Polymer

Download as pdf or txt
Download as pdf or txt
You are on page 1of 27

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/284158672

Synthetic Polymer-based Nanomaterials

Chapter · November 2015


DOI: 10.1007/978-1-4939-3121-7_1

CITATIONS READS

3 1,653

3 authors:

Swapnil Desale Jinjin Zhang


AMRI University of Nebraska Medical Center
15 PUBLICATIONS 708 CITATIONS 11 PUBLICATIONS 221 CITATIONS

SEE PROFILE SEE PROFILE

Tatiana Bronich
University of Nebraska Medical Center
135 PUBLICATIONS 9,808 CITATIONS

SEE PROFILE

All content following this page was uploaded by Swapnil Desale on 18 November 2015.

The user has requested enhancement of the downloaded file.


Chapter 1

Synthetic Polymer-based Nanomaterials


Swapnil S. Desale, Jinjin Zhang, and Tatiana K. Bronich

Abstract
Synthetic polymers play an essential role in the development of conventional pharmaceutical formulations
as well as devices for controlled drug delivery and represent the most commonly used “building blocks”
for engineering of various nanomedicines. In this chapter we review several classes of synthetic polymers
that are widely used for the design of drug delivery systems, synthetic methodologies to tailor their physi-
cochemical properties, and therapeutic applications and developments.

Key words Nanomedicines, Polymers, Polymerization, Nanoparticles, Drug delivery

1 Introduction

Nanomedicine is a rapidly developing area of biomedical research


that uses devices of nanoscale size to address urgent needs for effec-
tively detecting diseases and improving drug and gene delivery. Such
nanomaterial (NM)-based delivery vehicles have the ability to
improve drug pharmacokinetics, biodistribution, cell- or tissue-spe-
cific targeting, and drug exposure kinetics, resulting in enhanced
efficacy and improved tolerability [1–3]. The most sophisticated
nanocarriers can simultaneously deliver multiple therapeutic and/or
imaging agents and thus enable both diagnosis and therapy. Polymers
are playing an essential role in the development of conventional
pharmaceutical formulations as well as devices for controlled drug
delivery and represent the most commonly used “building blocks”
for engineering of the NM-based drug delivery systems. The
advances in synthetic polymer chemistry led to the exceptional diver-
sity and control over the composition, architecture and functionality
of the polymers, which in turn enables the building of NM with
tunable properties for various biomedical applications. This chapter
first highlights several classes of synthetic polymers, which are cur-
rently the most widely used for the design of NM-based drug deliv-
ery systems. The remainder of the chapter focuses on some examples
of polymeric NMs and their applications in drug and gene delivery.

Zheng-Rong Lu and Shinji Sakuma (eds.), Nanomaterials in Pharmacology, Methods in Pharmacology and Toxicology,
DOI 10.1007/978-1-4939-3121-7_1, © Springer Science+Business Media New York 2016

1
2 Swapnil S. Desale et al.

2 Synthetic Polymers

Polymers can be classified as homopolymers and copolymers based


on their composition. Homopolymer is defined as the polymer con-
taining a single type of repeating units, while copolymer is com-
posed of two or more different monomers. Both homopolymers
and copolymers have been used extensively for drug delivery appli-
cations. The development of these polymer-based drug delivery sys-
tems relies on novel polymeric architectures and appropriate
synthetic methodologies to tailor their physicochemical properties.

3 Polyesters

Among all degradable polymers, aliphatic polyester-based poly-


meric structures are receiving special attention because of their
compatibility with the natural environment and their ability to
undergo hydrolytic and biological degradation [4]. Among the
wide variety of aliphatic polyesters, poly(D,L-lactide-co-glycolide)
(PLGA), poly(lactic acid) (PLA), poly(caprolactone) (PCL) are
the most commonly used polymers for controlled drug release
applications. There are two methods of the synthesis of aliphatic
polyesters: polycondensation of diols and dicarboxylic acids or
hydroxycarboxylic acids and ring-opening polymerization (ROP)
of cyclic monomers (Scheme 1) [5, 6]. The polycondensation is
hampered by drawbacks such as typically required high tempera-
tures and long reaction times that favor side reactions, necessity of
continuous water removal to achieve high conversions and high
molecular weights. The polymers obtained in this process are char-
acterized by high polydispersity. ROP yields polymer products
with high molecular weight, lower polydispersity and, therefore, is
a preferred route to obtain aliphatic polyesters [7, 8]. Various
organometallic compounds, such as oxides, carboxylates, and
alkoxides are used as effective catalysts for the controlled synthesis

a
O O O O
+ n HO R2 OH C C R2 + 2n H2O
n HO R1 OH R1 O O n

b O O
Sn(Oct)2 O
O O
ROH + n + m Heating
RO
O
O
H
O O 2n 2m
O
O O
DL-lactide glycolide PLGA

Scheme 1 (a) Synthesis of polyesters by polycondensation of dicarboxylic acids and diols; (b) synthesis of
PLGA copolymer by ROP using alcohol as an initiator and stannous octoate as the catalyst
Synthetic Polymer-based Nanomaterials 3

of polyesters using ROP [9]. Alternately, polyesters without toxic


metallic residuals, which is an important requirement for biomedi-
cal and pharmaceutical applications, can be prepared by using
enzyme-catalyzed polymerizations [10]. Unlike chemical catalysts,
enzymes function under mild conditions, enable high enantiose-
lectivity and regioselectivity, and are recyclable. Extensive research
efforts have been expended in the past years to refine the ROP
technique for the synthesis of the polyesters with controlled archi-
tecture and tailor-made properties. To obtain a polymer with a par-
ticular combination of desirable properties, copolymerization
techniques have been extensively used. Controlling the
composition, morphology, glass transition temperature, and degra-
dation rate of the copolymers allows regulation of the drug release
behavior from such matrices. Nanoparticles formulated from bio-
degradable polyesters are of great interest for drug delivery pur-
poses. A wide variety of hydrophilic or hydrophobic drugs and
biological macromolecules can be encapsulated into biodegradable
polyesters-based nanoparticles and delivered to specific organs or
cells [11]. A number of techniques are available for the formula-
tion of polyester-based nanoparticles (or nanocapsules). The choice
of a particular approach mainly depends on the physicochemical
properties of the polymer (i.e., solubility and molecular weight)
and the drug physicochemical properties (i.e., hydrophobicity/
hydrophilicity, sensitivity to the solvent). Currently, the most pop-
ular methods are emulsion/solvent evaporation or diffusion, dou-
ble emulsion, nanoprecipitation, and the salting out procedure.
The emulsion/solvent evaporation method involves the prepara-
tion of an oil-in-water emulsion, where a small quantity of water-
immiscible organic solvent containing polymer and hydrophobic
drug (oil phase) is emulsified in an aqueous phase containing a
stabilizer [12, 13]. The most common stabilizers are hydrophilic
molecules such as polyvinyl alcohol (PVA), polysorbates
(TWEEN®), or sodium cholate. Stable nanoparticles are formed in
the aqueous phase by organic solvent evaporation under increased
temperature or reduced pressure. In the emulsion/diffusion
method, the oil phase containing polymer is dissolved in a partially
water-miscible solvent (e.g., ethyl acetate, propylene carbonate)
[14, 15]. The addition of a certain volume of water to the oil-in-
water emulsion induces a change in the equilibrium of the system
and causes the partially water-miscible solvent to diffuse from the
droplets into the aqueous phase. This reduces the polymer’s solu-
bility and results in particle formation. Another approach is the
double-emulsion, W/O/W, method [16, 17]. The main benefit of
the double-emulsion method is its ability to efficiently encapsulate
hydrophilic drugs and proteins. In this approach, the drug, dis-
solved in water, is added to an organic solvent containing polymer,
forming a water-in-oil emulsion. Then a small quantity of this ini-
tial emulsion is added to a second aqueous phase containing an
4 Swapnil S. Desale et al.

emulsifier, such as PVA, to stabilize the particles. Nanoparticles are


then obtained by evaporation of the organic solvent. Typically this
method yields nanoparticles with larger sizes than single emulsion
method with moderate drug loading and encapsulation efficiency.
Nanoprecipitation (or solvent displacement) method utilizes inter-
facial polymer deposition to form nanoparticles [18–20]. In this
simple process, polymer and a drug are dissolved in a water-miscible
solvent and this solution is then added dropwise to an aqueous
solution (non-solvent) with or without stabilizer. Solvent diffusion
results in polymer precipitation on the interface between the
aqueous phase and finely dispersed oil droplets, resulting in the
formation of solid particles. Depending on the solvent choice and
solvent–non-solvent ratio, this method is suitable for encapsulation
of both hydrophilic and hydrophobic drugs as well as protein-
based therapeutics. In general, nanoparticles of smaller sizes but
with lower entrapment efficiencies are obtained through this
method when compared to other methods. Finally, the salting out
method is an oil-in-water emulsion comprised of a primary aque-
ous phase containing stabilizer and a high concentration of salt
(e.g., magnesium chloride hexahydrate) and polymer dissolved in a
water-miscible solvent, such as acetone or tetrahydrofuran [21].
Due to the presence of a high concentration of salt, there is no dif-
fusion of the solvent into the aqueous phase. Fast addition of a
large amount of water to this oil-in-water emulsion reduces the
ionic strength and leads to the migration of the water-miscible
organic solvent into the aqueous phase, inducing formation of
solid particles. In this method, particles must be purified to remove
residual salt and solvent prior to use. A central challenge in the
development of drug-encapsulated polymeric nanoparticles is the
inability to control the mixing processes required for their synthe-
sis, which results in variable nanoparticle physicochemical proper-
ties (size, surface composition, and drug loading). To address this
challenge the Langer group [22] developed a rapid and tunable
mixing procedure utilizing hydrodynamic flow focusing in micro-
channels to control nanoprecipitation and prepare nanoparticles in
a reproducible manner. The microfluidic technique allows tuning
the size of the resulting particles by varying flow rates, polymer
composition, molecular weight or polymer concentration in
organic solution. Remarkably, higher drug encapsulation was also
reported for the polymeric nanoparticles prepared through micro-
fluidics [22]. Recently, DeSimone and colleagues have developed a
unique nanofabrication process called Particle Replication In
Nonwetting Templates (PRINT) [23]. Using soft-lithography
techniques adopted from the semiconductor industry, PRINT
enables the production of monodisperse polymeric nanoparticles
with well-defined control over particle size, shape, composition,
and surface chemistry, and permits the loading of a wide range of
cargoes with high loading efficiency [24, 25].
Synthetic Polymer-based Nanomaterials 5

4 Polyanhydrides

Polyanhydrides, polymers containing hydrolytically labile anhy-


dride linkages in hydrophobic backbone, have been investigated
for more than three decades as important biomaterials used for
short-term release of drugs [26]. Polyanhydrides are characterized
by their fast degradation followed by rapid erosion of material, but
at the same time can be designed to release drugs that last from
days to weeks by suitable choice of monomers. Numerous poly-
mers have been synthesized in this class of material by various
polymerization techniques such as melt condensation, ROP, inter-
facial condensation, dehydrochlorination, and dehydrative cou-
pling agents [27]. The most convenient method of synthesizing
high molecular weight polyanhydride copolymer is by the melt
polycondensation of anhydride prepolymers (Scheme 2) [28, 29].
Solution polymerizations at ambient temperature are typically
utilized to prepare polyanhydrides from heat-sensitive monomers
and generally yield low molecular weight polymers. The degrada-
tion of polyanhydrides is a hydrolytically triggered process and
depends on the uptake of water into the polymer matrix and pH of
the medium. The rate of water uptake is dependent on factors such
as crystallinity, polymer composition, and molecular weight. It was
reported that polyanhydrides derived from monomers containing
aromatic moieties degrade much slower than aliphatic polyanhy-
drides [30]. Thus, the degradation rate can be tuned by control-
ling the composition of the corresponding copolymers. They
degrade in vitro as well as in vivo to the nontoxic consistent dicar-
boxylic acids. Importantly, the materials based on the polyanhy-
drides show no evidence of inflammatory reaction [31].
Polyanhydrides constitute the only class of surface eroding poly-
mers approved for clinical trial use by the Food and Drug
Administration (FDA). One of the commonly used techniques for
the preparation of polyanhydride-based nanoparticles is the solvent
displacement method followed by freeze or spray drying [32–34].
Another method known as an anti-solvent nanoprecipitation tech-
nique utilized by Ulery et al. [35] and others [36–38] allows fab-
rication of the nanoparticles with relatively uniform size and shape.
In biomedical research, polyanhydride-based nanoparticles have
been widely explored as a vaccine adjuvant because of their ability
to induce potent immune response in a pathogen-mimicking man-
ner without side effects [37, 39–42].

