You are on page 1of 11

Review

Poly(glycoamidoamine)s: a broad class


of carbohydrate-containing
polycations for nucleic acid delivery
Nilesh P. Ingle1, Brett Malone2 and Theresa M. Reineke1
1
Department of Chemistry and Macromolecules and Interfaces Institute, Virginia Tech, Blacksburg, VA 24061, USA
2
Techulon Inc., 2200 Kraft Drive, Blacksburg, VA 24060, USA

In the era of nucleic acid therapeutics, there is an urgent as the delivery of specific nucleic acid sequences that
need for non-viral delivery vehicles that can cross the can cause inhibition of human epidermal growth factor
extracellular and intracellular barriers and deliver nucleic receptor 2 (HER-2) in breast and ovarian cancer cells [1].
acids to specific intracellular regions. This paper reviews Although there are many variables that need to be consid-
the development of a subclass of polymer-based delivery ered in developing specific delivery methods, the clear
vehicles termed poly(glycoamidoamine)s (PGAAs). The trend is toward more specificity and intent of function.
general design of this family consists of carbohydrate In other words, the special nature of nucleic acids is that
residues copolymerized with oligoethyleneamine units, they have the ability to selectively target a gene in a cell
which have proven to be an effective motif that pro- that is responsible for expressing a specific protein related
motes polyplex formation, efficient cellular internaliza- to a disease. A primary research aim for this field is to
tion, high gene expression and low cytotoxicity with increase the amount of nucleic acid delivered to the target
cultured cell lines and primary cell types. We then dis- tissue at a low dose profile while maintaining a minimal
cuss the structure–property relationships of the PGAA level of cellular toxicity and off-target effects. Herein, we
class of delivery vehicles and studies aimed at under- provide a brief review of a subclass of glycopolymer tech-
standing the mechanisms involved in cellular internali- nologies available for polynucleotide delivery. First, we
zation and trafficking. discuss the requirements that are shaping the develop-
ment of delivery methods. Second, we examine structure–
Intracellular delivery of nucleic acids property relationships of novel polymers termed poly(gly-
The intracellular delivery of nucleic acids has far-reaching coamidoamine)s (PGAAs). These materials are created by
implications in both health-related research and thera- coupling a variety of mono-, di- and oligo-saccharide mono-
peutic development. The ability to treat a disease target in mers with oligoethyleneamine units to create a new family
a specific and selective manner by utilizing the ever-grow- of non-viral nucleic acid delivery vehicles. Finally, we
ing genetic and epigenetic information obtained from discuss recent progress in the understanding of the cellu-
biomedical research could lead to novel research tools, lar interaction and intracellular trafficking behavior of
diagnostic methods and treatment strategies. However, to these materials.
advance this field toward this end goal, promoting delivery
into cells and being able to specifically target the delivery
to diseased cells is a major obstacle that needs to be Factors shaping development
overcome. Negatively-charged nucleic acids are repelled Several key factors are contributing to the need for im-
by the plasma membrane, which is typically coated with proved delivery of nucleic acids. First, research tools and
sulfated glycosaminoglycans (GAGs) that are also nega- therapies are more highly specialized and the payloads for
tive in charge; therefore, a means of masking the charge on the delivery vehicles are more diverse. For example, plas-
the nucleic acid, protecting these sensitive biomacromo- mid DNA (pDNA), small interfering RNA (siRNA) and
lecules from enzymatic degradation, and compacting microRNA have all become important components in the
these large structures into small structures that can tra- arsenal of cellular modification for therapeutic and bio-
verse the cell membrane are the key hurdles to overcome to technology purposes [2]. In addition, the cells of interest for
make this therapeutic method feasible. Nucleic acid de- certain diseases are sensitive and expensive. Primary cells,
livery vehicles perform this crucial function of transport. such as cortical neurons or induced pluripotent stem cells
However, the discovery of effective and benign methods of (IPSCs), are key to advanced cell therapies for many
polynucleotide delivery has emerged as the primary chal- indications. To be an effective long-term transfection solu-
lenge to this field. Nucleic acid delivery methods are tion, the intended carrier must be able to withstand the
continuously evolving to meet the many specialized rigors of clinical and regulatory protocol. Clinical solutions
requirements in disease research and treatment, such require safe, non-toxic delivery approaches that can be
easily manufactured at a low cost and have demonstrated
Corresponding author: Reineke, T.M. (treineke@vt.edu). low toxicity profiles.
0167-7799/$ – see front matter ß 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.tibtech.2011.04.012 Trends in Biotechnology, September 2011, Vol. 29, No. 9 443
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Structure and properties of nucleic acid delivery should be released from the vehicle. For example, pDNA
vehicles can be transported to the nucleus of cells by some non-viral
Non-viral delivery vehicles consist of complexes between vehicles; however, the mechanisms of this process are still
nucleic acids and various macromolecules such as poly- unclear and under investigation [13,14].
mers (to form polyplexes) or lipids (to form lipoplexes). Complexes between polymers and nucleic acids are
Furthermore, multifaceted assemblies consisting of typically characterized by their charge or N/P ratio. This
nucleic acids and one or more polymer types and/or lipids number is defined by the molar ratio of the number of
to form composite–polyplexes [3] are also being extensively nitrogen (N) or cationic groups in the polymer repeat unit
researched in this field. All of these delivery techniques to the number of phosphate groups (P) on the nucleic acid
being utilized for the delivery of nucleic acid drugs have cargo that is used to form the nanoparticle complexes. For
revealed promising characteristics. The structure and in vitro transfection purposes, a positive N/P or charge
unique function of these macromolecules allows for the ratio is most commonly utilized because this promotes
encapsulation, protection and stabilization of their nucleic strong and non-specific interaction of the polyplexes with
acid cargo. In addition, these delivery modalities can be the surface of cultured cells. Also, in pure water, this
readily modified via synthetic methods to promote effective overall cationic surface (or Zeta) potential promotes colloi-
delivery that is specific for the desired target [4]. The dal stabilization as a result of the charge repulsion of the
surface of most cell types is negatively-charged [5], which nanoparticle complexes. However, once introduced into
is mostly due to sulfated proteoglycans and negatively- salt- or serum-containing media, these complexes rapidly
charged phospho-lipids displayed on the plasma mem- aggregate. For in vitro or ex vivo delivery methods, this is
brane surface [6]. Nucleic acids, the cargo of the carriers, not of concern because it has been found that a number of
also have a net negative charge on their surface from the nanoparticle sizes and shapes can be readily taken up by
phosphodiester backbone. For this reason, the molecules cultured cells [15]. However, for in vivo purposes, this lack
used to complex nucleic acids, for the most part, are of colloidal stability is of high concern and must be avoided
cationic. This serves to facilitate electrostatic interactions for systemic delivery success because it may cause aggre-
with nucleic acids, which ultimately drives the complexa- gation with blood components, facilitate rapid systemic
tion and compaction of the polynucleotide cargo. In addi- clearance by the reticuloendothelial system and, in turn,
tion, the polycation vehicles also serve to charge neutralize significantly reduce dose and target specificity. To this end,
this macromolecular cargo to promote complex interaction the amount of nucleic acid reaching the target cell site
with the cell surface and internalization. Studies have (such as a tumor) is significantly reduced. The modification
shown that with naked pDNA (and other nucleic acids), of the polymers to avoid aggregation is briefly discussed
delivery is marginal at best and yields poor results, in part later in this review.
due to a charge-repulsion from the negative surface of cells
[7]. In this review, we will focus on polymeric routes of Effect of size and shape
nucleic acid delivery because of their complex structures The size and shape that these polyplexes form can vary
and unique ability to be synthetically modified for optimiz- significantly based on the formulation conditions of the
ing delivery. polyplexes and the structure of the vehicle [16]. In general,
it is most desirable to obtain polyplexes of 50–100 nm to
Effect of surface charge promote cellular internalization via endocytosis; in vivo
Polymers (usually polycations) are used as nucleic acid use requires polyplexes near the smaller end of this range.
delivery vehicles because they can assemble with various To this end, physicochemical characterization of the
sizes of polynucleotides (Figure 1) in aqueous solution and polyplexes is very important in understanding polymer–
compact their cargo into colloidal nanoparticle complexes. polynucleotide binding affinity, complex stability (in phys-
This process neutralizes the polyanionic charge and, iological conditions), size, shape and colloidal behavior in
in most cases, these complexes are overcharged, which the media used for delivery (or in the blood for in vivo
results in a net positive charge on the surface (typically applications) because these parameters can significantly
20–40 mV, as determined by Zeta potential measurements affect efficacy and side effects. There are many analytical
[8,9]). The positive charge facilitates interaction of poly- methods currently used to characterize polyplex formation,
plexes and lipoplexes with the plasma membrane of the cell and the field is generally in need of enhancing the knowl-
and subsequent cellular uptake of these complexes [10,11]. edge base of how the polymer structure influences polyplex
In addition, these macromolecular materials also perform characteristics. Generally, the polyplexes are first charac-
a very important function of shielding the nucleic acid from terized for their binding to nucleic acids via gel electropho-
the detrimental extracellular and intracellular nucleases resis shift assays, which reveals the N/P ratio of complex
that can degrade the cargo as the complexes traffic into and neutralization and is generally a qualitative assessment of
through the cell [12]. The vehicle is also responsible for polyplex formation [17,18]. Dynamic light scattering (DLS)
promoting cell surface interaction, internalization, traf- and Zeta potential measurements are typically completed
ficking and release of the cargo within the cell. Of course to assess the general nanoparticle hydrodynamic radius
the cargo type (i.e. pDNA, oligonucleotides or siRNA) and surface charge, respectively. These two techniques are
significantly determines the desired intracellular destina- the primary methods of characterizing nanoparticle size
tion, whether it be the nucleus (pDNA or some oligo types) and charge and are also used to monitor polyplex colloidal
or the cytoplasm (siRNA or other oligo forms), and thus this stability upon the addition of salt and serum media. How-
factor also significantly dictates where the nucleic acid ever, a drawback of these techniques is that they do not

