146
Views
63
CrossRef citations to date
0
Altmetric
Review

Polyethylenimine-based micro/nanoparticles as vaccine adjuvants

, , , , , & show all
Pages 5443-5460 | Published online: 31 Jul 2017

Abstract

Vaccines have shown great success in treating and preventing tumors and infections, while adjuvants are always demanded to ensure potent immune responses. Polyethylenimine (PEI), as one of the well-studied cationic polymers, has been used as a transfection reagent for decades. However, increasing evidence has shown that PEI-based particles are also capable of acting as adjuvants. In this paper, we briefly review the physicochemical properties and the broad applications of PEI in different fields, and elaborate on the intracellular processes of PEI-based vaccines. In addition, we sum up the proof of their in vivo and clinical applications. We also highlight some mechanisms proposed for the intrinsic immunoactivation function of PEI, followed by the challenges and future perspectives of the applications of PEI in the vaccines, as well as some strategies to elicit the desirable immune responses.

View correction statement:
Polyethylenimine-based micro/nanoparticles as vaccine adjuvants [Corrigendum]

Introduction

Due to their weak immunogenicity, conventional vaccines, especially the subunit vaccines, are always combined with adjuvants to ensure potent immune responses. Polyethylenimine (PEI), as a kind of cationic polymer, has been extensively applied as a nucleotide delivery reagent for decades. In recent years, the robust adjuvanticity of PEI has been continuously documented. Increasing evidence has shown that PEI-based particles are capable of improving the efficiency of conventional vaccines against infections and tumors. These efficiencies are characterized by direct indicators, such as enhanced maturation rates of antigen-presenting cells (APCs) and increased proliferation of effector cells, as well as amplified production of antigen-specific antibodies and various cytokines and chemokines.

In this review, we first introduce the physicochemical properties of PEI and its general applications in distinct areas. Then, we focus on the effects of neat PEI itself and PEI-based nanoparticles/microparticles (NPs/MPs) in antigen uptake and presentation, which is the foundation for understanding the interplay between PEI and APCs. We then extend the focus on the effects of PEI-relevant adjuvant potency from the cell level to preclinical studies or clinical trials. Subsequently, we are fascinated to figure out some possible underlying mechanisms of its intrinsic immunoactivation functions. Finally, we discuss the challenges and future perspectives of the applications of PEI in vaccines, and explore the promising approaches to optimize it’s immune responses while manipulating the toxicity properly.

Synthesis and broad applications of PEI

PEI is a kind of synthesized cationic polymer with topologies of linear or branched forms, and its molecular weight ranges from 1 kDa to 1,000 kDa.Citation1 The most common and essential characteristic of PEI is its hydrophilic cationic polymeric structure. The strong positive-charged PEI condenses negative particles (such as the DNA, negative antigens) or plasma membranes in vivo. Moreover, the PEI backbone contains one nitrogen atom in every three atoms,Citation2 forming amorphous net structures to work powerfully in lysosomes, as a “proton sponge”.Citation3,Citation4 These active amino groups, especially the primary and secondary amines, provide numerous possibilities for structural modifications, which enable them to target the agents and attenuate the potential toxicity.Citation5

Synthesis and physiochemical properties of PEI

In general, branched PEI, which contains primary, secondary and tertiary amine groups, can be synthesized through the cationic ring-opening polymerization of aziridine (). Linear PEI (LPEI), however, includes secondary amines only, commonly derived from acidic hydrolysis of polyoxazoline (). Generally, the synthetic processes of gaining LPEI with narrow distribution of the molecular weight could be rather challenging.Citation5 In PEI, amino groups with different sorts possess different properties. It is evidenced that primary and secondary amines have strong abilities to bind to nucleic acids, as well as targeting agents, drugs and other functional moieties. Yet, although with less binding capacity, tertiary amines efficiently buffer the pH decline in acidic conditions.Citation6,Citation7 In general, branched PEI is in the liquid state and water-soluble, whereas LPEI is solid at room temperature and less soluble in cold water, phenol, ethyl, ether, acetone and other solvants, and it turns more soluble in hot water, acidic aqueous solution and organic solutions (such as the methanol, ethanol or chloroform). Though PEIs of these two topologies are quite different, they both possess active amino groups, and the over-positive-charged nitrogens are always linked to the toxicity of PEI.Citation5

Figure 1 (A) Molecular structure of branched PEI. (B) Polymerization of aziridine to branched PEI. (C) Mechanism of the cationic ring-opening polymerization of aziridine. (D) Molecular structure of linear PEI. (E) Synthesis of linear PEI from substituted 2-oxazolines. Reproduced from Jäger M, Schubert S, Ochrimenko S, Fischer D, Schubert US. Branched and linear poly(ethylene imine)-based conjugates: syn thetic modification, characterization, and application. Chem Soc Rev. 2012;41(13):4755–4767. With permission of The Royal Society of Chemistry.Citation5

Abbreviations: PEI, polyethylenimine; conc, concentrate.

Figure 1 (A) Molecular structure of branched PEI. (B) Polymerization of aziridine to branched PEI. (C) Mechanism of the cationic ring-opening polymerization of aziridine. (D) Molecular structure of linear PEI. (E) Synthesis of linear PEI from substituted 2-oxazolines. Reproduced from Jäger M, Schubert S, Ochrimenko S, Fischer D, Schubert US. Branched and linear poly(ethylene imine)-based conjugates: syn thetic modification, characterization, and application. Chem Soc Rev. 2012;41(13):4755–4767. With permission of The Royal Society of Chemistry.Citation5Abbreviations: PEI, polyethylenimine; conc, concentrate.

Broad applications of PEI

On the basis of the physicochemical properties, PEI is widely used in broad fields, including effluent treatments, carbon dioxide absorption, separation and purification of proteins, antibacterial operations and other procedures.Citation8Citation12 In 1995, Boussif et al prepared a groundbreaking type of PEI/DNA NPs and successfully transferred DNA into nerve stem cells.Citation2 Subsequently, PEI has drawn more attention in biomedical field, especially as drug carriers,Citation13Citation15 biological labelsCitation16,Citation17 and vaccine adjuvants ().Citation18

Table 1 Broad applications of PEI

Among all the delivered drugs, the most common ones are nucleic acids. PEI is the second acceptable nonviral nucleic acid transfer agent, besides poly-L-lysine (PLL).Citation2 Nowadays, PEI transfection reagents are commercially available, which include ExGen500®, jetPEI® and PEIpro™.Citation19 Besides nucleic acids, proteins or peptides are also included in the category.Citation20,Citation21 PEI also works as a biological label when conjugated with imaging agents (such as the Fe3O4 and fluorescent NPs), and reflects the specific cell or sub-cell information, such as migrations and other behaviors, under certain conditions.Citation16,Citation22,Citation23 The adjuvant effect of PEI is an emerging area. In a series of recent studies, vaccines composed of PEI as the immunostimulants are quite competent in treating infections or tumors.Citation24,Citation25

Intracellular process of PEI/modified PEI as vaccine adjuvants

To better manipulate its adjuvanticity, strategies to construct the PEI-based vaccines are highly desirable. PEI could be incorporated into vaccine structures through different ways: directly binding with antigens, coating on antigen-loaded NPs/MPs, coating existing particles with antigens absorbed on the surface or co-encapsulated NPs/MPs with antigens and other constructed forms (). During these processes, plenty of cytokines and ligands could also be added in the vaccines based on the desirable applications.

Figure 2 Schematic illustration of PEI-based NPs/MPs. (A) Electrostatic nanosized complexes of cationic PEI and anionic peptides or DNA. (B) PEI-coated MPs encapsulated with peptides or DNA. (C) PEI-coated MPs peptides or DNA adsorbed on the surface by static electricity. (D) PEI MPs encapsulated with DNA or peptides.

Abbreviations: PEI, polyethylenimine; NPs, nanoparticles; MPs, microparticles.

Figure 2 Schematic illustration of PEI-based NPs/MPs. (A) Electrostatic nanosized complexes of cationic PEI and anionic peptides or DNA. (B) PEI-coated MPs encapsulated with peptides or DNA. (C) PEI-coated MPs peptides or DNA adsorbed on the surface by static electricity. (D) PEI MPs encapsulated with DNA or peptides.Abbreviations: PEI, polyethylenimine; NPs, nanoparticles; MPs, microparticles.

Cell uptake of PEI-based vaccines

The first step for a vaccine to take its effect is the endocytosis process by APCs. Similar to traditional vaccines, the factors influencing the cell uptake of PEI-based vaccines include size,Citation33 shape,Citation34 chargeCitation35 and surface chemistry of the particles.Citation36,Citation37

General factors influencing PEI-based polyplex uptake

With broad range of molecular weights, different topologies and plentiful modification possibilities, PEI-based polyplexes are rather versatile in size, shape and properties.Citation5 Therefore, PEI-based vaccines demonstrate broad possibilities in applications.