O O
O O O O O O O O O O
O Vacuum
HO R OH Reflux O R O Heating O R O n

Scheme 2 Synthesis of polyanhydride prepolymer and polymer from a dicarboxylic acid


6 Swapnil S. Desale et al.

5 N-(2-Hydroxypropyl) Methacrylamide (HPMA) Copolymers

Among synthetic polymeric drug carriers, the water-soluble HPMA


copolymers are the most studied for the last 40 years [43]. HPMA
polymer–drug conjugates have been developed as nanomedicines
for delivery of a number of therapeutics, including anticancer and
anti-inflammatory agents [44–46]. HPMA polymers and their drug
conjugates were initially synthesized by conventional free radical
polymerization techniques. Recent advances in living radical polym-
erization methods, including atom transfer radical polymerization
(ATRP) and reversible addition-fragmentation chain transfer
(RAFT), allowed for the controlled synthesis of well-defined HPMA
copolymers with narrow molecular weight distributions (Scheme 3)
[47, 48].
HPMA copolymers have many attributes that make them ide-
ally suited for the development as drug carriers. These polymers
are biocompatible, non-immunogenic and can be tailored to the
characteristics of the specific target. The molecular weight of
HPMA copolymers can be adjusted to alter biodistribution, tissue
localization and elimination from the body. The proper selection
of the spacer between the drug and HPMA carrier (e.g., oligopep-
tide sequence, hydrazone bond, cis-aconityl spacer) allowed for
controlled drug release within the tissue [49–52]. The HPMA
copolymer conjugates containing doxorubicin were the first syn-
thetic polymer-based therapeutics to enter clinical trials. Since
then, numerous conjugates have been synthesized to contain drug
or drug combinations, proteins and peptides, as well as targeting
moieties and/or imaging probes. In addition, HPMA polymers
were used for the modification of proteins, surface modification of
biomaterials, masking and retargeting of therapeutic viruses and as
macromolecular platforms for contrast agents.

O
CH2 C n
O O N Br
O O
HN HN
CuCl, bpy, 50 oC
80:20 IPA/H2O
OH OH

b
O S

HO CH2 Cn S
O
CTP, V501 O
HN CN
Acetic Buffer, pH 5.2,70 oC HN

OH
OH

Scheme 3 Synthesis of HPMA polymer by (a) ATRP and (b) RAFT polymerization methods
Synthetic Polymer-based Nanomaterials 7

6 Polyethylenimine (PEI)

PEIs are synthetic cationic polymers that have been widely employed
as the most prominent polynucleotide delivery systems [53].
Depending on the linkage of the repeating ethylenimine units, PEI
possesses either branched or linear topology (Scheme 4). Branched
PEI is synthesized by polymerization of aziridine either in aqueous
or alcoholic solutions [54–56], where the reaction is controlled by
adjusting the temperature and initiator concentration, or in a rather
vigorous bulk polymerization of anhydrous aziridine at a lower tem-
perature [57]. Linear PEI has been synthesized via cationic ROP of
either N-(2-tetrahydropyranyl)azidirine [58] or unsubstituted and
2-substituted 2-oxazolines followed by acid or base-catalyzed hydro-
lysis of the corresponding N-substituted polymer [59, 60].
The efficiency of PEI as transfection agent as well as its cyto-
toxicity is closely linked to the polymer characteristics such as
molecular weight, the degree of branching, charge density and
buffering capacity. Generally, the low molecular weight linear or
branched PEIs have low cytotoxicity but display poor transfection
efficiency. In order to solve the efficiency versus toxicity problems,
cross-linked PEIs were synthesized to include biodegradable cross-
links that facilitate an increase in molecular weight but then follow-
ing cross-link degradation, allowing the release of lower molecular
weight components to reduce cytotoxicity [61]. Alternatively,
star-like PEI derivatives with high charge density have been
designed; these polymers utilize low molecular weight PEIs that
are conjugated to a central multivalent polymer core [62–64].

7 Amino Acid-based Polymers

Polymers based on natural L-amino acids are an attractive class of


materials given their biocompatibility, controlled biodegradability,
and metabolizable degradation products. Incorporation of amino

a
H2N NH
N NH2
H
N
N N
H2N N N
H H n

b
N MeOTs N Hydrolysis
n N
O n
O H

Scheme 4 Synthesis of (a) branched PEI by acid catalyzed polymerization of


aziridine and (b) linear PEI by ROP of 2-ethyl-2-oxazoline
8 Swapnil S. Desale et al.

H2N CO2H O O O O
Triphosgene R'-NH2
CH R' NH C CH NH n
HN
R R R

Scheme 5 Synthesis of polypeptides via α-amino acid N-carboxyanhydride ROP

acids as a building block into synthetic polymers not only allows


for adjusting hydrophilic/hydrophobic properties of the resulting
polymers and tune their degradability but also imparts chemical
functionalities to facilitate further modification with bioactive mol-
ecules (e.g., drugs, imaging probes, or targeting ligands). The
most common and efficient method to produce polypeptides of
sufficient molecular weight is ROP of N-carboxyanhydrides
(NCAs) [65]. NCAs are easily synthesized through a reaction
between amino acids and phosgene or a phosgene derivative (e.g.,
triphosgene), and the polymerization is commonly initiated using
amino-functional molecules (Scheme 5).
In recent years, the advances in NCA polymerizations, either
using metal initiators or improved conventional initiators, allowed
for the synthesis of broad range of block copolymers and side-chain
functionalized polypeptides with controlled characteristics (molec-
ular weight, sequence, composition, and molecular weight distri-
bution) [66]. In addition, the diversity of synthetic polymer
chemistries and “click” reactions offer the ability to covalently link
a variety of different polymers together and prepare hybrid materi-
als with functional macromolecular architectures and tunable phys-
icochemical properties to meet various requirements of biomedical
applications.

8 Dendrimers

Dendrimers are a unique class of polymeric materials most fre-


quently synthesized using divergent or convergent strategies by a
series of controlled polymerization reactions [67]. Divergent syn-
thesis grows outwards from a multifunctional core molecule and
comprises of two steps: first is the activation of functional surface
groups, and second is the addition of branching monomer units.
The process is repeated for several generations and a dendrimer is
built layer after layer. While this method is successful for the pro-
duction of relatively large quantities of dendrimers, problems
occur from side reactions and incomplete reactions of the end
groups that lead to structural defects. Convergent synthesis, on
the other hand, starts at the surface and proceeds inwards before
the attachment of the synthesized dendrons to the core [68]. In
the convergent growth, a small number of reactive sites are func-
tionalized in each step resulting in less number of side reactions
and more precise control over molecular weight of the dendrimer.
Synthetic Polymer-based Nanomaterials 9

However, convergent synthesis strategy is generally limited to the


construction of lower-generation dendrimers due to steric difficul-
ties upon attachment of the dendrons to the core molecule. In
contrast to other synthetic polymers, dendrimers are nearly mono-
disperse in molecular weight and size. They are highly branched
macromolecules of nanoscale molecular size with multiple surface
group functionalities and practically perfect spherical topology in
case of higher generations. Functionalization of their terminal
groups provides an exceptional opportunity for the immobilization
of multiple bioactive molecules and solubilizing groups in a spe-
cific and controllable manner. Alternatively, the numerous internal
cavities in the dendrimer cores can be loaded with various drugs via
hydrophobic interaction, hydrogen bonds or chemical conjuga-
tion. Surface modification of the dendrimer by a modification with
polyethylene glycol (PEG), acetylation or esterification is often
used to improve the drug loading capacity of the resulting den-
drimers. It also renders them more biocompatible, less immuno-
genic, and less toxic, especially those decorated by free amino
functionalities [69–71]. Because of these characteristics, den-
drimers have been widely studied as nanoscale containers for deliv-
ery of therapeutic payloads. The flexibility to tailor both the core
and surface of these systems allows optimizing the properties of
drug carriers for the specific applications. Dendrimers of various
structure, amphiphilicity, and architecture were explored as carriers
for anticancer, antimalarial, antiviral, antitubercular, antimicrobial,
and antihypertensive drugs [72]. Their application as scaffolds of
prodrugs is particularly interesting [73–75]. Furthermore, several
groups addressed the strategies for the synthesis of the degradable
dendrimers that can achieve high accumulation and retention in
diseased tissues, but allow rapid and safe elimination of nontoxic
dendrimer fragments [76]. Biodegradable dendrimers are com-
monly prepared by inclusion of ester groups in the polymer back-
bone, which will be chemically hydrolyzed and/or enzymatically
cleaved by esterases in physiological solutions [77]. For example,
Frechet and colleagues reported efficient synthesis of robust and
biodegradable PEGylated dendrimer based on a polyester–polyam-
ide hybrid core. The architecture has been designed to avoid
destructive side reactions during the synthesis while maintaining
the dendrimer’s degradability [78]. Polycationic dendrimers can
be complexed with nucleic acids and used for gene therapy.
Applications of dendrimers as protein mimics [79], biomimetic
regeneration of hydroxyapatite [80], and mimicking in angiogenesis
[81] has also been reported. Furthermore, dendrimers can act as
“nano-drugs” themselves and their therapeutic potential has been
explored against various diseases [82]. As an example, dendrimer-
based nanomedicine mixed in carbomer gel (VivaGel®) is being
developed as a vaginal microbicide gel to prevent the transmission
of genital herpes and human immunodeficiency virus [83].
10 Swapnil S. Desale et al.

The active ingredient of this gel is a dendrimer comprising a divalent


benzhydrylamine core and four generations of lysine branches
capped with sulfonated naphthyl groups that impart hydrophobicity,
and a high anionic charge to the dendrimer surface. Initial human
trials have shown VivaGel to be safe and well-tolerated, and Phase II
clinical studies for its efficacy are ongoing. Despite the promise of
dendrimers-based drug delivery systems, their translation into actual
therapies is challenging due to their lengthy synthesis and the need
to develop nontoxic and biocompatible dendrimers.

