  • [33].Kim SW, Polylysine copolymers for gene delivery, Cold Spring Harbor protocols, 2012 (2012) 433–438. [DOI] [PubMed] [Google Scholar]
  • [34].Sonawane ND, Szoka FC Jr., Verkman AS, Chloride accumulation and swelling in endosomes enhances DNA transfer by polyamine-DNA polyplexes, The Journal of biological chemistry, 278 (2003) 44826–44831. [DOI] [PubMed] [Google Scholar]
  • [35].Haensler J, Szoka FC Jr., Polyamidoamine cascade polymers mediate efficient transfection of cells in culture, Bioconjug Chem, 4 (1993) 372–379. [DOI] [PubMed] [Google Scholar]
  • [36].Araujo RV, Santos SDS, Igne Ferreira E, Giarolla J, New Advances in General Biomedical Applications of PAMAM Dendrimers, Molecules, 23 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Malik N, Wiwattanapatapee R, Klopsch R, Lorenz K, Frey H, Weener JW, Meijer EW, Paulus W, Duncan R, Dendrimers:: Relationship between structure and biocompatibility in vitro, and preliminary studies on the biodistribution of 125I-labelled polyamidoamine dendrimers in vivo, Journal of Controlled Release, 65 (2000) 133–148. [DOI] [PubMed] [Google Scholar]
  • [38].Roberts JC, Bhalgat MK, Zera RT, Preliminary biological evaluation of polyamidoamine (PAMAM) StarburstTM dendrimers, Journal of Biomedical Materials Research, 30 (1996) 53–65. [DOI] [PubMed] [Google Scholar]
  • [39].Shen Y, Zhou Z, Sui M, Tang J, Xu P, Kirk EAV, Murdoch WJ, Fan M, Radosz M, Charge-reversal polyamidoamine dendrimer for cascade nuclear drug delivery, Nanomedicine, 5 (2010) 1205–1217. [DOI] [PubMed] [Google Scholar]
  • [40].Milhem OM, Myles C, McKeown NB, Attwood D, D’Emanuele A, Polyamidoamine Starburst® dendrimers as solubility enhancers, International Journal of Pharmaceutics, 197 (2000) 239–241. [DOI] [PubMed] [Google Scholar]
  • [41].Sharma A, Porterfield JE, Smith E, Sharma R, Kannan S, Kannan RM, Effect of mannose targeting of hydroxyl PAMAM dendrimers on cellular and organ biodistribution in a neonatal brain injury model, J Control Release, 283 (2018) 175–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Ma Y, Sha M, Cheng S, Yao W, Li Z, Qi XR, Construction of Hyaluronic Tetrasaccharide Clusters Modified Polyamidoamine siRNA Delivery System, Nanomaterials (Basel), 8 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Altwaijry N, Somani S, Parkinson JA, Tate RJ, Keating P, Warzecha M, Mackenzie GR, Leung HY, Dufes C, Regression of prostate tumors after intravenous administration of lactoferrin-bearing polypropylenimine dendriplexes encoding TNF-alpha, TRAIL, and interleukin-12, Drug Deliv, 25 (2018) 679–689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Luo D, Haverstick K, Belcheva N, Han E, Saltzman WM, Poly(ethylene glycol)-Conjugated PAMAM Dendrimer for Biocompatible, High-Efficiency DNA Delivery, Macromolecules, 35 (2002) 3456–3462. [Google Scholar]
  • [45].Urban-Klein B, Werth S, Abuharbeid S, Czubayko F, Aigner A, RNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivo, Gene Therapy, 12 (2004) 461. [DOI] [PubMed] [Google Scholar]
  • [46].Ogris M, Brunner S, Schuller S, Kircheis R, Wagner E, PEGylated DNA/transferrin-PEI complexes: reduced interaction with blood components, extended circulation in blood and potential for systemic gene delivery, Gene Ther, 6 (1999) 595–605. [DOI] [PubMed] [Google Scholar]
  • [47].Wightman L, Kircheis R, Rossler V, Carotta S, Ruzicka R, Kursa M, Wagner E, Different behavior of branched and linear polyethylenimine for gene delivery in vitro and in vivo, The journal of gene medicine, 3 (2001) 362–372. [DOI] [PubMed] [Google Scholar]
  • [48].Schlosser K, Taha M, Deng Y, Stewart DJ, Systemic delivery of MicroRNA mimics with polyethylenimine elevates pulmonary microRNA levels, but lacks pulmonary selectivity, Pulm Circ, 8 (2018) 2045893217750613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Wagner E, Polymers for siRNA delivery: inspired by viruses to be targeted, dynamic, and precise, Accounts of chemical research, 45 (2012) 1005–1013. [DOI] [PubMed] [Google Scholar]
  • [50].Akinc A, Thomas M, Klibanov AM, Langer R, Exploring polyethylenimine-mediated DNA transfection and the proton sponge hypothesis, The journal of gene medicine, 7 (2005) 657–663. [DOI] [PubMed] [Google Scholar]
  • [51].Benjaminsen RV, Mattebjerg MA, Henriksen JR, Moghimi SM, Andresen TL, The Possible Proton Sponge; Effect of Polyethylenimine (PEI) Does Not Include Change in Lysosomal pH, Molecular Therapy, 21 (2013) 149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Doyle SR, Chan CK, Differential intracellular distribution of DNA complexed with polyethylenimine (PEI) and PEI-polyarginine PTD influences exogenous gene expression within live COS-7 cells, Genetic Vaccines and Therapy, 5 (2007) 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Godbey WT, Wu KK, Hirasaki GJ, Mikos AG, Improved packing of poly(ethylenimine)/DNA complexes increases transfection efficiency, Gene Ther, 6 (1999) 1380–1388. [DOI] [PubMed] [Google Scholar]
  • [54].Lee SH, Choi SH, Kim SH, Park TG, Thermally sensitive cationic polymer nanocapsules for specific cytosolic delivery and efficient gene silencing of siRNA: swelling induced physical disruption of endosome by cold shock, J Control Release, 125 (2008) 25–32. [DOI] [PubMed] [Google Scholar]
  • [55].Choi SH, Lee SH, Park TG, Temperature-Sensitive Pluronic/Poly(ethylenimine) Nanocapsules for Thermally Triggered Disruption of Intracellular Endosomal Compartment, Biomacromolecules, 7 (2006) 1864–1870. [DOI] [PubMed] [Google Scholar]
  • [56].Peng L, Wagner E, Polymeric Carriers for Nucleic Acid Delivery: Current Designs and Future Directions, Biomacromolecules, 20 (2019) 3613–3626. [DOI] [PubMed] [Google Scholar]
  • [57].Moghimi SM, Symonds P, Murray JC, Hunter AC, Debska G, Szewczyk A, A two-stage poly(ethylenimine)-mediated cytotoxicity: implications for gene transfer/therapy, Molecular therapy : the journal of the American Society of Gene Therapy, 11 (2005) 990–995. [DOI] [PubMed] [Google Scholar]
  • [58].Grandinetti G, Ingle NP, Reineke TM, Interaction of Poly(ethylenimine)–DNA Polyplexes with Mitochondria: Implications for a Mechanism of Cytotoxicity, Molecular Pharmaceutics, 8 (2011) 1709–1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Engineering Polymer Systems for Improved Drug Delivery, John Wiley & Sons, Inc; 2014. [Google Scholar]
  • [60].Grandinetti G, Ingle NP, Reineke TM, Interaction of poly(ethylenimine)-DNA polyplexes with mitochondria: implications for a mechanism of cytotoxicity, Mol Pharm, 8 (2011) 1709–1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Halman JR, Kim KT, Gwak SJ, Pace R, Johnson MB, Chandler MR, Rackley L, Viard M, Marriott I, Lee JS, Afonin KA, A cationic amphiphilic co-polymer as a carrier of nucleic acid nanoparticles (Nanps) for controlled gene silencing, immunostimulation, and biodistribution, Nanomedicine, 23 (2020) 102094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Gwak SJ, Lee JS, Suicide Gene Therapy By Amphiphilic Copolymer Nanocarrier for Spinal Cord Tumor, Nanomaterials (Basel), 9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Park JS, Yang HN, Yi SW, Kim JH, Park KH, Neoangiogenesis of human mesenchymal stem cells transfected with peptide-loaded and gene-coated PLGA nanoparticles, Biomaterials, 76 (2016) 226–237. [DOI] [PubMed] [Google Scholar]