444
(Figure_1)TD$IG][ Review Trends in Biotechnology September 2011, Vol. 29, No. 9

+
GlycofectTM + +
pDNA
OH OH O + +
R

O OH OH
N
H
( N
)
4
N
H
+ +
14
Polymer Plasmid DNA

Trehalose polycations
N N O OH
N HO OH O H O
O N N
HO OH N N
HO O N N 1-4
H H 55 or 77
+ + +
+ +
+ +
+
++ + ++
+
Polyplex
TRENDS in Biotechnology

Figure 1. Polycations can readily assemble with polyanionic nucleic acids and form colloidal complexes termed polyplexes that facilitate cellular uptake and entry. The
two examples of glycopolymers developed by Reineke et al. that are displayed have shown high efficiency for delivering nucleic acids into cells. Adapted, with permission,
from [17,44].

discriminate different morphologies of polyplexes (rod-like groups and oligoethyleneamines (between one and six)
or torroidal) because most DLS instrumentation assumes that are linked together by amide bonds [31]. These struc-
that the complexes are spherical in nature. For this reason, tures were originally designed to encompass the most
direct visualization techniques, such as transmission elec- promising and active structural attributes of the two most
tron microscopy (TEM), scanning electron microscopy commonly-studied polymeric delivery vehicles, polyethyle-
(SEM) and atomic force microscopy (AFM) are also used neimine (PEI), a highly active yet toxic polymer, and
in conjunction with light scattering to observe general chitosan, a benign polysaccharide that generally yields
polyplex size and shape [19]. Many other characterization low delivery efficiency. The first example of this strategy
tools are currently in use such as isothermal titration was examined by creating a series of 16 structures by
calorimetry and multi-angle laser light scattering; indeed, copolymerizing monosaccharide monomers, L-tartarate
this subfield of analytical characterization of polyplex (T), D-glucarate (D), meso-galactarate (G) and D-manna-
substructure is certainly important and in need of further rate (M) with four different oligoethyleneamine monomers
research and development [20–22]. containing between one and four ethyleneamines (four
examples are shown in Figure 2) [17,32,33]. These glyco-
Structure and bioactivity relationships of nucleic acid polymers were found to bind and compact pDNA into
delivery vehicles polyplexes and successfully deliver pDNA into various cell
A primary challenge in this field is finding materials that types without toxic effects. Interestingly, these PGAAs
promote both safe and effective delivery efficiency because have been found to rapidly hydrolyze under physiological
some of the most effective delivery vehicles tend to damage conditions, which probably contributes to their nontoxic
the cell, cause high cell death and/or can illicit an immune nature [31]. The number and stereochemistry of the hy-
response [23]. Also, understanding the structure–bioactiv- droxyl groups was found to alter the efficiency, cell surface
ity relationships is key to the development of successful binding and internalization, and the mechanisms of intra-
vehicles because many seemingly trivial attributes within cellular trafficking; the galactarate polymers with four
identical chemical structures of the polymer repeat unit ethyleneamine units (G4), in general, yielded the highest
such as different counterions [24,25], linear versus efficiency with most cell types [7,34]. In addition, the
branched [26,27] and the polymer length [16,28–30] can biological effect of increasing the number of ethylenea-
play a large role in the observed toxicity and efficiency. To mines to five and six and the effects of linear versus
examine this, many creative structures continue to be branched analogs of these PGAA structures have also been
developed and examined for their structure–activity rela- investigated. In general, increasing the number of ethyle-
tionships, and some of these systems are summarized in neamines does not significantly enhance transfection effi-
Table 1. For the purposes of this review, we focus on the ciency whereas it does increase toxicity. Linear analogs
structure–activity studies that our group has completed on were also found to be more toxic than branched analogs of
a subclass of structures we have designed and developed the same molecular weight and chemical structure [26].
termed PGAAs. Polymer G4 has recently been licensed by Techulon and is
being sold in a novel formulation under the trade name
Linear monosaccharides GlycofectTM; it shows high activity in delivering pDNA to
PGAAs (Figure 2) were designed to embody a multifunc- many primary cell types without causing high toxicity,
tional structure platform consisting of carbohydrate which is a drawback of other leading transfection reagents

