Spicer2018. Peptide and Protein NPs Conjugates. 2018
Spicer2018. Peptide and Protein NPs Conjugates. 2018
Peptide– and protein–nanoparticle conjugates have emerged as powerful tools for biomedical applications,
enabling the treatment, diagnosis, and prevention of disease. In this review, we focus on the key roles played
by peptides and proteins in improving, controlling, and defining the performance of nanotechnologies.
Within this framework, we provide a comprehensive overview of the key sequences and structures
utilised to provide biological and physical stability to nano-constructs, direct particles to their target and
Received 16th December 2017 influence their cellular and tissue distribution, induce and control biological responses, and form
DOI: 10.1039/c7cs00877e polypeptide self-assembled nanoparticles. In doing so, we highlight the great advances made by the field, as
well as the challenges still faced in achieving the clinical translation of peptide- and protein-functionalised
rsc.li/chem-soc-rev nano-drug delivery vehicles, imaging species, and active therapeutics.
3574 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
the difficulties that arise as a result of these considerations. drugs often suffer from poor pharmacokinetics, exhibiting rapid
These hybrid materials enable the favourable characteristics clearance and difficulties reaching the desired site-of-action
of nano-sized structures to be combined with the biological in vivo.1 As a result, severe side-effects may accompany any
activity, biocompatibility, and versatility of both naturally derived therapeutic benefit, while it is common for in vitro efficacy to be
and synthetic polypeptides. poorly translated to a clinical setting.7 The ability of NPs
In this review, we provide a comprehensive overview of to solubilise therapeutic molecules, enhance retention and
the development and use of peptide/protein–NP conjugates in circulation, and promote accumulation in the target tissue
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
biomedicine. While many reviews have previously been pub- makes them attractive vehicles for overcoming these limita-
lished on methods to create NP conjugates, or their subsequent tions, which hinder the drug discovery process.8 These effects
applications within the body, the specific features imparted can be further enhanced through the incorporation of peptide
by the peptide or protein on the NP conjugate have been or protein coatings, as will be the focus of this review, by further
less widely discussed. We will therefore focus on the distinct improving pharmacokinetics, enabling tissue targeting, and
roles played by the peptide/protein in improving, controlling, or promoting cell and tissue penetration.9 Similarly, NP drug
defining the performance of nano-technologies (Fig. 1). In doing delivery vehicles offer several benefits over the use of protein–
so, we will deliver a detailed reference for both experts and those and antibody–drug conjugates (ADCs), which have emerged over
new to the field of NP technologies alike. Furthermore, we hope the last 10 years as promising clinical tools for the treatment of a
to stimulate discussion and innovation within the field, in order range of diseases.10 In particular, NP platforms offer the possibility
to overcome many of the difficulties that continue to hinder the to incorporate multiple functionalities within a single con-
clinical translation of these potentially powerful tools. struct. As a result, problems such as the poor tumour penetra-
tion often exhibited by ADCs can be overcome, as described in
Section 5.10,11 Furthermore, the encapsulation of the therapeutic
2. The benefits of peptide/protein–NP agent allows the need for a cleavable linkage to be avoided, and
conjugates in biomedicine enables high levels of drug to be delivered for every recognition
event.12
The unique properties and size regime of NPs offer many benefits The benefits of both bare NPs and those decorated by
over small molecules and larger micrometre sized particles. These polypeptides, have also been widely exploited in the field of
have been widely exploited within the biomedical field and in vivo imaging. When compared to small molecule imaging
reviewed extensively elsewhere.1–6 By way of context for this agents, NPs often offer the advantages of greatly improved
review, we will briefly summarise here some of the key factors signal-to-noise ratios, stable signal generation, high spectral
that make NP-based technologies particularly attractive, and resolution for multiplexed detection, and the ability to display
the roles in which they have predominantly been applied. multimodal signal generation.13,14 As a result, NPs have found
One of the most prominent realms in which NP systems have increasing utility for imaging in a range of modalities, including
found utility is as vehicles for drug delivery. Small molecule near-infrared (NIR) fluorescence,15 magnetic resonance imaging
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3575
View Article Online
Fig. 1 Polypeptides can play an important role in determining NP functionality and fate. In this review, we will focus on the features imparted by the
peptide/protein and their influence on NP behaviour.
(MRI),16 and positron emission tomography (PET).17 Indeed, others have also been utilised in more niche settings, such
many NP-based imaging technologies are now routinely used in as peptide-based NPs as discussed in Section 8). Each will be
a clinical setting.18 briefly introduced in order to contextualise the discussion that
Stimuli responsive NPs are also finding increasing utility follows (Table 1):
within the biomedical field, often leading to the localised destruc- (i) Gold nanoparticles (AuNPs). AuNPs are attractive struc-
tion of pathological tissue.19 Whether as a result of magnetic or tures for biomedical applications.22 As one of the most stable
light induced hyperthermia, or NP-mediated photoablation, such and least toxic metal NP formulations, AuNPs offer a safe and
technologies are strongly reliant on adequate accumulation at the effective diagnostic and therapeutic tool.23 The ability of thiols to
site of treatment. Many systems rely on inorganic cores, from form stable linkages with the surface of AuNPs allows versatile
which it is vital to reduce the leaching of metal components due surface chemistry to be achieved.24 Decoration of the particle
to their associated toxicity. As a result, peptide or protein coatings surface with a wide range of biomolecules enables the biodistribu-
that can direct, stabilise, and eventually lead to the clearance tion, physical properties, and intracellular fate to be modulated.
of therapeutic NPs are a key component of these emerging Furthermore, AuNPs exhibit unique optical and electronic
technologies as they approach clinical application.20 properties, with concerted electron oscillation following excitation
leading to strong light emission via localised surface plasmon
resonance (LSPR).25 Their interaction with light is strongly depen-
3. Commonly utilised nanoparticles dent on size, shape, surface chemistry, aggregation and environ-
ment. These properties enable the use of AuNPs in a range of
NPs have found widespread use across the material, physical, imaging modalities, including NIR fluorescence, photoacoustic
engineering, and biological sciences, with the number of new imaging, and X-ray computational tomography, as well as finding
technologies increasing at an exponential rate.21 The particle utility in photoablation and hyperthermic therapies.22,26
structures which enable these applications are equally diverse, (ii) Magnetic nanoparticles (MNPs). MNPs are composed
with precise control over material, architecture, and design predominantly of magnetically-responsive elements, such as iron,
enabling a broad spectrum of tunable properties dependent on nickel, and cobalt in various different forms, and are finding
the end application.4 Despite this large variation, a number of increasing utility in the biomedical field.27,28 Iron oxide particles
NP formats have found particular utility in the biomedical field. have found most widespread use, particularly as high contrast
Much of the research to be discussed within this review has imaging agents for MRI.29 The application of alternating magnetic
focussed on the use of 6 main categories of NPs (though many fields can also be utilised as a minimally invasive stimuli for
3576 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
Magnetic NPs Active for high-contrast MRI imaging Cytotoxic and poor biocompatibility
Can be guided magnetically Non-biodegradable
Can be utilised for photodynamic therapy
Semi-conducting Size tunable, high quantum yield emission Fluorescence sensitive to surface functionalisation
NPs and QDs Resistant to photo-bleaching and degradation Toxicity of metal components
Broad wavelength excitation Non-biodegradable
Narrow emission allows multiplexing
Liposomes Low immunogenicity and high biocompatibility Low loading efficiency of valuable cargo
Ease of functionalization Poor stability and prone to leakage
Flexibility of formulation for tuning of structure
Can encapsulate hydrophilic and hydrophobic cargoes
Polymer NPs Versatile function and structure Potential toxicity of both NP and degradation products
Ease of modification and tunability
Can be made to be degradable or stimuli responsive
generating hyperthermia as described above.30 Finally, the to their applications in drug delivery, MSNPs have also found
inherent magnetism of MNPs enables their spatial distribution widespread use as imaging agents, as a consequence of their
to be easily manipulated, allowing the guided delivery of drug- ability to encapsulate and concentrate contrast agents for a wide
containing vehicles.31 range of modalities.38
(iii) Semi-conducting NPs and quantum dots (QDs). In order (v) Liposomes. Liposomes are spherical vesicles, most commonly
to undertake fluorescence imaging in vivo it is advantageous composed of at least one bilayer of self-assembled phospholipids.
for emission to be in the NIR to allow tissue penetration.32 The similarity of the liposome bilayer to that of the cell membrane is
Conventional small molecule NIR fluorophores suffer from particularly attractive for drug delivery applications, and indeed
poor photo-stability, high hydrophobicity which hinders dis- liposomes are perhaps the most widely used and investigated NP
tribution, and weak signal-to-noise generation. In contrast, carriers for drug delivery, and were amongst the first NP-based
semi-conducting NPs (and in particular QDs) possess narrow, technologies to enter the clinic.2 Liposomes are able to encapsulate
size-tunable, and high quantum yield emission.33 Furthermore, both hydrophilic cargo, within the aqueous interior, and hydro-
they can be excited with broad wavelength light and display phobics, within the membrane bilayer. Furthermore, liposomes
greatly improved resistance to photobleaching and chemical can be readily functionalised with lipid, or hydrocarbon func-
degradation. They are therefore highly attractive structures for tionalised ligands, making them a stable, cost-effective, and
undertaking biomedical imaging.34 However, the high toxicity attractive tool for biomedical applications.
of cadmium metal, common in many QD formulations, neces- (vi) Polymer nanoparticles. The versatility of synthetic polymers
sitates the use of stable coatings able to prevent leaching into enables a wide range of particle architectures and end-
the biological milieu. applications to be explored. Polymer vesicles (commonly referred
(iv) Mesoporous silica nanoparticles (MSNPs). MSNPs possess to as polymersomes) and micelles, composed of self-assembled
tunable pore sizes, large surface areas, and high pore volume. amphiphilic block copolymers, are particularly attractive due to
They are therefore attractive drug-delivery vehicles, with the their ability to encapsulate cargoes in an analogous fashion to
ability to encapsulate a large payload of cargo ranging from their corresponding lipidic analogues.39,40 The NP size, membrane
small molecules up to large proteins.35,36 The ease of surface thickness, porosity, and many other factors can be tuned by
modification, and cost-effective and scalable production adds to adjusting block length and pendant group functionalisation.
the attractiveness of MSNPs. Furthermore, the classification of Furthermore, the design flexibility enabled by synthetic polymer
silica as a ‘Generally Recognized as Safe’ substance by the United chemistry enables the introduction of stimuli-responsive or
States Food and Drug Administration (FDA) greatly facilitates biologically active functionalities, which are able to modulate
regulatory approval of MSNP technologies, though questions on particle properties in a smart, predictable manner.41 As such,
the long-term effects, biodegradability, and biocompatibility of polymer NPs are finding increasing use across a number of
many silica nanotechnologies remain unanswered.37 In addition biomedical disciplines.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3577
View Article Online
It is striking that the number of NP systems that have found In order to reach the desired site-of-action, NPs must navigate their
successful application in biomedical applications pales in way around an ensemble of biological obstacles. These include
insignificance when compared to the vast body of literature defined biological surfaces, endothelial and cell membrane barriers
on the use of NP constructs under controlled model settings.42 to permeation, and circulating monocytes and macrophages of
This is predominantly due to the significant challenges the body’s immune system.44 To achieve their function, NPs
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
associated with translating NP stability under idealised condi- must circulate in the bloodstream for long enough to reach
tions to the complex environments of biologically relevant their target cells, tissues, or organs, whilst avoiding removal by
scenarios.43 Peptide and protein coatings play a major role in active phagocytosis or renal clearance.44,45 The nanoscale
enabling NP based technologies, and their ability to provide dimensions in themselves can lead to greatly altered pharmaco-
both biological and physical stability will be summarised in kinetic properties when compared to small molecules in vivo.
this section (Fig. 2, Tables 2 and 3). Importantly, these two For example, for ‘hard’ inorganic particles, renal clearance is
factors are not necessarily independent or complementary. As greatly reduced above a diameter of B5.5 nm due to the size
will be discussed, a coating which prevents NP aggregation may cut-off of the kidneys for urinary excretion (Fig. 3).46 This ability
also impact on the rate at which the construct is cleared from of NPs to modulate pharmacokinetics has been widely utilised as a
the circulation. It is therefore important to carefully consider means to improve the retention, biodistribution, or in vivo stability
design criteria in order to ensure that the end-application can of an encapsulated small molecule.47 However, these differences
be achieved. do not come without their own challenges – dependent on the
Fig. 2 Methods by which peptide/protein–NP coatings can be designed to influence biological stability and decrease susceptibility to clearance:
(a) recognition and clearance can be limited by providing balanced charge or surface hydrophobicity; (b) ‘self-peptides’ can be recognised by
macrophages, and used to inhibit phagocytosis; and (c) the formation of a protein corona can be modulated by providing a stable peptide/protein
coating, promoting the absorption of dysopsonins, or by tuning NP properties such as size, shape, or charge (i–iii).