9 Polymer Therapeutics

The term “Polymer Therapeutics” was coined by Prof. Ruth Duncan


to define a family of new chemical entities that comprises a variety of
complex macromolecular systems containing a water-soluble poly-
meric carrier covalently bound to the bioactive molecule(s). These
polymeric drugs include polymer–drug and polymer–protein conju-
gates, polymeric micelles where drug is covalently bound to the
polymer, and polyplexes (containing covalent linkers) developed as
nonviral vectors for the delivery of nucleic acids [84]. Polymer is an
integral and functional part of such multifunctional systems for
improved drug, protein and gene delivery [30, 85, 86]. The linkers
are typically sensitive to the conditions that are unique for the tar-
geted site (e.g., an acidic environment, presence of specific enzyme),
which increases the specificity of drug delivery and release. In addi-
tion, a targeting moiety may also be introduced into the conjugate
to increase its therapeutic index. PEGylation has become a well-
established technology for the use of proteins as drugs. When
attached to a protein, peptide and more recently to an aptamers,
typically via conjugation of a monomethoxylated PEG segment
bearing a reactive moiety at one of the polymer termini, PEG imparts
prolonged blood residency and diminished immunogenicity of the
bioconjugate. The most studied polymer–drug conjugates are based
on HPMA copolymer and PEG backbones and, initially, their design
was focused on cancer treatments and incorporated common che-
motherapeutic agents (e.g., doxorubicin, taxanes, camptothecin, or
platinates) [87]. Over the years, numerous conjugates have been
synthesized which contain drugs that act on the emerging targets for
cancer therapy such as angiogenesis, apoptotic pathways, kinases and
others. As an example, HPMA–fumagillol, the first antiangiogenic
conjugate, proved to be effective at inhibiting tumor growth and
exhibited a significantly better toxicity as compared to free drug
[88]. It was demonstrated that HPMA copolymer–wortmannin
conjugate retained the ability to inhibit type I PI3-kinase activity
[89], and HPMA–geldanamycin conjugate inhibited the capacity of
heat shock proteins such as HSP-90 to form complexes with client
oncoproteins [90, 91]. Therapies focusing on the activation of apop-
Synthetic Polymer-based Nanomaterials 11

totic pathways are also promising anticancer strategies [92]. The


PEGylation of curcumin, a Jab1 inhibitor, and the conjugation of
Bcl2-inhibitor HA14 to HPMA, are two examples of this proapop-
totic approach [93, 94]. In addition, polymer–drug conjugates were
actively explored for the treatment of other diseases including infec-
tions [95], inflammation [96], rheumatoid arthritis [97] and diabe-
tes [98]. Several preclinical studies have already illustrated the
potential of conjugates in regenerative medicine, for example, for
wound healing [99], ischemia [100], or osteoporosis [101]. The
new concepts such as delivering drug combinations via one polymer
carrier [102], exploring new polymer architectures (branched,
grafted, and star polymers and dendrimers) or use of coiled-coil pep-
tides motifs as linkers [103] are the recent developments in the field
of polymer therapeutics. To address the concerns regarding the pos-
sible accumulation of the nondegradable polymers in the body, an
increasing number of biodegradable polymers such as poly(glutamic
acid) (PGA) [104], polyacetal Fleximer [105], poly(malic acid)
[106], dextrin [107] have been investigated as platforms for the
design of new polymer therapeutics. Translational research in poly-
mer therapeutics has yielded ten marketed PEG-protein/aptamer
conjugates [105, 108]. In the case of polymer–drug conjugates
progression to regulatory approval has been slower. The closest to
market is Opaxio™ (PGA-paclitaxel conjugate, also known as
Xyotax or CT-2103) developed by Cell Therapeutics Inc. [http://
www.celltherapeutics.com/opaxio], which is in advanced Phase III
clinical trials. Opaxio™ sister conjugate CT-2106, PGA-
camptothecin conjugate, is also in Phase II clinical trial. Promising
results are emerging from a number of ongoing clinical studies
involving anticancer conjugates such as ProLindac™, an HPMA
copolymer DACH-platinate from Access Pharmaceuticals that has
successfully completed a European Phase II clinical trial in patients
with ovarian cancer [109]. PEG-irinotecan conjugate (NKTR-
102) is undergoing Phase III evaluation in patients with metastatic
breast cancer and is also being studied in Phase II clinical trials in
ovarian and colorectal cancer [http://Nektar.com]. Mersana is
commercializing XMT1001 (Fleximer®-camptothecin conjugate)
as its lead candidate but has also a potent antiangiogenic conjugate
XMT1107 (Fleximer®-fumagillin) in Phase I studies. Overall, the
design and clinical development of new polymer therapeutics hold
a significant potential of this group of NMs for targeting a wide
range of the diseases.

10 Polymeric Micelles

Amphiphilic block or graft copolymers comprised of two or more


chains with different hydrophobicity have been used extensively
in pharmaceutical applications ranging from sustained-release
12 Swapnil S. Desale et al.

technologies to gene delivery. These copolymers are known to


spontaneously self-assemble in an aqueous solution into nano-
scopic polymeric micelles (10–100 nm) having fairly narrow size
distribution. The nature of the self-assembly process allows for sig-
nificant versatility in the chemical composition of the polymeric
micelles and thus permits fine tuning of the material properties,
morphology and sizes. These micelles have unique core–shell
architectures with hydrophobic polymer chains segregating into a
micelle core surrounded by a shell of hydrophilic chains.
Hydrophobic drugs can be entrapped into the micelle core non-
specifically through hydrophobic interactions or specifically by
chemical conjugation to the core-forming block of the copolymer
via a carefully designed pH- or enzyme-sensitive linker that can be
cleaved to release a drug in its active form. A variety of drugs with
diverse physicochemical properties can be incorporated into the
core by engineering the structure of the core-forming segment of
the copolymer to attain sufficiently strong interaction with drug
molecules. These polymeric micelle systems can also be used for
co-delivery of two or more drugs with similar or different proper-
ties for combination therapy, or to combine multiple modalities
within a single carrier [110, 111]. Hydrophilic shell serves as a
stabilizing interface between the hydrophobic core and external
milieu and most commonly consists of PEG chains with a molecu-
lar weight ranging from 2 to 15 kDa. In addition, shell can inhibit
protein binding and opsonization during systemic administration,
which allows them to remain in the circulation longer by evading
the mononuclear phagocytic system. Also, modification of the shell
with various ligands using different surface chemistries enables the
micelle to be targeted to a specific site. A much wider range of
hydrophobic blocks have been explored as drug loading cores.
Examples include polyesters, polyanhydrides, poly(L-amino acids),
poly(methyl methacrylate), phospholipids/long chain fatty acids,
polypropylene oxide (in Pluronics/poloxamers). The choice of
hydrophobic block is largely dictated by drug compatibility with
the hydrophobic core (when drug is physically loaded) and the
stability of the micelle. Micelles must be stable enough to retain
drug cargo upon administration and remain intact long enough to
accumulate in sufficient concentrations at the target site. The ther-
modynamic tendency for micelles to dissociate is primarily con-
trolled by the length of the hydrophobic block while the kinetic
(rate of dissociation) stability depends on many factors, including
the size of a hydrophobic block, the mass ratio of hydrophilic to
hydrophobic blocks, and physical state of the micelle core [112].
The incorporation of hydrophobic drugs may also further enhance
micelle stability. Among the different strategies to enhance the
kinetic stability of polymeric micelles, core- or shell-cross-linking
have been shown to limit the premature disassembly and slow
down the release of the encapsulated drug [113]. The resulting
Synthetic Polymer-based Nanomaterials 13

cross-linked micelles are, in essence, nanoscale single molecules


that are stable upon dilution and can withstand environmental
challenges and shear forces without structural deterioration. For
example, Iijima et al. [114] have shown that core cross-linked
micelles of PEG-b-poly(D,L-lactide) possess high stability against
dilution, temperature change, and even dissolution of surfactants.
Wooley and coworkers [115, 116] cross-linked the micellar corona
and obtained the so-called shell-cross-linked knedel-like micelles—
robust nanostructures with a permeable cross-linked shell.
However, the reversible stabilization of the micellar structure will
be more desirable to achieve efficient intracellular drug release and
circumvent micelle accumulation in the body. To this end, the use
of degradable polymer components or the introduction of revers-
ible cross-links allowed development in situ degradable micelles
[117–119]. High loading capacity and controlled drug release
profile are key features for the potential drug delivery system. The
loading capacity and loading efficiency of the polymeric micelles is
influenced by several factors, including both structure of core-
forming block and a drug, molecular characteristics of the copoly-
mer, such as composition, molecular weight, and the solution
temperature. The maximum loading level is largely influenced by
the interaction between the drug and core-forming block, and
stronger interactions enable saturation to be reached at a lower
polymer concentration. The drug loading can also be improved by
enhancing the local environment of the micelle core through the
means of conjugation of small amount of drug [120] or conjuga-
tion of side chains with similar structure to the drug [121]. In
addition, the location of the incorporated molecules within poly-
meric micelles (micelle core or the core–shell interface) determines
the extent of solubilization as well as the rate of drug release [122,
123]. In general, for drugs physically incorporated in polymeric
micelles, release is controlled by the rate of diffusion of the drug
from the micellar core, stability of the micelles, and the rate of
biodegradation of the copolymer. If the micelle is stable and the
rate of polymer biodegradation is slow, the diffusion rate of the
drug will be mainly determined by the compatibility between the
drug and core-forming block of copolymer, amount of drug
loaded, the molecular volume of drug, length of the core forming
block, and physical state of the core. Moreover, external conditions
such as change in pH, temperature or application of ultrasound
[124] can also trigger drug release from polymeric micelles.
Another important but less investigated factor that can affect dif-
ferent characteristics of polymeric micelles is the morphology and
dimensions of micellar aggregates. Indeed, filamentous polymeric
micelles (“filomicelles,” also referred to as “worm micelles”) with
single dimensions as long as 18 μm (the diameter is ~57 nm) syn-
thesized from amphiphilic PEG-b-PCL copolymer were reported
to solubilize twice as much paclitaxel as spherical micelles [125].
14 Swapnil S. Desale et al.