445
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

[35]. Figure 3 shows the expression profile and human the effects of carbohydrate size and polymer length
dermal fibroblast cell (HDFn) morphology for G4 compared (Figure 4).
to Lipofectamine [35]. These structures were created by polymerizing, via the
click reaction (azide–alkyne cycloaddition), a diazide–car-
Larger carbohydrate-based monomers bohydrate monomer (either b-cyclodextrin or trehalose)
In addition to these original structures that incorporate with a set of dialkyne–ethyleneamine monomers that con-
linear monosaccharides, larger carbohydrate-based mono- tain between one and four secondary amines. This poly-
mers have been used to synthesize glycopolymer delivery merization reaction forms a 1,2,3-triazole linkage between
vehicles with structures similar in nature to the original the monomers and allowed a variety of polymers with
linear monosaccharide structures but with some chemical different molecular weights to be created. This structural
differences. Both trehalose [16,17,36] and b-cyclodextrin versatility allowed the assessment of how structural
monomers [37] have been incorporated into new related changes affect both toxicity and delivery efficiency. In
architectures by our research team to further investigate general, the trehalose and b-cyclodextrin families of struc-

Table 1. Nucleic acid delivery vehicles based on carbohydrate and synthetic polymeric derivatives
Abbreviation Chemical name Refs.
Carbohydrate-based polymers
PGAAs
D1–D6 Poly(D-glucaramidoamine)s [17,31,32]
G1–G6 Poly(galactaramidoamine)s; trade name for G4: Glycofect TM [17,31]
M1–M6 Poly(D-mannaramidoamine)s [17,31]
T1–T6 Poly(L-tartaramidoamine)s [31,48]
O4 Poly(oxalamidopentaethylenehexamine) [31,43]
S4 Poly(succinamidopentaethylenetetramine) [31,43]
A4 Poly(adipamidopentaethylenetetramine) [31,43]
Disaccharide-containing polymers
Tr1–Tr4 Trehalose and a dialkyne-pentaethylene tetramine monomer [44]
AP2, AP3 D-trehalose amidine-based comonomers [49]
Glycopolymer chelates
Eu3a, Eu3b Lanthanide chelate with europium [50]
Gd3a, Gd3b Lanthanide chelate with gadolinium [50]
Cyclodextrin polymers
Cd1–Cd4 b-cyclodextrin ‘click’ polymers [37]
Gd4, Gd10 Gadolinium conjugated b-cyclodextrin [51,52]
Eu4, Eu10 Europium conjugated b-cyclodextrin [51,52]
QP4 Quaternary ammonium polycation conjugated b-cyclodextrin [53]
Tf–PEG–AD Transferrin and adamantane conjugated poly(ethylene glycol) [54]
6a–6d; 7a–7d g-Cyclodextrin series [18]
CD–IPEI, CD–lPEI+AD–PEG, CD–bPEI+AD–PEG Linear and branched poly(ethylenimine) grafted with b-cyclodextrin and conjugated [55]
with adamantane–poly(ethylene glycol)
a, b and g CDE conjugates Poly(amidoamine) dendrimer conjugates with a, b and g cyclodextrin [56]
AP1–CD, AP2–CD, IM1–CD to IM3–CD, Pyridylamino, alkylimidazole, methoxyethylamino or primary amine group [57]
AM1–CD, ME1–CD modified cyclodextrins
Hist–bCDP6 Histidylated–b cyclodextrin polymer [58]
bCDP4–bCDP8, bCDP10 b-Cyclodextrin-containing polycations [59]
paCD Polycationic amphiphilic cyclodextrins [60]
Malonic-based polymers
Ma-x-y-Gal-z: Ma, malonic groups; x, spacer length between polymer backbone and triazole [61]
Ma-1-3-Gal-40, Ma-1-2-Gal-18, Ma-1-2-Gal-13, ring; y, spacer length between triazole ring and carbohydrate; Gal, b-D-galactose;
Ma-1-6-Gal-37, Ma-1-6-Gal-22, Ma-1-6-Gal-09, Glu, b-D-glucose; Man, a-D-mannose; z, degree of carbohydrate substitution
Ma-1-9-Gal-44, Ma-1-9-Gal-24, Ma-1-9-Gal-08,
Ma-1-3-Glu-10, Ma-1-3-Man-22
Chitosan-based polymers
Chitosan–g–PEI Poly(ethylenimine) grafted on chitosan [62]
Gal–PEG–chitosan–g–PEI Galactosylated poly(ethylene glycol)–chitosan–g–poly(ethylenimine) [63]
TMC Trimethylchitosan [64]
Schizophyllan-based polymers
SPG–DNA–polycation ternary complex Schizophyllan–DNA–poly(ethylenimine) [65]
Dextran-based polymers
DEAE–dextran 2-Diethylaminoethyl–dextran [66]
DDMC 2-Diethylaminoethyl–dextranmethyl methacrylate graft copolymer [66]
PAA–DS–DNA Poly(allylamine)–dextran sulfate–DNA [67]

446
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Table 1 (Continued )
Abbreviation Chemical name Refs.
Lysine-based polymers
PLL Poly(L-lysine) [68,69]
PEG–poly(Lys) Poly(ethylene glycol)-conjugated poly(lysine) [70]
cRGD–PEG–P(Lys-SH) Thiolated c(RGDfK)–poly(ethylene glycol)–block–poly(L-lysine) [71]
PDL Poly(D-lysine) [69]
Polyethylenimine-based polymers
pLPEI Short linear poly(ethylenimine) [72]
PEI–CD b-cyclodextrin-conjugated poly(ethylenimine) [73]
bPEI Branched poly(ethylenimine) [74]
Polymethacrylate-based polymers
Poly(BMDO-co-DMAEMA) 5,6-Benzo-2-methylene-1,3-dioxepane-co-N,N-dimethylaminoethyl methacrylate [75]
p(CBMA)–EE Poly(carboxybetaine methacrylate–ethyl ester [39]
PAEMA Poly[N-(2-aminoethyl)methacrylamide trifluoroacetate] [76,77]
PAHPMA Poly(3-amino-2-hydroxypropyl methacrylate) [77]
PAEAEMA Poly(2-(2-aminoethylamino)ethyl methacrylate) [77]
PAEAHPMA Poly(3-(2-aminoethylamino) 2-hydroxypropyl methacrylate) [77]
PDMAEMA Poly(2-dimethylamino) ethyl methacrylate [78]