  • [64].Ulkoski D, Bak A, Wilson JT, Krishnamurthy VR, Recent advances in polymeric materials for the delivery of RNA therapeutics, Expert Opin Drug Deliv, 16 (2019) 1149–1167. [DOI] [PubMed] [Google Scholar]
  • [65].Raup A, Stahlschmidt U, Jerome V, Synatschke CV, Muller AHE, Freitag R, Influence of Polyplex Formation on the Performance of Star-Shaped Polycationic Transfection Agents for Mammalian Cells, Polymers (Basel), 8 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Olden BR, Cheng Y, Yu JL, Pun SH, Cationic polymers for non-viral gene delivery to human T cells, J Control Release, 282 (2018) 140–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Jeon O, Yang HS, Lee TJ, Kim BS, Heparin-conjugated polyethylenimine for gene delivery, J Control Release, 132 (2008) 236–242. [DOI] [PubMed] [Google Scholar]
  • [68].Saito G, Swanson JA, Lee K-D, Drug delivery strategy utilizing conjugation via reversible disulfide linkages: role and site of cellular reducing activities, Advanced Drug Delivery Reviews, 55 (2003) 199–215. [DOI] [PubMed] [Google Scholar]
  • [69].Cheng R, Feng F, Meng F, Deng C, Feijen J, Zhong Z, Glutathione-responsive nano-vehicles as a promising platform for targeted intracellular drug and gene delivery, Journal of Controlled Release, 152 (2011) 2–12. [DOI] [PubMed] [Google Scholar]
  • [70].Bulmus V, Woodward M, Lin L, Murthy N, Stayton P, Hoffman A, A new pH-responsive and glutathione-reactive, endosomal membrane-disruptive polymeric carrier for intracellular delivery of biomolecular drugs, Journal of Controlled Release, 93 (2003) 105–120. [DOI] [PubMed] [Google Scholar]
  • [71].Ganta S, Devalapally H, Shahiwala A, Amiji M, A review of stimuli-responsive nanocarriers for drug and gene delivery, Journal of Controlled Release, 126 (2008) 187–204. [DOI] [PubMed] [Google Scholar]
  • [72].Meng F, Hennink WE, Zhong Z, Reduction-sensitive polymers and bioconjugates for biomedical applications, Biomaterials, 30 (2009) 2180–2198. [DOI] [PubMed] [Google Scholar]
  • [73].Tsarevsky NV, Matyjaszewski K, Combining Atom Transfer Radical Polymerization and Disulfide/Thiol Redox Chemistry:  A Route to Well-Defined (Bio)degradable Polymeric Materials, Macromolecules, 38 (2005) 3087–3092. [Google Scholar]
  • [74].Sunshine JC, Peng DY, Green JJ, Uptake and transfection with polymeric nanoparticles are dependent on polymer end-group structure, but largely independent of nanoparticle physical and chemical properties, Molecular pharmaceutics, 9 (2012) 3375–3383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Bus T, Traeger A, Schubert US, The great escape: how cationic polyplexes overcome the endosomal barrier, Journal of Materials Chemistry B, 6 (2018) 6904–6918. [DOI] [PubMed] [Google Scholar]
  • [76].Scherer F, Schillinger U, Putz U, Stemberger A, Plank C, Nonviral vector loaded collagen sponges for sustained gene delivery in vitro and in vivo, The journal of gene medicine, 4 (2002) 634–643. [DOI] [PubMed] [Google Scholar]
  • [77].Capito RM, Spector M, Collagen scaffolds for nonviral IGF-1 gene delivery in articular cartilage tissue engineering, Gene Therapy, 14 (2007) 721–732. [DOI] [PubMed] [Google Scholar]
  • [78].Curtin CM, Tierney EG, McSorley K, Cryan S-A, Duffy GP, O’Brien FJ, Combinatorial Gene Therapy Accelerates Bone Regeneration: Non-Viral Dual Delivery of VEGF and BMP2 in a Collagen-Nanohydroxyapatite Scaffold, Advanced Healthcare Materials, 4 (2015) 223–227. [DOI] [PubMed] [Google Scholar]
  • [79].Mageed RA, Adams G, Woodrow D, Podhajcer OL, Chernajovsky Y, Prevention of collagen-induced arthritis by gene delivery of soluble p75 tumour necrosis factor receptor, Gene Therapy, 5 (1998) 1584–1592. [DOI] [PubMed] [Google Scholar]
  • [80].Wang XT, Liu PY, Xin K-Q, Tang JB, Tendon Healing In Vitro: bFGF Gene Transfer to Tenocytes by Adeno-Associated Viral Vectors Promotes Expression of Collagen Genes, The Journal of Hand Surgery, 30 (2005) 1255–1261. [DOI] [PubMed] [Google Scholar]
  • [81].Cohen-Sacks H, Elazar V, Gao J, Golomb A, Adwan H, Korchov N, Levy RJ, Berger MR, Golomb G, Delivery and expression of pDNA embedded in collagen matrices, Journal of Controlled Release, 95 (2004) 309–320. [DOI] [PubMed] [Google Scholar]
  • [82].Erbacher P, Zou S, Bettinger T, Steffan AM, Remy JS, Chitosan-based vector/DNA complexes for gene delivery: biophysical characteristics and transfection ability, Pharmaceutical research, 15 (1998) 1332–1339. [DOI] [PubMed] [Google Scholar]
  • [83].Park IK, Kim TH, Park YH, Shin BA, Choi ES, Chowdhury EH, Akaike T, Cho CS, Galactosylated chitosan-graft-poly(ethylene glycol) as hepatocyte-targeting DNA carrier, J Control Release, 76 (2001) 349–362. [DOI] [PubMed] [Google Scholar]
  • [84].Fernandez Fernandez E, Santos-Carballal B, Weber WM, Goycoolea FM, Chitosan as a non-viral co-transfection system in a cystic fibrosis cell line, Int J Pharm, 502 (2016) 1–9. [DOI] [PubMed] [Google Scholar]
  • [85].Lallana E, Rios de la Rosa JM, Tirella A, Pelliccia M, Gennari A, Stratford IJ, Puri S, Ashford M, Tirelli N, Chitosan/Hyaluronic Acid Nanoparticles: Rational Design Revisited for RNA Delivery, Mol Pharm, 14 (2017) 2422–2436. [DOI] [PubMed] [Google Scholar]
  • [86].Louw AM, Kolar MK, Novikova LN, Kingham PJ, Wiberg M, Kjems J, Novikov LN, Chitosan polyplex mediated delivery of miRNA-124 reduces activation of microglial cells in vitro and in rat models of spinal cord injury, Nanomedicine, 12 (2016) 643–653. [DOI] [PubMed] [Google Scholar]
  • [87].Gaur S, Wen Y, Song JH, Parikh NU, Mangala LS, Blessing AM, Ivan C, Wu SY, Varkaris A, Shi Y, Lopez-Berestein G, Frigo DE, Sood AK, Gallick GE, Chitosan nanoparticle-mediated delivery of miRNA-34a decreases prostate tumor growth in the bone and its expression induces non-canonical autophagy, Oncotarget, 6 (2015) 29161–29177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Cosco D, Cilurzo F, Maiuolo J, Federico C, Di Martino MT, Cristiano MC, Tassone P, Fresta M, Paolino D, Delivery of miR-34a by chitosan/PLGA nanoplexes for the anticancer treatment of multiple myeloma, Sci Rep, 5 (2015) 17579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Chen X, Gu S, Chen BF, Shen WL, Yin Z, Xu GW, Hu JJ, Zhu T, Li G, Wan C, Ouyang HW, Lee TL, Chan WY, Nanoparticle delivery of stable miR-199a-5p agomir improves the osteogenesis of human mesenchymal stem cells via the HIF1a pathway, Biomaterials, 53 (2015) 239–250. [DOI] [PubMed] [Google Scholar]