Recently, Wooley’s group [126] demonstrated that shell-cross-


linked, rod-shaped micelles prepared from pH-sensitive poly(acrylic
acid)-b-poly(p-hydroxystyrene) block copolymer exhibited a
higher doxorubicin-loading capacity and rate of release compared
to their spherical counterparts derived from the same copolymer
precursor. Importantly, it was also demonstrated that rational
design of polymeric micelles and other NMs of a given geometry
(size and shape) offers an unprecedented control of their longevity
in circulation and targeting to selective cellular and subcellular
locations [127–129]. Further understanding of structure–activity
relationships of such complex multicomponent nanomedicines is
essential to determine the criteria for the successful development of
polymeric micelle therapeutics. Currently several micelle formula-
tions based on amphiphilic block copolymers are in clinical trials
for treating a variety of cancers [130–133]. In addition, the first
targeted micellar formulation (BIND-014) has recently reached
clinical development [134].
The field of polymeric micelles was significantly advanced by
employing charge-driven self-assembly of block copolymers con-
taining water-soluble ionic and nonionic block (double hydrophilic
block copolymers or block ionomers). The micellization of these
copolymers can be induced by adding oppositely charged molecules
such as synthetic [135, 136] or natural (DNA, proteins) [137–139]
polyelectrolytes, surfactants/lipids [140, 141], or metal ions [142,
143]. These molecules form electrostatic complexes with the
charged blocks of block ionomers, and prompt spontaneous segre-
gation of the resulting complexes into micelle-like structures with
electrostatically neutralized polyion cores and hydrophilic polymer
shells. Ionic block lengths, charge density, and ionic strength of a
solution affect the formation of stable block ionomer complexes
and, therefore, control the amount of the drug that can be incorpo-
rated within the micelles [139, 144]. As an example, the metal-
complex formation of ionic block copolymer, PEG-PGA, was
explored to prepare polymeric micelles incorporating anticancer
drug cisplatin. In preclinical studies, they exhibited remarkably pro-
longed blood circulation and effective accumulation in solid tumors
[145]. This formulation is currently being evaluated in Phase III
clinical trial as a treatment for pancreatic cancer under the name
NC-6004 (Nanoplatin; NanoCarrier Co., Ltd.; Japan) [146].
Since being proposed independently by Kabanov and Kataoka
[147, 148] in 1995, this approach has been widely used for devel-
oping nonviral gene delivery systems using cationic block or graft
copolymers. In this case, the neutralization of positive charges on
the polycation block by negatively charged DNA or siRNA leads to
the formation of polyion micelles. Advanced and tunable charac-
teristics of the cationic copolymers such as chemical structure and
length of the segments, rigidity, hydrophilicity, charge density, bio-
degradability allow to modulate gene-delivery properties such as
Synthetic Polymer-based Nanomaterials 15

DNA binding, colloidal stability of the complexes, toxicity, endo-


somal escape, vector unpacking, and transfection efficiency [149–
151]. Similar to nucleic acid, charged proteins and peptides can be
entrapped into a polyion micelle core [137, 152–154]. Recently,
this strategy was successfully used for the delivery of antioxidant
enzymes to the central nervous system [155–157]. There has also
been a focus on the development of micellar carriers that are able
to deliver both a therapeutic drug and a nucleic acid [158]. As an
example, Wang and coworkers reported micelles composed of bio-
degradable PEG-poly(ε-caprolactone)-b-poly(2-aminoethylethylene
phosphate) triblock copolymers, PEG-PCL-PPEEA, with the capac-
ity to encapsulate hydrophobic paclitaxel into PCL core and com-
plex siRNA against polo-like kinase 1 to the cationic PPEEA block.
These micelles simultaneously delivered two payloads into the same
tumor cells both in vitro and in vivo, and inhibited tumor growth in
a synergistic manner following systemic administration [159].

11 Polymersomes

Polymersomes (polymeric vesicles) is another class of supramolecu-


lar assemblies formed by amphiphilic block copolymers in diluted
aqueous solutions. Typically the formation of vesicular structures is
favored for the copolymers with relatively low weight fraction of
hydrophilic block (~20–40 %) [160]. Polymersome sizes vary from
50 nm to 10 μm depending on the chemical composition and the
length of polymer blocks, the preparation method, as well as reac-
tion conditions [161]. The copolymers of various architectures
(diblock, triblock, graft, and dendritic) have been utilized as build-
ing blocks for design and preparation of polymersomes [162–165].
Nonbiodegradable poly(ethyl ethylene), poly(butadiene),
poly(dimethylsiloxane), poly(propylene oxide), polystyrene as well
as biodegradable PLA, PCL, and poly(trimethylene carbonate)
have been used as a hydrophobic part of the block copolymers
[166–171]. PEG, PGA and poly(acrylic acid) have been frequently
selected as water-soluble blocks. More recently, so-called PICsomes
(polyion complex vesicles) have been developed by electrostatic
self-assembly of oppositely charged block- and homoionomers
[172]. The key distinction between polymersomes and spherical
micelles is that the former have an interior aqueous cavity sur-
rounded by a wall that consists of entangled chains [173]. Due to
the higher molecular weight of the polymers, polymersome mem-
branes are generally thicker (8–22 nm), stronger, and hence,
intrinsically more stable and less permeable than conventional lipo-
somes. The thickness of the membrane is determined by the length
of the hydrophobic block and can be tuned through fine control of
the polymer chemical composition [167, 174–176]. The enhanced
structural integrity and much denser hydrophilic polymer brush on
16 Swapnil S. Desale et al.

the surface of the polymersomes lead to their more persistent cir-


culation in the bloodstream [177]. Polymersomes are able to
encapsulate hydrophilic, hydrophobic and amphiphilic molecules
like any other vesicular structure, but their membrane tunability
and superior stability are unique and undoubtedly beneficial for
potential applications in drug delivery as well as medical diagnos-
tics. Examples of drug-loaded polymersomes are increasingly broad
and now include anticancer drugs, oligonucleotides, therapeutic
proteins, diagnostic probes, or their combinations [178–182]. In
addition, targetability of the polymersomes with various ligands
attached to the surface has been demonstrated [183–187]. Recently,
modulation of the membrane stability or permeability in response
to specific stimulus has received a lot of attention as a strategy for
controlled drug release from polymersomes. Numerous stimuli
(e.g., pH, redox potential, temperature, magnetic field, light, and
ultrasound) have been exploited in the design of stimuli-responsive
polymersomes [188]. While these systems are still at the level of
research and development, their structural versatility and improved
properties in terms of stability and multifunctionality make them
excellent candidates for potential biomedical applications.

12 Polymeric Nanogels

The term “nanogels” usually defines hydrogel particles of nanoscale


size formed by physically or chemically cross-linked polymer net-
works [85]. Polymeric nanogels can be prepared by physical self-
assembly of interactive polymers [189] or by cross-linking reaction
of preformed polymers [190]. Nanoemulsion or microemulsion
polymerization methods are often used to obtain nanogels with
well-controlled sizes [191–193]. Another attractive set of synthetic
techniques for preparation of nanogel particles is based on a
template-assisted nanofabrication and exploit the internal water
phase of liposomes [194], cross-linking of polymeric micelles [195,
196] or lithographic PRINT process [24]. Various chemical cross-
linking reactions have now been developed, including photocross-
linking, carbodiimide-mediated amide bond cross-linking,
quaternization of amino groups, and “click” chemistry [143, 197–
199]. Labile bonds are frequently introduced into nanogels during
their synthesis to make them degradable and facilitate drug release.
Dispersed in the water, the nanogels are highly swollen and can
incorporate 30 % wt. and more of biological molecules and drugs
through electrostatic, van der Waals and/or hydrophobic interac-
tions or covalent bonding with the nanogel chains. These loading
capacities are unusually high and exceed those of liposomes and
polymeric micelles [85, 200]. As a result of drug loading, the
nanogels collapse forming stable nanoparticles, in which biological
agent becomes entrapped. Introducing dispersing hydrophilic
Synthetic Polymer-based Nanomaterials 17

polymers (e.g., PEG) in a nanogel structure can prevent their


aggregation. During the collapse of the drug-nanogel complex
hydrophilic polymer chains become exposed at the surface and
form a protective layer around the nanogel. The control and versa-
tility of polymer chemistry allows designing a broad range of drug
formulations and inclusion of multiple therapeutic cargos within
the same nanogel carrier [85, 196, 201]. Stimuli-responsive drug
release via temperature or pH-induced volume collapse can also be
very attractive for drug delivery applications. The functionalization
of the nanogel surface can further facilitate their selective accumu-
lation in the target tissue or cells [202–204]. Development of
nanogels that can carry, protect, target and release therapeutic
agents in spatially and temporally controlled manner is actively
ongoing and their rational design can provide a platform for mul-
tiple applications.
Over the last decade, the field of polymer-based biomaterials
for delivery of low molecular drugs, proteins and nucleic acids has
seen exponential growth. Only selected examples were reviewed
here due to the large number of contributions on this topic. Indeed,
the recent advances in polymer chemistry have allowed the develop-
ment of a diverse range of NMs of various sizes, shapes, surface
chemistries, and targeting properties. Polymer composition also
plays an essential role in function and application of such macromo-
lecular carriers. There is growing evidence that some synthetic poly-
mers can display biological response-modifying activity and can
influence the molecular mechanism of action of a drug. Several of
the polymer-based nanocarriers are already in clinical use and
numerous of the others are undergoing various stages of preclinical
and clinical evaluation. Although considerable progress has been
made, further synthetic improvements are needed to design safe,
more “intelligent” and efficient drug delivery platforms.

References

1. Brigger I, Dubernet C, Couvreur P (2002) 6. Labet M, Thielemans W (2009) Synthesis of


Nanoparticles in cancer therapy and diagno- polycaprolactone: a review. Chem Soc Rev
sis. Adv Drug Deliv Rev 54(5):631–651 38(12):3484–3504
2. Lavan DA, McGuire T, Langer R (2003) 7. Lou X, Detrembleur C, Jérôme R (2003)
Small-scale systems for in vivo drug delivery. Novel aliphatic polyesters based on functional
Nat Biotechnol 21(10):1184–1191 cyclic (di) esters. Macromol Rapid Commun
3. Brannon-Peppas L, Blanchette JO (2012) 24(2):161–172
Nanoparticle and targeted systems for cancer 8. Williams CK (2007) Synthesis of functional-
therapy. Adv Drug Deliv Rev 64:206–212 ized biodegradable polyesters. Chem Soc Rev
4. Vert M (2005) Aliphatic polyesters: great 36(10):1573–1580
degradable polymers that cannot do every- 9. Penczek S, Cypryk M, Duda A, Kubisa P,
thing. Biomacromolecules 6(2):538–546 Słomkowski S (2007) Living ring-opening
5. Jérôme C, Lecomte P (2008) Recent advances polymerizations of heterocyclic monomers.
in the synthesis of aliphatic polyesters by ring- Prog Polym Sci 32(2):247–282
opening polymerization. Adv Drug Deliv Rev 10. Gross RA, Ganesh M, Lu W (2010) Enzyme-
60(9):1056–1076 catalysis breathes new life into polyester
18 Swapnil S. Desale et al.

condensation polymerizations. Trends platform for controlled synthesis of polymeric