[(Figure_2)TD$IG]

(i) OH OH O (ii)

Luciferase Gene Expression (RLU/mg)


R O 10
D4 N H 10
N N O4 N
N
N
O OH OH H 4 H H 4 H
O
12 16 9
10

OH OH O O H 8
R 10
G4 N S4 N
N
N
N N H 4 H
O OH OH H H O 11
4
11 107

OH OH O
O H 106
M4 R N
N A4 N N
N N O H 4 H
O OH OH H 4 H 11 105
Y EI ine 10 20 30 10 20 30 10 20 30 10 20 30 10 20 30 10 20 30 10 20 30
14 NL P
O Jet tam
NA fec D4 G4 M4 T4 A4 S4 O4
pD po
OH O Li
T4 R
N
N N
O OH H 4 H
14

TRENDS in Biotechnology

Figure 2. (i) Novel poly(glycoamidoamine) (D4, G4, M4 and T4) and poly(amidoamine) (O4, S4 and A4) polymers for non-viral nucleic acid delivery. (ii) Luciferase gene
expression (RLU/mg protein) in human adenocervical carcinoma (HeLa) cells. Transfections were completed in reduced serum media (OptiMEM). The molecular weights of
the polymers shown and examined in this study are as follows: D4 = 4.9 kDa, G4 = 4.6 kDa, M4 = 5.6 kDa, T4 = 4.8 kDa, A4 = 3.8 kDa, S4 = 3.3 kDa, O4 = 4.5 kDa. Adapted, with
permission from [31].
[(Figure_3)TD$IG]

DNA only G4 (GlycofectTM) Lipofectamine 2000


TRENDS in Biotechnology

Figure 3. GlycofectTM (a novel formulation of G4) yields significantly higher green fluorescent protein (GFP) gene expression in human dermal fibroblast cells (HDFn) after
48 hours post transfection. Both the differential interference contrast images (DIC) (top row) and wide field microscopy images (bottom row) are displayed. Adapted, with
permission from [35].

447
(Figure_4)TD$IG][ Review Trends in Biotechnology September 2011, Vol. 29, No. 9

(i) (ii) OH
O
HO O
O O OH
OH HO OH
O OH HO O
HO
N N O OH O OH O O O
N HO OH O O N HO H
O H N OH N N
N N N N N
HO OH N N HO
H 1-4 H
HO O N N O OH OH O N
H 1-4 H OH HO O
N
nw O nw
HO O O
(a) (b) (c) (d) HO

Key:
(iii) 1010 (iv) Jet-PEI Cd349 Cd447
(a) 1010
(a) Cd246 Cd146

RLU/mg of protien
RLU/mg protien

109
108 108
107
106 106
Key:
9a 9b
105
9c Jet-PEI
104 104
0 5 10 15 20 25 0 5 10 15 20 25
N/P Ratio N/P Ratio
Transfection in Opti-MEM Transfection in Opti-MEM

Key:
1010 1010 Jet-PEI Cd493 Cd4200
(b) (b) Cd447 Cd427
RLU/mg of protien

109
RLU/mg protien

108 108
107

106 106
Key: 105
9a 9b
9c Jet-PEI
104 104
0 5 10 15 20 25 0 5 10 15 20 25
N/P Ratio N/P Ratio
Transfection in DMEM Transfection in Opti-MEM
TRENDS in Biotechnology

Figure 4. (i) Trehalose and b-cyclodextrin polymers for non-viral nucleic acid delivery. (a) The trehalose carbohydrate unit that helps to increase cell viability, solubility and
stability in serum-containing media. (b) Heterocycle-amide groups to increase binding affinity. (c) An oligoethyleneamine unit to aid electrostatic binding of the polymer
with the phosphodiester backbone of the nucleic acids and enhance cellular uptake. (d) The degree of polymerization that can be tuned easily to optimize delivery
performance for various applications. (ii) b-Cyclodextrin-based polymers with similar structure to the trehalose polymers (i) have also been created and examined. (iii) Gene
expression (RLU/mg protein) for trehalose polymers with ethyleneamines units = 1, where the degree of polymerization (n) = 34, 2 (n = 39), and 3 (n = 40) at: 0 (DNA only), 3,
5, 7, 10, 15 and 25 N/P ratios. (iv) Gene expression (RLU/mg protein) for b-cyclodextrin polymers at: 0, 5, 7, 10, 15, 20 and 25 N/P ratios. The control is Jet-PEI (N/P = 5), and
the cell type used was human adenocervical carcinoma (HeLa) cells. Abbreviation: RLU, relative light units. Adapted, with permission from [37,46].

tures followed similar biological trends with both HeLa With similar structural features to these carbohydrate-
and H9c2(2-1) cells. As the number of ethyleneamines based polymers, our research team has also created com-
increased, the intracellular delivery of pDNA and trans- plimentary structures termed ‘click clusters’ that contain
gene expression increased. However, having either three or seven ethyleneamine arms coupled to a b-cyclodextrin
four ethyleneamines in the repeat units (with both fami- core via triazole linkage chemistry. Unlike the previous
lies) yielded similar results for toxicity and transfection polymer-based systems that have a dispersity index asso-
efficiency. Interestingly, in some cases the structures with ciated with the molecular weight, these macromolecules
three ethyleneamines yielded higher efficiency than four are completely monodisperse, as supported by mass spec-
(transgene expression similar to Jet-PEI). A major differ- trometry data. As published, these structures facilitate
ence between these structures and the original monosac- nucleic acid binding and compaction [38] (Figure 5). In-
charide PGAAs is that polyplexes formed at low N/P ratios terestingly, these macromolecules were similar to the b-
(N/P = 5–7) and facilitated high gene expression, whereas cyclodextrin polymers in that the structures that contain
high N/P ratios of 20–30 were needed to promote high gene three and four secondary amines both yielded high trans-
expression levels with the original set of monosaccharide fection results. However, unlike the b-cyclodextrin poly-
PGAAs. As the polymer molecular weight increased, the mers that yield high gene expression at low N/P ratios, a
delivery efficiency increased significantly, yet with the cost much higher N/P ratio of 20–50 was required of the click
of also increasing the toxicity [16,17,36]. clusters to observe similar high transfection efficiency

448
(Figure_5)TD$IG][ Review Trends in Biotechnology September 2011, Vol. 29, No. 9

106

Relative light units (RLU)