Also, the strong positive charge in PEI is beneficial for cellular internalization. PEI can bind to the negative proteoglycan on cell membranes and mediate the uptake process through the electrostatic interaction.Citation33,Citation35 Generally, phagocytosis by immune cells of foreign particles is an actin-dependent process.Citation34 The loosely organized PEI in neat state will disturb the actin remodeling when initiating the internalization. Yet, PEI compresses negative substances to condense small particles, or PEI-coating strategy will result in a smooth surface.Citation25,Citation38 These spherical particles are symmetrical and easily uptaken from any point of attachment.Citation34

Size is one of the useful factors in the toolbox affecting the uptake of PEI-based particles, as well as other polymeric particulate carriers. Champion et al demonstrated that particle size played a role in phagocytosis only if the volume was larger than the cell size.Citation34,Citation39 It is relatively easy to achieve small-sized PEI-based polymers. For the PEI-coated large MPs, the uptake process is restricted; however, the electrostatic forces attach the huge particles to the cell membranes, and perform as a continuous depot of the antigens.Citation40,Citation41

Effect of surface properties of PEI-based polyplex on cellular uptake

In addition to the general parameters, significant attention has been paid to the chemical modifications of the surface chemistry.Citation34 Some modifications would largely influence the cellular uptake of the PEI-based polyplexes.

In APCs, phagocytosis is frequently mediated by receptors (via mannose receptor-, complement receptor-, Fcγ receptor- and scavenger receptor-mediated pathways or other pathways).Citation34 Targeted modifications can obviously improve the cellular uptake efficiency and the subsequent biological effects of the polyplexes.Citation36,Citation37,Citation42 For MPs or less positively charged particles, the tethering of targeting moieties on the particles’ surface is even more important for endocytosis.Citation43,Citation44 Hu et al prepared mannose-modified PEI-cell-penetrating peptide (CPP)/DNA particles and found an improved DC2.4-consuming efficiency.Citation45 It is worth noting that Toll-like receptors (TLRs) are not included in the phagocytosis-related receptors, but they initiate the phagosome-mediated APCs maturation and inflammatory secretion,Citation46 which is discussed in this review. Cho et al reported that the maltosylated PEI-mediated vaccines for cervical cancers were more effective than neat human papillomavirus antigens. Presumably, receptor-mediated endocytosis of PEI-based particles by maltose subsequently enhanced the transfection efficiency.Citation18 For PEI-coated NPs/MPs, similar patterns of receptor-mediated gene delivery have also been demonstrated. Mesoporous silica NPs coupled with PEI also demonstrated high transfection efficiency due to the more effective targeting into APCs.Citation47

Besides the specific receptor-mediated targeting strategy, another nonspecific targeting method is the hydrophobic modification of PEI. Given the lipophilic nature of cell membranes, the lipid-like structure facilitates the interactions between the hydrophobic species and plasma membranes (eg, lysosomal membranes and nuclear membranes).Citation7 It is demonstrated that 76% and 96% of acetylated branched PEI (BPEI) enhanced the cellular uptake ability by four-fold, corresponding to the hydrophobic interaction theory.Citation48,Citation49 When the lipid components were incorporated, though possessing a relatively decreased positive charge, the electrostatic interaction could still be maintained and aid in binding.Citation48,Citation49 Recently, Parhiz et al reported that hexanoated-PEI vector was also more effective than the corresponding non-hydrophobic PEI in enhancing transfection rates.Citation19 Besides the transfection benefits, Wang et al proved that hydrophobic modification of alkyl chains to PEI vaccines optimized the cross-presentation of antigens and upregulated IL-2 secretion. Yet, correlation between the properties of the synthesized hydrophobic-modified PEI and the vaccine potency still needs further investigation.Citation50

CPPs are another valuable species that can optimize the interactions between cell membranes and the PEI-based polyplex. CPPs, characterized by the polycationic amino acid residues (ie, arginine and lysine), have been known for their specialized ability to penetrate the cellular membranes. Until now, most studies demonstrated that CPPs take effect by electrostatic binding to negatively charged cell membranes and induction of vesicle rupture by osmotic change and/or membrane lysis.Citation19 Truncated Tat peptide and penetratin are the most popular CPPs, and both are arginine-rich peptides.Citation19 Recently, Morris and Sharma constructed arginine-modified oligo-(alkylaminosiloxanes)-grafted PEI and found that the modified PEI/pDNA exhibited 98% cell viability and 150% more gene transfection efficiency than neat BPEI in human nasopharyngeal epidermoid carcinoma.Citation51 Inhibitor studies, by use of various cellular uptake inhibitors such as wortmannin and genistein, indicated the role of arginine moiety in promoting internalization of the polyplex, and the process possibly contained a combination of multiple pathways.Citation51

More modification strategies will be developed to magnify the advantages of PEI-based vaccines in the initiation process. Yet, after certain modifications, the particle size, charge and other parameters of the polymers may vary correspondingly. Though each of these areas can be studied separately, the interplay among these parameters must be considered as a whole to identify the best performance in boosting the uptake.Citation34,Citation52

Antigen presentation of PEI-based vaccines

After being uptaken, antigens are processed and transported through different cell compartments and are finally presented within the peptide-binding groove of a major histocompatibility complex (MHC) molecule (MHC class I molecules or class II molecules). T cell receptors can only recognize antigens presented in this pattern. Mostly, MHC class I molecules present endogenous peptides (such as transformed or infected cell components) and elicit the cell-mediated immunity (CMI), while class II molecules acquire exogenous peptides (extracellular pathogens or vaccine peptides) and induce abundance of antibodies.Citation53 PEI-based vaccines with antigen peptides mainly follow the exogenous/class II pathway and stimulate the humoral immunity.Citation54 PEI-based DNA vaccines provide the opportunities to synthesize multiple antigens in APCs and stimulate effective cellular immunity and long-lasting memory immunity. However, most DNA is actually delivered to bystander cells (such as myocytes and fibroblasts). In this case, newly synthesized antigen peptides are presented on or secreted out of bystander cells. They are shuttled to APCs, acting as the exogenous antigens.Citation55,Citation56

The “proton sponge effect” of PEI makes cross-presentation possible

As mentioned above, MHC class I molecules present endogenous peptides synthesized in the cells, while class II molecules present the exogenous internalized antigens. Occasionally, MHC class I molecules also participate in presenting exogenous antigens, known as the cross-presentation, which is very meaningful in biological evolution. In human beings, since the virus invasion does not commonly occur in professional APCs and the CMI is not naturally generated,Citation57 virus antigens released from the bystander cells turn into exogenous antigens for APCs. The cross-presentation mechanism in APCs largely increases the efficiency of the immune system to generate CMI and eliminate all the infected cells. Yet, in application of vaccines for treating cancers and infections, which needs the CMI instead of mere humoral immunity, the cross-presentation is also of great importance.Citation57 In the processes, phagocytosed or endocytosed antigens escape from the vacuole and gain entry to the cytosol (known as the “lysosomal escape”). Then, they become qualified clients for ubiquitination and subsequent degradation by the proteasome, followed by the transporter associated with antigen processing (TAP)-mediated transfer into the endoplasmic reticulum, and presentation by MHC class I molecules.Citation57Citation59

There are two representative mechanisms of the “lysosomal escape”, that are, the variation of osmotic pressure and membrane lytic activity.Citation19 Some pH-sensitive biomaterials possess the membrane-destabilizing property; thus, they are good candidates.Citation60,Citation61 PEI has the unique “proton sponge effect”, which buffers under acidic conditions.Citation2,Citation62 When involved in acidic conditions, the ATP-mediated pH- dependent proton pumps open, followed by passive influx of chloride ions and water molecules resulting in hyperosmolar state instantaneously, causing the vesicles to burst.Citation2,Citation62 The “lysosomal escape” process of PEI-based polyplex was well recorded by Merdan et al ().Citation6,Citation7 PEI/ribozyme and PLL/ribozyme migrations in the living cells were identified under confocal laser scanning microscope, and they were found to first gather in acidic vesicles, most probably lysosomes. Unlike the PLL-based complex, the vesicles containing the PEI-based complex met with a sudden burst while letting out the contents throughout the cytoplasm.Citation6,Citation7 To verify the function of the pH change in the “lysosomal escape” process of PEI, they prepared the PEI/ribozyme group with bafilomycin A, a selective inhibitor of endosomal/lysosomal acidification. It showed no lysosomal rupture, suggesting the major role of acidification in the procedure.Citation6,Citation7 The escaped antigens (peptides or DNA) in the PEI-based vaccines in the cytoplasm are then prepared to undergo a cross-presentation process. The process has recently been verified again by Song et al. They found that PEI-coated poly(lactide-co-glycolide) (PLGA) (OVA) NPs induced efficient cross-presentation of antigens on MHC class I molecules through the endosome escape and lysosomal processing.Citation63