Biotechnol 28(8):435–443 nanoparticles. Nano Lett 8(9):2906–2912
11. Soppimath KS, Aminabhavi TM, Kulkarni 23. Rolland JP, Maynor BW, Euliss LE, Exner
AR, Rudzinski WE (2001) Biodegradable AE, Denison GM, DeSimone JM (2005)
polymeric nanoparticles as drug delivery Direct fabrication and harvesting of monodis-
devices. J Control Release 70(1):1–20 perse, shape-specific nanobiomaterials. J Am
12. Desgouilles S, Vauthier C, Bazile D, Vacus J, Chem Soc 127(28):10096–10100
Grossiord J-L, Veillard M et al (2003) The 24. Gratton SE, Pohlhaus PD, Lee J, Guo J, Cho
design of nanoparticles obtained by solvent MJ, DeSimone JM (2007) Nanofabricated par-
evaporation: a comprehensive study. ticles for engineered drug therapies: a prelimi-
Langmuir 19(22):9504–9510 nary biodistribution study of PRINT™
13. Lai M-K, Tsiang R-C (2004) Encapsulating nanoparticles. J Control Release 121(1):10–18
acetaminophen into poly (L-lactide) micro- 25. Enlow EM, Luft JC, Napier ME, DeSimone
capsules by solvent-evaporation technique in JM (2011) Potent engineered PLGA
an O/W emulsion. J Microencapsul 21(3): nanoparticles by virtue of exceptionally high
307–316 chemotherapeutic loadings. Nano Lett
14. Hariharan S, Bhardwaj V, Bala I, Sitterberg J, 11(2):808–813
Bakowsky U, Kumar MR (2006) Design of 26. Chasin M, Lewis D, Langer R (1988)
estradiol loaded PLGA nanoparticulate for- Polyanhydrides for controlled drug delivery.
mulations: a potential oral delivery system for Biopharm Manufact 1:33–46
hormone therapy. Pharm Res 23(1):184–195 27. Kumar N, Langer RS, Domb AJ (2002)
15. Messai I, Delair T (2005) Adsorption of chi- Polyanhydrides: an overview. Adv Drug Deliv
tosan onto poly (D, L-lactic acid) particles: a Rev 54(7):889–910
physico-chemical investigation. Macromol 28. Conix A (1966) Poly [1, 3-bis
Chem Phys 206(16):1665–1674 (p-carboxyphenoxy)-propane anhydride],
16. Keegan ME, Falcone JL, Leung TC, Saltzman macromolecular synthesis, vol 2. Wiley,
WM (2004) Biodegradable microspheres with New York, NY
enhanced capacity for covalently bound surface 29. Domb A, Langer R (1987) Polyanhydrides.
ligands. Macromolecules 37(26):9779–9784 I. Preparation of high molecular weight poly-
17. Zambaux M, Bonneaux F, Gref R, Maincent anhydrides. J Polym Sci A Polym Chem
P, Dellacherie E, Alonso M et al (1998) 25(12):3373–3386
Influence of experimental parameters on the 30. Leong K, Brott B, Langer R (1985)
characteristics of poly (lactic acid) nanoparti- Bioerodible polyanhydrides as drug-carrier
cles prepared by a double emulsion method. matrices. I: Characterization, degradation,
J Control Release 50(1):31–40 and release characteristics. J Biomed Mater
18. Paiphansiri U, Tangboriboonrat P, Landfester Res 19(8):941–955
K (2006) Polymeric nanocapsules containing 31. Katti D, Lakshmi S, Langer R, Laurencin C
an antiseptic agent obtained by controlled (2002) Toxicity, biodegradation and elimina-
nanoprecipitation onto water-in-oil miniemul- tion of polyanhydrides. Adv Drug Deliv Rev
sion droplets. Macromol Biosci 6(1):33–40 54(7):933–961
19. Govender T, Stolnik S, Garnett MC, Illum L, 32. Rebouças JDS, Irache JM, Camacho AI,
Davis SS (1999) PLGA nanoparticles pre- Esparza I, del Pozo V, Sanz ML et al (2012)
pared by nanoprecipitation: drug loading and Development of poly (anhydride) nanoparti-
release studies of a water soluble drug. cles loaded with peanut proteins: the influ-
J Control Release 57(2):171–185 ence of preparation method on the
20. Jung T, Breitenbach A, Kissel T (2000) immunogenic properties. Eur J Pharm
Sulfobutylated poly (vinyl alcohol)-graft-poly Biopharm 82(2):241–249
(lactide-co-glycolide) s facilitate the prepara- 33. Agüeros M, Ruiz-Gatón L, Vauthier C,
tion of small negatively charged biodegrad- Bouchemal K, Espuelas S, Ponchel G et al
able nanospheres. J Control Release (2009) Combined hydroxypropyl-β-
67(2):157–169 cyclodextrin and poly (anhydride) nanoparti-
21. Allémann E, Leroux J-C, Gurny R, Doelker E cles improve the oral permeability of
(1993) In vitro extended-release properties of paclitaxel. Eur J Pharm Sci 38(4):405–413
drug-loaded poly (DL-lactic acid) nanoparti- 34. Arbos P, Campanero M, Arangoa M, Renedo
cles produced by a salting-out procedure. M, Irache J (2003) Influence of the surface
Pharm Res 10(12):1732–1737 characteristics of PVM/MA nanoparticles on
22. Karnik R, Gu F, Basto P, Cannizzaro C, Dean their bioadhesive properties. J Control
L, Kyei-Manu W et al (2008) Microfluidic Release 89(1):19–30
Synthetic Polymer-based Nanomaterials 19

35. Ulery BD, Phanse Y, Sinha A, Wannemuehler and its potential application in treatment of
MJ, Narasimhan B, Bellaire BH (2009) rheumatoid arthritis. Arthritis Res Ther
Polymer chemistry influences monocytic 9(1):R2
uptake of polyanhydride nanospheres. Pharm 47. Teodorescu M, Matyjaszewski K (1999) Atom
Res 26(3):683–690 transfer radical polymerization of (meth) acryl-
36. Petersen L, Sackett C, Narasimhan B (2010) amides. Macromolecules 32(15):4826–4831
High-throughput analysis of protein stability 48. Scales CW, Vasilieva YA, Convertine AJ, Lowe
in polyanhydride nanoparticles. Acta Biomater AB, McCormick CL (2005) Direct, con-
6(10):3873–3881 trolled synthesis of the nonimmunogenic,
37. Chavez-Santoscoy A, Carrillo-Conde B, Song hydrophilic polymer, poly (N-(2-
E-H, Phanse Y, Ramer-Tait AE, Pohl NL et al. hydroxypropyl) methacrylamide) via RAFT in
(2011) Targeted activation of antigen present- aqueous media. Biomacromolecules
ing cells with mannose-modified polyanhy- 6(4):1846–1850
dride nanoparticles. Trans Soc Biomater 34 49. Duncan R, Kopeckova-Rejmanova P,
38. Haughney SL, Petersen LK, Schoofs AD, Strohalm J, Hume I, Cable H, Pohl J et al
Ramer-Tait AE, King JD, Briles DE et al (1987) Anticancer agents coupled to N-(2-
(2013) Retention of structure, antigenicity, hydroxypropyl) methacrylamide copolymers.
and biological function of pneumococcal sur- I. Evaluation of daunomycin and puromycin
face protein A (PspA) released from polyan- conjugates in vitro. Br J Cancer 55(2):165
hydride nanoparticles. Acta Biomater 50. Šubr V, Kopeček J, Pohl J, Baudyš M, Kostka
9(9):8262–8271 V (1988) Cleavage of oligopeptide side-chains
39. Petersen LK, Ramer-Tait AE, Broderick SR, in N-(2-hydroxypropyl) meth-acrylamide
Kong C-S, Ulery BD, Rajan K et al (2011) copolymers by mixtures of lysosomal enzymes.
Activation of innate immune responses in a J Control Release 8(2):133–140
pathogen-mimicking manner by amphiphilic 51. Subr V, Strohalm J, Ulbrich K, Duncan R,
polyanhydride nanoparticle adjuvants. Hume I (1992) Polymers containing enzy-
Biomaterials 32(28):6815–6822 matically degradable bonds, XII. Effect of
40. Chavez-Santoscoy AV, Roychoudhury R, spacer structure on the rate of release of dau-
Pohl NL, Wannemuehler MJ, Narasimhan B, nomycin and adriamycin from poly [N-(2-
Ramer-Tait AE (2012) Tailoring the immune hydroxypropyl)-methacrylamide] copolymer
response by targeting C-type lectin receptors drag carriers in vitro and antitumour activity
on alveolar macrophages using “pathogen- measured in vivo. J Control Release
like” amphiphilic polyanhydride nanoparti- 18(2):123–132
cles. Biomaterials 33(18):4762–4772 52. Liu X-M, Quan L-D, Tian J, Alnouti Y, Fu K,
41. Petersen L, Phanse Y, Ramer-Tait A, Thiele GM et al (2008) Synthesis and evalua-
Wannemuehler M, Narasimhan B (2012) tion of a well-defined HPMA copolymer–
Amphiphilic polyanhydride nanoparticles sta- dexamethasone conjugate for effective
bilize Bacillus anthracis protective antigen. treatment of rheumatoid arthritis. Pharm Res
Mol Pharm 9(4):874–882 25(12):2910–2919
42. Ulery BD, Petersen LK, Phanse Y, Kong CS, 53. Lemkine G, Demeneix B (2001)
Broderick SR, Kumar D et al (2011) Rational Polyethylenimines for in vivo gene delivery.
design of pathogen-mimicking amphiphilic Curr Opin Mol Ther 3(2):178–182
materials as nanoadjuvants. Sci Rep 1:198 54. von Harpe A, Petersen H, Li Y, Kissel T
43. Kopeček J, Kopečková P (2010) HPMA (2000) Characterization of commercially
copolymers: origins, early developments, available and synthesized polyethylenimines
present, and future. Adv Drug Deliv Rev for gene delivery. J Control Release
62(2):122–149 69(2):309–322
44. Duncan R (2006) Polymer conjugates as anti- 55. Kunath K, von Harpe A, Fischer D, Petersen
cancer nanomedicines. Nat Rev Cancer H, Bickel U, Voigt K et al (2003) Low-
6(9):688–701 molecular-weight polyethylenimine as a non-
45. Kopeček J, Kopečková P, Minko T, Lu Z-R viral vector for DNA delivery: comparison of
(2000) HPMA copolymer–anticancer drug physicochemical properties, transfection effi-
conjugates: design, activity, and mechanism ciency and in vivo distribution with high-
of action. Eur J Pharm Biopharm molecular-weight polyethylenimine.
50(1):61–81 J Control Release 89(1):113–125
46. Wang D, Miller SC, Liu X, Anderson B, Wang 56. Fischer D, Bieber T, Li Y, Elsässer H-P, Kissel
XS, Goldring SR (2007) Novel T (1999) A novel non-viral vector for DNA
dexamethasone-HPMA copolymer conjugate delivery based on low molecular weight,
20 Swapnil S. Desale et al.