(i) (ii) (a) (In HeLa cells)
105
H2N
X NH 104
2
NH
X 103
NH

HN HN 102
O N N
N N O 101
H2N N N O O
NH N N H
O O N 100
O HO OH N NH 9a 9b 9c 9d 9e

ct
X

NA

EI
N 2
OH

erfe
O

t-P
N O OH HO

pD
H O H

Je
HOO X

Sup
N N O OH
N OH HO N N
OH HO O N 106
O OH HO

Relative light units (RLU)


HO O (b) (In H9C2 cells)
O NH
O N O O O 105
N
NH N N N
HN 104
O N HN
NH 103
H2N X NH2
X
102
HN X=0-4
101
X
NH2 100
9a 9b 9c 9d 9e

ct
I
NA

-PE

erfe
pD

Jet

Sup
Key: N/P 5 N/P 10 N/P 20 N/P 50

TRENDS in Biotechnology

Figure 5. (i) Schematic structures of the b-cyclodextrin ‘click cluster’ delivery vehicles. (ii) Gene expression for analogues 9a (x = 0), 9b (x = 1), 9c (x = 2), 9d (x = 3) and 9e
(x = 4) in human adenocervical carcinoma (HeLa) cells and rat cardiomyoblast H9C2(2-1) cells in OptiMEM at N/P ratios of 5, 10, 20 and 50. The controls are pDNA, Jet-PEI (N/
P = 5) and Superfect (N/P = 5). Abbreviation: RLU, relative light units. Adapted, with permission from [38].

(comparable to the commercial controls) with HeLa and GAG structures), the internalization was unaffected,
H9c2(2-1) cells [38]. which demonstrates that the other GAGs are as important
in the uptake of PGAA polyplexes. As a final note, although
Cellular interactions of nucleic acid delivery vehicles charge certainly plays a role in polyplex internalizations
Surface proteins on plasma membrane (desulfating the GAGs on CHO cells nearly abolished
As discussed earlier, the cell surface derives its negative PGAA polyplex uptake), the internalization was not solely
charge from glycoprotein-based structures termed GAGs charge-dependent. The GAG with the lowest anionic den-
[34]. Primarily, there are six different types of GAGs: sity, HA, revealed the highest binding affinity to the PGAA
heparin (H), heparan sulfate (HS), dermatan sulfate polyplexes. Likewise, CSA yielded a higher PGAA polyplex
(DS), chondroitin sulfate C (CSC), chondroitin sulfate A binding affinity than CSC despite the fact that the two
(CSA) and hyaluronate (HA), with decreasing anionic structures have the same sulfation density (but different
charge density from the former (H) to the later (HA), where sulfation sites on the polysaccharide structure). From
CSC = CSA [34]. To promote cellular entry, PGAA poly- these studies, it was concluded that the carbohydrate
plexes are formulated by combining the cationic polymers residues within the PGAA structures are probably playing
with anionic nucleic acids that have an overall positive a role in promoting GAG binding via H-bonding [34], and
charge due to the higher stoichiometric ratio of amines. these effects are still widely being studied in our group.
The cationic polyplexes interact with anionic GAGs, and
this interaction is in part driven via electrostatic interac- Internalization pathways
tions, which trigger subsequent internalization via endo- After the polyplexes adhere to the GAGs on the outer
cytosis. Our research has shown that the presence of GAGs surface of cellular plasma membrane, the next step in
on the cell surface is necessary to promote cellular inter- their transit route is trafficking across the plasma mem-
nalization because studies with GAG-deficient CHO cells brane into the cell. The interaction of polyplexes with cell
(pgsA-745 cell line) show a dramatic decrease in PGAA surface receptors triggers an uptake mechanism called
polyplex uptake compared to CHO cells expressing normal endocytosis. Endocytosis is a general term that encom-
GAG levels. It was found that HS yields the highest passes several mechanisms of active intracellular trans-
binding affinity to the PGAA polyplexes with HA coming port such as clathrin, caveolae-mediated endocytosis,
in a close second. Interestingly, if polyplex solutions were macropinocytosis and some other lesser-known active
exposed to excessive concentrations of the various GAGs transport pathways. Our research team has examined both
free in solution, HA bound to the surface of the polyplexes the internalization pathways and the routes that lead to
to form ternary complexes, but HS dissociates the polyplex. nuclear delivery for gene expression with a combination of
However, if transfections were completed with CHO cells both confocal microscopy and pharmacological inhibitor
lacking only HS on the surface (but retaining the other experiments. Interestingly, the PGAA polyplexes were

449
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

found to be internalized into cells by a combination of these ing of PGAA polyplexes because their knockdown resulted
pathways primarily involving caveolae, whereas about in a significant drop in uptake (80%) (Figure 6). The
80% of PGAA polyplex internalization was inhibited with knockdown of dynamin resulted in a 99% reduction in gene
cellular exposure to filipin III (Figure 6). However, if the expression [7]. This work also revealed that caveolae
cells were exposed to chlorpromazine (clathrin inhibitor) or appears to be the primary route that leads to nuclear
dimethylameloride (macropinocytosis inhibitor), only ap- delivery of pDNA complexed with the PGAAs for transgene
proximately 50% and 20% internalization knockdown was expression because inhibiting both clathrin and macropi-
observed, respectively, for the PGAA polyplexes. [7]. Actin, nocytosis results in a negligible decrease in gene expres-
a primary intracellular highway, and dynamin, a protein sion. Actin and dynamin were also found to play very large
essential for pinching off endocytic vesicles from the plas- roles in nuclear delivery of pDNA with the PGAAs because
ma membrane to promote internalization, were also found knockdown of these key proteins decreased gene expres-
to be essential in both cellular internalization and traffick- sion [7].
[(Figure_6)TD$IG]

PGAA DNA Polyplex


+
OH OH O H +
H3CO N H + +
N N
O OH OH H 4 H + +
n

Macropinocytosis Clathrin Caveolae Direct membrane


penetration

Key: Actin
Dynamin-1
Cholesterol
Nucleus
Caveolin-1 (for gene expression)

Proteoglycan

(a) Endocytosis pathways for PGAA nucleic acid delivery vehicles

100.0 108
90.0
Luciferase expression (RLU)

#
107
80.0
Decrease in Cy5-pDNA
fluorescence (%)

70.0 # ##
106
60.0
*
∧ ∧
50.0 105
# # # #
40.0
104
30.0 **
#
20.0 103
10.0
102
0.0
Cells only DNA only G4 D4 M4 T4 Jet-PEI
G4 D4 M4 T4 Jet-PEI
Key: Untreated Clathrin Caveolae
Key: Clathrin Caveolae Macropinocytosis Macropinocytosis Macropinocytosis Actin
Macropinocytosis Actin Dynamin Dynamin

(b) Uptake inhibition (c) Endocytosis inhibition


TRENDS in Biotechnology

Figure 6. (a) Different endocytosis mechanisms that are commonly examined as transfection routes for polyplexes. (b) Decrease in cellular uptake at 2.5 hours post
transfection for the PGAA polyplexes. All values are significantly different from pDNA at P < 0.05; except when marked by #. (c) Luciferase gene expression for the PGAA
polyplexes at 48 hours post transfection in cells inhibited for various endocytosis pathways. All values are significantly different from uninhibited control at P < 0.001;
where, *: P < 0.001; **: P < 0.005; ^: P < 0.1; and #: not significant. Inhibition method: Various routes involved in polyplex uptake were knocked down in the cultured cells
using the following inhibitors: chlorpromazine for clathrin; filipin III for caveolae; dimethylamiloride (DMA) for macropinocytosis; dynasore for dynamin; cytochalasin D to
depolymerize actin. Phorbol myristate acetate (PMA) was used to stimulate macropinocytosis. Experiments were performed with human adenocervical carcinoma (HeLa)
cells. Adapted, with permission, from [7].