Figure 3 (A) Living cell microscopic visualization of the “proton sponge effect” of polyethylenimine (PEI). PEI is green, while ribozyme is red in this observation. Before burst, the vesicle has a yellow core and green PEI corona, while afterwards, the faint green-yellow fluorescence can be seen evenly distributed throughout the entire cell. The remnant of the vesicles is significantly smaller and deeper red. Images were recorded 28, 31, 37, and 40 min after incubation. Yellow arrows indicate a single vesicle undergoing the process of swelling and bursting. The scale bar in the upper-left image is 10 μm and can be applied in the other images. (B) The same confocal layer of 1 nm thickness before and after lysosomal burst (37 and 40 min after incubation). Clearly, fluorescence intensity increases throughout the whole cell after burst, whereas there seems to be an area where it is slightly weaker which might be the nucleus. The right image is an optical microscopy image showing the positions and numbers of the cells. Reproduced from Pharm Res, Intracellular pro cessing of poly(ethylene imine)/ribozyme complexes can be observed in living cells by using confocal laser scanning microscopy and inhibitor experiments. , 2002;19(2):140–146, Merdan T, Kunath K, Fischer D, Kopecek J, Kissel T, (copyright 2002 Springer). With permission of Springer.Citation6

Figure 3 (A) Living cell microscopic visualization of the “proton sponge effect” of polyethylenimine (PEI). PEI is green, while ribozyme is red in this observation. Before burst, the vesicle has a yellow core and green PEI corona, while afterwards, the faint green-yellow fluorescence can be seen evenly distributed throughout the entire cell. The remnant of the vesicles is significantly smaller and deeper red. Images were recorded 28, 31, 37, and 40 min after incubation. Yellow arrows indicate a single vesicle undergoing the process of swelling and bursting. The scale bar in the upper-left image is 10 μm and can be applied in the other images. (B) The same confocal layer of 1 nm thickness before and after lysosomal burst (37 and 40 min after incubation). Clearly, fluorescence intensity increases throughout the whole cell after burst, whereas there seems to be an area where it is slightly weaker which might be the nucleus. The right image is an optical microscopy image showing the positions and numbers of the cells. Reproduced from Pharm Res, Intracellular pro cessing of poly(ethylene imine)/ribozyme complexes can be observed in living cells by using confocal laser scanning microscopy and inhibitor experiments. , 2002;19(2):140–146, Merdan T, Kunath K, Fischer D, Kopecek J, Kissel T, (copyright 2002 Springer). With permission of Springer.Citation6

Effect of surface properties of PEI-based polyplex on antigen presentation

There is a group of membrane-destabilizing peptides, which have shown the possibility of not only promoting cell uptake but also improving “lysosomal escape” and even nuclear translocation.Citation64,Citation65 Although PEI has the proton sponge effect to carry out the “lysosomal escape” itself, sometimes it is incompetent, and addition of membrane-destabilizing peptides (such as CPPs) in the vaccines can improve the cross-presentation effect. Ogris et al have explained the conditions in their study.Citation52 The transfection efficiency was 10-fold (in B16F10 cells) to more than 100-fold (in Neuro2A cells, K562 cells) lower in small PEI/DNA particles compared with the large ones. Introduction of the lysosomotropic drug chloroquine or the pH-specific, membrane-active peptide (INF5) really promoted a substantial increase in antigen genes expression, which confirmed the hypothesis and importance of adding CPP moieties.Citation52 Tan et al bounded two truncated peptides with penetrating properties to BPEI, resulting in higher transfection efficiency without causing cytotoxicity in CHO-K1, B16F10 and 293FT cell lines.Citation66 These results can be ascribed to the enhanced endosomal disrupting activity of CPP-bound PEI carriers than to their parent carriers.

In PEI/DNA vaccines, PEI helps with the nucleic acids protection and endosomal escape. Yet, the transfection efficiency may still be limited in these vaccines.Citation62 Particles of over 30 nm diameter or over 40 kDa molecular weight will require the active aid of the coupled nuclear localization signal (NLS) in nuclear translocation.Citation60 NLS peptides are usually short conservative sequences, which bind to cytoplasmic importins and dock to the nuclear pore complex, thus aiding the nucleic acids transcription. The most commonly used NLS contains the PKKKRKV sequence.Citation67 In addition, the well-known arginine-rich peptides, such as Tat and penetratin, also possess such nuclear transport activities.Citation67 Besides, many other viral-origin peptides and even histone H1, protamine, ribonucleoprotein A1, high-motility-group proteins and others containing the above polycationic amino acids also act as effective NLS.Citation60 Parhiz et al investigated the efficiency of PEI/DNA vaccines by attaching different arginine-rich sequences. It was found that the arginine-rich derivatives of PEI induced higher DNA and siRNA transfection efficiency than the groups without modifications. Moreover, PEI/DNA vaccines conjugated with arginine-rich peptides and hydrophobic derivatives demonstrated the highest DNA transfection efficiency, indicating that rational designs of two or more modifications are promising, to get a better transfection efficiency (antigen DNA expression) and lower cytotoxicity.Citation68 All the efforts to elevate DNA transfection promise antigenic DNA translation and presentation. The advantages and modification strategies of PEI-based vaccines related to antigen uptake and presentation are illustrated in .

Figure 4 Illustration showing the uptake and presentation processes of PEI-based vaccines and the modified forms. Positive charge, round shape and controllable size of PEI-based MPs and NPs are all benefits contributing to the uptake process. Besides, they could be modified with PRR ligands, hydrophobic moieties or CPPs, which facilitate the polyplex internalization by receptor-mediated routes, membrane fusions, penetration process and other unknown routes. After polyplex degradation in the endosomes and lysosomes, the released antigens are presented onto MHC molecules directly by transmission or indirectly by gene expression process. PEI itself has the “lysosomal escape” feature, while vaccines with CPPs-modified PEI possess more potent “lysosomal escape” property. They assist with the cross-presentation of delivered antigens. NLS is the specialized sequence added with nucleic acids to assist nuclear translocation of the polymers.

Abbreviations: PEI, polyethylenimine; MPs, microparticles; NPs, nanoparticles; PRR, pattern recognition receptor; CPPs, cell-penetrating peptides; MHC, major histo-compatibility complex; NLS, nuclear localization signal; iDC, immature dendritic cells; mDC, mature dendritic cells.

Figure 4 Illustration showing the uptake and presentation processes of PEI-based vaccines and the modified forms. Positive charge, round shape and controllable size of PEI-based MPs and NPs are all benefits contributing to the uptake process. Besides, they could be modified with PRR ligands, hydrophobic moieties or CPPs, which facilitate the polyplex internalization by receptor-mediated routes, membrane fusions, penetration process and other unknown routes. After polyplex degradation in the endosomes and lysosomes, the released antigens are presented onto MHC molecules directly by transmission or indirectly by gene expression process. PEI itself has the “lysosomal escape” feature, while vaccines with CPPs-modified PEI possess more potent “lysosomal escape” property. They assist with the cross-presentation of delivered antigens. NLS is the specialized sequence added with nucleic acids to assist nuclear translocation of the polymers.Abbreviations: PEI, polyethylenimine; MPs, microparticles; NPs, nanoparticles; PRR, pattern recognition receptor; CPPs, cell-penetrating peptides; MHC, major histo-compatibility complex; NLS, nuclear localization signal; iDC, immature dendritic cells; mDC, mature dendritic cells.

In vivo and clinical applications of PEI and modifiers as vaccine adjuvants

To assess the performance of the PEI-based vaccines, we must consider the following three levels: in vitro, in vivo and clinical trials. Clearly, the clinical trials of large samples are the most convincing data for evaluating the efficiency of newly developed vaccines. Although we have got much more data from animal trials supporting the superior effects of PEI-based vaccines, only a few trials of PEI-based vaccines have been processed into clinical level. We summarize the application of PEI-based vaccines in animal experiments against infections and cancers, in and , respectively.