branched polyethylenimine: effect of molecu- 69. Jevprasesphant R, Penny J, Jalal R, Attwood


lar weight on transfection efficiency and cyto- D, McKeown N, D’emanuele A (2003) The
toxicity. Pharm Res 16(8):1273–1279 influence of surface modification on the cyto-
57. Dick C, Ham G (1970) Characterization of toxicity of PAMAM dendrimers. Int J Pharm
polyethylenimine. J Macromol Sci Pure Appl 252(1):263–266
Chem 4(6):1301–1314 70. Choi JS, Nam K, Park J-Y, Kim J-B, Lee J-K,
58. Weyts KF, Goethals EJ (1988) New synthesis Park J-S (2004) Enhanced transfection effi-
of linear polyethyleneimine. Polym Bull ciency of PAMAM dendrimer by surface
19(1):13–19 modification with L-arginine. J Control
59. Saegusa T, Ikeda H, Fujii H (1972) Release 99(3):445–456
Isomerization polymerization of 2-oxazoline. 71. Kim Y, Klutz AM, Jacobson KA (2008)
I. Preparation of unsubstituted 2-oxazoline Systematic investigation of polyamidoamine
polymer. Polym J 3(1):35–39 dendrimers surface-modified with poly (ethyl-
60. Saegusa T, Ikeda H, Fujii H (1972) ene glycol) for drug delivery applications:
Isomerization polymerization of 2-oxazoline. synthesis, characterization, and evaluation of
IV. Kinetic study of 2-methyl-2-oxazoline cytotoxicity. Bioconjug Chem
polymerization. Macromolecules 5(4): 19(8):1660–1672
359–362 72. Kesharwani P, Jain K, Jain NK (2014)
61. Peng Q, Zhong Z, Zhuo R (2008) Disulfide Dendrimer as nanocarrier for drug delivery.
cross-linked polyethylenimines (PEI) pre- Prog Polym Sci 39(2):268–307
pared via thiolation of low molecular weight 73. Kukowska-Latallo JF, Candido KA, Cao Z,
PEI as highly efficient gene vectors. Bioconjug Nigavekar SS, Majoros IJ, Thomas TP et al
Chem 19(2):499–506 (2005) Nanoparticle targeting of anticancer
62. Nakayama Y, Kakei C, Ishikawa A, Zhou drug improves therapeutic response in animal
Y-M, Nemoto Y, Uchida K (2007) Synthesis model of human epithelial cancer. Cancer Res
and in vitro evaluation of novel star-shaped 65(12):5317–5324
block copolymers (blocked star vectors) for 74. McCarthy TD, Karellas P, Henderson SA,
efficient gene delivery. Bioconjug Chem Giannis M, O’Keefe DF, Heery G et al (2005)
18(6):2037–2044 Dendrimers as drugs: discovery and preclini-
63. Banerjee P, Weissleder R, Bogdanov A (2006) cal and clinical development of dendrimer-
Linear polyethyleneimine grafted to a hyper- based microbicides for HIV and STI
branched poly (ethylene glycol)-like core: a prevention. Mol Pharm 2(4):312–318
copolymer for gene delivery. Bioconjug Chem 75. Sampathkumar S-G, Yarema KJ (2005)
17(1):125–131 Targeting cancer cells with dendrimers. Chem
64. Namgung R, Kim J, Singha K, Kim CH, Kim Biol 12(1):5
WJ (2009) Synergistic effect of low cytotoxic 76. Medina SH, El-Sayed ME (2009) Dendrimers
linear polyethylenimine and multiarm poly- as carriers for delivery of chemotherapeutic
ethylene glycol: study of physicochemical agents. Chem Rev 109(7):3141–3157
properties and in vitro gene transfection. Mol 77. Twibanire JAK, Grindley TB (2014) Polyester
Pharm 6(6):1826–1835 dendrimers: smart carriers for drug delivery.
65. Hadjichristidis N, Iatrou H, Pitsikalis M, Polymers 6(1):179–213
Sakellariou G (2009) Synthesis of well- 78. van der Poll DG, Kieler-Ferguson HM, Floyd
defined polypeptide-based materials via the WC, Guillaudeu SJ, Jerger K, Szoka FC et al
ring-opening polymerization of α-amino acid (2010) Design, synthesis, and biological eval-
N-carboxyanhydrides. Chem Rev uation of a robust, biodegradable dendrimer.
109(11):5528–5578 Bioconjug Chem 21(4):764–773
66. Cheng J, Deming TJ (2012) Synthesis of 79. Liu L, Breslow R (2003) Dendrimeric pyri-
polypeptides by ring-opening polymerization doxamine enzyme mimics. J Am Chem Soc
of α-amino acid N-carboxyanhydrides, 125(40):12110–12111
Peptide-based materials. Springer, New York, 80. Wu D, Yang J, Li J, Chen L, Tang B, Chen X
NY, pp 1–26 et al (2013) Hydroxyapatite-anchored den-
67. Hodge P (1993) Polymer science branches drimer for in situ remineralization of human
out. Nature 362:18–19 tooth enamel. Biomaterials 34(21):5036–5047
68. Hawker CJ, Frechet JM (1990) Preparation 81. Kasai S, Nagasawa H, Shimamura M, Uto Y,
of polymers with controlled molecular archi- Hori H (2002) Design and synthesis of anti-
tecture. A new convergent approach to den- angiogenic/heparin-binding arginine den-
dritic macromolecules. J Am Chem Soc drimer mimicking the surface of endostatin.
112(21):7638–7647 Bioorg Med Chem Lett 12(6):951–954
Synthetic Polymer-based Nanomaterials 21

82. Gajbhiye V, Palanirajan VK, Tekade RK, Jain 95. Vicent MJ, Cascales L, Carbajo RJ, Cortés N,
NK (2009) Dendrimers as therapeutic agents: Messeguer A, Pérez Payá E (2010)
a systematic review. J Pharm Pharmacol Nanoconjugates as intracorporeal neutralizers
61(8):989–1003 of bacterial endotoxins. J Control Release
83. Rupp R, Rosenthal SL, Stanberry LR (2007) 142(2):277–285
VivaGel™(SPL7013 Gel): a candidate den- 96. Santamaría B, Benito-Martin A, Ucero AC,
drimer–microbicide for the prevention of Aroeira LS, Reyero A, Vicent MJ et al (2009)
HIV and HSV infection. Int J Nanomedicine A nanoconjugate Apaf-1 inhibitor protects
2(4):561 mesothelial cells from cytokine-induced
84. Duncan R (2003) The dawning era of poly- injury. PLoS One 4(8):e6634
mer therapeutics. Nat Rev Drug Discov 2(5): 97. Liu X-M, Quan L-D, Tian J, Laquer FC,
347–360 Ciborowski P, Wang D (2010) Syntheses of
85. Kabanov AV, Vinogradov SV (2009) click PEG–dexamethasone conjugates for the
Nanogels as pharmaceutical carriers: finite treatment of rheumatoid arthritis.
networks of infinite capabilities. Angew Chem Biomacromolecules 11(10):2621–2628
Int Ed Engl 48(30):5418–5429 98. Ikumi Y, Kida T, Sakuma S, Yamashita S,
86. Vicent MJ, Duncan R (2006) Polymer conju- Akashi M (2008) Polymer–phloridzin conju-
gates: nanosized medicines for treating can- gates as an anti-diabetic drug that Inhibits
cer. Trends Biotechnol 24(1):39–47 glucose absorption through the Na+/glucose
87. Vicent MJ, Ringsdorf H, Duncan R (2009) cotransporter (SGLT1) in the small intestine.
Polymer therapeutics: clinical applications J Control Release 125(1):42–49
and challenges for development. Adv Drug 99. Shaunak S, Thomas S, Gianasi E, Godwin A,
Deliv Rev 61(13):1117–1120 Jones E, Teo I et al (2004) Polyvalent den-
88. Satchi-Fainaro R, Puder M, Davies JW, Tran drimer glucosamine conjugates prevent scar tis-
HT, Sampson DA, Greene AK et al (2004) sue formation. Nat Biotechnol 22(8):977–984
Targeting angiogenesis with a conjugate of 100. Mondragon L, Orzaez M, Sanclimens G,
HPMA copolymer and TNP-470. Nat Med Moure A, Arminan A, Sepulveda P et al
10(3):255–261 (2008) Modulation of cellular apoptosis with
89. Varticovski L, Lu Z-R, Mitchell K, de Aos I, apoptotic protease-activating factor 1 (Apaf-
Kopeček J (2001) Water-soluble HPMA 1) inhibitors. J Med Chem 51(3):521–529
copolymer–wortmannin conjugate retains 101. Pan H, Sima M, Kopečková P, Wu K, Gao S
phosphoinositide 3-kinase inhibitory activity et al (2008) Biodistribution and pharmacoki-
in vitro and in vivo. J Control Release netic studies of bone-targeting N-(2-
74(1):275–281 hydroxypropyl) methacrylamide
90. Larson N, Ray A, Malugin A, Pike DB, copolymer–alendronate conjugates. Mol
Ghandehari H (2010) HPMA copolymer- Pharm 5(4):548–558
aminohexylgeldanamycin conjugates target- 102. Greco F, Vicent MJ (2009) Combination
ing cell surface expressed GRP78 in prostate therapy: opportunities and challenges for
cancer. Pharm Res 27(12):2683–2693 polymer–drug conjugates as anticancer nano-
91. Kasuya Y, Lu Z-R, Kopečková P, Tabibi SE, medicines. Adv Drug Deliv Rev
Kopeček J (2002) Influence of the structure 61(13):1203–1213
of drug moieties on the in vitro efficacy of 103. Deacon SP, Apostolovic B, Carbajo RJ, Schott
HPMA copolymer–geldanamycin derivative A-K, Beck K, Vicent MJ et al (2010) Polymer
conjugates. Pharm Res 19(2):115–123 coiled-coil conjugates: potential for develop-
92. Vicent MJ (2007) Polymer-drug conjugates ment as a new class of therapeutic “molecular
as modulators of cellular apoptosis. AAPS switch”. Biomacromolecules 12(1):19–27
J 9(2):E200–E207 104. Chipman SD, Oldham FB, Pezzoni G, Singer
93. Li J, Wang Y, Yang C, Wang P, Oelschlager JW (2006) Biological and clinical character-
DK, Zheng Y et al (2009) Polyethylene gly- ization of paclitaxel poliglumex (PPX,
cosylated curcumin conjugate inhibits pancre- CT-2103), a macromolecular polymer–drug
atic cancer cell growth through inactivation of conjugate. Int J Nanomedicine 1(4):375
Jab1. Mol Pharmacol 76(1):81–90 105. Sausville E, Garbo L, Weiss G, Shkolny D,
94. Oman M, Liu J, Chen J, Durrant D, Yang Yurkovetskiy A, Bethune C et al (2010) Phase
H-S, He Y et al (2006) Using N-(2- I study of XMT-1001 given IV every 3 weeks
hydroxypropyl) methacrylamide copolymer to patients with advanced solid tumors. J Clin
drug bioconjugate as a novel approach to Oncol 28:13121
deliver a Bcl-2-targeting compound HA14-1 106. Lee B-S, Fujita M, Khazenzon NM,
in vivo. Gene Ther Mol Biol A 10:113–122 Wawrowsky KA, Wachsmann-Hogiu S, Farkas
22 Swapnil S. Desale et al.