Table 2 Peptides mediating biological and physical stability discussed in this review
3578 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
a
UniProt; PDB – protein data bank; pI estimated using http://isoelec
tric.ovh.org/.
Fig. 4 Upon entering the body, NPs quickly acquire a protein corona. The
exact composition of the corona is dynamic and highly dependent on the
environment: (a) an initial corona is formed of typically highly abundant
proteins; (b) weakly bound proteins are gradually removed by proteins with
a higher affinity for the NP surface; (c) adsorption of proteins can be
dependent on the already existing coating, with inter-protein as well as
protein–NP interactions determining affinity; and (d) gradually a stable
coating of strongly adsorbed proteins is formed, creating a ‘hard’ protein
corona. Reproduced from Monopoli et al. with permission from Nature
Fig. 3 Retention of different size QDs 4 h after intravenous injection. Publishing Group.49
Particles of o5 nm diameter are rapidly cleared to the kidney, while larger
particles are retained. Adapted from Soo Choi et al. with permission from
Nature Publishing Group.46 lipoproteins (often referred to as dysopsonins) recognition is
blocked and circulation times are increased.53,58,59 Amongst the
other NP properties known to promote clearance, surface charge
size, structure, or functionalisation of a NP, detrimental tissue and hydrophobicity are particularly important. Although conflicting
accumulation can occur, while the uptake of particles by macro- reports exist on the relative clearance rates of positively or
phages can not only lead to rapid removal from the circulation, but negatively charged particles, it has become apparent that a near-
also the induction of an unfavourable inflammatory response.48 neutral charge may in fact be most favourable.60 At the same
As soon as NPs are introduced into a biological environment time, a reduction in hydrophobicity has also shown to be a key
further complications arise, as a mixture of biomacromolecules parameter in reducing the uptake of NPs by macrophages.45,60
interacts with the particle surface, often masking the effect While beneficial from a biological perspective, it is important to
of the particle and strongly influencing the pharmacokinetic note that particles which exhibit neutral or low surface charge
properties.49–51 In protein-rich media, such as blood, this new often exhibit reduced colloidal stability, as discussed in the
coating is referred to as the protein corona. The ‘hard’ corona is subsequent section.61 It is therefore important to consider
made up of tightly-bound proteins, forming thermodynami- the delicate balance that determines NP fate when designing
cally favoured interactions with the NP surface, followed by a peptide/protein coatings.
‘soft’ corona of rapidly exchanging proteins (Fig. 4).52,53 The One widely utilised method to limit opsonisation and NP
nature of this biomolecule coating is highly dependent on the clearance from the circulation is the use of stealth polymers,
environment and thus difficult to predict, often resulting in a such as poly(ethylene glycol) (PEG), which form a protective
dramatic loss of activity or function and strongly influencing layer around the NP, neutralizing surface charge, conferring
the nanomaterial fate in vivo.45,54,55 hydrophilicity, and providing a steric barrier to adsorption.56,62
Peptide and protein coatings can be used to improve upon However, there is a fine line between providing a coating which
the biological stability of delivered NPs and address these improves biodistribution and diminishing interactions with target
difficulties. By altering the interactions with host cells and cells and tissues.61 The coating of NPs with PEG has been associated
circulating biomolecules, peptide/protein coatings can modulate with reduced cellular uptake and thus therapeutic efficacy,63,64
and control tissue accumulation to ensure NP activity (Fig. 2). In the generation of PEG-specific antibodies which accelerate
order to understand how this can be achieved, it is important to clearance from the blood,65,66 and preferential accumulation
first understand the factors which induce clearance. The adsorp- in the liver and spleen.67 The versatility of peptide design thus
tion of opsonins (plasma proteins such as immunoglobulin G, offers an attractive alternative to such coatings (Fig. 2a). The use of
complement factors, and fibrinogen) to the NP surface plays a non-ionic peptides, or those offering zwitterionic balanced charge
prominent role in triggering clearance, by inducing macrophage may be particularly useful for reducing clearance. Following this
recognition and subsequent elimination via phagocytosis.56,57 By strategy, Guerrero et al. conjugated the amphipathic peptide
inhibiting this process, so too can the recognition and removal CLPFFD, previously shown to mediate transport across the
of NPs by the immune system be reduced.56 In contrast, blood–brain barrier, to AuNPs with the aim of increasing particle
when the protein corona is enriched with serum albumins or delivery to the central nervous system.68 By reducing the negative
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3579
View Article Online
Morais et al. reported that the coating of AuNPs with CALNN led
to increased clearance and liver accumulation compared to
citrate-capped particles.70 These results highlight the difficulty
in predicting pharmacokinetic properties – often apparently
similar peptide sequences can have drastically different effects
on clearance. This conclusion is strongly supported by the
research of Poon et al., who revealed the complexity of designing
peptide–NP conjugates for escaping the body’s reticuloendothelial
system.71 By coating AuNPs with a mixture of PEG and either
the negatively charged therapeutic peptide Myx or the positively
charged cRGD targeting peptide, it was found that the effect
of the PEG/peptide coating could act independently or syner-
gistically, depending on the sequence employed. As such, it
is important to evaluate NP pharmacokinetics on a case-by-
case basis.
An alternative approach to avoiding clearance is to rely on
biological signalling to enhance retention. This has been most
prominently achieved by using the membrane protein CD47. By
interacting with the signal-regulatory protein alpha (SIRPa) and
triggering downstream anti-phagocytic processes, CD47 acts in
effect as a ‘marker of self’, sending out a ‘do not eat me signal’
Fig. 5 (a) Near-infrared imaging of mice injected with control NPs, or
(Fig. 2b).72–75 Peptides derived to mimic the effect of CD47 have thus those coated with human CD47 or a CD47-derived peptide. An increase
been designed and shown to reduce phagocytosis.76 Importantly, in tumour accumulation is observed as a result of reduced clearance;
peptide coated NPs displayed greatly reduced accumulation in (b) quantification of tumour fluorescence intensity. Adapted from Rodriguez
the liver and spleen, and enhanced accumulation in cancerous et al. with permission from The American Association for the Advancement
of Science.78
tissues following intravenous injection (Fig. 5). Similarly,
Qie et al. recently demonstrated that NPs functionalised with
full-length CD47 were also able to modulate clearance time, surfaces and can also alter the composition of the protein
enabling the evasion of different macrophage populations.77 corona when NPs are exposed to bodily fluids.52 The high
However, this work also highlighted the need to better under- affinity of hydrophobin enables the protein to remain strongly
stand the interaction of nanomaterials with macrophages dis- associated with the NP surface, even when in competition with
playing distinct phenotypes, in order to pave for the way for other plasma proteins. Apolipoprotein coatings have also been
truly immunologically inert nanomaterials.77 shown to increase NP circulation time. A study of polystyrene–NP
The adsorption of dysopsonins is known to enhance retention. coatings demonstrated that amino and sulfonate functionalised
A third approach to improve NP pharmacokinetics is therefore to particle coatings led to the accumulation of high levels of
promote the formation of a favourable dysopsonin-protein corona apolipoprotein.59 These results highlight the importance of
(Fig. 2c). This can be achieved either through the pre-coating of surface charge and functional groups on influencing protein
NPs prior to in vivo application,79 or by tuning the NP surface corona formation. Notably, the binding of specific apolipoprotein
properties to promote the enrichment of dysopsonins over subtypes was also shown to strongly influence subsequent cell
opsonins in situ.53,80 Serum albumin, the most abundant uptake in vitro.59 Schöttler et al. demonstrated that the apolipo-
protein in blood, plays a wide range of roles including acting protein clusterin (also known as ApoJ) reduced the non-specific
as a molecular and protein transporter, maintaining oncotic cellular uptake of sterically protected polymer-NPs.80 A recent
pressure, and buffering blood pH. Pre-formation of an albumin study highlighted the effect of initial polymer coverage on the
corona is also able to act to prevent the attachment of alter- downstream effects of clusterin, which was able to shield NPs
native proteins and improve NP stability.53 Peng et al. showed from opsonisation at low PEG densities but did not effect NP
that such a strategy was able to limit phagocytosis and prolong clearance at higher PEG coverages.82 The downstream effects of
circulation time in vivo, through simple NP pre-incubation in a protein corona formation must also therefore be considered.
solution of bovine serum albumin (BSA).58,81 Hydrophobin, an Fibrinogen is a circulating glycoprotein in the bloodstream
exogenous protein expressed by fungi has high affinity for NP and a well known opsonin able to inhibit cell adhesion at
3580 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
suitable concentrations. When low fibrinogen concentrations for which the high susceptibility to aggregation must be care-
are adsorbed on NP surfaces, a single highly adhesive monolayer fully mitigated. For example, depending on the capping layer
is formed.83 In contrast, dense coatings at high concentrations utilised, unfunctionalised AuNPs are incredibly sensitive to
reduce cell adhesion under both static and flow conditions, via environmental changes, with small changes in pH, salt concen-
the formation of a nanoscale multilayer matrix.84 However, tration, temperature, or the presence of biomolecules often
translating fibrinogen to designed NP coatings for increased leading to rapid and irreversible aggregation.26,33,99–103 Through
circulation remains challenging, as fibrinogen can also lead to combinatorial design, peptide sequences have been identified
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
NP aggregation as a consequence of the formation of inter- that are able to find a careful balance of charge, polarity,
particle bridges.85 functionality, hydrophobicity, and length.104 In 2004, Lévy et al.
identified the pentapeptide CALNN through such libraries, as a
4.2 Physical stability water-soluble peptide able to provide extremely stable AuNPs
The physical stability of a NP construct is an important con- following surface functionalisation.105 The presence of the
sideration when designing a biomedical technology. Not only N-terminal cysteine enables facile functionalisation of the particle
are suitable stabilities required to ensure that the particle is surface, with the rest of the sequence providing a densely packed,
able to fulfil its function, but the leaching of small molecules negatively charged peptide corona which is able to withstand
from the particle structure or the formation and accumulation aggregation.