  • [90].Köping-Höggård M, Tubulekas I, Guan H, Edwards K, Nilsson M, Vårum KM, Artursson P, Chitosan as a nonviral gene delivery system. Structure–property relationships and characteristics compared with polyethylenimine in vitro and after lung administration in vivo, Gene Therapy, 8 (2001) 1108–1121. [DOI] [PubMed] [Google Scholar]
  • [91].Kievit FM, Veiseh O, Bhattarai N, Fang C, Gunn JW, Lee D, Ellenbogen RG, Olson JM, Zhang M, PEI–PEG–Chitosan-Copolymer-Coated Iron Oxide Nanoparticles for Safe Gene Delivery: Synthesis, Complexation, and Transfection, Advanced Functional Materials, 19 (2009) 2244–2251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Ping Y, Liu C, Zhang Z, Liu KL, Chen J, Li J, Chitosan-graft-(PEI-β-cyclodextrin) copolymers and their supramolecular PEGylation for DNA and siRNA delivery, Biomaterials, 32 (2011) 8328–8341. [DOI] [PubMed] [Google Scholar]
  • [93].Wong K, Sun, Zhang G, Dai H, Liu, He Y, Leong KW, PEI-g-chitosan, a Novel Gene Delivery System with Transfection Efficiency Comparable to Polyethylenimine in Vitro and after Liver Administration in Vivo, Bioconjugate Chemistry, 17 (2006) 152–158. [DOI] [PubMed] [Google Scholar]
  • [94].Jiang H-L, Kim Y-K, Arote R, Nah J-W, Cho M-H, Choi Y-J, Akaike T, Cho C-S, Chitosan-graft-polyethylenimine as a gene carrier, Journal of Controlled Release, 117 (2007) 273–280. [DOI] [PubMed] [Google Scholar]
  • [95].Gao J-Q, Zhao Q-Q, Lv T-F, Shuai W-P, Zhou J, Tang G-P, Liang W-Q, Tabata Y, Hu Y-L, Gene-carried chitosan-linked-PEI induced high gene transfection efficiency with low toxicity and significant tumor-suppressive activity, International Journal of Pharmaceutics, 387 (2010) 286–294. [DOI] [PubMed] [Google Scholar]
  • [96].Mellet CO, Fernández JMG, Benito JM, Cyclodextrin-based gene delivery systems, Chemical Society Reviews, 40 (2011) 1586–1608. [DOI] [PubMed] [Google Scholar]
  • [97].Li J, Loh XJ, Cyclodextrin-based supramolecular architectures: Syntheses, structures, and applications for drug and gene delivery, Advanced Drug Delivery Reviews, 60 (2008) 1000–1017. [DOI] [PubMed] [Google Scholar]
  • [98].Pun SH, Bellocq NC, Liu A, Jensen G, Machemer T, Quijano E, Schluep T, Wen S, Engler H, Heidel J, Davis ME, Cyclodextrin-Modified Polyethylenimine Polymers for Gene Delivery, Bioconjugate Chemistry, 15 (2004) 831–840. [DOI] [PubMed] [Google Scholar]
  • [99].Bellocq NC, Pun SH, Jensen GS, Davis ME, Transferrin-Containing, Cyclodextrin Polymer-Based Particles for Tumor-Targeted Gene Delivery, Bioconjugate Chemistry, 14 (2003) 1122–1132. [DOI] [PubMed] [Google Scholar]
  • [100].Wada K, Arima H, Tsutsumi T, Chihara Y, Hattori K, Hirayama F, Uekama K, Improvement of gene delivery mediated by mannosylated dendrimer/α-cyclodextrin conjugates, Journal of Controlled Release, 104 (2005) 397–413. [DOI] [PubMed] [Google Scholar]
  • [101].Lai W-F, Cyclodextrins in non-viral gene delivery, Biomaterials, 35 (2014) 401–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Li J, Yang C, Li H, Wang X, Goh SH, Ding JL, Wang DY, Leong KW, Cationic Supramolecules Composed of Multiple Oligoethylenimine-Grafted β-Cyclodextrins Threaded on a Polymer Chain for Efficient Gene Delivery, Advanced Materials, 18 (2006) 2969–2974. [Google Scholar]
  • [103].Challa R, Ahuja A, Ali J, Khar RK, Cyclodextrins in drug delivery: an updated review, AAPS PharmSciTech, 6 (2005) E329–E357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Gidwani B, Vyas A, A Comprehensive Review on Cyclodextrin-Based Carriers for Delivery of Chemotherapeutic Cytotoxic Anticancer Drugs, Biomed Res Int, 2015 (2015) 198268–198268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Hu QD, Tang GP, Chu PK, Cyclodextrin-based host-guest supramolecular nanoparticles for delivery: from design to applications, Accounts of chemical research, 47 (2014) 2017–2025. [DOI] [PubMed] [Google Scholar]
  • [106].Feng R, Deng P, Teng F, Song Z, Recent Development of Copolymeric Delivery System for Anticancer Agents Based on Cyclodextrin Derivatives, Anti-Cancer Agents in Medicinal Chemistry- Anti-Cancer Agents), 16 (2016) 299–308. [DOI] [PubMed] [Google Scholar]
  • [107].Park I-K, von Recum HA, Jiang S, Pun SH, Supramolecular Assembly of Cyclodextrin-Based Nanoparticles on Solid Surfaces for Gene Delivery, Langmuir, 22 (2006) 8478–8484. [DOI] [PubMed] [Google Scholar]
  • [108].Mishra S, Webster P, Davis ME, PEGylation significantly affects cellular uptake and intracellular trafficking of non-viral gene delivery particles, European Journal of Cell Biology, 83 (2004) 97–111. [DOI] [PubMed] [Google Scholar]
  • [109].Pun SH, Davis ME, Development of a Nonviral Gene Delivery Vehicle for Systemic Application, Bioconjugate Chemistry, 13 (2002) 630–639. [DOI] [PubMed] [Google Scholar]
  • [110].Luo D, Woodrow-Mumford K, Belcheva N, Saltzman WM, Controlled DNA delivery systems, Pharmaceutical research, 16 (1999) 1300–1308. [DOI] [PubMed] [Google Scholar]
  • [111].Kapoor DN, Bhatia A, Kaur R, Sharma R, Kaur G, Dhawan S, PLGA: a unique polymer for drug delivery, Therapeutic delivery, 6 (2015) 41–58. [DOI] [PubMed] [Google Scholar]
  • [112].Woodrow KA, Cu Y, Booth CJ, Saucier-Sawyer JK, Wood MJ, Saltzman WM, Intravaginal gene silencing using biodegradable polymer nanoparticles densely loaded with small-interfering RNA, Nature materials, 8 (2009) 526–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Devalliere J, Chang WG, Andrejecsk JW, Abrahimi P, Cheng CJ, Jane-wit D, Saltzman WM, Pober JS, Sustained delivery of proangiogenic microRNA-132 by nanoparticle transfection improves endothelial cell transplantation, FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 28 (2014) 908–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Babar IA, Cheng CJ, Booth CJ, Liang X, Weidhaas JB, Saltzman WM, Slack FJ, Nanoparticle-based therapy in an in vivo microRNA-155 (miR-155)-dependent mouse model of lymphoma, Proceedings of the National Academy of Sciences of the United States of America, 109 (2012) E1695–1704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Wang T, Upponi JR, Torchilin VP, Design of multifunctional non-viral gene vectors to overcome physiological barriers: dilemmas and strategies, Int J Pharm, 427 (2012) 3–20. [DOI] [PubMed] [Google Scholar]
  • [116].Jain AK, Das M, Swarnakar NK, Jain S, Engineered PLGA nanoparticles: an emerging delivery tool in cancer therapeutics, Critical reviews in therapeutic drug carrier systems, 28 (2011) 1–45. [DOI] [PubMed] [Google Scholar]
  • [117].Oude Blenke E, Mahakena S, Fens M, van den Dikkenberg J, Holkers M, Mastrobattista E, Impact of chemistry and nanoformulation parameters on cellular uptake and airway distribution of RNA oligonucleotides, J Control Release, 317 (2020) 154–165. [DOI] [PubMed] [Google Scholar]