DL et al (2006) Polycefin, a new prototype of delivery of chemotherapeutic agents.


a multifunctional nanoconjugate based on Biomacromolecules 11(4):919–926
poly (β-L-malic acid) for drug delivery. 119. Li YL, Zhu L, Liu Z, Cheng R, Meng F, Cui
Bioconjug Chem 17(2):317–326 JH et al (2009) Reversibly stabilized multi-
107. Hreczuk-Hirst D, Chicco D, German L, functional dextran nanoparticles efficiently
Duncan R (2001) Dextrins as potential carri- deliver doxorubicin into the nuclei of cancer
ers for drug targeting: tailored rates of dextrin cells. Angew Chem Int Ed Engl 48(52):
degradation by introduction of pendant 9914–9918
groups. Int J Pharm 230(1):57–66 120. Nakanishi T, Fukushima S, Okamoto K,
108. Becker R, Dembek C, White LA, Garrison LP Suzuki M, Matsumura Y, Yokoyama M et al
(2012) The cost offsets and cost-effectiveness (2001) Development of the polymer micelle
associated with pegylated drugs: a review of carrier system for doxorubicin. J Control
the literature. Expert Rev Pharmacoecon Release 74(1):295–302
Outcomes Res 12:775 121. Adams ML, Kwon GS (2003) Relative aggre-
109. Duncan R, Vicent MJ (2010) Do HPMA gation state and hemolytic activity of ampho-
copolymer conjugates have a future as clini- tericin B encapsulated by poly (ethylene
cally useful nanomedicines? A critical over- oxide)- block–poly (N-hexyl-l-aspartamide)-
view of current status and future opportunities. acyl conjugate micelles: effects of acyl chain
Adv Drug Deliv Rev 62(2):272–282 length. J Control Release 87(1):23–32
110. Godsey ME, Suryaprakash S, Leong KW 122. Teng Y, Morrison M, Munk P, Webber S,
(2013) Materials innovation for co-delivery Procházka K (1998) Release kinetics studies
of diverse therapeutic cargos. RSC Adv of aromatic molecules into water from block
3(47):24794–24811 polymer micelles. Macromolecules
111. Zhu L, Perche F, Wang T, Torchilin VP 31(11):3578–3587
(2014) Matrix metalloproteinase 2-sensitive 123. Choucair A, Eisenberg A (2003) Interfacial
multifunctional polymeric micelles for tumor- solubilization of model amphiphilic molecules
specific co-delivery of siRNA and hydropho- in block copolymer micelles. J Am Chem Soc
bic drugs. Biomaterials 35(13):4213–4222 125(39):11993–12000
112. Allen C, Maysinger D, Eisenberg A (1999) 124. Husseini GA, Christensen DA, Rapoport NY,
Nano-engineering block copolymer aggre- Pitt WG (2002) Ultrasonic release of doxoru-
gates for drug delivery. Colloids Surf B bicin from Pluronic P105 micelles stabilized
Biointerfaces 16(1):3–27 with an interpenetrating network of N,N-
113. O’Reilly RK, Hawker CJ, Wooley KL (2006) diethylacrylamide. J Control Release
Cross-linked block copolymer micelles: func- 83(2):303–305
tional nanostructures of great potential and 125. Cai S, Vijayan K, Cheng D, Lima EM, Discher
versatility. Chem Soc Rev 35(11):1068–1083 DE (2007) Micelles of different morpholo-
114. Iijima M, Nagasaki Y, Okada T, Kato M, gies—advantages of worm-like filomicelles of
Kataoka K (1999) Core-polymerized reactive PEO-PCL in paclitaxel delivery. Pharm Res
micelles from heterotelechelic amphiphilic 24(11):2099–2109
block copolymers. Macromolecules 32(4): 126. Lee NS, Lin LY, Neumann WL, Freskos JN,
1140–1146 Karwa A, Shieh JJ et al (2011) Influence of
115. Thurmond KB, Kowalewski T, Wooley KL nanostructure morphology on host capacity
(1996) Water-soluble knedel-like structures: and kinetics of guest release. Small
the preparation of shell-cross-linked small par- 7(14):1998–2003
ticles. J Am Chem Soc 118(30):7239–7240 127. Geng Y, Dalhaimer P, Cai S, Tsai R, Tewari
116. Elsabahy M, Wooley KL (2012) Strategies M, Minko T et al (2007) Shape effects of fila-
toward well-defined polymer nanoparticles ments versus spherical particles in flow and
inspired by nature: chemistry versus versatil- drug delivery. Nat Nanotechnol 2(4):
ity. J Polym Sci A Polym Chem 50(10): 249–255
1869–1880 128. Simone EA, Dziubla TD, Muzykantov VR
117. Li Y, Lokitz BS, Armes SP, McCormick CL (2008) Polymeric carriers: role of geometry
(2006) Synthesis of reversible shell cross- in drug delivery. Expert Opin Drug Deliv
linked micelles for controlled release of bioac- 5:1283
tive agents. Macromolecules 39(8): 129. Zhang K, Fang H, Chen Z, Taylor J-SA,
2726–2728 Wooley KL (2008) Shape effects of nanopar-
118. Kim JO, Sahay G, Kabanov AV, Bronich TK ticles conjugated with cell-penetrating pep-
(2010) Polymeric micelles with ionic cores tides (HIV Tat PTD) on CHO cell uptake.
containing biodegradable cross-links for Bioconjug Chem 19(9):1880–1887
Synthetic Polymer-based Nanomaterials 23

130. Valle JW, Armstrong A, Newman C, Alakhov compounds. Adv Drug Deliv Rev
V, Pietrzynski G, Brewer J et al (2011) A 54(2):203–222
phase 2 study of SP1049C, doxorubicin in 140. Bronich TK, Kabanov AV, Kabanov VA, Yu K,
P-glycoprotein-targeting pluronics, in Eisenberg A (1997) Soluble complexes from
patients with advanced adenocarcinoma of poly (ethylene oxide)-block-polymethacrylate
the esophagus and gastroesophageal junction. anions and N-alkylpyridinium cations.
Invest New Drugs 29(5):1029–1037 Macromolecules 30(12):3519–3525
131. Kim T-Y, Kim D-W, Chung J-Y, Shin SG, Kim 141. Kabanov AV, Bronich T, Kabanov V, Yu K,
S-C, Heo DS et al (2004) Phase I and phar- Eisenberg A (1998) Spontaneous formation
macokinetic study of Genexol-PM, a of vesicles from complexes of block ionomers
cremophor-free, polymeric micelle-formulated and surfactants. J Am Chem Soc
paclitaxel, in patients with advanced malignan- 120(38):9941–9942
cies. Clin Cancer Res 10(11):3708–3716
142. Yokoyama M, Okano T, Sakurai Y, Suwa S,
132. Hamaguchi T, Matsumura Y, Suzuki M, Kataoka K (1996) Introduction of cisplatin
Shimizu K, Goda R, Nakamura I et al (2005) into polymeric micelle. J Control Release
NK105, a paclitaxel-incorporating micellar 39(2):351–356
nanoparticle formulation, can extend in vivo
antitumour activity and reduce the neurotox- 143. Bronich TK, Keifer PA, Shlyakhtenko LS,
icity of paclitaxel. Br J Cancer Kabanov AV (2005) Polymer micelle with
92(7):1240–1246 cross-linked ionic core. J Am Chem Soc
127(23):8236–8237
133. Chin K, Kato K, Yoshikawa T, Yamaguchi K,
Esaki T, Tsuji Y et al (2010) Phase II study of 144. Kabanov AV, Kabanov VA (1998)
NK105, a paclitaxel-incorporating micellar Interpolyelectrolyte and block ionomer com-
nanoparticle as second-line treatment for plexes for gene delivery: physico-chemical
advanced or recurrent gastric cancer. J Clin aspects. Adv Drug Deliv Rev 30(1):49–60
Oncol 28(15S):4041 145. Nishiyama N, Okazaki S, Cabral H, Miyamoto
134. Hrkach J, Von Hoff D, Ali MM, Andrianova M, Kato Y, Sugiyama Y et al (2003) Novel
E, Auer J, Campbell T et al (2012) Preclinical cisplatin-incorporated polymeric micelles can
development and clinical translation of a eradicate solid tumors in mice. Cancer Res
PSMA-targeted docetaxel nanoparticle with a 63(24):8977–8983
differentiated pharmacological profile. Sci 146. Heinemann V, Quietzsch D, Gieseler F,
Transl Med 4(128):128ra39 Gonnermann M, Schönekäs H, Rost A et al
135. Kabanov AV, Bronich TK, Kabanov VA, Yu K, (2006) Randomized phase III trial of gem-
Eisenberg A (1996) Soluble stoichiometric citabine plus cisplatin compared with gem-
complexes from poly (N-ethyl-4- citabine alone in advanced pancreatic cancer.
vinylpyridinium) cations and poly (ethylene J Clin Oncol 24(24):3946–3952
oxide)-block-polymethacrylate anions. 147. Kabanov AV, Vinogradov SV, Suzdaltseva
Macromolecules 29(21):6797–6802 YG, Alakhov VY (1995) Water-soluble block
136. Harada A, Nakanishi K, Ichimura S, Kojima polycations as carriers for oligonucleotide
C, Kono K (2009) Spontaneous formation of delivery. Bioconjug Chem 6(6):639–643
narrowly-distributed self-assembly from poly- 148. Harada A, Kataoka K (1995) Formation of
amidoamine dendron-poly (L-lysine) block polyion complex micelles in an aqueous
copolymers through helix-coil transition of milieu from a pair of oppositely-charged block
poly (L-lysine) block. J Polym Sci A Polym copolymers with poly (ethylene glycol) seg-
Chem 47(4):1217–1223 ments. Macromolecules 28(15):5294–5299
137. Batrakova EV, Li S, Reynolds AD, Mosley 149. Miyata K, Nishiyama N, Kataoka K (2012)
RL, Bronich TK, Kabanov AV et al (2007) A Rational design of smart supramolecular
macrophage-nanozyme delivery system for assemblies for gene delivery: chemical chal-
Parkinson’s disease. Bioconjug Chem lenges in the creation of artificial viruses.
18(5):1498–1506 Chem Soc Rev 41(7):2562–2574
138. Kishimura A, Koide A, Osada K, Yamasaki Y, 150. Itaka K, Yamauchi K, Harada A, Nakamura K,
Kataoka K (2007) Encapsulation of myoglobin Kawaguchi H, Kataoka K (2003) Polyion
in PEGylated polyion complex vesicles made complex micelles from plasmid DNA and poly
from a pair of oppositely charged block iono- (ethylene glycol)–poly (l-lysine) block copoly-
mers: a physiologically available oxygen carrier. mer as serum-tolerable polyplex system: phys-
Angew Chem Int Ed Engl 46(32):6085–6088 icochemical properties of micelles relevant to
139. Kakizawa Y, Kataoka K (2002) Block copoly- gene transfection efficiency. Biomaterials
mer micelles for delivery of gene and related 24(24):4495–4506
24 Swapnil S. Desale et al.