of larger aggregates must also be controlled to minimise The ability of thiols to bind to metal surfaces has led to
toxicity. In order to demonstrate the ability of NPs to be applied cysteine capped peptides being widely utilised as capping agents
both in vitro and in vivo they must form a dispersed, stable for metal NPs.106–109 Indeed, peptides such as CALNN can be
suspension under physiological conditions. Several reports used as a core peptide to which additional biological function-
have demonstrated that agglomerated NPs have drastically ality can be attached, providing a stable peptide-functionalised
altered properties, in particular toxicity, which can lead to large inner corona bearing pendant peptides which can then direct
discrepancies in experimental output.86–90 Indeed, aggregated and influence NP activity.110–112 However, although cysteine–
particles display a greatly decreased surface area and reduced metal binding is sufficient to provide stabilised NPs in vitro, the
cellular uptake when compared to dispersed particles, often reversibility of thiol–metal bond formation means that ligand
leading to an underestimation of toxicity prior to application exchange can occur.113–115 The concentration of free thiols in the
in vivo. In contrast, sedimentation and decreased diffusion can blood is far lower than that observed intracellularly, but remains
significantly impact the ‘effective dose’ experienced by cells high enough to induce loss of monothiol coatings during
in vitro, leading to large discrepancies in experimental readout circulation.116 This is in part due to the possibility for dissocia-
and significantly decreased efficacy upon in vivo translation.91,92 tive ligand exchange (SN1-like pathway), in which ligands are
NP aggregation can also trigger opsonisation in the bloodstream, rapidly diluted within serum upon dissociation, and replaced by
as discussed above, making particles more visible to the phago- circulating small molecule thiols.117 In addition, the high levels
cytotic system.63,64 Thus, physical stability and biological stability of serum albumin in the blood, containing a single reduced
are intimately linked. cysteine residue, have been shown to mediate ligand exchange in
The flocculation of particles in aqueous solution is governed by biological samples.118,119 This loss of NP coating can lead to two,
the Derjaguin–Landau–Verwey–Overbeek theory (DLVO theory).93,94 often concurrent outcomes in the dilute regime provided by
The stability of the dispersion depends on the balance between in vivo settings: (i) replacement of the initial thiol coating by
attractive and repulsive forces – a particle becomes unstable and thiolated biomolecules, leading to a loss of any function imparted
starts to agglomerate when the repulsive energy is not sufficient to by the capping layer, as discussed throughout this review; and
counteract the van der Waals attractive energy.88 At biological salt (ii) loss of any stabilization effects provided by the capping layer as
concentrations the situation becomes more complicated, however ligands are gradually lost.120 As a result, increasingly stabilised
the basics for stability remain similar.95 Sufficient electrostatic peptide coatings have been explored in recent years. This is often
and steric stabilization is required to provide the repulsive achieved through the use of multi-dentate binding sites – that is,
energy to prevent agglomeration.88 For biomedical applica- to use multiple cysteine residues or unnatural amino acids
tions, coatings must not only provide colloidal stability, but bearing dithiol motifs, to provide multiple anchoring points to
also aqueous solubility, while conserving the NP functionality. the metal surface for each individual peptide (Fig. 6).121–124 In
The formation of a protein corona upon application in vivo may doing so, even with reversible desorption of one thiol group from
either enhance or compromise NP colloidal stability. Most the metal surface, the other is still attached maintaining stability.
commonly, the steric bulk provided by the protein coating Due to the proximity of the unbound thiol, this will then simply
can provide significant stabilisation, limiting inter-particle reattach and reform the stabilised NP construct.
attractive forces.49,96,97 This phenomena is able to offset the low An alternative approach, particularly widely employed for the
contribution of stabilising electrostatic repulsion forces which are functionalisation of QD surfaces, is the utilisation of poly-histidine
largely negated in the presence of the high electrolyte concentra- sequences, able to form multi-dentate NP interactions. The ability of
tions of physiological conditions.97,98 histidine residues to bind a variety of metals is well established
Peptides have been particularly widely used to provide throughout biology, and the hexa-histidine motif in particular
physical stabilisation to metal and semi-conducting particles, has been found to form stable metal coordinates (Fig. 6).126–128
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3581
View Article Online
Fig. 6 Monothiol ligands are prone to ligand exchange on the surface of metal NPs, and coatings can therefore be rapidly lost in biological
environments. In contrast, bidentate dithiol ligands offer increased stability, as two simultaneous exchange events are required in order to disrupt the
coating. Alternatively, hexahistidine ligands offer strong and stable metal binding.121,125
Hexahistidine tagged peptides and proteins have therefore Furthermore, even when NPs are taken up by cells, for example via
been widely used to provide stable biomolecule AuNP and QD endocytosis, the challenges are not over – up to 99% of particles
coatings which are resistant to desorption for a wide variety of may be sequestered from the cytosol or nucleus in endosomes and
applications.125,129–132 organelles, depending on their exact properties, the target cell type,
and the uptake mechanism.135,136 There is therefore a pressing
need to develop NP constructs that can be directed to the desired
5. Promoting cell and tissue site-of-action, in order to maintain functionality.
penetration In recent years the decoration of NP surfaces with peptides able
to promote uptake has come to prominence, with many so-called
For a NP to be useful in biomedical applications, it is vital that ‘cell-penetrating peptides’ (CPPs) and sequences able to trigger
the particle is able to reach the necessary site of action – the receptor-mediated endocytosis being studied in detail.137,138
ability of a NP to fulfil its specified purpose is not sufficient. Furthermore, sequences able to efficiently promote endosomal
This is particularly true for particles that are delivered systemically, escape following internalisation have also been identified and
rather than being applied directly at the desired site of action. Even used to great effect. We will here first briefly discuss the key
once a particle has overcome the significant challenge of avoiding mechanisms of peptide-mediated NP uptake, before highlighting
rapid clearance, the body places formidable barriers in the way the main sequences that have been most widely utilised in
of subsequent distribution and delivery of NPs at both the biomedical applications. As a word of caution, it is important
tissue and cellular level.1 These restrictions are the body’s way to note that the efficacy of both penetration and subsequent
of protecting and maintaining homeostasis, preventing the endosomal escape is highly dependent on the experimental
uncontrolled transport of material into and out of sensitive conditions.139,140 While a particular sequence may successfully
environments. Penetrating these barriers remains one of the mediate the transport of one NP cargo, it may fail to enable
biggest challenges in biomedicine, both for small molecule penetration of another.141 Similarly, while uptake may be effi-
therapeutics and the NPs that are the focus of this review. The cient with a cell line in vitro, such results may not be readily
decoration of particles with peptides or proteins able to mediate replicated in the corresponding tissue upon in vivo translation. It
transport or penetration is at the forefront of efforts to enable NP is therefore paramount to test and characterise a newly designed
technologies to reach the areas they need to perform their NP–CPP construct under biologically relevant conditions, rather
function. In this section, we will discuss each barrier in turn than relying on literature precedence for the transport of related,
and give a critical overview of the key sequences that have been but essentially distinct NP cargoes.
utilised to modulate NP transport across them (Tables 4 and 5). Mechanism of uptake. In general, penetration mechanisms
can be split into two major categories: (i) direct, energy-
5.1 Cell penetration independent pathways; and (ii) those relying on active uptake
The cell membrane is a selectively permeable barrier, which via endocytosis (Fig. 7). Often, multiple uptake pathways will be
efficiently protects the cell interior from its environment. Some exploited in parallel, with the exact environmental conditions
specific small molecules may be efficiently transported across the determining which process dominates.142 As such, it can prove
membrane, however the transfer of NPs into the intracellular space complicated to deduce the exact mechanism of internalisation
is far more challenging.133 Although in some cases the uptake of and conflicting reports are therefore commonplace.143
unfunctionalised particles is possible, cell membrane imperme- Direct uptake mechanisms are typically initiated by the
ability often results in a highly detrimental lack of activity.134 interaction of the peptide–NP conjugate with the cell membrane,
3582 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
Table 5 Proteins mediating penetration discussed in this review This is followed by permanent or transient destabilization of the
cell membrane, for example through pore or inverted micelle
Protein name PDB # Mw pI Role
formation.142,145 Although many CPPs were at first thought to be
Transferrin 1D3K 76 000 5.5 BBB penetration uptaken via such means, subsequent reports have demonstrated
OX26 N/A N/A N/A BBB penetration
Lactoferrin 1LFG 77 302 8.7 BBB penetration that experimental artefacts in fact led to initial discrepancies and
it is no longer thought that direct uptake is the major contributing
PDB – protein data bank; pI estimated at http://isoelectric.ovh.org/.
factor in the penetration of CPP-coated NPs.145,146 In contrast,
during endocytotic uptake, interactions with cell membrane
most commonly via electrostatic interactions of positively components lead to the engulfment of the peptide–NP construct,
charged surfaces with negatively charged phospholipids.110,144 which is then transported intracellularly in endosomes.
Fig. 7 Schematic demonstrating the main mechanisms by which NPs are uptaken and subsequently intracellularly processed by cells. The exact
mechanism taken by a particular particle is highly dependent on the precise NP characteristics, polypeptide coating layers, target cell type, and
environment. The situation is further complicated by the ability of particles to exploit multiple different uptake pathways in parallel. Adapted with
permission from Zhu et al.149 Copyright 2013 American Chemical Society.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3583
View Article Online
Endocytosis occurs predominantly via macropinocytosis, or peptides are able to deliver their cargo – it is difficult to
clathrin- or caveolin-dependent receptor-mediated uptake, improve upon the penetration efficiency, even if other proper-
but in all cases subsequent escape of the cargo from their ties leave a lot to be desired, as described below.