  • [118].Choi KY, Correa S, Min J, Li J, Roy S, Laccetti KH, Dreaden E, Kong S, Heo R, Roh YH, Lawson EC, Palmer PA, Hammond PT, Binary Targeting of siRNA to Hematologic Cancer Cells In Vivo using Layer-by-Layer Nanoparticles, Adv Funct Mater, 29 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Haque A, Dewerth A, Antony JS, Riethmuller J, Schweizer GR, Weinmann P, Latifi N, Yasar H, Pedemonte N, Sondo E, Weidensee B, Ralhan A, Laval J, Schlegel P, Seitz C, Loretz B, Lehr CM, Handgretinger R, Kormann MSD, Chemically modified hCFTR mRNAs recuperate lung function in a mouse model of cystic fibrosis, Sci Rep, 8 (2018) 16776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Lynn DM, Langer R, Degradable Poly(β-amino esters):  Synthesis, Characterization, and Self-Assembly with Plasmid DNA, Journal of the American Chemical Society, 122 (2000) 10761–10768. [Google Scholar]
  • [121].Lim Y.-b., Kim C.-h., Kim K, Kim SW, Park J.-s., Development of a Safe Gene Delivery System Using Biodegradable Polymer, Poly[α-(4-aminobutyl)-l-glycolic acid], Journal of the American Chemical Society, 122 (2000) 6524–6525. [Google Scholar]
  • [122].Bishop CJ, Abubaker-Sharif B, Guiriba T, Tzeng SY, Green JJ, Gene delivery polymer structure-function relationships elucidated via principal component analysis, Chem Commun (Camb), 51 (2015) 12134–12137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Kaczmarek JC, Kauffman KJ, Fenton OS, Sadtler K, Patel AK, Heartlein MW, DeRosa F, Anderson DG, Optimization of a Degradable Polymer-Lipid Nanoparticle for Potent Systemic Delivery of mRNA to the Lung Endothelium and Immune Cells, Nano Lett, 18 (2018) 6449–6454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Choi J, Rui Y, Kim J, Gorelick N, Wilson DR, Kozielski K, Mangraviti A, Sankey E, Brem H, Tyler B, Green JJ, Jackson EM, Nonviral polymeric nanoparticles for gene therapy in pediatric CNS malignancies, Nanomedicine, 23 (2020) 102115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Zamboni CG, Kozielski KL, Vaughan HJ, Nakata MM, Kim J, Higgins LJ, Pomper MG, Green JJ, Polymeric nanoparticles as cancer-specific DNA delivery vectors to human hepatocellular carcinoma, J Control Release, 263 (2017) 18–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Sunshine JC, Sunshine SB, Bhutto I, Handa JT, Green JJ, Poly(beta-amino ester)-nanoparticle mediated transfection of retinal pigment epithelial cells in vitro and in vivo, PLoS One, 7 (2012) e37543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Tros de Ilarduya C, Sun Y, Duzgunes N, Gene delivery by lipoplexes and polyplexes, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences, 40 (2010) 159–170. [DOI] [PubMed] [Google Scholar]
  • [128].Lv H, Zhang S, Wang B, Cui S, Yan J, Toxicity of cationic lipids and cationic polymers in gene delivery, Journal of Controlled Release, 114 (2006) 100–109. [DOI] [PubMed] [Google Scholar]
  • [129].Fields RJ, Cheng CJ, Quijano E, Weller C, Kristofik N, Duong N, Hoimes C, Egan ME, Saltzman WM, Surface modified poly(β amino ester)-containing nanoparticles for plasmid DNA delivery, Journal of Controlled Release, 164 (2012) 41–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Zhou J, Liu J, Cheng CJ, Patel TR, Weller CE, Piepmeier JM, Jiang Z, Saltzman WM, Biodegradable poly(amine-co-ester) terpolymers for targeted gene delivery, Nature materials, 11 (2011) 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Sunshine J, Bhise N, Green JJ, Degradable polymers for gene delivery, Conf Proc IEEE Eng Med Biol Soc, 2009 (2009) 2412–2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Kauffman AC, Piotrowski-Daspit AS, Nakazawa KH, Jiang Y, Datye A, Saltzman WM, Tunability of Biodegradable Poly(amine-co-ester) Polymers for Customized Nucleic Acid Delivery and Other Biomedical Applications, Biomacromolecules, 19 (2018) 3861–3873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Jiang Y, Gaudin A, Zhang J, Agarwal T, Song E, Kauffman AC, Tietjen GT, Wang Y, Jiang Z, Cheng CJ, Saltzman WM, A “top-down” approach to actuate poly(amine-co-ester) terpolymers for potent and safe mRNA delivery, Biomaterials, 176 (2018) 122–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Cui J, Piotrowski-Daspit AS, Zhang J, Shao M, Bracaglia LG, Utsumi T, Seo Y-E, DiRito J, Song E, Wu C, Inada A, Tietjen GT, Pober JS, Iwakiri Y, Saltzman WM, Poly(amine-co-ester) nanoparticles for effective Nogo-B knockdown in the liver, Journal of Controlled Release, 304 (2019) 259–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Pinheiro VB, Holliger P, The XNA world: progress towards replication and evolution of synthetic genetic polymers, Curr Opin Chem Biol, 16 (2012) 245–252. [DOI] [PubMed] [Google Scholar]
  • [136].Hollenstein M, Nucleoside triphosphates--building blocks for the modification of nucleic acids, Molecules, 17 (2012) 13569–13591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Rogers JM, Suga H, Discovering functional, non-proteinogenic amino acid containing, peptides using genetic code reprogramming, Org Biomol Chem, 13 (2015) 9353–9363. [DOI] [PubMed] [Google Scholar]
  • [138].Boyer C, Bulmus V, Davis TP, Ladmiral V, Liu J, Perrier S, Bioapplications of RAFT polymerization, Chemical reviews, 109 (2009) 5402–5436. [DOI] [PubMed] [Google Scholar]
  • [139].McClellan AK, Hao T, Brooks TA, Smith AE, RAFT Polymerization for the Synthesis of Tertiary Amine-Based Diblock Copolymer Nucleic Acid Delivery Vehicles, Macromol Biosci, 17 (2017). [DOI] [PubMed] [Google Scholar]
  • [140].Chen Z, Lichtor PA, Berliner AP, Chen JC, Liu DR, Evolution of sequence-defined highly functionalized nucleic acid polymers, Nat Chem, 10 (2018) 420–427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Hartmann L, Krause E, Antonietti M, Borner HG, Solid-phase supported polymer synthesis of sequence-defined, multifunctional poly(amidoamines), Biomacromolecules, 7 (2006) 1239–1244. [DOI] [PubMed] [Google Scholar]
  • [142].Wang Y, Luo J, Truebenbach I, Reinhard S, Klein PM, Höhn M, Kern S, Morys S, Loy DM, Wagner E, Zhang W, Double Click-Functionalized siRNA Polyplexes for Gene Silencing in Epidermal Growth Factor Receptor-Positive Tumor Cells, ACS Biomaterials Science & Engineering, 6 (2020) 1074–1089. [DOI] [PubMed] [Google Scholar]
  • [143].Kuhn J, Lin Y, Krhac Levacic A, Al Danaf N, Peng L, Höhn M, Lamb DC, Wagner E, Lächelt U, Delivery of Cas9/sgRNA Ribonucleoprotein Complexes via Hydroxystearyl Oligoamino Amides, Bioconjug Chem, 31 (2020) 729–742. [DOI] [PubMed] [Google Scholar]
  • [144].He D, Müller K, Krhac Levacic A, Kos P, Lächelt U, Wagner E, Combinatorial Optimization of Sequence-Defined Oligo(ethanamino)amides for Folate Receptor-Targeted pDNA and siRNA Delivery, Bioconjugate chemistry, 27 (2016) 647–659. [DOI] [PubMed] [Google Scholar]
  • [145].Zhang F, Trent Magruder J, Lin YA, Crawford TC, Grimm JC, Sciortino CM, Wilson MA, Blue ME, Kannan S, Johnston MV, Baumgartner WA, Kannan RM, Generation-6 hydroxyl PAMAM dendrimers improve CNS penetration from intravenous administration in a large animal brain injury model, J Control Release, 249 (2017) 173–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Mandl HK, Quijano E, Suh HW, Sparago E, Oeck S, Grun M, Glazer PM, Saltzman WM, Optimizing biodegradable nanoparticle size for tissue-specific delivery, J Control Release, 314 (2019) 92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Zhang M, Elango J, Guo X, Fan H, Cui M, Wang M, Liu K, Fe(3+)-Coordinated Multifunctional Elastic Nanoplatform for Effective in Vivo Gene Transfection, ACS Appl Mater Interfaces, 12 (2020) 3453–3464. [DOI] [PubMed] [Google Scholar]