Table 2 The infection applications of PEI-based vaccines in vivo

Table 3 The anticancer applications of PEI-based vaccines in vivo

In vivo applications of vaccines

The in vivo applications of vaccines are confronted with bottlenecks sometimes, for two main reasons. One is the safety issue, as PEI is widely known for its toxicity. The other is the complicated microenvironment in vivo.Citation61,Citation69 In the microenvironment, every immune-active material has its corresponding targeting immune cells. These cells secrete various chemokines, attracting monocytes, granulocytes and other immune-promoting cells, while the attracted cells secrete more cytokines to form a positive feedback. As frequently depicted for many traditional adjuvants, the monocytes ultimately uptake the complexes, differentiate into dendritic cells (DCs) or phagocytes and then migrate to secondary lymphoid tissues.Citation70,Citation71 Intriguingly in PEI, the local inflammation seems beneficial to the recruitment and activation of APCs.Citation72

Though the general process is somewhat clear, the detailed signal communications of one specific vaccine in in vivo applications are still puzzling. For example, it was found that intramuscularly injected MF59 and aluminum adjuvants targeted different cell types, while all the recruited cells had similar potential to engulf carrier–antigen complexes. All events occurred in draining lymph nodes (DLNs) at certain time intervals, probably due to a bystander effect.Citation70,Citation71 Further research indicated that, different from lipopolysaccharide (LPS), MF59 and aluminum took a TLR-independent mechanism to activate immunization and bias the monocytes differentiation to DCs rather than macrophages.Citation70,Citation71 Yet, as for the PEI-based vaccines, several issues, such as the local activation of cells and the roles of PEI-based vaccines in it, the secretion of cytokines and the uptake and differentiation processes, along with the lymph drainage situations, need further investigation.

Progress of in vivo applications of PEI-based vaccines

Different types of PEI, antigens, constructed strategies and immune routes are chosen in constructing the PEI-based vaccines in vivo. Frequently researched diseases include AIDS,Citation24,Citation73Citation76 Listeria monocytogenes infection,Citation56,Citation77 respiratory diseases,Citation54,Citation78Citation80 B cell lymphoma,Citation38 herpes simplex virus attacks,Citation81 viral B hepatitis,Citation82 renal cell carcinoma,Citation83 melanomaCitation84 and others.Citation85 The PEI-based vaccines work through almost all the known immune routes, and usually mimic the pathogenesis of the diseases themselves. For example, in respiratory infection, intranasal administration was prone to elicit mucosal secretory antibodies, as well as more serum antibodies.Citation54 A similar trend was found for the HIV vaccines which elicited more antibodies in the vagina.Citation74

The detected markers prove the efficacy of these vaccines usually through two main aspects: (1) intermediate indicators, such as antigen DNA expression, APCs maturation rates, humoral and cellular immunity responses in serum, lymphoid organs or tissues, the T helper type 1 (Th1)/T helper type 2 (Th2) response bias, the memory T and B cells production and secretion levels of the related cytokines;Citation56,Citation76,Citation86 (2) comprehensive effects, such as higher animal survival rates, less weight loss and less tumor metastasis rates.Citation38,Citation54,Citation77 The vaccines have demonstrated positive results in a series of in vivo experiments, while only a few clinical trials have been performed.Citation40 The only satisfactory example was the DermaVir (mannosylated-PEI/plasmid Simian HIV DNA). Lisziewicz et al performed the ex vivo DC-based vaccination of DermaVir and found that DermaVir-transduced autolo-gous monocyte-derived DCs induced HIV-specific T cells in rhesus macaques.Citation87 Moreover, the same group improved the function of the ex vivo DC vaccination strategy with topical administration. Compared with the previous ex vivo DC-based vaccinations, the vaccination strategy proposed by Lisziewicz et al resulted in a similar number of transduced DCs in the lymph node and induced a similar quantity and quality of HIV-specific Th1-type T cell responses. The DermaVir Patch has been tested in Phase I/II clinical trials at present, and is the new promising agent in clinical applications.Citation73

Intrinsic immune-activating function of PEI

Though many studies showed the performance of PEI-based vaccines in vitro and in vivo, the immune-activating properties of PEI still need to be revisited. It is not clear from these studies whether the improved immune efficiency was solely due to antigen protection (vaccine delivery vehicle) or due to the intrinsic adjuvant property (immunostimulator) of PEI.Citation25,Citation56,Citation78,Citation95 Moreover, in the case of PEI-coated polyplex, PEI together with the NPs/MPs should be regarded as a whole to take effects. When encapsulated inside, it was even more difficult to prove the adjuvanticity of PEI. Thus, the observed immunoactivation effects of PEI need further confirmation, and the revelation of the possible molecular pathways of the PEI-based vaccines would help us in understanding and developing new PEI-based vaccines.

Notably, some biomaterials have been regarded as well-known immunostimulants. Such immunologically active materials have been continuously discovered, and the underlying mechanisms were partially explained.Citation35,Citation96 Among them, the clinically approved adjuvants (aluminum and MF59) are good examples.Citation97 Besides, 2,000-kDa poly(γ-glutamic acid) (γ-PGA) NPs were proved to activate DCs through TLR4 pathway, similar to γ-PGA-Phenol NPs.Citation98Citation100 The activation by polyanhydride NPs and fullerene NPs was mediated by multiple TLRs on DCs.Citation101Citation104 Inspiring in vitro and in vivo results have also suggested the immunoactivating functions of PEI, especially in DNA vaccines. With the understanding of the above-stated characteristics of PEI and PEI-based polyplex, it is necessary to provide an overview of the adjuvants’ properties and their underlying mechanisms.

Effect of immune activation of PEI

A well-established mode of an adjuvant taking effects is the direct activation of APCs, resulting in costimulatory molecules expression and cytokines secretion in vitro.Citation63 Compared to bare Au nanorods (AuNRs), PEI-coated AuNRs induced a higher level of phenotypic maturation of DCs () and cytokines release. The immune-promoting effects almost matched LPS to some extent, while the subsequent activation of specific immunization and the interferon (IFN)-γ production were strictly Env-dependent.Citation105 Similarly, when conjugating hepatitis B surface antigen (HBsAg), cationic chitosan and PEI-coated poly(lactic acid) (PLA) MPs also had a stronger capability than PLA MPs or bare antigens to promote macrophages internalization and maturation, followed by stronger humoral immune and Th1 responses in vivo.Citation82 The anti-B cell lymphoma vaccines made of 70-kDa BPEI and 25-kDa LPEI-functionalized PLGA MPs also resulted in higher CD80 and MHC II expression in RAW264.7 cells in vitro than bare MPs.Citation38 Moreover, the immunoactivating effects of PEI should also be assessed to understand if better in vivo effect can be achieved without adding PEI in polyplex compared to control groups.

Figure 5 Significant increase in the percentage of mature DCs (CD11c+ MHCII+CD80+CD86+DCs) when DCs were treated with PEI-coated AuNRs polyplex. *P<0.05. Reprinted with permission from Xu L, Liu Y, Chen Z, et al. Surface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. Nano Lett. 2012;12(4):2003–2012. Copyright 2012 American Chemical Society.Citation105

bbreviations: DCs, dendritic cells; PEI, polyethylenimine; AuNRs, Au nanorods; CTAB, cetyl trimenthyl ammonium bromide; PDDAC, poly(diallydimethyl ammonium chloride); LPS, lipopolysaccharide.

Figure 5 Significant increase in the percentage of mature DCs (CD11c+ MHCII+CD80+CD86+DCs) when DCs were treated with PEI-coated AuNRs polyplex. *P<0.05. Reprinted with permission from Xu L, Liu Y, Chen Z, et al. Surface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. Nano Lett. 2012;12(4):2003–2012. Copyright 2012 American Chemical Society.Citation105bbreviations: DCs, dendritic cells; PEI, polyethylenimine; AuNRs, Au nanorods; CTAB, cetyl trimenthyl ammonium bromide; PDDAC, poly(diallydimethyl ammonium chloride); LPS, lipopolysaccharide.

Immune-activating mechanisms of PEI

To reveal the immunoactivating property of PEI, gene expression profiles of in vivo immunization of mice with PEI were investigated.Citation86 Compared with the control group, immuno-related genes of two injected groups were activated at considerably high levels regardless of the antigens (). Based on the results and relations analyzed by PubGene, Regnström et al speculated that PEI has important immune-activating effects, presumably through the granulocyte colony-stimulating factor, and the next step will be the exploration of their interplay.Citation86

Figure 6 Immune-related gene expressions in spleen cells of bare polyethylenimine (PEI)-treated mice as compared to reporter plasmid-conjugated PEI-immunized mice. The mRNA levels are shown as adjusted intensities for each gene. (A) Upregulated markers for the Th1 and Th2 responses. (B) Genes of the adaptive and innate immune responses. (C) Upregulated genes involved in immunogenicity and immunostimulation. Adapted by permission from Macmillan Publishers Ltd: Gene Ther. Regnström K, Ragnarsson EG, Köping-Höggård M, Torstensson E, Nyblom H, Artursson P. PEI – a potent, but not harmless, mucosal immuno-stimulator of mixed T-helper cell response and FasL-mediated cell death in mice. Gene Ther. 2003;10(18):1575–1583. copyright 2003.Citation86