resultant endosomal location is a key step, as described in the The most commonly utilised method to mediate internalisa-
subsequent section. tion is to rely on highly cationic peptide sequences, able to first
The exact mechanism by which a particular NP is internalised is strongly bind negatively charged cell membranes and then
a vast area of research, and one which often remains contentious. induce transport via either a direct or endocytic pathway. This
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
Exact details are therefore outside the scope of this review and group includes the now ubiquitous human immunodeficiency
readers are instead directed to a number of excellent reviews on virus (HIV) derived TAT peptide, as well as synthetic polyarginine
the topic.133,139,147,148 It has become increasingly evident that it and penetratin (derived from the Drosophila Antennapedia
is difficult to draw generalisations. Importantly, the precise homeodomain) sequences. Indeed, the CPPs utilised for the
means by which a particular construct is taken up is highly in vivo delivery of NP substrates fall almost exclusively into this
dependent on not only the peptide sequence used to promote category. In 1999 Schwarze et al. demonstrated that the con-
uptake, but also on the target cell type, and perhaps most jugation of TAT to a protein substrate could be used to deliver
strongly the exact nature of the NP cargo, including size, the cargo to all parts of the body.152 Since this initial report, a
structure, and surface properties. number of NP cargoes have been systemically delivered to all cells
Key peptide sequences. By some estimates, in excess of indiscriminately, with preferential localisation determined merely
800 unique peptides have been identified which are able to by the NP pharmacokinetics.153,154 The passive accumulation of
promote cell penetration in in vitro experiments.150 Of these, NPs in tumours has also been exploited to allow the preferential
only a handful of prominent sequences or closely related delivery of TAT-labelled, drug-loaded micelles155 and chitosan
analogues have been translated to biomedical applications. NPs,156 as well as anti-tumour silver NPs157 to tumour cells in
Although there are many possible explanations for the paucity animal in vivo models (Fig. 8). Similarly, arginine-rich peptides158,159
of peptides that have been used in such settings, 2 factors are of and penetratin160,161 have been used to both penetrate tumour
particular importance. Firstly, many sequences induce toxicity, cells, following passive accumulation, and to target the brain as
particularly amphipathic peptides which often induce membrane covered in Section 5.3. Interestingly, the non-natural D-enantiomer
disruption in order to promote uptake.151 Secondly, and perhaps of TAT has also been shown to mediate cell penetration. This
more importantly, is the efficiency with which the best CPP strongly suggests that NP uptake as a result of TAT decoration
Fig. 8 Cellular uptake of: (a) phosphate-buffered saline (PBS); (b) bare silver NPs; and (c) TAT-functionalised silver NPs. TAT-particle accumulation
in tumours enables a reduction in tumour growth when compared to unfunctionalised particles, due to increased intracellular delivery. Adapted from
Liu et al. with permission from Elsevier.157
3584 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
is not due to a specific biological interaction – rather it is the environment, the TAT peptide was unmasked enabling cell
density or distribution of positive charges along the peptide uptake. Alternatively, ultra-violet (UV) light-cleavable groups
backbone that are responsible for internalisation.154,162 Impor- can be used to trigger cleavage of appended lipids with spatial
tantly, despite the ability of cationic peptides to increase uptake precision.168 Finally, hydrazone-linked PEGs that are cleaved in
efficiency, cytosolic delivery of the NP cargo often remains the mildly acidic tumour environment have also been reported
low.163 The strong affinity of positively charged peptides towards for the selective unblocking of TAT peptides at the desired site-
negatively charged endosomal membrane components may of-action.169,170 Importantly, hydrazone structure has been
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
hinder endosomal escape. As a result, additional factors able shown to play a vital role in the rate of hydrolysis, and thus
to mediate this process may need to be incorporated during NP the ability of the NP construct to undergo acid-mediated
design, as discussed in Section 5.2. cleavage. By suitable choice of coupling partners, hydrazone
Increasing cell and tissue specificity. In all of the cases noted half-life can be tuned from a matter of minutes, all the way up
above, the low specificity of CPPs for a particular cell type or to months at neutral pH.171,172 This therefore represents an
target organ leads to widespread tissue delivery. CPPs cross cell important consideration during construct design. For example,
membranes in a largely indiscriminate manner, leading to the aliphatic hydrazones may display insufficient stability to allow
uptake of NPs by almost all cells that are encountered. As such, application in vivo,170 while diaryl linkages may prove too stable
technologies relying on these sequences are often associated to allow acid responsive behaviour to be displayed.173
with significant side-effects, particularly when they are used to Role of peptide/protein density. The density of CPP surface
deliver a therapeutic payload. In order to address this limitation, coverage is an important determinant of NP behaviour. Sufficient
a number of methods for CPP ‘screening’ have been reported ligands must be presented to enable efficient penetration, while
that enable NP targeting prior to unmasking of the penetrative also avoiding overcrowding which can diminish bioactivity and
sequence. In an important early demonstration of this principle prevent the presentation of dual-functionalities.174 In the early
the Tsien group attached a complementary polyanionic peptide, 2000s, Zhao et al. demonstrated that the efficiency of MNP uptake
able to electrostatically bind and thus block the activity of a was highly dependent on the number of conjugated TAT
polyarginine CPP, via a matrix metalloproteinase (MMP) cleavable peptides.175 Penetration was greatly enhanced when more than
linker region.164 In the presence of the corresponding protease, 10 peptides were displayed on the particle surface, with a non-
typically upregulated within the tumour environment, peptide linear response suggesting multi-valent interactions were at least
cleavage resulted in the unmasking of the cationic CPP and partially responsible for the increase in efficiency. This has been
intracellular payload delivery. Such systems have been subsequently validated by a number of subsequent reports – a lower coverage of
utilised for the intracellular delivery of QDs165 and more recently peptides bearing multiple CPP arms often results in a higher cell
for the selective in vivo delivery of PEG–polycaprolactone (PCL) uptake than that observed for particles coated with a high density
NPs to tumours166 (Fig. 9). of mono-valent peptides, though the effect is not universal.176,177
A related approach is to screen the activity of the CPP using Indeed, in a recent report Breger et al. demonstrated that QDs
steric bulk. Harris et al. reported the coating of TAT-functionalised decorated with a single ligand could be efficiently uptaken if a
iron oxide NPs with a shield of PEG, via a MMP-2 cleavable multivalent dendrimeric CPP was presented.177 A number of
peptide linker.167 Upon action of the protease within the tumour hypotheses have been made to explain the origins of these effects,
Fig. 9 (a) Schematic demonstrating the electrostatic blocking of polyarginine cell-penetrating ability. An R8 peptide is blocked by electrostatic binding
of an octa-glutamic acid peptide, connected via an MMP cleavable linker. Upon protease activity in the tumour environment, cleavage of the linker will
lead to release of the glutamic acid blocking group, and activation of R8-mediated penetration. (b) NIR imaging of mice injected with NPs coated with R8
or E8–R8 peptides, in the presence or absence of an angiopep-2 glioma targeting sequence (see Section 5.4). Targeted R8–E8 functionalised NPs display
the greatest enhancement at the tumour site. Adapted with permission from Gao et al.166 Copyright 2014 American Chemical Society.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3585
View Article Online
and, as is often the case, the exact answer is probably depen- and the presence of highly active efflux pumps makes even such
dent on the precise cell type, NP cargo, and construct design. transcellular movement difficult.188
While receptor clustering and crosslinking is known to play a Four main strategies have been exploited in order to allow
major role in many cell–surface recognition events and may BBB penetration, each of which can be exploited by peptide or
also be important in mediating cell uptake, an increase in protein decorated NPs: (i) receptor-mediated transport, whereby
membrane curvature and pore formation as a result of greater NP interactions with over-expressed receptors at the BBB trigger
localised electrostatic binding has also been proposed to play a internalisation; (ii) transporter-mediated movement, hijacking
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
role.177,178 Intriguingly, the Jana group has recently demon- the natural uptake of nutrients such as glutathione by the brain;
strated that the valency itself is a key determinant in not only (iii) adsorptive transport, as a result of the strong binding of
uptake mechanism, but also subsequent sub-cellular location, positively charged particles to the negatively charged BBB; and
and indeed the rate at which particles are subsequently ejected (iv) the exploitation of CPPs, which have also been demon-
by exocytosis.179 A high valency of TAT peptides on the surface of strated to mediate transport across the BBB (Fig. 10). For an in
QDs was found to lead to an increased rate of initial cell uptake. depth discussion on these processes and the mechanisms by
However, the same particles were then rapidly processed and which NPs can be transported across the BBB the reader is
exocytosed, leading to an overall drop in penetration efficiency.180 referred to excellent recent overviews by the groups of Teixidó,
Gao, and Tosi.187,188,190
5.2 Endosomal escape Importantly, the ‘leakiness’ of the BBB is often increased during
As described above, the endocytosis of NPs (both with and pathogenesis. This is particularly true for tumours originating in
without peptide functionalisation) leads to internalisation within the brain, more commonly referred to as gliomas.191 Gliomas are
endosomes. Unless for tissue imaging purposes, this location is therefore commonly used as a model system on which to
typically not the end target, with delivery to the cytosol or nucleus demonstrate the ability of a particular NP construct to penetrate
usually necessary for activity. As such, the endosomal escape of the BBB. While this is not in itself problematic, it is important to
the NP cargo is an essential yet often overlooked factor in have in mind the possible effects of impaired cellular junctions
enabling function. While a number of small molecule additives on NP transport in such systems.
such as chloroquine may act as transduction enhancers, their use Key peptide sequences. The key CPP sequences utilised for the
is not a plausible solution for cell penetration in vivo. As an in vivo delivery of NPs, as outlined in Section 5.1, have all also been
alternative, a number of peptide sequences have been identified utilised to deliver NP cargoes across the BBB. Their ability to
that are able to promote escape, typically through the formation penetrate the cells of the brain endothelium enables transcellular
of disruptive a-helices, which can be triggered selectively within transport into brain tissue. QDs,192 peptide self-assemblies,193 and
the acidic environment of endosomal compartments.181 The AuNPs186,194 have all been shown to penetrate the brain following
N-terminal domain of influenza virus haemagglutinin HA2 TAT functionalisation. Similarly, the delivery of polyarginine-
was one of the earliest sequences identified, with Plank et al. functionalised liposomes195 and penetratin-labelled PEG–PLA
demonstrating that HA2 functionalisation of DNA-based NPs particles160 has also been reported. However, as described above,
greatly enhanced cell transduction.146,181,182 Other sequences, the promiscuity of CPPs limits any specificity for brain targeting
such as oligomers of the tetrapeptide GALA183 and Penetration and these reports are also associated with systemic delivery
Accelerating Sequence (Pas)184,185 have also been utilised to to other organs and tissues. As a result, strategies improving
promote the endosomal escape of functionalised liposomes and specificity for brain targeting represent a more attractive solution.
QDs respectively. An alternative approach to enhance escape has The most common technique to promote BBB specific trans-
recently been reported by Morshed and co-workers. By attaching port is to target receptors at the blood–brain interface, which are
TAT peptides to AuNPs via an acid labile hydrazone linkage, they able to induce receptor-mediated endocytosis. The transferrin-
were able to promote endosomal escape through cleavage of the receptor (TfR) is overexpressed on brain endothelial cells, playing
CPP. Using a non-cleavable linkage resulted in the strong binding a crucial role in the transport of iron across the BBB by mediating
of the peptide to the negatively charged endosomal membrane the endocytosis and transport of the iron-binding protein trans-
being retained and thus hindered particle escape.186 ferrin (Tf). The attachment of Tf itself to NPs has therefore been
widely used as a means to mediate BBB penetration, transporting
5.3 Crossing the blood–brain barrier a range of structures including drug-loaded serum albumin
The blood–brain barrier (BBB) is a formidable and restrictive NPs,196 liposomes,158,197,198 polymersomes,199 and polymer
obstacle, able to exclude over 98% of small molecule drugs dendrimer particles,200 as well as RNA-based structures.201
and almost all nanoscale objects in order to maintain brain Alternatively, anti-TfR antibodies, such as OX26 can be utilised
homeostasis.187,188 The endothelial cells of the brain capillaries to mediate transport.202–205 However, possible difficulties
form continuous tight junctions that preclude paracellular with the species-specificity of antibodies may partially hinder
transport.188,189 While it is possible to disrupt this barrier such technologies. For example, since OX26 is targeted to the
and allow passage between cells, more commonly strategies rat TfR, NPs coated with this antibody are unable to be readily
which aim to overcome the BBB rely on transport through translated into human systems.206
the cells that make up the barrier (Fig. 10). However, the The use of the TfR as a target for BBB penetration has
downregulation of receptors that mediate vesicular transport, significant limitations in spite of its attractiveness as a target.
3586 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
Fig. 10 Schematic demonstrating the key mechanisms by which peptides and proteins can mediate the transport of cargo across the BBB. NPs are most
commonly transported via transcellular mechanisms, rather than passing through the tight endothelial cell junctions. For a detailed overview, readers are
directed to the excellent recent review by Oller-Salvia et al. from which this graphic is reproduced, published by The Royal Society of Chemistry.188
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3587
View Article Online
3588 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3589
View Article Online
experiments against a target tissue, receptor, or cell type.272–275 multiplexed imaging of both the blood and lymphatic vessels
First reported in 1985 by Smith, phage display relies on the within the same tumour (Fig. 14). The exquisite sensitivity and
presentation of short amino acid sequences on the protein target specificity offered by these sequences is in part due to
coating of filamentous phage.276 Random peptide libraries of a their multivalent presentation on the QD surface. This highlights
defined length can be expressed, and then screened for binding the power of combining peptide display with nanomaterials for
against the target. Following several rounds of increasingly biomedical imaging. In a similar manner, the tumour-homing
stringent screening, coupled with the amplification of binding pentapeptide CREKA was also identified from phage libraries,
phage, peptides with high target affinity can be identified.277 and found to target the clotted plasma proteins that accumulate
Phage display has been most widely used to screen binding to a within the interstitial tissue and vessel walls in cancerous
target protein or cell–surface receptor. However it is also masses (Fig. 15).285,286 This peptide was able to deliver both
possible to systemically deliver phage libraries in vivo. Using iron oxide NPs and liposomes to the desired site, where they
this technique, phage binding to a target tissue or organ have were observed to induce additional clotting and thus amplified
been isolated and peptide sequences mediating cargo transport accumulation.
to the desired location identified.274,278–280 Poor perfusion of the tumour vasculature has been shown
The Ruoslahti group have utilised in vivo bio-panning to to limit the penetration of NPs into the tumour tissue and
identify a number of sequences able to target NP delivery, presents a major obstacle to effective tumour treatment. In
particularly for cancerous tissue.264,281–283 Åkerman et al. demon- Section 5 we introduced a number of sequences which address
strated that 3 of these sequences, GFE (targeting membrane these difficulties, mediating transcellular transport deep into
dipeptidase on the endothelial cells of lung vasculature),281 the tumour mass. The Lyp-1 peptide described above is one
F3 (binding blood vessels and various tumour cell types),282 such sequence, playing a dual-functional role in mediating
and Lyp-1 (targeting tumour lymphatic vessels, as discussed in both targeting and penetration. As discussed in Section 5.4,
Section 5.4),264 could be used to selectively deliver QDs in vivo.284 Karmali et al. demonstrated that combining the CREKA and
Interestingly, F3 and Lyp-1 labelled particles can be used for the Lyp-1 peptide sequences within a single NP construct enabled
Fig. 14 Fluorescence imaging of breast cancer xenografts in mice, following intravenous injection of peptide labelled QDs. (a) F3 labelled QDs (red)
colocalise with tumour vasculature (green); (b) Lyp-1 (red) labelled particles have a distinct, lymphatic distribution from the vasculature (green); (c) F3 (red)
and Lyp-1 (green) labelled QDs label different portions of the tumour. Original magnifications are 400 (a) and 600 (b and c). Adapted from Åkerman
et al., copyright 2002 National Academy of Sciences.284
3590 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3591
View Article Online
reported that the overexpression of MMPs within the heart As described in Section 5.3, the iron-binding proteins Tf and
following infarction could be used to direct the aggregation of Lf mediate the transport of this key nutrient around the body.