450
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

Perinuclear release of nucleic acid structures play a role in the binding affinity, and polymer
Many further investigations are ongoing to better under- G4 was found to yield the highest binding affinity to pDNA
stand their transport pathways and the release mecha- [43]. This effect has also been studied with the trehalose
nisms of the polymer from the nucleic acid. It should be polymers; interestingly, increasing the length of the oli-
noted that escape of the endosomal/lysosomal pathway is goethyleneamine units within this subclass of structures
still currently thought to be an important step in the was found to promote direct interactions within the
overall delivery process of polyplexes to facilitate nucleic grooves of pDNA [16,44–46].
acid delivery and drug efficacy [39]. Although much work
has been completed to understand this process, much work Concluding remarks
still remains because the trafficking pathway and delivery The field of non-viral nucleic acid delivery has the poten-
efficiency is dependent on both the structures of the poly- tial to yield significant advances in biotechnology research
mer and the nucleic acid. For example, as discussed above, and therapeutic development. As discussed herein, glyco-
for pDNA, the nucleic acid must be transported to and polymer-based vehicles have been developed and continue
enter the nucleus to promote transgene expression. How- to be thoroughly studied for their bioactivity and mecha-
ever, for siRNA delivery, the polyplexes must escape these nistic properties. Our research team found that carbohy-
membrane-bound vesicles and enter the cytoplasm for drate-based PGAA polymers have the ability to bind and
transport to the RNA-induced silencing complex (RISC) compact nucleic acids into polyplexes that facilitate effec-
for knockdown of mRNA expression [40]. The high trans- tive intracellular pDNA delivery with low toxicity. Poly-
fection efficiency of some commercial vehicles such as mer G4 is currently being sold in a novel formulation
polyamidoamine (PAMAM) dendrimer and PEI has been under the name GlycofectTM; it shows exceptional pDNA
attributed to the ability of these polymers to escape the delivery efficiency with many primary cell types. This
endosomes. This escape mechanism may be due to the exciting class of polymers has unique degradation profiles,
presence of a large number of positively-charged amines therefore they may also be promising for controlled release
that are thought to cause a ‘proton sponge’-like effect applications [47].
resulting in endosomal swelling, rupture and ultimately
polyplex release [39,41]. References
After the polyplex reaches its destination within the 1 Oh, Y-K. and Park, T.G. (2009) siRNA delivery systems for cancer
cell, it should also dissociate to expose the nucleic acid to treatment. Adv. Drug Deliv. Rev. 61, 850–862
perform its therapeutic effect [42]. This dissociation can be 2 Alvarez-Salas, L.M. (2008) Nucleic acids as therapeutic agents. Curr.
Top. Med. Chem. 8, 1379–1404
influenced by several factors such as pH, ionic strength of
3 Debus, H. et al. (2010) Delivery of messenger RNA using poly(ethylene
the surrounding cytoplasmic fluid, temperature [42] and imine)-poly(ethylene glycol)-copolymer blends for polyplex formation:
the N/P ratio [31]. In particular, many systems are biophysical characterization and in vitro transfection properties. J.
designed to degrade under biological conditions to promote Control. Release 148, 334–343
nucleic acid release. Through a comparative degradation 4 Rajendran, L. et al. (2010) Subcellular targeting strategies for drug
design and delivery. Nat. Rev. Drug Discov. 9, 29–42
analysis of the PGAA polymers, it was discovered that they 5 Gao, K. and Huang, L. (2008) Nonviral methods for siRNA delivery.
rapidly degrade under physiological temperature and pH Mol. Pharm. 6, 651–658
[31]. Interestingly, at acidic pH 5.0, the M4 and T4 poly- 6 Tarahovsky, Y. (2009) Cell transfection by DNA–lipid complexes-
mers degrade slower than the G4 and D4 polymers [31]. lipoplexes. Biochemistry (Moscow) 74, 1293–1304
This degradation is currently attributed to hydrolysis of 7 McLendon, P.M. et al. (2010) Poly(glycoamidoamine) vehicles promote
pDNA uptake through multiple routes and efficient gene expression via
the amide bond, which is unusual because amides are caveolae-mediated endocytosis. Mol. Pharm. 7, 738–750
normally hydrolytically stable under these mild conditions 8 Herringson, T.P. et al. (2009) Targeting of plasmid DNA–lipoplexes to
[31]. This preliminary study found that the synergistic cells with molecules anchored via a metal chelator lipid. J. Gene Med.
presence of both the hydroxyl groups (from the carbohy- 11, 1048–1063
9 Geusens, B. et al. (2009) Ultradeformable cationic liposomes for
drate unit) and the amines promote the rapid hydrolysis
delivery of small interfering RNA (siRNA) into human primary
event. Polymer hydrolysis was shown to aid gene expres- melanocytes. J. Control. Release 133, 214–220
sion because models with related structures that contain 10 Rao, N.M. (2010) Cationic lipid-mediated nucleic acid delivery: beyond
alkyl chains in place of the carbohydrates (A4 and S4) do being cationic. Chem. Phys. Lipids 163, 245–252
not rapidly degrade under the experimental conditions and 11 Lu, J.J. et al. (2009) A novel mechanism is involved in cationic lipid-
mediated functional siRNA delivery. Mol. Pharm. 6, 763–771
were found to yield much lower gene expression profiles
12 De Laporte, L. et al. (2009) Local gene delivery from ECM-coated
(Figure 2). poly(lactide-co-glycolide) multiple channel bridges after spinal cord
Both fundamental and applied work on these PGAA injury. Biomaterials 30, 2361–2368
vehicles is still ongoing in our lab. For example, we are 13 Cohen, R.N. et al. (2009) Quantification of plasmid DNA copies in the
interested in understanding how the polymer structure nucleus after lipoplex and polyplex transfection. J. Control. Release
135, 166–174
affects the nucleic acid binding affinity, polyplex stability 14 Wolff, J.A. and Budker, V. (2005) The mechanism of naked DNA
and nucleic acid release. Fundamental studies have been uptake and expression. In Advances in Genetics (Leaf Huang, M.-
performed to reveal that the binding affinity of the polymer C.H. and Ernst, W., eds), pp. 1–20, Academic Press
to the nucleic acid is driven by a combination of both 15 Gratton, S.E.A. et al. (2008) The effect of particle design on cellular
electrostatics and H-bonding, and the presence of the internalization pathways. Proc. Natl. Acad. Sci. U.S.A. 105, 11613–
11618
carbohydrates enhance the H-bonding interaction over 16 Srinivasachari, S. et al. (2007) Effects of trehalose click polymer length
models lacking hydroxyls [43]. Furthermore, the stereo- on pDNA complex stability and delivery efficacy. Biomaterials 28,
chemistry of the hydroxyl groups within the polymer 2885–2898