Figure 6 Immune-related gene expressions in spleen cells of bare polyethylenimine (PEI)-treated mice as compared to reporter plasmid-conjugated PEI-immunized mice. The mRNA levels are shown as adjusted intensities for each gene. (A) Upregulated markers for the Th1 and Th2 responses. (B) Genes of the adaptive and innate immune responses. (C) Upregulated genes involved in immunogenicity and immunostimulation. Adapted by permission from Macmillan Publishers Ltd: Gene Ther. Regnström K, Ragnarsson EG, Köping-Höggård M, Torstensson E, Nyblom H, Artursson P. PEI – a potent, but not harmless, mucosal immuno-stimulator of mixed T-helper cell response and FasL-mediated cell death in mice. Gene Ther. 2003;10(18):1575–1583. copyright 2003.Citation86

TLR pathways

With many immune-related genes being upregulated in the treatment of PEI, it is interesting to reveal the precise molecular pathways behind this upregulation. The TLRs and their downstream myeloid differentiation primary response gene 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-β (TRIF) pathways are the most classical immuno-activation routes, which stimulate the nuclear transcription factors, nuclear factor-κB (NF-κB), to activate the expression of a series of cytokines and maturation markers (such as B7 molecules).Citation57 Shokouhi et al studied a series of biomaterials and found that many of them modulated the maturation and cytokine secretion of DCs to different extents in vitro. The TLRs cascade effects may play an important role in sensing the existence of these materials, and the hydrophobic domains may be the “danger signals” which start the APCs activation, which are similar to “pattern antigens”.Citation106 Huang et al and Chen et al found that cationic polymers (such as cationic dextrans and PEI) promoted the maturation of macrophages via TLR4, and secreted type-I cytokines (). Surprisingly, further investigation revealed that PEI reversed the differentiation of tumor-associated macrophages and modulated the activity of natural killers (NKs) to kill the tumor cells.Citation107,Citation108 Similarly, cationic dextran and PEI have also been found to repolarize myeloid-derived suppressor cells (MDSCs) into the antitumor phenotype. Knock-out mice experiments further proved the requirement of TLR4 signaling.Citation109 Other results found that in in vitro investigation, LPEI produced the TLR5-inducible cytokines in wild-type mice instead of TLR5−/− littermates. Therefore, LPEI was considered as a TLR5 agonist, and it is speculated that LPEI structurally resembled the flagellin.Citation110 Ma et al found that the PEI/DNA vaccines were much more effective than naked DNA vaccines in antitumor trial. The co-cultivation of PEI with BMDCs in vitro revealed the activation and expression of important nuclear translocation factors (such as NF-κB p50 and p65), which were important nuclear transcription molecules of TLRs. Therefore, the enhanced type I-mediated cytotoxic T lymphocyte activation, type II-mediated Th1/Th2 response and interferon-γ-activated NKs might relate to the NF-κB-dependent inflammation and apoptosis induced by PEI.Citation94

Figure 7 PEI-induced IL-12 secretion of macrophages was inhibited by TLR4 antibody. In contrast, the addition of isotype control antibody did not inhibit such IL-12 production significantly. Reprinted from Biomaterials, 2010;31(32), Chen H, Li P, Yin Y, et al, The promotion of type 1 T helper cell responses to cationic polymers in vivo via toll-like receptor-4 mediated IL-12 secretion. 8172–8180, Copyright (2010), with permission from Elsevier.Citation108

Abbreviations: PEI, polyethylenimine; IL-12, interleukin-12; TLR4, Toll-like receptor 4.

Figure 7 PEI-induced IL-12 secretion of macrophages was inhibited by TLR4 antibody. In contrast, the addition of isotype control antibody did not inhibit such IL-12 production significantly. Reprinted from Biomaterials, 2010;31(32), Chen H, Li P, Yin Y, et al, The promotion of type 1 T helper cell responses to cationic polymers in vivo via toll-like receptor-4 mediated IL-12 secretion. 8172–8180, Copyright (2010), with permission from Elsevier.Citation108Abbreviations: PEI, polyethylenimine; IL-12, interleukin-12; TLR4, Toll-like receptor 4.

NLR family pyrin domain-containing 3 (NLRP3) inflammasome pathways

Wegmann et al also supported the adjuvanticity of PEI, which promoted DCs trafficking to DLNs and led to cytokines secretion.Citation25 They proved the immunoactivation mechanisms of PEI related to the interferon regulatory factor 3 (IRF3)-dependent signaling and NLRP3 inflammasome activation, especially the in vivo process.Citation25 The NLRP3 inflammasome is among the nod-like receptors (NLRs), a kind of pattern recognition receptors found on APCs. The NLRP3 inflammasome was known to be activated by various stresses, including K+ efflux, reactive oxygen species (ROS) generation and lysosome rupture.Citation111 Thus, the NLRP3 inflammasome activation by PEI-antigen NPs in the experiment is potentially through the lysosomal-destabilizing activity or other damage-associated molecular patterns (DAMPs) caused by the toxicity of PEI. Interestingly, NLRP3 inflammasome function only biased adaptive immunity toward a Th2 response, instead of affecting the overall PEI adjuvant activity.Citation25

ROS

ROS generation is always related to tissue or cell damage, which is a kind of “danger signal”. The ROS and innate-mediated pro-inflammatory cytokines (such as the tumor necrosis factor-α and interleukin-1β) were signals in arming the adaptive immunity, sufficiently demonstrating the adjuvant effects of PEI.Citation112 Mulens-Arias coated superparamagnetic iron oxide NPs (SPIONs) with PEI and used them to stimulate macrophages. They found that the TLR4 and ROS signaling were involved. Intriguingly, though PEI-coated SPIONs and LPS both induced macrophages activation and M1 phenotype genes upregulation, the gene expression profiles were different.Citation113 It was also reported that the OVA-loaded PLGA MPs with a PEI-DS polyelectrolyte multilayer induced ROS in attachment with APCs. More positive charges on the outermost layer and higher molecular weight resulted in higher ROS production.Citation114 Many nonpathogenic adjuvants, including PEI, are toxic to tissues, under which circumstances ROS is generated. Although ROS are of extensive interest in the exploration of PEI-based vaccine mechanisms, the specific role and relationship with other immunity-related molecules still merit further investigations.

Other factors in the “danger signals”

The “danger signals” elicited from PEI seem to be the main cause of its adjuvant effects. Regnström et al explored the gene expression profiles of PEI/DNA bronchial vaccines and found that bare PEI upregulated the expressions of genes involved in cell cycle regulation, oncogenesis and differentiation. These findings indicated the cytotoxicity and risks involved with PEI-based vaccines.Citation86 The hypothesis went along with the previous explanations of aluminum adjuvants and that the immunoactivation potency may be caused by the physiological reaction toward the sensed danger signals.Citation115 Meanwhile, the toxicity from adjuvants would affect the cell states to exhibit various activities, which will impair the outcome of overall effects of PEI. Recently, Palumbo et al constructed an in vitro co-culture system and demonstrated the feasibility to transfect the PEI/DNA vaccine via fibroblasts. The vaccination surprisingly resulted in the cross-presentation of antigens and DC maturation. By comparing the high- and less-toxic PEI/DNA compositions and corresponding DC maturation states, as well as MHC I-restricted OVA presentation, the results highlighted that polymer-induced cytotoxicity probably benefited the immune activation.Citation116 Many endogenous antigens from the apoptotic or necrotic cells (such as the dsDNA, HMGB1, heat shock proteins and uric acids) have long been known to recruit and activate immune systems.Citation116 In Wegmann et al’s report, they proved the strong dependence of double-stranded DNA (dsDNA)-mediated IRF3-triggered adjuvant effect of PEI. The free cellular dsDNA was released from the apoptotic or dead cells.Citation25 It is a good representative of the DAMPs signal to investigate an immune response.

Summary of the immunoactivation mechanisms of PEI

Generally, PEI or PEI-based particles can stimulate the immune system, accompanied by the induction of various sorts of cell stress and immune response-related transcription factors.Citation117 Yet, it is notoriously difficult to elucidate the mechanisms clearly. The evidence for adjuvant effect of PEI or PEI-based particles in different research is generalized and drawn out by our own understanding (). Most current evidence supports the “danger signals” or “damage-associated molecular patterns” theory. Both TLRs and NLRs are receptors in APCs that recognize the danger signals. PEI-associated cytotoxicity may be one manifested pattern of such danger signals.Citation24,Citation25 Some nontoxic biomaterials also have profound effects on immune cells.Citation106 In some experiments, PEI was also found to be nontoxic in vivo, but still possessed the adjuvanticity.Citation25 Thus, other mechanisms may exist to stimulate immunity in addition to the direct toxicity of PEI. Some products reflecting the danger or damage signals have been tested continuously, such as the ROS and the uric acids, while the roles involved have not been clarified.Citation97,Citation116,Citation118 Moreover, all the above-explained factors are actually not independent but work as a network that affects mutually. For example, the activation of NLRP3 inflammasome may be caused by the uric acids, which formed under necrosis or ROS.Citation25,Citation97 Besides, ROS can be induced by the primary generation, or the product from cell stress, while NF-κB is the key suppressor of ROS-induced apoptosis.Citation117 Other intermediates (such as the dectin-1) intermediate bioactive cytokines and chemokines (such as the CCL family), and accessory immune cells (such as neutrophils and invariant NK T cells) are also involved in the network.Citation119

Figure 8 Intrinsic immunoactivation properties of PEI-based vaccines. PEI-based particles bind the PRRs and trigger the NF-κB and IRF3 factors to elicit a series of APCs maturation manifestation. They are also observed to elicit ROS in APCs, generating the “danger signal” by various routes. Besides, PEI-based particles cause the bystander cells death, and release a series of products, including uric acid, released DNA, HMGB1 and others. These materials activate IRF3, inflammasome and other pathways to generate the “danger signals” and APCs maturation.