drug-loaded NPs, following proteolytic degradation of hydro- NPs coated with these structures can therefore be targeted to
philic surface peptides and thus accumulation at the infarct sites where their relevant receptors are expressed, as epitomised
site.306 In order to mediate NP delivery to the brain, Mann et al. by their ability to deliver particles to, and indeed induce trans-
recently developed the short tetrapeptide CAQK, which despite port across, the BBB.187 The TfR has a particularly widespread
its relatively short length, is remarkably able to target the distribution throughout the body – almost all cells are thought to
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
delivery of both small molecule and NP cargoes.307 Importantly, express cell surface TfR, albeit at significantly varying levels.312
this peptide displayed affinity for the extravascular tissue, However, the over-expression of TfR in certain cancers as a
rather than the blood vessels themselves. As a result, delivery result of rapid cell growth kinetics, and thus a demanding need
was enabled throughout the site of acute brain injury for the for iron, enables the use of Tf as a targeting agent when
distribution of therapeutic payloads. combined with potential particle accumulation within the
In addition to peptides which can direct the delivery of particles tumour vasculature via the EPR effect.313 Interestingly, a number
to certain parts of the body, several sequences able to drive NP of papers have demonstrated that TfR targeting does not occur
accumulation within specific subcellular locations following cell as a result of altered biodistribution, with similar accumulation
uptake have also been identified. Distinct from peptides that kinetics being observed for both functionalised and non-
induce cell penetration or endosomal escape, these sequences functionalised NPs. Instead, it is the ability of the targeting
enable intracellular trafficking to the site at which their action is group to induce penetration and uptake within the target tissue
required.271 Although such sequences have been most commonly that leads to an enhancement of the therapeutic efficiency of NP
utilised for the in vitro study or targeting of intracellular processes, drug-delivery vehicles.314,315
more recent studies have also demonstrated their application in a The degree of Tf modification plays an important role in
biomedical setting.271 Agemy et al. combined the mitochondrial determining the level of NP uptake, whether through multi-
targeting peptide (KLAKLAK)2 with tumour-targeting CGKRK valent interactions or increased chance of recognition.315,316
and penetrating iRGD sequences, to form a tri-functional iron Salvati et al. have also highlighted the importance of carefully
oxide NP coating, able to efficiently deliver particles within designing NP coatings in order to maintain maximal activity
mouse glioblastoma models. In addition to its targeting role, in vivo. Following the incubation of Tf-functionalised silica NPs
(KLAKLAK)2 also promoted apoptosis through disruption of the in serum, they showed that rapid loss of TfR targeting occurred
mitochondrial membrane, increasing the therapeutic effect.308 due to the accumulation of a blocking protein corona.317
In another example, the endoplasmic reticulum (ER) retention Despite this sensitivity, Tf functionalisation has been the most
signal ‘Eriss’ (derived from the adenovirus E13-19K protein) has widely utilised strategy for directing tumour delivery. Such
been used to direct NPs to the ER of lymphocytes, improving the systems have been shown to mediate the transport of a wide
processing and presentation of NP-displayed antigens. By doing range of NP cargoes, as well as being at the core of several
so, the efficiency of synthetic vaccines can be greatly enhanced liposomal based therapeutic technologies currently in clinical
in vivo as discussed in Section 7, leading to an improvement in trials (Fig. 17).313,314,316,318–321
the generation of immunity.309,310 Many other protein ligands have also found widespread use
for the selective targeting of NPs to cell surface receptors within
6.2 Protein targeting motifs a specific tissue or organ. An important consideration is the
The recognition of protein binding partners by cell–surface ability of many such ligands to trigger cellular responses and
receptors is one of the most important interactions in biology. downstream effects. While this may be favourable in certain
The vast number of hormones, growth factors, cytokines, cell applications, in such cases the protein substrate is not merely
adhesion proteins, and cell-signalling structures are involved in acting as a targeting motif but is also controlling or inducing
virtually all intracellular interactions that enable the formation a biological effect.322 For example, growth factors such as
and function of complex tissues.311 It is therefore not surpris- epidermal growth factor (EGF) and VEGF have been used to
ing that protein–NP coatings have become an important means target NPs to their relevant receptors, when overexpressed on
to drive targeted delivery to a certain area of the body where a the surface of tumour cells and tissues.270,322–327 However, the
particular receptor is likely to be overexpressed (Table 7).270,275 potency of growth factors as cell signalling moieties and their
3592 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
Fig. 18 AuNP templated HDLP particles were injected into xenograft tumour bearing mice: (a) particles selectively target and are uptaken by B-cell
lymphoma cells. Disruption of cholesterol flux as a result limits tumour growth; (b) in contrast, particles are not targeted to T-cell lymphoma cells and
have no effect on tumour growth. Reproduced from Yang et al., copyright 2013 National Academy of Sciences.329
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3593
View Article Online
3594 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
the Ab on the NP surface is an important consideration – non- NP vaccines can offer the benefits of increasing antigen
specific protein modification techniques can lead to Ab con- stability over soluble delivery. Furthermore, their nanoscale
jugates with many different NP binding points, some of which size may promote scavenging by dendritic cells and therefore
may result in an antigen-binding domain that is positioned in a improve T-cell presentation, minimising the activation of alter-
hindered or blocked orientation. Indeed, a recent study by native immune response pathways.382,385 Reddy et al. demon-
Herda et al. demonstrated that as little as 5% grafted protein strated that when NPs were of a sufficiently small size (o100 nm)
possessed an accessible epitope following non-specific conju- preferential drainage and accumulation in the lymph nodes was
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
gation to silica NPs.374 The use of site-specific modification enabled.386 However, other reports have suggested that particles
strategies able to selectively form Ab–NP conjugates away from up to 1 mm in diameter may also preferentially accumulate under
the active recognition site are likely to improve the efficiency of certain delivery conditions, highlighting the need for further
NP targeting in vivo.342,375 To this extent, Greene et al. recently investigation into this phenomena.387 Preferential exploitation
reported a strategy to functionally re-bridge distal disulphide of the lymphatic system brings particles into closer contact with
bonds within a trastuzumab fragment–Ab, enabling orientated the residing dendritic cells and leads to an enhanced activation
Ab display on the surface of PLGA–PEG NPs and a corresponding of the immune system maximising vaccine efficiency. However,
increase in binding efficiency when compared to non-specific Ab the major advantage of NP-based vaccines lies in their ability to
conjugation.376 enable the multivalent display of antigens, promoting the
In a related manner, the Ab loading density on the NP interaction of multiple ligand–receptor pairs. Such processes
surface can play an important role in targeting efficiency. At often play an essential role during T-cell activation, rather than
high levels, hindered antigen-binding may result leading to an relying on individual recognition events.388 As such, the pre-
initially counter-intuitive drop in targeting efficiency. Indeed, sentation of multiple antigens on the NP surface can play a far
Jiang et al. demonstrated that the size of the NP template plays more effective role in modulating the immune response than
an important role in determining hindrance and thus binding presentation of free peptide or protein motifs. Interestingly,
efficiency, with increasing curvature leading to an increase in it has been shown that the antigen patterning plays a crucial
separation and thus decrease in steric inhibition of antigen role during activation. As a result, antigen grafting is a key
recognition.377 Although multi-valent Ab display may be desirable design feature during the production of NP vaccines. The Yu
in some cases in order to promote receptor recognition, recent group demonstrated that enhanced antigen clustering elicits a
papers by the groups of Davis and Prosperi have highlighted stronger immune response than uniformly distributed ligands
that this may not always be the case.378,379 In both cases, a at both the micron and nano scale.388,389 Similarly, particle
singly Ab labelled NP was found to be sufficient, and in the case surfaces that closely mimic the natural context of the antigen,
of Colombo et al. superior to multivalent Ab display for effective such as liposomes able to mimic cell membranes, have been
tumour targeting. found to improve activation efficiency.390,391
Amongst the NPs used for synthetic vaccines, ‘hard’ NPs have
generally been more widely applied for the surface presentation of
7. Mimicking biological species biomimetic peptide/protein antigens. In contrast, ‘soft’ particles
able to encapsulate a payload have commonly been utilised for
Many biological entities fall within the nanometre size regime, the delivery of antigen following particle recognition and sub-
displaying multi-valent peptide or protein motifs on their surface. sequent release.384 AuNPs in particular have found widespread
NP–polypeptide conjugates are able to effectively mimic the use as NP antigen carriers due to their ease of surface function-
behaviour of these structures, stimulating signalling pathways and alisation, high surface-area, biocompatibility, and tunable size.392
eliciting cellular responses. The interactions induced by these parti- Importantly, recent research suggests that AuNPs may also act as
cles are responsible for many of the applications discussed in this size- and shape-dependent adjuvants, stimulating the immune
review. The promotion of receptor clustering is often implicated in system and enhancing antigen recognition.393,394 As a result, the
receptor-mediated endocytosis and cell or tissue penetration,380,381 attachment of peptide and protein ligands to AuNPs has emerged
and NP targeting may be enhanced by multi-valent interactions at as an effective means to stimulate the production of Abs against a
the nanoscale. As these topics have already been covered in detail wide range of pathologies and pathogens, including malaria,395
above, in this section we will focus on the ability of NP–polypeptide foot and mouth disease,396,397 the Yersinia pestis bacteria
constructs to mimic the multi-valent display of antigens present on responsible for plague,398 cancers via the mucin-1 (MUC-1) glyco-
the surfaces of pathogens and tumour cells. By doing so these protein,399,400 influenza A virus,401 Streptococcus pneumoniae,402
conjugates are able to induce a controlled immune response and respiratory syncytial virus,403 encephalitis causing viruses,404
thus generate immunity in the recipient. Technologies which rely on and HIV via gp120 derived peptides.405
the encapsulation, rather than surface presentation of antigens, are Both single- and multi-walled carbon nanotubes (CNTs) have
outside the scope of this review and the reader is instead also been widely explored as antigen carriers due to their largely
directed to a number of comprehensive reviews on this biologically inert nature, facile surface modification, and ability
topic.382–384 Furthermore, the use of self-assembling peptides/ to induce cell penetration. However, recent reports on their
protein NPs as vaccine candidates will be detailed in Section 8 toxicity may limit further development towards applications in
below and so will also not be discussed in detail here. patients.406 Since being first introduced by Pantarotto et al.,407
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3595
View Article Online
peptide- and protein-modified CNTs have been utilised known to be targeted by auto-immune responses in patients
to generate immune responses against a range of antigens suffering from multiple sclerosis, with tolerogenic small mole-
including tumour lysate,408 Plasmodium vivax apical membrane cules that stimulate Treg proliferation on the surface of AuNPs,
antigen-1 derived peptide,409 Mycobacterium tuberculosis protein has subsequently been shown to suppress symptoms within
derivatives,410 Wilm’s tumour antigen,411 and foot and mouth mouse models of the disease.425 More recently, Hess et al.
virus derived antigens.412 ‘Hard’ NPs such as polystyrene nano- utilised QDs as a scaffold for the display of myelin peptides
beads,413,414 polyacrylate dendrimers,415 and calcium phosphate allowing the authors to monitor the distribution and activated
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
particles416,417 have all also been shown to enable Ab generation pathways of the NP–peptide complexes in vivo.426
in vivo. The Baneyx group have recently reported that a calcium
phosphate binding peptide epitope can be attached to the
termini of the desired antigen, inducing biomineralization and 8. Playing a structural role
particle assembly at a late stage of the formulation process.416,417
By doing so, the low stability of calcium phosphate particles can In much of this review, we have focussed on systems in which
be mitigated, allowing ‘on-demand’ production and application the peptide or protein defines or modulates NP function or
to take place. performance. In addition to these roles, peptide/proteins have
Although ‘soft’ organic NPs have been more rarely used for emerged as key structural motifs, which are integral to the
the formulation of vaccines, a number of examples exist and in formation as well as eventual end application of the NP. Indeed,
many cases they reveal interesting facets of vaccine design that in many of the scenarios to be discussed in this section the
should be carefully considered, regardless of the particle type. particle is composed entirely of peptide or protein components.