451
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

17 Liu, Y. and Reineke, T.M. (2005) Hydroxyl stereochemistry and amine 42 Blacklock, J. et al. (2010) DNA release dynamics from bioreducible
number within poly(glycoamidoamine)s affect intracellular DNA layer-by-layer films. Langmuir 26, 8597–8605
delivery. J. Am. Chem. Soc. 127, 3004–3015 43 Prevette, L.E. et al. (2010) Amide spacing influences pDNA binding of
18 Popielarski, S.R. et al. (2003) Structural effects of carbohydrate- poly(amidoamine)s. Biomacromolecules 11, 326–332
containing polycations on gene delivery. 3. Cyclodextrin type and 44 Prevette, L.E. et al. (2008) Correlation of amine number and pDNA
functionalization. Bioconjug. Chem. 14, 672–678 binding mechanism for trehalose-based polycations. Langmuir 24,
19 Shcharbin, D. et al. (2009) How to study dendriplexes I: 8090–8101
characterization. J. Control. Release 135, 186–197 45 Kizjakina, K. and Reineke Theresa, M. (2007) Effects of secondary
20 Prevette, L.E. et al. (2007) Deciphering the role of hydrogen bonding in amine number in trehalose click polymers for transfection. Polym.
enhancing pDNA–polycation interactions. Langmuir 23, 9773–9784 Mater. Sci. Eng. 96, 646
21 Bartlett, D.W. and Davis, M.E. (2007) Physicochemical and biological 46 Srinivasachari, S. et al. (2006) Trehalose click polymers inhibit
characterization of targeted, nucleic acid-containing nanoparticles. nanoparticle aggregation and promote pDNA delivery in serum. J.
Bioconjug. Chem. 18, 456–468 Am. Chem. Soc. 128, 8176–8184
22 Lai, E. and van Zanten, J.H. (2001) Monitoring DNA/poly-l-lysine 47 Taori, V.P. et al. (2009) DNA delivery in vitro via surface release from
polyplex formation with time-resolved multiangle laser light multilayer assemblies with poly(glycoamidoamine)s. Acta Biomat. 5,
scattering. Biophys. J. 80, 864–873 925–933
23 Li, C. et al. (2009) PAMAM nanoparticles promote acute lung injury by 48 Liu, Y. et al. (2006) Gene delivery with novel poly(l-tartaramidoamine)s,
inducing autophagic cell death through the Akt-TSC2-mTOR signaling In Polymeric Drug Delivery I, American Chemical Society, pp. 217–227
pathway. J. Mol. Cell Biol. 1, 37–45 49 Reineke, T.M. and Davis, M.E. (2002) Structural Effects of
24 Aberle, A.M. et al. (1996) The counterion influence on cationic lipid- Carbohydrate-Containing Polycations on Gene Delivery. 1.
mediated transfection of plasmid DNA. Biochim. Biophys. Acta 1299, Carbohydrate Size and Its Distance from Charge Centers. Bioconj.
281–283 Chem. 14, 247–254
25 Asgatay, S. et al. (2004) Polynorbornene polycationic polymers as gene 50 Bryson, J.M., et al. Polymer beacons for luminescence and magnetic
transfer agents: influence of the counterion for in vitro transfection. Int. resonance imaging of DNA delivery. Proc. Natl. Acad. Sci. U.S.A.
J. Pharm. 285, 121–133 51 Bryson, J.M. et al. (2008) A b-Cyclodextrin ‘‘Click Cluster’’ Decorated
26 Lee, C-C. et al. (2008) General structure–activity relationship for with Seven Paramagnetic Chelates Containing Two Water Exchange
poly(glycoamidoamine)s: the effect of amine density on cytotoxicity Sites. Bioconj. Chem. 19, 1505–1509
and DNA delivery efficiency. Bioconjug. Chem. 19, 428–440 52 Lucas, R.L. et al. (2007) Comparison of a Tartaric Acid Derived
27 Vicennati, P. et al. (2008) Polyethylenimine in medicinal chemistry. Polymeric MRI Contrast Agent to a Small Molecule Model Chelate.
Curr. Med. Chem. 15, 2826–2839 Bioconj. Chem. 19, 24–27
28 Fischer, D. et al. (1999) A novel non-viral vector for DNA delivery based 53 Reineke, T.M. and Davis, M.E. (2002) Structural Effects of
on low molecular weight, branched polyethylenimine: effect of Carbohydrate-Containing Polycations on Gene Delivery. 2. Charge
molecular weight on transfection efficiency and cytotoxicity. Pharm. Center Type. Bioconj. Chem. 14, 255–261
Res. 16, 1273–1279 54 Heidel, J.D. et al. (2007) Administration in non-human primates of
29 Godbey, W.T. et al. (1999) Size matters: molecular weight affects the escalating intravenous doses of targeted nanoparticles containing
efficiency of poly(ethylenimine) as a gene delivery vehicle. J. Biomed. ribonucleotidereductase subunit M2 siRNA. Proc. Natl. Acad. Sci.
Mater. Res. 45, 268–275 U.S.A. 104, 5715–5721
30 Huang, M. et al. (2005) Transfection efficiency of chitosan vectors: effect 55 Pun, S.H. et al. (2004) Cyclodextrin-Modified Polyethylenimine
of polymer molecular weight and degree of deacetylation. J. Control. Polymers for Gene Delivery. Bioconj. Chem. 15, 831–840
Release 106, 391–406 56 Arima, H. et al. (2001) Enhancement of Gene Expression by
31 Liu, Y. and Reineke, T.M. (2010) Degradation of Polyamidoamine Dendrimer Conjugates with a-,b-, and g-
poly(glycoamidoamine) DNA delivery vehicles: polyamide hydrolysis Cyclodextrins. Bioconj. Chem. 12, 476–484
at physiological conditions promotes DNA release. Biomacromolecules 57 Cryan, S-A. et al. (2004) Cell transfection with polycationic cyclodextrin
11, 316–325 vectors. Eur. J. Pharm. Sci. 21, 625–633
32 Liu, Y. et al. (2004) New poly(d-glucaramidoamine)s induce DNA 58 Davis, M.E. et al. (2004) Self-Assembling Nucleic Acid Delivery
nanoparticle formation and efficient gene delivery into mammalian Vehicles via Linear, Water-Soluble, Cyclodextrin-Containing
cells. J. Am. Chem. Soc. 126, 7422–7423 Polymers. Curr. Med. Chem. 11, 179–197
33 Reineke, T.M. (2006) Poly(glycoamidoamine)s: cationic 59 Hwang, S.J. et al. (2001) Effects of Structure of b-Cyclodextrin-
glycopolymers for DNA delivery. J. Polym. Sci. Part A: Polym. Containing Polymers on Gene Delivery. Bioconj. Chem. 12, 280–290
Chem. 44, 6895–6908 60 Dı́az-Moscoso, A., et al. Insights in cellular uptake mechanisms of
34 McLendon, P.M. et al. (2010) Interaction of poly(glycoamidoamine) pDNA-polycationic amphiphilic cyclodextrin nanoparticles
DNA delivery vehicles with cell-surface glycosaminoglycans leads to (CDplexes). J. Control. Release, in press, corrected proof
polyplex internalization in a manner not solely dependent on charge. 61 Lee, C-C. et al. (2010) A Polycation Scaffold Presenting Tunable ‘‘Click’’
Mol. Pharm. 7, 1757–1768 Sites: Conjugation to Carbohydrate Ligands and Examination of
35 Bryson, J. (2010) Novel twist on primary cell transfection. Genet. Eng. Hepatocyte-Targeted pDNA Delivery. Macromol. Biosci. 10, 585–598
Biotechnol. News 30 62 Kim, T-H. et al. (2007) Chemical modification of chitosan as a gene
36 Liu, Y. and Reineke, T.M. (2006) Poly(glycoamidoamine)s for gene carrier in vitro and in vivo. Prog. Polym. Sci. 32, 726–753
delivery: stability of polyplexes and efficacy with cardiomyoblast 63 Jiang, H-L. et al. (2008) Galactosylatedpoly(ethylene glycol)-chitosan-
cells. Bioconjug. Chem. 17, 101–108 graft-polyethylenimine as a gene carrier for hepatocyte-targeting. J.
37 Srinivasachari, S. and Reineke, T.M. (2009) Versatile supramolecular Control. Release 131, 150–157
pDNA vehicles via ‘click polymerization’ of [beta]-cyclodextrin with 64 Dehousse, V. et al. (2010) Comparison of chitosan/siRNA and
oligoethyleneamines. Biomaterials 30, 928–938 trimethylchitosan/siRNA complexes behaviour in vitro. Int. J. Biol.
38 Srinivasachari, S. et al. (2008) Polycationic b-cyclodextrin ‘click Macromol. 46, 342–349
clusters’: monodisperse and versatile scaffolds for nucleic acid 65 Takeda, Y. et al. (2007) Ternary Complex Consisting of DNA,
delivery. J. Am. Chem. Soc. 130, 4618–4627 Polycation, and a Natural Polysaccharide of Schizophyllan to Induce
39 Carr, L.R. and Jiang, S. (2010) Mediating high levels of gene transfer Cellular Uptake by Antigen Presenting Cells. Biomacromolecules 8,
without cytotoxicity via hydrolytic cationic ester polymers. 1178–1186
Biomaterials 31, 4186–4193 66 Eshita, Y. et al. (2009) Mechanism of Introduction of Exogenous Genes
40 Reynolds, A. et al. (2004) Rational siRNA design for RNA interference. into Cultured Cells Using DEAE-Dextran-MMA Graft Copolymer as
Nat. Biotechnol. 22, 326–330 Non-Viral Gene Carrier. Molecules 14, 2669–2683
41 Boussif, O. et al. (1995) A versatile vector for gene and oligonucleotide 67 Nimesh, S. et al. (2006) Novel polyallylamine-dextran sulfate-DNA
transfer into cells in culture and in vivo: polyethylenimine. Proc. Natl. nanoplexes: Highly efficient non-viral vector for gene delivery. Int. J.
Acad. Sci. U.S.A. 92, 7297–7301 Pharm. 320, 143–149