Abbreviations: PEI, polyethylenimine; PRRs, pattern recognition receptors; NF-κB, nuclear factor-κB; IRF3, interferon regulatory factor 3; APCs, antigen-presenting cells; ROS, reactive oxygen species; MPs, microparticles; NPs, nanoparticles; TLRs, Toll-like receptors; MRs, mannose receptors; TRIF, TIR-domain-containing adapter-inducing interferon-β; MyD88, myeloid differentiation primary response gene 88; MHC, major histocompatibility complex; IL, interleukin; IFN, interferon.

Figure 8 Intrinsic immunoactivation properties of PEI-based vaccines. PEI-based particles bind the PRRs and trigger the NF-κB and IRF3 factors to elicit a series of APCs maturation manifestation. They are also observed to elicit ROS in APCs, generating the “danger signal” by various routes. Besides, PEI-based particles cause the bystander cells death, and release a series of products, including uric acid, released DNA, HMGB1 and others. These materials activate IRF3, inflammasome and other pathways to generate the “danger signals” and APCs maturation.Abbreviations: PEI, polyethylenimine; PRRs, pattern recognition receptors; NF-κB, nuclear factor-κB; IRF3, interferon regulatory factor 3; APCs, antigen-presenting cells; ROS, reactive oxygen species; MPs, microparticles; NPs, nanoparticles; TLRs, Toll-like receptors; MRs, mannose receptors; TRIF, TIR-domain-containing adapter-inducing interferon-β; MyD88, myeloid differentiation primary response gene 88; MHC, major histocompatibility complex; IL, interleukin; IFN, interferon.

Conclusion and future perspectives

Although adjuvants are capable of increasing the efficacy of clinical vaccines and have been extensively studied, the pace of their development is relatively slow. Till now, only five adjuvants have been licensed for use in clinical applications by the US Food and Drug Administration and European Medicines Agency, including aluminum (1926), MF59 (1997), virosome (2000), AS04 (2005) and AS03 (2009). Among them, even aluminum, the most widely used adjuvant in vaccination for almost one century with many advantages such as good safety profile, antigen stabilization and augmentation of high-titer antibody production, does not have the ability to elicit CMI. Furthermore, only a few adjuvants have been licensed for cancer immunotherapy, indicating a need for novel adjuvants that can induce robust antitumor CMI. Therefore, it is desirable to develop new immunostimulant adjuvants in vaccine formulations to induce robust immune responses including both humoral immunity and CMI.

Interestingly, PEI confers the ability to induce the “danger signals” of immune cells and the production of pro-inflammatory cytokines, which forms the basis of its adjuvanticity. Given limited research and publications related to the effect of PEI on immunity, an in-depth understanding of its immune mechanism of action, especially the relationship with the cytotoxicity, is of great significance. Moreover, the biodistribution and biodegradation of PEI-based vaccines in vivo, with different sizes and shapes, different administration routes and different modification strategies, still need further investigation.Citation85,Citation120,Citation121 With breakthrough advances in multidisciplinary fields including chemistry, materials science, immunology and nanotechnology, it is believed that PEI-based vaccines, as well as other new adjuvants that improve the potency of the vaccine with precise modulations, will appear on the list of licensed adjuvants in the near future.

Acknowledgments

The authors gratefully acknowledge funding for this work provided by the National Natural Science Foundation of China (81573047 and 81502367) and Clinical Research Physician Program of Tongji Medical College, HUST (5001530014).

Disclosure

The authors report no conflicts of interest in this work.