  • [148].Stylianopoulos T, Jain RK, Design considerations for nanotherapeutics in oncology, Nanomedicine, 11 (2015) 1893–1907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Nakamura Y, Mochida A, Choyke PL, Kobayashi H, Nanodrug Delivery: Is the Enhanced Permeability and Retention Effect Sufficient for Curing Cancer?, Bioconjug Chem, 27 (2016) 2225–2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Kobayashi H, Watanabe R, Choyke PL, Improving conventional enhanced permeability and retention (EPR) effects; what is the appropriate target?, Theranostics, 4 (2013) 81–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Hoffman AS, The early days of PEG and PEGylation (1970s–1990s), Acta Biomaterialia, 40 (2016) 1–5. [DOI] [PubMed] [Google Scholar]
  • [152].Suk JS, Xu Q, Kim N, Hanes J, Ensign LM, PEGylation as a strategy for improving nanoparticle-based drug and gene delivery, Adv Drug Deliv Rev, 99 (2016) 28–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Carril M, Padro D, del Pino P, Carrillo-Carrion C, Gallego M, Parak WJ, In situ detection of the protein corona in complex environments, Nature Communications, 8 (2017) 1542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Walkey CD, Olsen JB, Guo H, Emili A, Chan WCW, Nanoparticle Size and Surface Chemistry Determine Serum Protein Adsorption and Macrophage Uptake, Journal of the American Chemical Society, 134 (2012) 2139–2147. [DOI] [PubMed] [Google Scholar]
  • [155].Hatakeyama H, Akita H, Harashima H, A multifunctional envelope type nano device (MEND) for gene delivery to tumours based on the EPR effect: a strategy for overcoming the PEG dilemma, Adv Drug Deliv Rev, 63 (2011) 152–160. [DOI] [PubMed] [Google Scholar]
  • [156].Tagami T, Uehara Y, Moriyoshi N, Ishida T, Kiwada H, Anti-PEG IgM production by siRNA encapsulated in a PEGylated lipid nanocarrier is dependent on the sequence of the siRNA, J Control Release, 151 (2011) 149–154. [DOI] [PubMed] [Google Scholar]
  • [157].Hamad I, Al-Hanbali O, Hunter AC, Rutt KJ, Andresen TL, Moghimi SM, Distinct polymer architecture mediates switching of complement activation pathways at the nanosphere-serum interface: implications for stealth nanoparticle engineering, ACS Nano, 4 (2010) 6629–6638. [DOI] [PubMed] [Google Scholar]
  • [158].Ban E, Kwon TH, Kim A, Delivery of therapeutic miRNA using polymer-based formulation, Drug Deliv Transl Res, 9 (2019) 1043–1056. [DOI] [PubMed] [Google Scholar]
  • [159].Sunder A, Hanselmann R, Frey H, Mülhaupt R, Controlled Synthesis of Hyperbranched Polyglycerols by Ring-Opening Multibranching Polymerization, Macromolecules, 32 (1999) 4240–4246. [Google Scholar]
  • [160].Deng Y, Ediriwickrema A, Yang F, Lewis J, Girardi M, Saltzman WM, A sunblock based on bioadhesive nanoparticles, Nature materials, 14 (2015) 1278–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Deng Y, Saucier-Sawyer JK, Hoimes CJ, Zhang J, Seo YE, Andrejecsk JW, Saltzman WM, The effect of hyperbranched polyglycerol coatings on drug delivery using degradable polymer nanoparticles, Biomaterials, 35 (2014) 6595–6602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Abbina S, Vappala S, Kumar P, Siren EMJ, La CC, Abbasi U, Brooks DE, Kizhakkedathu JN, Hyperbranched polyglycerols: recent advances in synthesis, biocompatibility and biomedical applications, Journal of Materials Chemistry B, 5 (2017) 9249–9277. [DOI] [PubMed] [Google Scholar]
  • [163].Wolfrum C, Shi S, Jayaprakash KN, Jayaraman M, Wang G, Pandey RK, Rajeev KG, Nakayama T, Charrise K, Ndungo EM, Zimmermann T, Koteliansky V, Manoharan M, Stoffel M, Mechanisms and optimization of in vivo delivery of lipophilic siRNAs, Nature biotechnology, 25 (2007) 1149–1157. [DOI] [PubMed] [Google Scholar]
  • [164].Ostergaard ME, Jackson M, Low A, Peralta ECA,GLR,RQ, Yu J, Kinberger GA, Dan A, Carty R, Tanowitz M, Anderson P, Kim TW, Fradkin L, Mullick AE, Murray S, Rigo F, Prakash TP, Bennett CF, Swayze EE, Gaus HJ, Seth PP, Conjugation of hydrophobic moieties enhances potency of antisense oligonucleotides in the muscle of rodents and non-human primates, Nucleic Acids Res, 47 (2019) 6045–6058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Tietjen GT, Bracaglia LG, Saltzman WM, Pober JS, Focus on Fundamentals: Achieving Effective Nanoparticle Targeting, Trends Mol Med, 24 (2018) 598–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Oishi M, Nagasaki Y, Nishiyama N, Itaka K, Takagi M, Shimamoto A, Furuichi Y, Kataoka K, Enhanced growth inhibition of hepatic multicellular tumor spheroids by lactosylated poly(ethylene glycol)-siRNA conjugate formulated in PEGylated polyplexes, ChemMedChem, 2 (2007) 1290–1297. [DOI] [PubMed] [Google Scholar]
  • [167].Mohammed AFA, Higashi T, Motoyama K, Ohyama A, Onodera R, Khaled KA, Sarhan HA, Hussein AK, Arima H, In Vitro and In Vivo Co-delivery of siRNA and Doxorubicin by Folate-PEG-Appended Dendrimer/Glucuronylglucosyl-beta-Cyclodextrin Conjugate, AAPS J, 21 (2019) 54. [DOI] [PubMed] [Google Scholar]
  • [168].Jones SK, Lizzio V, Merkel OM, Folate Receptor Targeted Delivery of siRNA and Paclitaxel to Ovarian Cancer Cells via Folate Conjugated Triblock Copolymer to Overcome TLR4 Driven Chemotherapy Resistance, Biomacromolecules, 17 (2016) 76–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Ohyama A, Higashi T, Motoyama K, Arima H, In Vitro and In Vivo Tumor-Targeting siRNA Delivery Using Folate-PEG-appended Dendrimer (G4)/alpha-Cyclodextrin Conjugates, Bioconjug Chem, 27 (2016) 521–532. [DOI] [PubMed] [Google Scholar]
  • [170].Li J, Chen L, Liu N, Li S, Hao Y, Zhang X, EGF-coated nano-dendriplexes for tumor-targeted nucleic acid delivery in vivo, Drug Deliv, 23 (2016) 1718–1725. [DOI] [PubMed] [Google Scholar]
  • [171].Mukalel AJ, Riley RS, Zhang R, Mitchell MJ, Nanoparticles for nucleic acid delivery: Applications in cancer immunotherapy, Cancer Lett, 458 (2019) 102–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Yu K, Wu S, Li H, A chitosan-graft-PEI-eprosartan conjugate for cardiomyocyte-targeted VEGF plasmid delivery in myocardial ischemia gene therapy, Journal of Experimental Nanoscience, 11 (2016) 81–96. [Google Scholar]
  • [173].Chen Q, Osada K, Ge Z, Uchida S, Tockary TA, Dirisala A, Matsui A, Toh K, Takeda KM, Liu X, Nomoto T, Ishii T, Oba M, Matsumoto Y, Kataoka K, Polyplex micelle installing intracellular self-processing functionalities without free catiomers for safe and efficient systemic gene therapy through tumor vasculature targeting, Biomaterials, 113 (2017) 253–265. [DOI] [PubMed] [Google Scholar]