151. Katayose S, Kataoka K (1997) Water-soluble 163. Ayen WY, Chintankumar B, Jain JP, Kumar N
polyion complex associates of DNA and poly (2011) Effect of PEG chain length and hydro-
(ethylene glycol)-poly (L-lysine) block copo- philic weight fraction on polymersomes pre-
lymer. Bioconjug Chem 8(5):702–707 pared from branched (PEG) 3-PLA co-polymers.
152. Harada A, Kataoka K (2001) Pronounced Polym Adv Technol 22(1):158–165
activity of enzymes through the incorporation 164. Zheng C, Qiu L, Zhu K (2009) Novel poly-
into the core of polyion complex micelles mersomes based on amphiphilic graft
made from charged block copolymers. polyphosphazenes and their encapsulation of
J Control Release 72(1):85–91 water-soluble anti-cancer drug. Polymer
153. Yuan X, Harada A, Yamasaki Y, Kataoka K 50(5):1173–1177
(2005) Stabilization of lysozyme-incorporated 165. del Barrio J, Oriol L, Sánchez C, Serrano JL,
polyion complex micelles by the ω-end Di Cicco A, Keller P et al (2010) Self-assembly
derivatization of poly (ethylene glycol)-poly (α, of linear–dendritic diblock copolymers: from
β-aspartic acid) block copolymers with hydro- nanofibers to polymersomes. J Am Chem Soc
phobic groups. Langmuir 21(7):2668–2674 132(11):3762–3769
154. Zhang J, Mulvenon A, Makarov E, Wagoner 166. Schillén K, Bryskhe K, Mel’Nikova YS
J, Knibbe J, Kim JO et al (2013) Antiviral (1999) Vesicles formed from a poly (ethyl-
peptide nanocomplexes as a potential thera- ene oxide)-poly (propylene oxide)-poly
peutic modality for HIV/HCV co-infection. (ethylene oxide) triblock copolymer in
Biomaterials 34(15):3846–3857 dilute aqueous solution. Macromolecules
155. Klyachko NL, Manickam DS, Brynskikh AM, 32(20):6885–6888
Uglanova SV, Li S, Higginbotham SM et al 167. Discher BM, Won Y-Y, Ege DS, Lee JC, Bates
(2012) Cross-linked antioxidant nanozymes FS, Discher DE et al (1999) Polymersomes:
for improved delivery to CNS. Nanomedicine tough vesicles made from diblock copoly-
(Lond) 8(1):119–129 mers. Science 284(5417):1143–1146
156. Manickam DS, Brynskikh AM, Kopanic JL, 168. Kukula H, Schlaad H, Antonietti M, Förster S
Sorgen PL, Klyachko NL, Batrakova EV et al (2002) The formation of polymer vesicles or
(2012) Well-defined cross-linked antioxidant “Peptosomes” by polybutadiene-b lock-poly
nanozymes for treatment of ischemic brain (l-glutamate) s in dilute aqueous solution.
injury. J Control Release 162(3):636–645 J Am Chem Soc 124(8):1658–1663
157. Savalia K, Manickam DS, Rosenbaugh EG, 169. Ding J, Liu G (1997) Polyisoprene-block-
Tian J, Ahmad TM, Kabanov AV et al (2014) poly (2-cinnamoylethyl methacrylate) vesicles
Neuronal uptake of nanoformulated superox- and their aggregates. Macromolecules
ide dismutase and attenuation of angiotensin 30(3):655–657
II-dependent hypertension following central 170. Nardin C, Hirt T, Leukel J, Meier W (2000)
administration. Free Radic Biol Med 73:299 Polymerized ABA triblock copolymer vesicles.
158. Jhaveri AM, Torchilin VP (2014) Langmuir 16(3):1035–1041
Multifunctional polymeric micelles for delivery 171. Meng F, Engbers GH, Feijen J (2005)
of drugs and siRNA. Front Pharmacol 5:77 Biodegradable polymersomes as a basis for
159. Sun T-M, Du J-Z, Yao Y-D, Mao C-Q, Dou S, artificial cells: encapsulation, release and tar-
Huang S-Y et al (2011) Simultaneous delivery geting. J Control Release 101(1):187–198
of siRNA and paclitaxel via a “two-in-one” 172. Chuanoi S, Kishimura A, Dong W-F, Anraku
micelleplex promotes synergistic tumor sup- Y, Yamasaki Y, Kataoka K (2013) Structural
pression. ACS Nano 5(2):1483–1494 factors directing nanosized polyion complex
160. Lee JCM, Bermudez H, Discher BM, vesicles (Nano-PICsomes) to form a pair of
Sheehan MA, Won YY, Bates FS et al (2001) block aniomer/homo catiomers: studies on
Preparation, stability, and in vitro perfor- the aniomer segment length and the catiomer
mance of vesicles made with diblock copoly- side-chain structure. Polym J 46:130
mers. Biotechnol Bioeng 73(2):135–145 173. Battaglia G, Ryan AJ (2005) Bilayers and
161. Yu G-e, Eisenberg A (1998) Multiple mor- interdigitation in block copolymer vesicles.
phologies formed from an amphiphilic ABC J Am Chem Soc 127(24):8757–8764
triblock copolymer in solution. 174. Meng F, Hiemstra C, Engbers GH, Feijen
Macromolecules 31(16):5546–5549 J (2003) Biodegradable polymersomes.
162. Jain JP, Kumar N (2010) Self assembly of Macromolecules 36(9):3004–3006
amphiphilic (PEG) 3-PLA copolymer as poly- 175. Dimova R, Seifert U, Pouligny B, Förster S,
mersomes: preparation, characterization, and Döbereiner H-G (2002) Hyperviscous
their evaluation as drug carrier. diblock copolymer vesicles. Eur J Phys E
Biomacromolecules 11(4):1027–1035 7(3):241–250
Synthetic Polymer-based Nanomaterials 25

176. Nardin C, Widmer J, Winterhalter M, Meier 187. Brož P, Benito SM, Saw C, Burger P, Heider
W (2001) Amphiphilic block copolymer H, Pfisterer M et al (2005) Cell targeting by a
nanocontainers as bioreactors. Eur J Phys E generic receptor-targeted polymer nanocon-
4(4):403–410 tainer platform. J Control Release
177. Lee JS, Ankone M, Pieters E, Schiffelers RM, 102(2):475–488
Hennink WE, Feijen J (2011) Circulation 188. Lee JS, Feijen J (2012) Polymersomes for
kinetics and biodistribution of dual-labeled drug delivery: design, formation and charac-
polymersomes with modulated surface charge terization. J Control Release
in tumor-bearing mice: comparison with stealth 161(2):473–483
liposomes. J Control Release 155(2):282–288 189. Sasaki Y, Akiyoshi K (2012) Self-assembled
178. Ghoroghchian PP, Li G, Levine DH, Davis nanogel engineering for advanced biomedical
KP, Bates FS, Hammer DA et al (2006) technology. Chem Lett 41(3):202–208
Bioresorbable vesicles formed through spon- 190. Vinogradov SV, Bronich TK, Kabanov AV
taneous self-assembly of amphiphilic poly (2002) Nanosized cationic hydrogels for
(ethylene oxide)-block-polycaprolactone. drug delivery: preparation, properties and
Macromolecules 39(5):1673–1675 interactions with cells. Adv Drug Deliv Rev
179. Ahmed F, Pakunlu RI, Brannan A, Bates F, 54(1):135–147
Minko T, Discher DE (2006) Biodegradable 191. McAllister K, Sazani P, Adam M, Cho MJ,
polymersomes loaded with both paclitaxel Rubinstein M, Samulski RJ et al (2002)
and doxorubicin permeate and shrink tumors, Polymeric nanogels produced via inverse
inducing apoptosis in proportion to accumu- microemulsion polymerization as potential
lated drug. J Control Release gene and antisense delivery agents. J Am
116(2):150–158 Chem Soc 124(51):15198–15207
180. Kim Y, Tewari M, Pajerowski JD, Cai S, Sen 192. Oh JK, Tang C, Gao H, Tsarevsky NV,
S, Williams J et al (2009) Polymersome deliv- Matyjaszewski K (2006) Inverse miniemul-
ery of siRNA and antisense oligonucleotides. sion ATRP: a new method for synthesis and
J Control Release 134(2):132–140 functionalization of well-defined water-
181. Chen W, Meng F, Cheng R, Zhong Z (2010) soluble/cross-linked polymeric particles.
pH-Sensitive degradable polymersomes for J Am Chem Soc 128(16):5578–5584
triggered release of anticancer drugs: a com- 193. Murthy N, Xu M, Schuck S, Kunisawa J,
parative study with micelles. J Control Release Shastri N, Fréchet JM (2003) A macromo-
142(1):40–46 lecular delivery vehicle for protein-based vac-
182. Hickey RJ, Koski J, Meng X, Riggleman RA, cines: acid-degradable protein-loaded
Zhang P, Park S-J (2014) Size-controlled self- microgels. Proc Natl Acad Sci U S A
assembly of superparamagnetic polymer- 100(9):4995–5000
somes. ACS Nano 8(1):495–502 194. Murphy EA, Majeti BK, Mukthavaram R,
183. Pangburn TO, Georgiou K, Bates FS, Kokkoli Acevedo LM, Barnes LA, Cheresh DA (2011)
E (2012) Targeted polymersome delivery of Targeted nanogels: a versatile platform for
siRNA induces cell death of breast cancer cells drug delivery to tumors. Mol Cancer Ther
dependent upon Orai3 protein expression. 10(6):972–982
Langmuir 28(35):12816–12830 195. Turner JL, Wooley KL (2004) Nanoscale
184. Lin JJ, Ghoroghchian PP, Zhang Y, Hammer cage-like structures derived from
DA (2006) Adhesion of antibody- polyisoprene-containing shell cross-linked
functionalized polymersomes. Langmuir nanoparticle templates. Nano Lett
22(9):3975–3979 4(4):683–688
185. Kim B-S, Yang W-Y, Ryu J-H, Yoo Y-S, Lee 196. Desale SS, Cohen SM, Zhao Y, Kabanov AV,
M (2005) Carbohydrate-coated nanocapsules Bronich TK (2013) Biodegradable hybrid
from amphiphilic rod–coil molecule: binding polymer micelles for combination drug ther-
to bacterial type 1 pili. Chem Commun apy in ovarian cancer. J Control Release
15:2035–2037 171(3):339–348
186. Yang X, Grailer JJ, Rowland IJ, Javadi A, 197. Wooley K, Remsen E (1998) Amphiphilic
Hurley SA, Matson VZ et al (2010) core–shell nanospheres obtained by intrami-
Multifunctional stable and pH-responsive cellar shell crosslinking of polymer micelles
polymer vesicles formed by heterofunctional with poly (ethylene oxide) linkers. Chem
triblock copolymer for targeted anticancer Commun 13:1415–1416
drug delivery and ultrasensitive MR imaging. 198. Bütün V, Lowe A, Billingham N, Armes S
ACS Nano 4(11):6805–6817 (1999) Synthesis of zwitterionic shell cross-
26 Swapnil S. Desale et al.

linked micelles. J Am Chem Soc 121(17): 202. Vinogradov SV, Batrakova EV, Kabanov AV
4288–4289 (2004) Nanogels for oligonucleotide delivery
199. Zhang J, Zhou Y, Zhu Z, Ge Z, Liu S (2008) to the brain. Bioconjug Chem 15(1):50–60
Polyion complex micelles possessing thermo- 203. Nukolova NV, Oberoi HS, Cohen SM,
responsive coronas and their covalent core Kabanov AV, Bronich TK (2011) Folate-
stabilization via “click” chemistry. decorated nanogels for targeted therapy of
Macromolecules 41(4):1444–1454 ovarian cancer. Biomaterials
200. Torchilin VP (2012) Multifunctional nano- 32(23):5417–5426
carriers. Adv Drug Deliv Rev 64:302–315 204. Nukolova NV, Oberoi HS, Zhao Y,
201. Kim JO, Kabanov AV, Bronich TK (2009) Chekhonin VP, Kabanov AV, Bronich TK
Polymer micelles with cross-linked polyanion (2013) LHRH-targeted nanogels as a deliv-
core for delivery of a cationic drug doxorubi- ery system for cisplatin to ovarian cancer. Mol
cin. J Control Release 138(3):197–204 Pharm 10(10):3913–3921

View publication stats

You might also like