452
Review Trends in Biotechnology September 2011, Vol. 29, No. 9

68 Kang, H.C. et al. (2010) Trafficking microenvironmentalpHs of 74 Sizovs, A. et al. (2010) Carbohydrate polymers for nonviral nucleic acid
polycationic gene vectors in drug-sensitive and multidrug-resistant delivery. In Nucleic Acid Transfection (Bielke, W. and Erbacher, C.,
MCF7 breast cancer cells. Biomaterials 31, 3071–3078 eds), pp. 131–190, Berlin/Heidelberg, Springer
69 O’Rorke, S. et al. (2010) Non-viral polyplexes: Scaffold mediated 75 Agarwal, S., et al. (2010) Synthetic Route and Characterization of Main
delivery for gene therapy. Prog. Polym. Sci. 35, 441–458 Chain Ester-Containing Hydrolytically Degradable Poly(N,N-
70 Shim, M.S. and Kwon, Y.J. (2010) Acid-transforming polypeptide dimethylaminoethyl methacrylate)-Based Polycations. Macromol.
micelles for targeted nonviral gene delivery. Biomaterials 31, 3404–3413 Chem. Phys. 9999, NA
71 Vachutinsky, Y., et al. Antiangiogenic gene therapy of experimental 76 Filippov, S.K., et al. (2010) Effect of Hydrophobic Interactions on
pancreatic tumor by sFlt-1 plasmid DNA carried by RGD-modified Properties and Stability of DNAPolyelectrolyte Complexes.
crosslinked polyplex micelles. J. Control. Release, in press, corrected proof Langmuir
72 Zhang, H. and Vinogradov, S.V. Short biodegradable polyamines for 77 Ma, M. et al. (2010) Influence of hydroxyl groups on the biological
gene delivery and transfection of brain capillary endothelial cells. J. properties of cationic polymethacrylates as gene vectors.
Control. Release, in press, corrected proof ActaBiomaterialia 6, 2658–2665
73 Zhang, J. et al. (2010) HostGuest Interaction Mediated Polymeric 78 Schallon, A. et al. (2010) Performance of three PDMAEMA-based
Assemblies: Multifunctional Nanoparticles for Drug and Gene polycation architectures as gene delivery agents in comparison to
Delivery. ACS Nano 4, 1049–1059 linear and branched PEI. React. Funct. Polym. 70, 1–10

453

You might also like