  • [174].Luo L, Zhang X, Hirano Y, Tyagi P, Barabas P, Uehara H, Miya TR, Singh N, Archer B, Qazi Y, Jackman K, Das SK, Olsen T, Chennamaneni SR, Stagg BC, Ahmed F, Emerson L, Zygmunt K, Whitaker R, Mamalis C, Huang W, Gao G, Srinivas SP, Krizaj D, Baffi J, Ambati J, Kompella UB, Ambati BK, Targeted intraceptor nanoparticle therapy reduces angiogenesis and fibrosis in primate and murine macular degeneration, ACS Nano, 7 (2013) 3264–3275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Wang J, Lei Y, Xie C, Lu W, Yan Z, Gao J, Xie Z, Zhang X, Liu M, Targeted gene delivery to glioblastoma using a C-end rule RGERPPR peptide-functionalised polyethylenimine complex, Int J Pharm, 458 (2013) 48–56. [DOI] [PubMed] [Google Scholar]
  • [176].Newman MR, Russell SG, Schmitt CS, Marozas IA, Sheu TJ, Puzas JE, Benoit DSW, Multivalent Presentation of Peptide Targeting Groups Alters Polymer Biodistribution to Target Tissues, Biomacromolecules, 19 (2018) 71–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Manunta MDI, Tagalakis AD, Attwood M, Aldossary AM, Barnes JL, Munye MM, Weng A, McAnulty RJ, Hart SL, Delivery of ENaC siRNA to epithelial cells mediated by a targeted nanocomplex: a therapeutic strategy for cystic fibrosis, Sci Rep, 7 (2017) 700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Wu LP, Ahmadvand D, Su J, Hall A, Tan X, Farhangrazi ZS, Moghimi SM, Crossing the blood-brain-barrier with nanoligand drug carriers self-assembled from a phage display peptide, Nat Commun, 10 (2019) 4635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Cai C, Xie Y, Wu L, Chen X, Liu H, Zhou Y, Zou H, Liu D, Zhao Y, Kong X, Liu P, PLGA-based dual targeted nanoparticles enhance miRNA transfection efficiency in hepatic carcinoma, Sci Rep, 7 (2017) 46250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Li S, Huang L, Nonviral gene therapy: promises and challenges, Gene Therapy, 7 (2000) 31–34. [DOI] [PubMed] [Google Scholar]
  • [181].Ogris M, Wagner E, Targeting tumors with non-viral gene delivery systems, Drug Discovery Today, 7 (2002) 479–485. [DOI] [PubMed] [Google Scholar]
  • [182].Sahoo SK, Labhasetwar V, Nanotech approaches to drug delivery and imaging, Drug Discovery Today, 8 (2003) 1112–1120. [DOI] [PubMed] [Google Scholar]
  • [183].Gao W, Zhang L, Coating nanoparticles with cell membranes for targeted drug delivery, J Drug Target, 23 (2015) 619–626. [DOI] [PubMed] [Google Scholar]
  • [184].Davis ME, Zuckerman JE, Choi CHJ, Seligson D, Tolcher A, Alabi CA, Yen Y, Heidel JD, Ribas A, Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles, Nature, 464 (2010) 1067–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Zuckerman JE, Gritli I, Tolcher A, Heidel JD, Lim D, Morgan R, Chmielowski B, Ribas A, Davis ME, Yen Y, Correlating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing siRNA, Proceedings of the National Academy of Sciences of the United States of America, 111 (2014) 11449–11454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Miele E, Spinelli GP, Miele E, Di Fabrizio E, Ferretti E, Tomao S, Gulino A, Nanoparticle-based delivery of small interfering RNA: challenges for cancer therapy, Int J Nanomedicine, 7 (2012) 3637–3657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Buscail L, Bournet B, Vernejoul F, Cambois G, Lulka H, Hanoun N, Dufresne M, Meulle A, Vignolle-Vidoni A, Ligat L, Saint-Laurent N, Pont F, Dejean S, Gayral M, Martins F, Torrisani J, Barbey O, Gross F, Guimbaud R, Otal P, Lopez F, Tiraby G, Cordelier P, First-in-man phase 1 clinical trial of gene therapy for advanced pancreatic cancer: safety, biodistribution, and preliminary clinical findings, Molecular therapy : the journal of the American Society of Gene Therapy, 23 (2015) 779–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Alvarez RD, Sill MW, Davidson SA, Muller CY, Bender DP, DeBernardo RL, Behbakht K, Huh WK, A phase II trial of intraperitoneal EGEN-001, an IL-12 plasmid formulated with PEG–PEI–cholesterol lipopolymer in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer: A Gynecologic Oncology Group study, Gynecologic Oncology, 133 (2014) 433–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Wong SY, Pelet JM, Putnam D, Polymer systems for gene delivery—Past, present, and future, Progress in Polymer Science, 32 (2007) 799–837. [Google Scholar]
  • [190].Hill AB, Chen M, Chen C-K, Pfeifer BA, Jones CH, Overcoming Gene-Delivery Hurdles: Physiological Considerations for Nonviral Vectors, Trends in Biotechnology, 34 (2016) 91–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Shanks N, Greek R, Greek J, Are animal models predictive for humans?, Philos Ethics Humanit Med, 4 (2009) 2–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].van der Worp HB, Howells DW, Sena ES, Porritt MJ, Rewell S, O’Collins V, Macleod MR, Can animal models of disease reliably inform human studies?, PLoS Med, 7 (2010) e1000245–e1000245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Ng CP, Pun SH, A perfusable 3D cell–matrix tissue culture chamber for in situ evaluation of nanoparticle vehicle penetration and transport, Biotechnology and Bioengineering, 99 (2008) 1490–1501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Ravi M, Paramesh V, Kaviya SR, Anuradha E, Solomon FDP, 3D Cell Culture Systems: Advantages and Applications, Journal of Cellular Physiology, 230 (2015) 16–26. [DOI] [PubMed] [Google Scholar]
  • [195].Fitzgerald KA, Malhotra M, Curtin CM, O’ Brien FJ, O’ Driscoll CM, Life in 3D is never flat: 3D models to optimise drug delivery, Journal of Controlled Release, 215 (2015) 39–54. [DOI] [PubMed] [Google Scholar]
  • [196].Simara P, Motl JA, Kaufman DS, Pluripotent stem cells and gene therapy, Transl Res, 161 (2013) 284–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Ma N, Zhang JZ, Itzhaki I, Zhang SL, Chen H, Haddad F, Kitani T, Wilson KD, Tian L, Shrestha R, Wu H, Lam CK, Sayed N, Wu JC, Determining the Pathogenicity of a Genomic Variant of Uncertain Significance Using CRISPR/Cas9 and Human-Induced Pluripotent Stem Cells, Circulation, 138 (2018) 2666–2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Yonamine Y, Hoshino Y, Shea KJ, ELISA-mimic screen for synthetic polymer nanoparticles with high affinity to target proteins, Biomacromolecules, 13 (2012) 2952–2957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Bracaglia LG, Piotrowski-Daspit AS, Lin CY, Moscato ZM, Wang Y, Tietjen GT, Saltzman WM, High-throughput quantitative microscopy-based half-life measurements of intravenously injected agents, Proceedings of the National Academy of Sciences of the United States of America, 117 (2020) 3502–3508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Dahlman JE, Kauffman KJ, Xing Y, Shaw TE, Mir FF, Dlott CC, Langer R, Anderson DG, Wang ET, Barcoded nanoparticles for high throughput in vivo discovery of targeted therapeutics, Proceedings of the National Academy of Sciences of the United States of America, 114 (2017) 2060–2065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Tavares AJ, Poon W, Zhang YN, Dai Q, Besla R, Ding D, Ouyang B, Li A, Chen J, Zheng G, Robbins C, Chan WCW, Effect of removing Kupffer cells on nanoparticle tumor delivery, Proceedings of the National Academy of Sciences of the United States of America, 114 (2017) E10871–E10880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Wilhelm S, Tavares AJ, Dai Q, Ohta S, Audet J, Dvorak HF, Chan WCW, Analysis of nanoparticle delivery to tumours, Nature Reviews Materials, 1 (2016) 16014. [Google Scholar]