References

  • NeuMFischerDKisselTRecent advances in rational gene transfer vector design based on poly(ethylene imine) and its derivativesJ Gene Med200578992100915920783
  • BoussifOLezoualc’hFZantaMAA versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimineProc Natl Acad Sci U S A19959216729773017638184
  • ListnerKBentleyLOkonkowskiJDevelopment of a highly productive and scalable plasmid DNA production platformBiotechnol Prog20062251335134517022672
  • SteinmanRMBanchereauJTaking dendritic cells into medicineNature2007449716141942617898760
  • JägerMSchubertSOchrimenkoSFischerDSchubertUSBranched and linear poly(ethylene imine)-based conjugates: synthetic modification, characterization, and applicationChem Soc Rev201241134755476722648524
  • MerdanTKunathKFischerDKopecekJKisselTIntracellular processing of poly(ethylene imine)/ribozyme complexes can be observed in living cells by using confocal laser scanning microscopy and inhibitor experimentsPharm Res200219214014611883640
  • IncaniVLavasanifarAUludağHLipid and hydrophobic modification of cationic carriers on route to superior gene vectorsSoft Matter201061021242138
  • BoltoBASoluble polymers in water purificationProg Polym Sci1995209871041
  • KumarPKimSIdaJGuliantsVVPolyethyleneimine-modified MCM-48 membranes: effect of water vapor and feedInd Eng Chem Res2008471201208
  • BeythNHouri-HaddadYBaraness-HadarLYudovin-FarberIDombAJWeissEISurface antimicrobial activity and biocompatibility of incorporated polyethylenimine nanoparticlesBiomaterials200829314157416318678404
  • VanchaARGovindarajuSParsaKVJastiMGonzález-GarcíaMBallesteroRPUse of polyethyleneimine polymer in cell culture as attachment factor and lipofection enhancerBMC Biotechnol200442315485583
  • SahinerNSagbasSSahinerMAyyalaRSPolyethyleneimine modified poly(hyaluronic acid) particles with controllable antimicrobial and anticancer effectsCarbohydr Polym2017159293828038751
  • LeeSYHuhMSLeeSStability and cellular uptake of polymerized siRNA (poly-siRNA)/polyethylenimine (PEI) complexes for efficient gene silencingJ Control Release2010141333934619836427
  • Urban-KleinBWerthSAbuharbeidSCzubaykoFAignerARNAi-mediated gene-targeting through systemic application of polyethylenimine (PEI)-complexed siRNA in vivoGene Ther200512546146615616603
  • MoffattSWiehleSCristianoRJTumor-specific gene delivery mediated by a novel peptide-polyethylenimine-DNA polyplex targeting aminopeptidase N/CD13Hum Gene Ther2005161576715703489
  • HayekAErcelenSZhangXConjugation of a new two-photon fluorophore to poly(ethylenimine) for gene delivery imagingBioconjug Chem200718384485117402707
  • ChertokBDavidAEYangVCPolyethyleneimine-modified iron oxide nanoparticles for brain tumor drug delivery using magnetic targeting and intra-carotid administrationBiomaterials201031246317632420494439
  • ChoHJHanSEImSMaltosylated polyethylenimine-based triple nanocomplexes of human papillomavirus 16L1 protein and DNA as a vaccine co-delivery systemBiomaterials201132204621462921440296
  • ParhizHShierWTRamezaniMFrom rationally designed polymeric and peptidic systems to sophisticated gene delivery nano-vectorsInt J Pharm2013457123725924060371
  • MurataHFutamiJKitazoeMIntracellular delivery of glutathi-one S-transferase-fused proteins into mammalian cells by polyethylenimine-glutathione conjugatesJ Biochem2008144444745518603589
  • ZhangSKucharskiCDoschakMRSebaldWUludağHPolyethylenimine-PEG coated albumin nanoparticles for BMP-2 deliveryBiomaterials201031595296319878992
  • WangFChatterjeeDKLiZZhangYFanXWangMSynthesis of polyethylenimine/NaYF4 nanoparticles with upconversion fluorescenceNanotechnology2006172357865791
  • FengMLeeDLiPIntracellular uptake and release of poly (ethyleneimine)-co-poly(methyl methacrylate) nanoparticle/pDNA complexes for gene deliveryInt J Pharm20063111–220921416442245
  • SheppardNCBrinckmannSAGartlanKHPolyethyleneimine is a potent systemic adjuvant for glycoprotein antigensInt Immunol2014261053153824844701
  • WegmannFGartlanKHHarandiAMPolyethyleneimine is a potent mucosal adjuvant for viral glycoprotein antigensNat Biotechnol201230988388822922673
  • DengSTingYPPolyethylenimine-modified fungal biomass as a high-capacity biosorbent for Cr(VI) anions: sorption capacity and uptake mechanismsEnviron Sci Technol200539218490849616294892
  • LiYTianHXiaoCDingJChenXEfficient recovery of precious metal based on Au–S bond and electrostatic interactionGreen Chem2014161248754878
  • XuXSongCAndresenJMMillerBGScaroniAWNovel polyethylenimine-modified mesoporous molecular sieve of MCM-41 type as high-capacity adsorbent for CO2 captureEnergy Fuels200216614631469
  • GreeneGRadhakrishnaHTannenbaumRProtein binding properties of surface-modified porous polyethylene membranesBiomaterials200526305972598215890400
  • ChenFWanDChangZPuHJinMHighly efficient separation, enrichment, and recovery of peptides by silica-supported polyethylenimineLangmuir20143041122501225725262957
  • BeythNYudovin-FarberIBahirRDombAJWeissEIAntibacterial activity of dental composites containing quaternary ammonium polyethylenimine nanoparticles against Streptococcus mutansBiomaterials200627213995400216564083
  • ZhangSDoschakMRUludağHPharmacokinetics and bone formation by BMP-2 entrapped in polyethylenimine-coated albumin nanoparticlesBiomaterials200930285143515519540582
  • XiangSDScholzenAMinigoGPathogen recognition and development of particulate vaccines: does size matter?Methods20064011916997708
  • ChampionJAKatareYKMitragotriSParticle shape: a new design parameter for micro- and nanoscale drug delivery carriersJ Control Release20071211–23917544538
  • DobrovolskaiaMAMcNeilSEImmunological properties of engineered nanomaterialsNat Nanotechnol20072846947818654343
  • PackDWHoffmanASPunSStaytonPSDesign and development of polymers for gene deliveryNat Rev Drug Discov20054758159316052241
  • NishikawaMHuangLNonviral vectors in the new millennium: delivery barriers in gene transferHum Gene Ther200112886187011387052
  • Pai KasturiSQinHThomsonKSProphylactic anti-tumor effects in a B cell lymphoma model with DNA vaccines delivered on polyethylenimine (PEI) functionalized PLGA microparticlesJ Control Release2006113326127016793161
  • ChampionJAMitragotriSRole of target geometry in phagocytosisProc Natl Acad Sci U S A2006103134930493416549762
  • JilekSMerkleHPWalterEDNA-loaded biodegradable microparticles as vaccine delivery systems and their interaction with dendritic cellsAdv Drug Deliver Rev2005573377390
  • PackDWTiming is everythingNat Mater20043313313414991008
  • ReadMLLoganASeymourLWBarriers to gene delivery using synthetic vectorsAdv Genet200553194616240989
  • JoubranSZiglerMPessahNOptimization of liganded polyethylenimine polyethylene glycol vector for nucleic acid deliveryBioconjug Chem20142591644165425121341
  • LiDPingYXuFConstruction of a star-shaped copolymer as a vector for FGF receptor-mediated gene delivery in vitro and in vivoBiomacromolecules20101192221222920704346
  • HuYXuBJiQA mannosylated cell-penetrating peptide-graft-polyethylenimine as a gene delivery vectorBiomaterials201435134236424624529626
  • BlanderJMMedzhitovROn regulation of phagosome maturation and antigen presentationNat Immunol20067101029103516985500
  • ParkIYKimIYYooMKChoiYJChoMHChoCSMannosylated polyethylenimine coupled mesoporous silica nanoparticles for receptor-mediated gene deliveryInt J Pharm20083591–228028718490119
  • ForrestMLMeisterGEKoerberJTPackDWPartial acetylation of polyethylenimine enhances in vitro gene deliveryPharm Res200421236537115032320
  • GabrielsonNPPackDWAcetylation of polyethylenimine enhances gene delivery via weakened polymer/DNA interactionsBiomacromolecules2006782427243516903692
  • WangHChenJYingJXuYShengRHydrophobic chain modified low molecular weight polyethylenimine for efficient antigen deliveryRSC Adv20166171363613643
  • MorrisVBSharmaCPEnhanced in-vitro transfection and biocompatibility of L-arginine modified oligo (-alkylaminosiloxanes)-graft-polyethylenimineBiomaterials201031338759876920727580
  • OgrisMSteinleinPKursaMMechtlerKKircheisRWagnerEThe size of DNA/transferrin-PEI complexes is an important factor for gene expression in cultured cellsGene Ther1998510142514339930349
  • DelvesPJMartinSJBurtonDRRoittIMRoitt’s Essential Immunology12th edChichesterWiley-Blackwell20117980
  • ShimBSParkSMQuanJSIntranasal immunization with plasmid DNA encoding spike protein of SARS-coronavirus/polyethylenimine nanoparticles elicits antigen-specific humoral and cellular immune responsesBMC Immunol2010116521194475
  • SharmaAKKhullerGKDNA vaccines: future strategies and relevance to intracellular pathogensImmunol Cell Biol200179653754611903613
  • GrantEVThomasMFortuneJKlibanovAMLetvinNLEnhancement of plasmid DNA immunogenicity with linear polyethylenimineEur J Immunol201242112937294822886924
  • DelvesPJMartinSJBurtonDRRoittIMRoitt’s Essential Immunology12th edChichesterWiley-Blackwell20111012
  • DelvesPJMartinSJBurtonDRRoittIMRoitt’s Essential Immunology12th edChichesterWiley-Blackwell2011128131
  • JoffreOPSeguraESavinaAAmigorenaSCross-presentation by dendritic cellsNat Rev Immunol201212855756922790179
  • VazquezEFerrer-MirallesNVillaverdeAPeptide-assisted traffic engineering for nonviral gene therapyDrug Discov Today20081323–241067107418801457
  • GharwanHWightmanLKircheisRWagnerEZatloukalKNonviral gene transfer into fetal mouse livers (a comparison between the cationic polymer PEI and naked DNA)Gene Ther200310981081712704421
  • AkincAThomasMKlibanovAMLangerRExploring polyethylenimine-mediated DNA transfection and the proton sponge hypothesisJ Gene Med20057565766315543529
  • SongCNohYWLimYTPolymer nanoparticles for cross-presentation of exogenous antigens and enhanced cytotoxic T-lymphocyte immune responseInt J Nanomedicine2016113753376427540289
  • BolhassaniAGhasemiNServisCTaghikhaniMRafatiSThe efficiency of a novel delivery system (PEI600-Tat) in development of potent DNA vaccine using HPV16 E7 as a model antigenDrug Deliv200916419620419514980