For example, liposome formulations have been commonly Peptide self-assembly into complex nano-architectures can
utilised for the encapsulation of peptide or protein antigens, be instigated by a combination of intra- and inter-molecular
enabling delivery to immune cells typically following targeted non-covalent interactions, including hydrogen bonding, electro-
delivery. Virosomes, liposomes functionalised with virus static or hydrophobic interactions, and p–p stacking.427 Although
components (often influenza virus derived) able to mimic viral individually these interactions may be weak, cumulatively they are
envelopes, have been particularly widely used as safe, self- able to define the secondary and tertiary structures of complex
adjuvanted, and stable antigen delivery vehicles.418,419 However, native protein architectures, and can be exploited to produce a vast
Guan et al. have demonstrated that the surface-presentation of array of self-assembled structures on both the nano- and micro-
ligands within liposome formulations is able to mediate activa- scale. The formation of architectures, ranging from fibres and
tion of alternative branches of the immune system in vivo, when tubes, through to vesicles and micelles, and on to more elaborate
compared to encapsulated antigen.420 Careful NP design is structures such as crystals and donuts can all be instigated.427–431
therefore key in ensuring that the desired response is triggered. While the fundamental driving forces behind the formation of a
Similarly, peptide–lipid amphiphiles have also been studied, particular architecture are becoming increasingly well understood,
as discussed in Section 8, for their ability to self-assemble in many cases the route of assembly remains a dynamic process
into peptide-decorated micelles. The use of peptides as a key which can be affected by seemingly minor modifications of
structural component results in an extremely high density of peptide primary structure or growth conditions.432–436
surface antigen coverage, enhancing recognition and activation The formation of self-assembled peptide architectures has
efficiency.421 Importantly, the crowded environment provided by found widespread application across the biomedical field, and
such a set-up has been found to induce peptides to adopt a further afield in the wider materials research community. Here, we
secondary structure more akin to their natural presentation will focus on the formation of 3D spherical NPs, particularly
within the parent protein.415,422 As a result, the Abs generated peptide/protein micelles and vesicles. The conditions for the
downstream are better able to produce an effective response formation of these structures are often strict, requiring precise
when subsequently challenged. control over composition, assembly conditions, and handling. For
While applications using peptide– and protein–NP conju- an in depth overview of the wider field, and in particular the use of
gates as vaccines to stimulate a protective immune response self-assembling peptides in the formation of 2D nano-fibres,
to previously unencountered antigens have been most widely hydrogels, and nanotubes, the reader is referred to excellent recent
studied, technologies which modulate the immune system, and reviews on the subject.427,429,430,437–440 It should be noted that we
in particular mitigate auto-immunity, have begun to emerge. In will not cover nano-sized aggregates of globular proteins in this
a healthy individual, tolerance of self-antigens is maintained review, such as those formed by serum albumins or gelatin, which
by the activity of regulatory T cells (Tregs). Modulation of have found increasing clinical use in recent years. Such systems do
Treg activity and addressing deficiencies is thus an attractive not rely on the specific self-assembly of polypeptide components,
target for the treatment of autoimmune diseases.423 Tsai et al. and thus fall outside the scope of this review. A number of reviews
demonstrated that the presentation of recombinant major focussed on these topics have recently been published.441–444
histocompatibility complexes bearing type 1-diabetes associated
peptides on the surface of iron oxide NPs resulted in the in vivo 8.1 Dipeptides
expansion of Tregs, and ultimately the restoration of normoglycemia Dipeptides represent the simplest self-assembling peptide
in diabetic mice.424 Furthermore, the co-delivery of myelin peptides, motif. Since the breakthrough discovery by Reches and Gazit
3596 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
that di-phenylalanine assembled into nanotubes at high was first reported by Vauthey et al.452 1 or 2 C-terminal aspartic
concentrations,445 a wide-range of nano-architectures have acid residues, bearing 2 or 3 negative charges respectively, were
been reported, making use of both natural and non-natural found to be sufficient to drive the assembly of an N-capped
amino acids.446 Often, the presence of a di-aromatic motif is hydrophobic peptide chain of 6 alanine, valine, or leucine
vital, providing the driving force for assembly via p–p stacking. residues. Dynamic heterogenous mixtures of nano-tubes,
The ease with which dipeptides can be accessed synthetically vesicles, and micelles were observed, depending on the exact
makes them particularly attractive structures for biomedical peptide sequence. Subsequent reports on the formation of
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
applications, with researchers from diverse backgrounds able vesicles from glycine-tail anionic peptides453 or cationic peptides
to exploit their use. bearing lysine or histidine termini432,433 validated this approach,
The first dipeptide shown to form spherical particles was the with the hydrophobic ‘tail’ and charged ‘head’ group in effect
unnatural structure di-phenylglycine, in stark contrast to the mimicking the structure of lipid surfactants. Interestingly, it has
nanotubes formed by the closely related di-phenylalanine.447 recently been reported that rather than forming tail-to-tail
The significance of seemingly minor differences and subtle arrangements analogous to those observed in lipid membranes,
changes in structure highlight the surprising versatility of such the hydrophobic regions of amphiphilic peptides form inter-
simple structures. The situation is further complicated by the digitated b-strand-like assemblies, leading to greatly reduced
dynamic nature of dipeptide nanostructures, which have been membrane thickness.454
observed to result in reversible transitions between architec- Despite these important fundamental studies on the ability
tures in response to stimuli or incubation conditions.446,448,449 of short amphiphilic native-peptides to form vesicles, bio-
Despite this apparent plasticity, dipeptide NPs exhibit remark- medical applications have to date been limited by the relative
able stability. Indeed, di-phenylglycine particles were shown to instability of self-assembled constructs. Linear peptides typi-
be stable to both acid and base treatment with no observed cally exhibit high critical aggregation concentrations (CACs) in
change in particle number.447 aqueous solution, below which particle formation does not
Despite the simplicity of dipeptide assemblies, their applica- occur, and a dynamic equilibrium with free peptide therefore
tion in biomedicine has been limited to just a few reports. In an often exists. This situation is further complicated in complex
early example, Alam et al. demonstrated that H2N–methionine– biological fluid.433–435 A number of different approaches have
dehydrophenylalanine–CO2H NPs could be loaded with the anti- been taken to address this issue, yet often CACs or dissociation
cancer drug curcumin and used to induce tumour regression in a constants are not reported, obscuring the analysis of particle
mouse melanoma model.450 Importantly the unnatural amino stability. Gudlar et al. showed that branched peptides derived
acid dehydrophenylalanine not only increased packing efficiency, from natural transmembrane helices can enhance vesicle stability
and thus enhanced physical stability through increased p–p as a result of enhanced hydrophobic interactions.455 These
stacking, but also promoted biological stability by providing branched structures were found to be a key driving force for the
protease resistance. More recently, Fan et al. reported the assembly preferential formation of vesicles over fibres, closely mimicking
of H2N–tryptophan–phenylalanine–CO2H, to produce fluorescent the di-hydrophobic tail of native lipids and enabling the delivery
NPs.451 By combining p–p stacking with peptide–zinc interactions of cargoes to cells in vitro. In order to stabilise vesicles post-
they were able to produce particles with visible light emission, formation, van Hell et al. incorporated cysteine residues into the
mimicking the red-shifted emission exhibited by fluorescent pro- primary structure of the amphiphilic peptide SA2.456,457 SA2 is a
teins upon metal-binding. Subsequent modification of the particle rationally-designed amphiphilic sequence containing sequential
surface with a MUC-1 binding aptamer enabled biocomptability to hydrophobic residues of decreasing bulk, leading to a cone-
be enhanced when compared to other fluorescent NPs such as shaped monomer that promotes the formation of spherical
quantum dots.451 Although this promising system has only so far architectures, with a CAC of 0.5 mM prior to crosslinking.434 The
been demonstrated in vitro, it offers an attractive means through formation of interchain disulphide linkages between adjacent
which to produce simple, photo-stable, biocompatible NPs for cysteines led to the production of stable vesicles, and enabled
in vivo imaging in the future. cellular delivery of encapsulated photosensitizers.457
An alternative approach to stabilise self-assembled peptide
8.2 Peptide amphiphiles architectures is to incorporate non-native functionalities. In its
Amphiphilic peptides, containing both a hydrophobic and simplest form, this can involve the incorporation of unnatural
hydrophilic domain, can be broadly split into three categories: amino acids, as reported by Tanisaka et al., providing increased
(i) those composed only of native amino acids (with or without hydrophobic interactions as well as resistance to proteases
minor modifications at the termini); (ii) those containing in vivo.458 Stable vesicles composed of hydrophilic sarcosine
unnatural amino acids; and (iii) lipid- or polymer-conjugated and hydrophobic methyl glutamate residues were shown to
peptide hybrids. In all three groups, self-assembly into spherical accumulate in cancer tissue as a result of the EPR effect in
particles is typically driven by a mixture of hydrophobic and animal xenografts. Taking this concept further, the formation
electrostatic interactions, with the formation of secondary struc- of lipid- or polymer-hybrid peptides has become an attractive
tures possible in some cases.427,431 means by which to drive peptide self-assembly.459,460 Liu et al.
The formation of nano-vesicles composed entirely of native demonstrated that addition of cholesterol to the end of a hydro-
amino acids, referred to in some instances as ‘peptosomes’, philic hexaarginine-TAT peptide block drove micelle formation
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3597
View Article Online
3598 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3599
View Article Online
Fig. 23 NIR fluorescence imaging of tumour xenograft bearing mice, injected with cisplatin loaded casein NPs. Gradual accumulation within the tumour
is enabled by the EPR effect and long circulation time of casein particles. Reproduced from Zhen et al. with permission from Elsevier.520
3600 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
AuNPs in commercially available lateral flow pregnancy tests.523 nanoworms.532 Due to their size, these particles were blocked
The development of NP-based biosensors has become a vast from renal clearance and displayed accumulation in the liver.
field of research, and a detailed overview of ex vivo technologies However, following protease activity, the peptide cleavage
is outside the scope of this review. The reader is instead referred products were excreted in the urine, and could subsequently
to a number of excellent comprehensive reviews on the topic for be detected by mass spectrometry, giving a panel-readout of
further details.33,524–527 Instead, we will here focus on the far in vivo protease levels. Subsequent iterations of this technology
smaller body of literature that focusses on the application of have demonstrated the detection of thrombosis and colorectal
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
NP-biosensors in vivo, and the challenges that have so far cancer using peptides terminated with recognition elements
limited their widespread implementation. Such systems allow that can subsequently be detected in urine by enzyme-linked
the true complexity of tissues to be captured in a way that immunosorbent assays, or point-of-care lateral flow detection
in vitro testing of biofluids cannot provide.528 systems.533,534 In order to overcome the challenges presented
Sensing complexes provide a means to interrogate biological by non-specific activation by circulating proteases, Dudani et al.