  • [203].Wang Y, Malcolm DW, Benoit DSW, Controlled and sustained delivery of siRNA/NPs from hydrogels expedites bone fracture healing, Biomaterials, 139 (2017) 127–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Tokatlian T, Cam C, Segura T, Porous hyaluronic acid hydrogels for localized nonviral DNA delivery in a diabetic wound healing model, Adv Healthc Mater, 4 (2015) 1084–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Gillooly AR, Perry J, Martins PN, First Report of siRNA Uptake (for RNA Interference) During Ex Vivo Hypothermic and Normothermic Liver Machine Perfusion, Transplantation, 103 (2019) e56–e57. [DOI] [PubMed] [Google Scholar]
  • [206].Tietjen GT, Hosgood SA, DiRito J, Cui J, Deep D, Song E, Kraehling JR, Piotrowski-Daspit AS, Kirkiles-Smith NC, Al-Lamki R, Thiru S, Bradley JA, Saeb-Parsy K, Bradley JR, Nicholson ML, Saltzman WM, Pober JS, Nanoparticle targeting to the endothelium during normothermic machine perfusion of human kidneys, Sci Transl Med, 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Chen YJ, Tsai CY, Cheng YM, Nieh SW, Yeh TK, Chen CP, Wang MH, Chou LH, Chiu TY, Liu L, Ho C, Chen CT, Liu TW, Impacts of Intralipid on Nanodrug Abraxane Therapy and on the Innate Immune System, Sci Rep, 10 (2020) 2838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Liu L, Ye Q, Lu M, Chen ST, Tseng HW, Lo YC, Ho C, A New Approach to Deliver Anti-cancer Nanodrugs with Reduced Off-target Toxicities and Improved Efficiency by Temporarily Blunting the Reticuloendothelial System with Intralipid, Sci Rep, 7 (2017) 16106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Liu T, Choi H, Zhou R, Chen IW, RES blockade: A strategy for boosting efficiency of nanoparticle drug, Nano Today, 10 (2015) 11–21. [Google Scholar]
  • [210].Cheng Q, Wei T, Farbiak L, Johnson LT, Dilliard SA, Siegwart DJ, Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing, Nature Nanotechnology, 15 (2020) 313–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Mathew AP, Cho KH, Uthaman S, Cho CS, Park IK, Stimuli-Regulated Smart Polymeric Systems for Gene Therapy, Polymers (Basel), 9 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Wang HB, Yang L, Wu J, Sun L, Wu J, Tian H, Weisel RD, Li RK, Reduced ischemic injury after stroke in mice by angiogenic gene delivery via ultrasound-targeted microbubble destruction, J Neuropathol Exp Neurol, 73 (2014) 548–558. [DOI] [PubMed] [Google Scholar]
  • [213].Neto MF, Letteri R, Chan-Seng D, Emrick T, Figueiredo ML, Sonodelivery Facilitates Sustained Luciferase Expression from an Episomal Vector in Skeletal Muscle, Materials (Basel), 8 (2015) 4608–4617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Epps TH, Vi T, Sullivan MO, Design and development of a robust photo-responsive block copolymer framework for tunable nucleic acid delivery and efficient gene silencing, Polymer Journal, 50 (2018) 711–723. [Google Scholar]
  • [215].He Y, Guo S, Wu L, Chen P, Wang L, Liu Y, Ju H, Near-infrared boosted ROS responsive siRNA delivery and cancer therapy with sequentially peeled upconversion nano-onions, Biomaterials, 225 (2019) 119501. [DOI] [PubMed] [Google Scholar]
  • [216].Fisher WG, Partridge WP Jr., Dees C, Wachter EA, Simultaneous Two-Photon Activation of Type-I Photodynamic Therapy Agents, Photochemistry and Photobiology, 66 (1997) 141–155. [DOI] [PubMed] [Google Scholar]
  • [217].Yin L, Tang H, Kim KH, Zheng N, Song Z, Gabrielson NP, Lu H, Cheng J, Light-Responsive Helical Polypeptides Capable of Reducing Toxicity and Unpacking DNA: Toward Nonviral Gene Delivery, Angewandte Chemie International Edition, 52 (2013) 9182–9186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Malcolm DW, Varghese JJ, Sorrells JE, Ovitt CE, Benoit DSW, The Effects of Biological Fluids on Colloidal Stability and siRNA Delivery of a pH-Responsive Micellar Nanoparticle Delivery System, ACS Nano, 12 (2018) 187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Wu C, Tian Y, Zhang Y, Xu J, Wang Y, Guan X, Li T, Yang H, Li S, Qin X, Liu Y, Acid-Triggered Charge-Convertible Graphene-Based All-in-One Nanocomplex for Enhanced Genetic Phototherapy of Triple-Negative Breast Cancer, Adv Healthc Mater, 9 (2020) e1901187. [DOI] [PubMed] [Google Scholar]
  • [220].Ping Y, Hu Q, Tang G, Li J, FGFR-targeted gene delivery mediated by supramolecular assembly between beta-cyclodextrin-crosslinked PEI and redox-sensitive PEG, Biomaterials, 34 (2013) 6482–6494. [DOI] [PubMed] [Google Scholar]
  • [221].Schmaljohann D, Thermo- and pH-responsive polymers in drug delivery, Advanced Drug Delivery Reviews, 58 (2006) 1655–1670. [DOI] [PubMed] [Google Scholar]
  • [222].Sethuraman VA, Na K, Bae YH, pH-Responsive Sulfonamide/PEI System for Tumor Specific Gene Delivery:  An in Vitro Study, Biomacromolecules, 7 (2006) 64–70. [DOI] [PubMed] [Google Scholar]
  • [223].Lynn DM, Amiji MM, Langer R, pH-Responsive Polymer Microspheres: Rapid Release of Encapsulated Material within the Range of Intracellular pH, Angewandte Chemie International Edition, 40 (2001) 1707–1710. [PubMed] [Google Scholar]
  • [224].Rasolonjatovo B, Illy N, Bennevault V, Mathe J, Midoux P, Le Gall T, Haudebourg T, Montier T, Lehn P, Pitard B, Cheradame H, Huin C, Guegan P, Temperature-Sensitive Amphiphilic Non-Ionic Triblock Copolymers for Enhanced In Vivo Skeletal Muscle Transfection, Macromol Biosci, 20 (2020) e1900276. [DOI] [PubMed] [Google Scholar]
  • [225].Takeda N, Nakamura E, Yokoyama M, Okano T, Temperature-responsive polymeric carriers incorporating hydrophobic monomers for effective transfection in small doses, Journal of Controlled Release, 95 (2004) 343–355. [DOI] [PubMed] [Google Scholar]
  • [226].Wang M, Cheng Y, Temperature-Responsive Gene Silencing by a Smart Polymer, Bioconjugate Chemistry, 27 (2016) 495–499. [DOI] [PubMed] [Google Scholar]
  • [227].Kim T.-i., Kim SW, Bioreducible polymers for gene delivery, Reactive and Functional Polymers, 71 (2011) 344–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Elzes MR, Akeroyd N, Engbersen JFJ, Paulusse JMJ, Disulfide-functional poly(amido amine)s with tunable degradability for gene delivery, Journal of Controlled Release, 244 (2016) 357–365. [DOI] [PubMed] [Google Scholar]
  • [229].Zhang P, Wagner E, History of Polymeric Gene Delivery Systems, in: Cheng Y (Ed.) Polymeric Gene Delivery Systems, Springer International Publishing, Cham, 2018, pp. 1–39. [Google Scholar]