  • ZhangHGersonTVarneyMLSinghRKVinogradovSVMultifunctional peptide-PEG intercalating conjugates: programmatic of gene delivery to the blood-brain barrierPharm Res201027122528254320824308
  • TanYXChenCWangYLTruncated peptides from melittin and its analog with high lytic activity at endosomal pH enhance branched polyethylenimine-mediated gene transfectionJ Gene Med201214424125022328546
  • HeitzFMorrisMCDivitaGTwenty years of cell-penetrating peptides: from molecular mechanisms to therapeuticsBr J Pharmacol2009157219520619309362
  • ParhizHHashemiMHatefiAShierWTAmel FarzadSRamezaniMArginine-rich hydrophobic polyethylenimine: potent agent with simple components for nucleic acid deliveryInt J Biol Macromol201360182723680600
  • KircheisRWightmanLWagnerEDesign and gene delivery activity of modified polyethyleniminesAdv Drug Delivery Rev2001533341358
  • SeubertAMonaciEPizzaMO’HaganDTWackAThe adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cellsJ Immunol200818085402541218390722
  • CalabroSTortoliMBaudnerBCVaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodesVaccine20112991812182321215831
  • NguyenDNGreenJJChanJMLangerRAndersonDGPolymeric materials for gene delivery and DNA vaccinationAdv Mater200921884786728413262
  • LisziewiczJTrocioJWhitmanLDermaVir: a novel topical vaccine for HIV/AIDSJ Invest Dermatol2005124116016915654970
  • Bivas-BenitaMBarLGillardGOEfficient generation of mucosal and systemic antigen-specific CD8+ T-cell responses following pulmonary DNA immunizationJ Virol201084115764577420335249
  • ZhouXLiuBYuXControlled release of PEI/DNA complexes from PLGA microspheres as a potent delivery system to enhance immune response to HIV vaccine DNA prime/MVA boost regimeEur J Pharm Biopharm200868358959517950586
  • GarzónMRBerraondoPCrettazJInduction of gp120-specific protective immune responses by genetic vaccination with linear polyethylenimine-plasmid complexVaccine200523111384139215661387
  • OsterCGKimNGrodeLCationic microparticles consisting of poly(lactide-co-glycolide) and polyethylenimine as carriers systems for parental DNA vaccinationJ Control Release2005104235937715907586
  • QinTYinYHuangLYuQYangQH9N2 influenza whole inactivated virus combined with polyethyleneimine strongly enhances mucosal and systemic immunity after intranasal immunization in miceClin Vaccine Immunol201522442142925673304
  • FirdousJIslamMAParkSMInduction of long-term immunity against respiratory syncytial virus glycoprotein by an osmotic polymeric nanocarrierActa Biomater201410114606461725110285
  • Bivas-BenitaMLinMYBalSMPulmonary delivery of DNA encoding Mycobacterium tuberculosis latency antigen Rv1733c associated to PLGA-PEI nanoparticles enhances T cell responses in a DNA prime/protein boost vaccination regimen in miceVaccine200927304010401719389445
  • HuKDouJYuFAn ocular mucosal administration of nanoparticles containing DNA vaccine pRSC-gD-IL-21 confers protection against mucosal challenge with herpes simplex virus type 1 in miceVaccine20112971455146221185849
  • ChenXLiuYWangLEnhanced humoral and cell-mediated immune responses generated by cationic polymer-coated PLA microspheres with adsorbed HBsAgMol Pharm20141161772178424738485
  • SunZLiuBRuanXLiuQAn enhanced immune response against G250, induced by a heterologous DNA prime-protein boost vaccination, using polyethyleneimine as a DNA vaccine adjuvantMol Med Rep20141052657266225190325
  • VellutoDThomasSNSimeoniESwartzMAHubbellJAPEG-b-PPS-b-PEI micelles and PEG-b-PPS/PEG-b-PPS-b-PEI mixed micelles as non-viral vectors for plasmid DNA: tumor immunotoxicity in B16F10 melanomaBiomaterials201132369839984721924769
  • ZhangMHongYChenWWangCPolymers for DNA vaccine deliveryACS Biomater Sci Eng201632108125
  • RegnströmKRagnarssonEGKöping-HöggårdMTorstenssonENyblomHArturssonPPEI – a potent, but not harmless, mucosal immuno-stimulator of mixed T-helper cell response and FasL-mediated cell death in miceGene Ther200310181575158312907949
  • LisziewiczJGabrilovichDIVargaGInduction of potent human immunodeficiency virus type 1-specific T-cell-restricted immunity by genetically modified dendritic cellsJ Virol200175167621762811462034
  • LiMJiangYXuCZhangZSunXEnhanced immune response against HIV-1 induced by a heterologous DNA prime-adenovirus boost vaccination using mannosylated polyethyleneimine as DNA vaccine adjuvantInt J Nanomedicine201381843185423690682
  • CherifMSShuaibuMNKodamaYNanoparticle formulation enhanced protective immunity provoked by PYGPI8p-transamidase related protein (PyTAM) DNA vaccine in Plasmodium yoelii malaria modelVaccine201432171998200624440206
  • Al-DeenFMXiangSDMaCMagnetic nanovectors for the development of DNA blood-stage malaria vaccinesNanomaterials (Basel)201772
  • ChenYZYaoXLRuanGXGene-carried chitosan-linked polyethylenimine induced high gene transfection efficiency on dendritic cellsBiotechnol Appl Biochem201259534635223586911
  • ZengQJiangHWangTZhangZGongTSunXCationic micelle delivery of Trp2 peptide for efficient lymphatic draining and enhanced cytotoxic T-lymphocyte responsesJ Control Release201420011225540903
  • MohitEBolhassaniAZahedifardFImmunomodulatory effects of IP-10 chemokine along with PEI600-Tat delivery system in DNA vaccination against HPV infectionsMol Immunol2013531–214916022926003
  • MaYFYangYWDelivery of DNA-based cancer vaccine with polyethylenimineEur J Pharm Sci2010402758320304049
  • SethAOhDBLimYTNanomaterials for enhanced immunity as an innovative paradigm in nanomedicineNanomedicine (Lond)201510695997525867860
  • BabenseeJEInteraction of dendritic cells with biomaterialsSemin Immunol200820210110818054498
  • LambrechtBNKoolMWillartMAHammadHMechanism of action of clinically approved adjuvantsCurr Opin Immunol2009211232919246182
  • UtoTWangXSatoKTargeting of antigen to dendritic cells with poly(γ-glutamic acid) nanoparticles induces antigen-specific humoral and cellular immunityJ Immunol200717852979298617312143
  • UtoTAkagiTYoshinagaKToyamaMAkashiMBabaMThe induction of innate and adaptive immunity by biodegradable poly(γ-glutamic acid) nanoparticles via a TLR4 and MyD88 signaling pathwayBiomaterials201132225206521221492934
  • MoonJJSuhHBershteynAInterbilayer-crosslinked multilamellar vesicles as synthetic vaccines for potent humoral and cellular immune responsesNat Mater201110324325121336265
  • KipperMJWilsonJHWannemuehlerMJNarasimhanBSingle dose vaccine based on biodegradable polyanhydride microspheres can modulate immune response mechanismJ Biomed Mater Res A200676479881016345084
  • AguerosMAresesPCampaneroMABioadhesive properties and biodistribution of cyclodextrin-poly(anhydride) nanoparticlesEur J Pharm Sci2009373–423124019491010
  • SalmanHHIracheJMGamazoCImmunoadjuvant capacity of flagellin and mannosamine-coated poly(anhydride) nanoparticles in oral vaccinationVaccine200927354784479019539576
  • XuLLiuYChenZMorphologically virus-like fullerenol nanoparticles act as the dual-functional nanoadjuvant for HIV-1 vaccineAdv Mater201325415928593623963730
  • XuLLiuYChenZSurface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatmentNano Lett20121242003201222372996
  • ShokouhiBCobanCHasirciVThe role of multiple toll-like receptor signalling cascades on interactions between biomedical polymers and dendritic cellsBiomaterials201031225759577120452017
  • HuangZYangYJiangYAnti-tumor immune responses of tumor-associated macrophages via toll-like receptor 4 triggered by cationic polymersBiomaterials201334374675523107297
  • ChenHLiPYinYThe promotion of type 1 T helper cell responses to cationic polymers in vivo via toll-like receptor-4 mediated IL-12 secretionBiomaterials201031328172818020692033
  • HeWLiangPGuoGRe-polarizing myeloid-derived suppressor cells (MDSCs) with cationic polymers for cancer immunotherapySci Rep201662450627074905
  • Cubillos-RuizJREngleXScarlettUKPolyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunityJ Clin Invest200911982231224419620771
  • OkadaMMatsuzawaAYoshimuraAIchijoHThe lysosome rupture-activated TAK1-JNK pathway regulates NLRP3 inflammasome activationJ Biol Chem201428947329263293625288801
  • TseHMMiltonMJSchreinerSProfozichJLTruccoMPiganelliJDDisruption of innate-mediated proinflammatory cytokine and reactive oxygen species third signal leads to antigen-specific hyporesponsivenessJ Immunol2007178290891717202352
  • Mulens-AriasVRojasJMPérez-YagüeSMoralesMPBarberDFPolyethylenimine-coated SPIONs trigger macrophage activation through TLR-4 signaling and ROS production and modulate podosome dynamicsBiomaterials20155249450625818455
  • YangYWHsuPYThe effect of poly(D,L-lactide-co-glycolide) microparticles with polyelectrolyte self-assembled multilayer surfaces on the cross-presentation of exogenous antigensBiomaterials200829162516252618329708
  • MarichalTOhataKBedoretDDNA released from dying host cells mediates aluminum adjuvant activityNat Med2011178996100221765404
  • PalumboRNZhongXWangCPolymer-mediated DNA vaccine delivery via bystander cells requires a proper balance between transfection efficiency and cytotoxicityJ Control Release20121571869321907252
  • LojkJStrojanKMišKBregarBVCell stress response to two different types of polymer coated cobalt ferrite nanoparticlesToxicol Lett201727010811828223195
  • PeterCWesselborgSHerrmannMLauberKDangerous attraction: phagocyte recruitment and danger signals of apoptotic and necrotic cellsApoptosis20101591007102820157780
  • De GregorioED’OroUWackAImmunology of TLR-independent vaccine adjuvantsCurr Opin Immunol200921333934519493664
  • OrsonFMKinseyBMHuaPJBhogalBSDensmoreCLBarryMAGenetic immunization with lung-targeting macroaggregated polyethyleneimine-albumin conjugates elicits combined systemic and mucosal immune responsesJ Immunol2000164126313632110843685
  • PalumboRNZhongXPanusDHanWJiWWangCTransgene expression and local tissue distribution of naked and polymer-condensed plasmid DNA after intradermal administration in miceJ Control Release2012159223223922300619