systems and probe differences in activity and temporal distribu- reported the use of photo-protected peptide–NP coatings which
tions of a desired analyte, as opposed to the spatial localisation could be activated towards protease sensitivity with spatial
provided by imaging modalities. Rather than being detected and temporal control, following the application of UV or two-
within a specific area, in vivo sensors offer a responsive platform photon light.535 Furthermore, the sub-cutaneous implantation
to monitor small molecules, biomacromolecules, and diseased of 8 nm peptide-functionalised PEG–NPs, which enable gradual
states in a continuous manner.528,529 Although implanted electro- leaching of a particle reservoir into the blood stream, has
chemical sensors have been widely described, in particular for the recently been shown to allow the continuous monitoring of
monitoring of blood glucose levels, problems with induced protease activity in vivo, although detection was still limited to a
fibrosis and foreign body responses can limit sensitivity and 24 h time period.535
accuracy.528 NP based systems offer a viable alternative, but also
have their own complications – in addition to the challenges of
generating a signal that can be actively transduced for detection, 10. Outlook and conclusions
NPs must overcome background signal generation within the
complex environment of the body, be retained at the desired site The past 20 years have seen a rapid increase in the development
of detection, and ideally provide a reversible and dynamic of nanotechnologies, and the exploitation of polypeptides in
response. The use of peptide/protein–NP conjugates in particular this context has also found increasing favour. The structural
must address the sensitivity of biological components to degra- and functional versatility of peptides and proteins allows
dation, clearance, and unfavourable interactions.529 As a result, them to play an important role in modulating, instigating,
systems in which peptides and proteins play an active sensing and defining the activity of NP constructs. In this review we
role, rather than mediating imaging or targeting, have only have outlined the key roles played by polypeptide coatings and
recently begun to emerge. structural components, and demonstrated how they can be
In one realisation of such technologies, environmentally utilised to improve the efficacy of NP tools in biomedical
sensitive polymer NPs have been exploited to detect differences applications. In many cases, both peptides and proteins can
in analyte concentration as a result of conjugated enzyme activity. fulfil the desired function. Each offers important advantages
More specifically, polymer NPs incorporating platinum based over the other (though exceptions to these generalisations exist)
fluorescent sensors have been shown to display phosphorescence that should be carefully considered during NP-conjugate design
dependent on environmental oxygen concentrations. Cash and (Fig. 24). For example, the ease with which peptides can be
Clark demonstrated that by conjugation of the histamine- produced via solid-phase peptide synthesis (SPPS) is highly
metabolising, oxygen-consuming enzyme diamine oxidase to the beneficial. As well as offering cost-effective, quick, and scalable
NP surface, biologically relevant levels of this key inflammatory- production, the versatility of SPPS enables unnatural chemical
and neuro-modulator could be detected by a change in space to be explored. As such, the facile introduction of reactive
phosphorescence.530 This response was reversible upon diffusion handles and protease resistant residues, or structures able to
of oxygen back into the biological milieu, with only a limited promote biological interactions, is enabled (Fig. 24b and c).
dropoff in polymer dynamic range observed upon repeated Furthermore, the small size of peptides enables the disruption of
applications. Similarly, Sun et al. monitored glucose levels NP hydrodynamic diameter and detrimental effects on activity to
in vivo using a glucose oxidase-functionalised oxygen–polymer be minimised, while creating a dense, accessible, and flexible
NP transducer.531 Subcutaneous implantation enabled reten- coverage of active sequences (Fig. 24a and d). In contrast, the
tion of the particles for up to a month, enabling reproducible increased structural complexity offered by proteins enables
signal generation over this extended period. NP-conjugates to typically attain increased activity and recogni-
As an alternative, the Bhatia group has pioneered the use of tion, albeit potentially at the cost of decreased biological
protease-sensitive NP coatings, which release peptide cleavage stability and increased recognition by the reticuloendothelial
products into the urine upon enzymatic activity in vivo. In their system (Fig. 24e). In addition, small structural modifications
original report, Kwong et al. conjugated peptides sensitive to a to proteins are usually well tolerated, without leading to major
range of common proteases onto the surface of iron oxide disruptions in activity. Corresponding changes in peptide
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3601
View Article Online
Fig. 24 Making the choice between a peptide– or protein–NP conjugate is an important decision during the design of biomedical nanotechnologies.
Both have significant advantages over the other, as well as drawbacks which may limit their applicability in certain scenarios. It is therefore imperative to
consider the interaction of the polypeptide component with the NP surface, core, and cargo, as well as the end application, interplay with natural
systems, and route of administration: (a) the small size of peptides allows a high surface density of coating to be achieved; (b) solid-phase peptide
synthesis enables the straightforward production of large volumes of short peptides, as well as (c) enabling the facile introduction of unnatural amino
acids, backbones, and architectures; (d) multiple peptides can be introduced within a single NP construct, enabling multi-functional coatings to be
accessed; (e) proteins typically display enhanced bioactivity and binding affinity when compared to peptide substrates, though many examples of potent
peptide substrates also exist; (f) the high molecular weight of proteins leads to strong non-covalent interactions which can drive the formation of stable
self-assemblies; (g) single amino acid alterations to protein sequences lead to significantly less structural disruption when compared to the analogous
peptide substrate.
sequence have a stronger influence on conformation and by the nanotechnology field.536 Within the realm of peptide/
activity, potentially leading to a significant drop in the efficacy protein–NP conjugates, we believe that addressing the follow-
of the nanotechnology (Fig. 24g). The situation is further ing issues is vital to enable translation of these naı̈ve systems to
complicated by the complex interplay between peptide/protein a biomedical setting:
components and the particular NP core, cargo, application, and (i) The use of native amino acids brings with it the threat of
delivery route. As such, consideration of the precise polypeptide protease sensitivity, and activity can often be rapidly lost in the
component is a vital step during NP-conjugate design. complex environments faced in vivo. In this review, a number of
Despite the advances that have been made in enabling approaches have been introduced towards addressing these
effective and targeted delivery to the desired site of action, difficulties. Unnatural amino acids are particularly effective,
providing an integral means to maintain activity, and acting to providing peptide bonds which are not recognised and processed
hijack and exploit native biological pathways, there remains by native enzymes. This is most easily achieved with synthetic
significant scope for the improvement of polypeptide–NP tech- peptides, as discussed above – relatively simple modifications
nologies. In a recent editorial, Leroux highlighted that while the such as N-methylation, enantio- or retro-enantio amino acids,
number of papers reporting increasingly creative and complex or extended b- or g-linkages can be introduced.217,537,538 While
NP systems increases exponentially, their therapeutic transla- increased stability is harder to achieve with protein substrates,
tion remains disappointing, highlighting the challenges faced the advent of technologies which allow the introduction of
3602 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
non-natural stabilising residues,539,540 the addition of protec- (v) Proper and standardised characterisation of NP–peptide/
tive polymer coatings,541,542 or the formation of additional protein conjugates is essential to not only allow progression in the
cross-links543,544 are now allowing such issues to be addressed. field, but to enable regulatory and clinical standards to be met.172
In order to fully facilitate the translation of NP-based systems, it This challenge must be addressed by both the development of new
is important that the research community begins to actively tools which are able to reflect the true nature of NP complexes, as
embrace such technologies. well as increased uptake by researchers of existing technologies, and
(ii) The small size of peptides allows them to form compact NP a more thorough approach to NP characterisation being taken.552,553
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
coatings, which lead to minimal disruption of desired function. At present, many systems reported in the literature are inadequately
They are therefore often the preferred choice when designing a NP– characterised, leading to large discrepancies in results, conflicting
polypeptide conjugate, with the major drawback being reduced reports, and incorrect interpretations that subsequently propagate
bioactivity (Fig. 24a and e). Nature has had millions of years to throughout the field. The inherent complexity of NP-bioconjugates
slowly evolve proteins with complex 3D structures and potent makes them undoubtedly challenging to study – even what appears
activity, and it remains challenging for designed, screened, or a simple measurement such as the concentration of particles in
derived peptides to fully recapitulate this. Nowhere is this more solution is often difficult.172 This becomes even more significant for
strongly felt than in the reduced ability of short synthetic peptides more complex calculations such as the density or orientation of a
(o30 amino acids) to target and bind a particular protein or cell type protein on a NP surface, or the origins of biological effects arising
when compared to Abs or other binding proteins. Although strongly from heterogeneous samples. However, novel tools that can answer
binding long peptides such as affibodies (450 amino acids) can be these questions are beginning to emerge, and although often highly
accessed synthetically, more typically they are produced recombi- specialised, are beginning to be readily accessible to researchers,
nantly and still result in a bulky coating on the NP surface.545 There through national and international facilities for NP research and
is a therefore a pressing need for technologies which can bring short characterisation.553–556
peptide binding affinities down from the mM to the nM range.546 A big leap towards enabling the clinical translation of many
Recent advances in the screening, design, and exploitation of peptide/protein–NP conjugate technologies will be taken as the
bicyclic peptides are enabling such levels to be reached, and will biomedical community addresses these issues. As a result, our
surely be exploited in the near future to enhance the ability of ability to treat, diagnose, and understand disease will be greatly
NP-targeting strategies to achieve their goals.547–549 enhanced, allowing nano-technology to fulfil its undoubted
(iii) The bioactivity of a peptide or protein–NP conjugate is promise in the biomedical sciences.
highly dependent on achieving an appropriate and accessible
orientation of the bioactive domain. Again, this can be easier to
achieve using peptides rather than protein substrates – tethering Conflicts of interest
residues can be easily introduced at the termini during solid There are no conflicts to declare.
phase peptide synthesis. Often protein substrates are attached
either by non-specific conjugation techniques or simple adsorp-
tion, leading to a detrimental loss of structure, heterogenous Acknowledgements
coatings, and steric hindrance of the active site.375 It is essential
that recent developments in the field of site-specific or selective Drs Mattias Björnmalm and Michael Thomas are thanked for
protein modification are exploited to overcome these problems, to critical evaluation of the manuscript. C. D. S. and M. M. S.
enable the controlled orientation of proteins on the NP surface. acknowledge support from the Swedish Research Council (VR
Strategies for genetically or chemically manipulating proteins to 4-478/2016) and the Swedish Foundation for Strategic Research
introduce and exploit uniquely reactive natural and non-natural (SSF 4-3713/2016). C. J. and M. M. S. acknowledge financial
amino acids, or to incorporate hexahistidine and other affinity support from the Rosetrees Trust. B. G. and M. M. S. acknowledge
tags, are becoming increasingly widespread and offer powerful support from GlaxoSmithKline through the Imperial College
tools to the biomedical research community.342,550,551 London Engineered Medicines Laboratory Project. M. M. S.
(iv) Many of the applications for peptide/protein–NP conjugates acknowledges support from the ERC Seventh Framework
discussed in this review rely on processes that are still poorly Programme Consolidator grant ‘‘Naturale CG’’ (616417), the
understood at a fundamental level. Controversy still exists about Engineering and Physical Science Research Council (EPSRC) grant
the precise mechanisms by which peptides mediate transport across ‘‘Bio-functionalised nanomaterials for ultrasensitive biosensing’’
biological barriers,145 coatings and particle components which were (EP/K020641/1), the i-sense EPSRC IRC in Early Warning Sensing
previously thought to be biologically inert are becoming increasingly Systems for Infectious Diseases (EP/K031953/1), and the Well-
found to induce an immunological response,66 and the design come Trust Senior Investigator Award (098411/Z/12/Z).
criteria required to produce stable self-assembled polypeptide NPs
are still being developed.436 In order to provide NP technologies that References
meet the challenging demands of biomedical applications it is
important that these topics, and many others within the field, 1 E. Blanco, H. Shen and M. Ferrari, Principles of nano-
continue to be investigated and that a deeper understanding particle design for overcoming biological barriers to drug
of the intertwined complexities that determine NP fate is provided. delivery, Nat. Biotechnol., 2015, 33, 941–951.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3603
View Article Online
2 G. Bozzuto and A. Molinari, Liposomes as nanomedical 21 I. Khan, K. Saeed and I. Khan, Nanoparticles: Properties,
devices, Int. J. Nanomed., 2015, 10, 975–999. applications and toxicities, Arabian J. Chem., 2017, DOI:
3 B. L. Banik, P. Fattahi and J. L. Brown, Polymeric nano- 10.1016/j.arabjc.2017.05.011.
particles: The future of nanomedicine, Wiley Interdiscip. 22 A. F. Versiani, et al., Gold nanoparticles and their applica-
Rev.: Nanomed. Nanobiotechnol., 2016, 8, 271–299. tions in biomedicine, Future Virol., 2016, 11, 293–309.