  • [230].Liou G-Y, Storz P, Reactive oxygen species in cancer, Free Radical Research, 44 (2010) 479–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Tsenoglou C, Molecular weight polydispersity effects on the viscoelasticity of entangled linear polymers, Macromolecules, 24 (1991) 1762–1767. [Google Scholar]
  • [232].Badi N, Lutz J-F, Sequence control in polymer synthesis, Chemical Society Reviews, 38 (2009) 3383–3390. [DOI] [PubMed] [Google Scholar]
  • [233].Paliwal R, Babu RJ, Palakurthi S, Nanomedicine scale-up technologies: feasibilities and challenges, AAPS PharmSciTech, 15 (2014) 1527–1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Sen S, Puskas JE, Green polymer chemistry: enzyme catalysis for polymer functionalization, Molecules (Basel, Switzerland), 20 (2015) 9358–9379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Hu Q, Katti PS, Gu Z, Enzyme-responsive nanomaterials for controlled drug delivery, Nanoscale, 6 (2014) 12273–12286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [236].Zhou J, Liu J, Cheng CJ, Patel TR, Weller CE, Piepmeier JM, Jiang Z, Saltzman WM, Biodegradable poly(amine-co-ester) terpolymers for targeted gene delivery, Nature materials, 11 (2012) 82–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Wang C-F, Lin Y-X, Jiang T, He F, Zhuo R-X, Polyethylenimine-grafted polycarbonates as biodegradable polycations for gene delivery, Biomaterials, 30 (2009) 4824–4832. [DOI] [PubMed] [Google Scholar]
  • [238].Kobayashi S, Kaplan DL, Ritter H, Kobayashi S, Kaplan D, Matsumura S, Ohmae M, Reihmann M, Singh A, Uyama H, Enzyme-Catalyzed Synthesis of Polymers, Springer Berlin Heidelberg 2006. [Google Scholar]
  • [239].Albertsson A-C, Srivastava RK, Recent developments in enzyme-catalyzed ring-opening polymerization, Advanced Drug Delivery Reviews, 60 (2008) 1077–1093. [DOI] [PubMed] [Google Scholar]
  • [240].Lutz J-F, Ouchi M, Liu DR, Sawamoto M, Sequence-Controlled Polymers, Science, 341 (2013) 1238149. [DOI] [PubMed] [Google Scholar]
  • [241].Gross RA, Ganesh M, Lu W, Enzyme-catalysis breathes new life into polyester condensation polymerizations, Trends in Biotechnology, 28 (2010) 435–443. [DOI] [PubMed] [Google Scholar]
  • [242].Gross RA, Kumar A, Kalra B, Polymer Synthesis by In Vitro Enzyme Catalysis, Chemical reviews, 101 (2001) 2097–2124. [DOI] [PubMed] [Google Scholar]
  • [243].Knani D, Gutman AL, Kohn DH, Enzymatic polyesterification in organic media. Enzyme-catalyzed synthesis of linear polyesters. I. Condensation polymerization of linear hydroxyesters. II. Ring-opening polymerization of ϵ-caprolactone, Journal of Polymer Science Part A: Polymer Chemistry, 31 (1993) 1221–1232. [Google Scholar]
  • [244].Lau SC, Lim HN, Basri M, Fard Masoumi HR, Ahmad Tajudin A, Huang NM, Pandikumar A, Chia CH, Andou Y, Enhanced Biocatalytic Esterification with Lipase-Immobilized Chitosan/Graphene Oxide Beads, PLOS ONE, 9 (2014) e104695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Pereira EB, Zanin GM, Castro HF, Immobilization and catalytic properties of lipase on chitosan for hydrolysis and esterification reactions, Brazilian Journal of Chemical Engineering, 20 (2003) 343–355. [Google Scholar]
  • [246].Kobayashi S, Uyama H, Kimura S, Enzymatic Polymerization, Chemical reviews, 101 (2001) 3793–3818. [DOI] [PubMed] [Google Scholar]
  • [247].Liu J, Jiang X, Xu L, Wang X, Hennink WE, Zhuo R, Novel Reduction-Responsive Cross-Linked Polyethylenimine Derivatives by Click Chemistry for Nonviral Gene Delivery, Bioconjugate Chemistry, 21 (2010) 1827–1835. [DOI] [PubMed] [Google Scholar]
  • [248].Lai W-F, Green DW, Jung H-S, Linear Poly(ethylenimine) Cross-Linked by Methyl-β-Cyclodextrin for Gene Delivery, Current Gene Therapy, 14 (2014) 258–268. [DOI] [PubMed] [Google Scholar]
  • [249].Xu FJ, Yang WT, Polymer vectors via controlled/living radical polymerization for gene delivery, Progress in Polymer Science, 36 (2011) 1099–1131. [Google Scholar]
  • [250].Wang ZH, Li WB, Ma J, Tang GP, Yang WT, Xu FJ, Functionalized Nonionic Dextran Backbones by Atom Transfer Radical Polymerization for Efficient Gene Delivery, Macromolecules, 44 (2011) 230–239. [Google Scholar]
  • [251].Leibfarth FA, Johnson JA, Jamison TF, Scalable synthesis of sequence-defined, unimolecular macromolecules by Flow-IEG, Proceedings of the National Academy of Sciences, 112 (2015) 10617–10622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Corrigan N, Manahan R, Lew ZT, Yeow J, Xu J, Boyer C, Copolymers with Controlled Molecular Weight Distributions and Compositional Gradients through Flow Polymerization, Macromolecules, 51 (2018) 4553–4563. [Google Scholar]
  • [253].Binauld S, Damiron D, Connal LA, Hawker CJ, Drockenmuller E, Precise Synthesis of Molecularly Defined Oligomers and Polymers by Orthogonal Iterative Divergent/Convergent Approaches, Macromolecular Rapid Communications, 32 (2011) 147–168. [DOI] [PubMed] [Google Scholar]
  • [254].Edwardson TGW, Carneiro KMM, Serpell CJ, Sleiman HF, An Efficient and Modular Route to Sequence-Defined Polymers Appended to DNA, Angewandte Chemie International Edition, 53 (2014) 4567–4571. [DOI] [PubMed] [Google Scholar]
  • [255].Kaligian KL, Sprachman MM, Controlled polymers: accessing new platforms for material synthesis, Molecular Systems Design & Engineering, 4 (2019) 144–161. [Google Scholar]
  • [256].Nanjan P, Porel M, Sequence-defined non-natural polymers: synthesis and applications, Polymer Chemistry, 10 (2019) 5406–5424. [Google Scholar]
  • [257].Zhao B, Gao Z, Zheng Y, Gao C, Scalable Synthesis of Positively Charged Sequence-Defined Functional Polymers, Journal of the American Chemical Society, 141 (2019) 4541–4546. [DOI] [PubMed] [Google Scholar]
  • [258].Yilgör I, McGrath JE, Novel supercritical fluid techniques for polymer fractionation and purification, Polymer Bulletin, 12 (1984) 499–506. [Google Scholar]
  • [259].Chang S-H, Park S-C, Shim J-J, Phase equilibria of supercritical fluid–polymer systems, The Journal of Supercritical Fluids, 13 (1998) 113–119. [Google Scholar]
  • [260].Kankala RK, Zhang YS, Wang S-B, Lee C-H, Chen A-Z, Supercritical Fluid Technology: An Emphasis on Drug Delivery and Related Biomedical Applications, Advanced Healthcare Materials, 6 (2017) 1700433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Yang W, Yu C, Wu C, Yao SQ, Wu S, Cell-penetrating poly(disulfide)-based star polymers for simultaneous intracellular delivery of miRNAs and small molecule drugs, Polymer Chemistry, 8 (2017) 4043–4051. [Google Scholar]
  • [262].Bus T, Englert C, Reifarth M, Borchers P, Hartlieb M, Vollrath A, Hoeppener S, Traeger A, Schubert US 3rd generation poly(ethylene imine)s for gene delivery, Journal of Materials Chemistry B, 5 (2017) 1258–1274. [DOI] [PubMed] [Google Scholar]
  • [263].Leung AKK, Tam YYC, Chen S, Hafez IM, Cullis PR, Microfluidic Mixing: A General Method for Encapsulating Macromolecules in Lipid Nanoparticle Systems, The Journal of Physical Chemistry B, 119 (2015) 8698–8706. [DOI] [PubMed] [Google Scholar]

RESOURCES