4 S. Bhatia, Nanoparticles Types, Classification, Characteriza- 23 E. C. Dreaden, A. M. Alkilany, X. Huang, C. J. Murphy and
tion, Fabrication Methods and Drug Delivery Applications, M. A. El-Sayed, The golden age: gold nanoparticles for
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
Natural Polymer Drug Delivery Systems, Springer, 2016, biomedicine, Chem. Soc. Rev., 2012, 41, 2740–2779.
pp. 33–93, DOI: 10.1007/978-3-319-41129-3. 24 J. Gao, X. Huang, H. Liu, F. Zan and J. Ren, Colloidal
5 J. J. Giner-Casares, M. Henriksen-Lacey, M. Coronado- stability of gold nanoparticles modified with thiol com-
Puchau and L. M. Liz-Marzán, Inorganic nanoparticles pounds: Bioconjugation and application in cancer cell
for biomedicine: Where materials scientists meet medical imaging, Langmuir, 2012, 28, 4464–4471.
research, Mater. Today, 2016, 19, 19–28. 25 P. D. Howes, S. Rana and M. M. Stevens, Plasmonic nano-
6 K. McNamara and S. A. M. Tofail, Nanoparticles in bio- materials for biodiagnostics, Chem. Soc. Rev., 2014, 43,
medical applications, Adv. Phys.: X, 2017, 2, 54–88. 3835–3853.
7 M. J. Waring, et al., An analysis of the attrition of drug 26 E. Boisselier and D. Astruc, Gold nanoparticles in nano-
candidates from four major pharmaceutical companies, medicine: preparations, imaging, diagnostics, therapies
Nat. Rev. Drug Discovery, 2015, 14, 475–486. and toxicity, Chem. Soc. Rev., 2009, 38, 1759.
8 J. Shi, P. W. Kantoff, R. Wooster and O. C. Farokhzad, 27 R. A. Frimpong and J. Z. Hilt, Magnetic nanoparticles in
Cancer nanomedicine: progress, challenges and opportu- biomedicine: synthesis, functionalization and applications,
nities, Nat. Rev. Cancer, 2017, 17, 20–37. Nanomedicine, 2010, 5, 1401–1414.
9 Y. Nakamura, A. Mochida, P. L. Choyke and H. Kobayashi, 28 A. Akbarzadeh, M. Samiei and S. Davaran, Magnetic nano-
Nanodrug Delivery: Is the Enhanced Permeability and particles: preparation, physical properties, and applica-
Retention Effect Sufficient for Curing Cancer?, Bioconjugate tions in biomedicine, Nanoscale Res. Lett., 2012, 7, 144.
Chem., 2016, 27, 2225–2238. 29 Z. Shen, A. Wu and X. Chen, Iron Oxide Nanoparticle Based
10 A. Beck, L. Goetsch, C. Dumontet and N. Corvaı̈a, Strategies Contrast Agents for Magnetic Resonance Imaging, Mol.
and challenges for the next generation of antibody–drug Pharmaceutics, 2017, 14, 1352–1364.
conjugates, Nat. Rev. Drug Discovery, 2017, 16, 315–337. 30 D.-E. Lee, et al., Multifunctional nanoparticles for multimodal
11 C. Vasalou, G. Helmlinger and B. Gomes, A Mechanistic imaging and theragnosis, Chem. Soc. Rev., 2012, 41, 2656–2672.
Tumor Penetration Model to Guide Antibody Drug Con- 31 K. Ulbrich, et al., Targeted Drug Delivery with Polymers
jugate Design, PLoS One, 2015, 10, e0118977. and Magnetic Nanoparticles: Covalent and Noncovalent
12 F. Fay and C. J. Scott, Antibody-targeted nanoparticles for Approaches, Release Control, and Clinical Studies, Chem.
cancer therapy, Immunotherapy, 2011, 3, 381–394. Rev., 2016, 116, 5338–5431.
13 Nanotechnology for Biomedical Imaging and Diagnostics: 32 R. G. Aswathy, Y. Yoshida, T. Maekawa and D. S. Kumar,
From Nanoparticle Design to Clinical Applications, ed. Near-infrared quantum dots for deep tissue imaging, Anal.
M. Y. Berezin, John Wiley & Sons, Inc., 2015. Bioanal. Chem., 2010, 397, 1417–1435.
14 M. Hembury, et al., Gold–silica quantum rattles for multi- 33 P. D. Howes, R. Chandrawati and M. M. Stevens, Colloidal
modal imaging and therapy, Proc. Natl. Acad. Sci. U. S. A., nanoparticles as advanced biological sensors, Science,
2015, 112, 1959–1964. 2014, 346, 1247390.
15 E. I. Altinoǧlu and J. H. Adair, Near infrared imaging with 34 P. Zhao, et al., Near infrared quantum dots in biomedical
nanoparticles, Wiley Interdiscip. Rev.: Nanomed. Nanobio- applications: current status and future perspective, Wiley
technol., 2010, 2, 461–477. Interdiscip. Rev.: Nanomed. Nanobiotechnol., 2017, e1483,
16 J. Estelrich, M. J. Sánchez-Martı́n and M. A. Busquets, Nano- DOI: 10.1002/wnan.1483.
particles in magnetic resonance imaging: From simple to 35 C. Argyo, V. Weiss, C. Bräuchle and T. Bein, Multifunc-
dual contrast agents, Int. J. Nanomed., 2015, 10, 1727–1741. tional mesoporous silica nanoparticles as a universal plat-
17 K. Stockhofe, J. M. Postema, H. Schieferstein and T. L. Ross, form for drug delivery, Chem. Mater., 2014, 26, 435–451.
Radiolabeling of nanoparticles and polymers for PET 36 C. Bharti, U. Nagaich, A. Pal and N. Gulati, Mesoporous
imaging, Pharmaceuticals, 2014, 7, 392–418. silica nanoparticles in target drug delivery system: A
18 A. S. Thakor, et al., Clinically Approved Nanoparticle review, Int. J. Pharm. Invest., 2015, 5, 124–133.
Imaging Agents, J. Nucl. Med., 2016, 57, 1833–1837. 37 J. G. Croissant, Y. Fatieiev and N. M. Khashab, Degradability
19 S. H. Yun and S. J. J. Kwok, Light in diagnosis, therapy and and Clearance of Silicon, Organosilica, Silsesquioxane,
surgery, Nat. Biomed. Eng., 2017, 1, 8. Silica Mixed Oxide, and Mesoporous Silica Nanoparticles,
20 G. Libralato, et al., Toxicity Effects of Functionalized Adv. Mater., 2017, 29, 1604634.
Quantum Dots, Gold and Polystyrene Nanoparticles on 38 A. Mehmood, H. Ghafar, S. Yaqoob, U. F. Gohar and
Target Aquatic Biological Models: A Review, Molecules, B. Ahmad, Mesoporous Silica Nanoparticles: A Review,
2017, 22, 1439–1454. J. Dev. Drugs, 2017, 6, 1000174.
3604 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
39 Z. Ahmad, A. Shah, M. Siddiq and H.-B. Kraatz, Polymeric 57 H. Gao and Q. He, The interaction of nanoparticles with plasma
micelles as drug delivery vehicles, RSC Adv., 2014, 4, proteins and the consequent influence on nanoparticles
17028–17038. behavior, Expert Opin. Drug Delivery, 2014, 11, 409–420.
40 X. Zhang and P. Zhang, Polymersomes in Nanomedicine - 58 Q. Peng, et al., Preformed albumin corona, a protective
A Review, Curr. Nanosci., 2017, 13, 124–129. coating for nanoparticles based drug delivery system,
41 A.-I. Moreno-Vega, T. Gómez-Quintero, R.-E. Nuñez-Anita, Biomaterials, 2013, 34, 8521–8530.
L.-S. Acosta-Torres and V. Castaño, Polymeric and ceramic 59 S. Ritz, et al., Protein Corona of Nanoparticles: Distinct
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
nanoparticles in biomedical applications, J. Nanotechnol., Proteins Regulate the Cellular Uptake, Biomacromolecules,
2012, 2012, 936041. 2015, 16, 1311–1321.
42 K. Park, Facing the truth about nanotechnology in drug 60 X. Duan and Y. Li, Physicochemical characteristics of
delivery, ACS Nano, 2013, 7, 7442–7447. nanoparticles affect circulation, biodistribution, cellular
43 M. Björnmalm, M. Faria and F. Caruso, Increasing the internalization, and trafficking, Small, 2013, 9, 1521–1532.
Impact of Materials in and beyond Bio-Nano Science, J. Am. 61 T. L. Moore, et al., Nanoparticle colloidal stability in cell
Chem. Soc., 2016, 138, 13449–13456. culture media and impact on cellular interactions, Chem.
44 M. Ferrari, Frontiers in cancer nanomedicine: directing Soc. Rev., 2015, 44, 6287–6305.
mass transport through biological barriers, Trends Biotechnol., 62 J. V. Jokerst, T. Lobovkina, R. N. Zare and S. S. Gambhir,
2010, 28, 181–188. Nanoparticle PEGylation for imaging and therapy,
45 K. Cho, X. Wang, S. Nie, Z. G. Chen and D. M. Shin, Nanomedicine, 2011, 6, 715–728.
Therapeutic nanoparticles for drug delivery in cancer, Clin. 63 M. K. Yu, J. Park and S. Jon, Targeting strategies for multi-
Cancer Res., 2008, 14, 1310–1316. functional nanoparticles in cancer imaging and therapy,
46 H. Soo Choi, et al., Renal clearance of quantum dots, Nat. Theranostics, 2012, 2, 3–44.
Biotechnol., 2007, 25, 1165–1170. 64 J. Conde, et al., Revisiting 30 years of biofunctionalization
47 S. Li and L. Huang, Pharmacokinetics and Biodistribution and surface chemistry of inorganic nanoparticles for nano-
of Nanoparticles, Mol. Pharmaceutics, 2008, 5, 496–504. medicine, Front. Chem., 2014, 2, 1–27.
48 H. H. Gustafson, D. Holt-Casper, D. W. Grainger and 65 F. Tang, L. Li and D. Chen, Mesoporous silica nano-
H. Ghandehari, Nanoparticle Uptake: The Phagocyte Problem, particles: synthesis, biocompatibility and drug delivery,
Nano Today, 2015, 10, 487–510. Adv. Mater., 2012, 24, 1504–1534.
49 M. P. Monopoli, C. Åberg, A. Salvati and K. A. Dawson, 66 Q. Yang and S. K. Lai, Anti-PEG immunity: emergence,
Biomolecular coronas provide the biological identity of characteristics, and unaddressed questions, Wiley Interdiscip.
nanosized materials, Nat. Nanotechnol., 2012, 7, 779–786. Rev.: Nanomed. Nanobiotechnol., 2015, 7, 655–677.
50 F. Chen, et al., Complement proteins bind to nanoparticle 67 W. S. Cho, et al., Size-dependent tissue kinetics of
protein corona and undergo dynamic exchange in vivo, PEG-coated gold nanoparticles, Toxicol. Appl. Pharmacol.,
Nat. Nanotechnol., 2016, 12, 387–393. 2010, 245, 116–123.
51 J. O’Brien and K. J. Shea, Tuning the Protein Corona of 68 S. Guerrero, et al., Improving the brain delivery of gold
Hydrogel Nanoparticles: The Synthesis of Abiotic Protein nanoparticles by conjugation with an amphipathic
and Peptide Affinity Reagents, Acc. Chem. Res., 2016, 49, peptide, Nanomedicine, 2010, 5, 897–913.
1200–1210. 69 P. M. Tiwari, et al., Enhanced intracellular translocation
52 M. S. Grunér, U. Kauscher, M. B. Linder and M. P. Monopoli, and biodistribution of gold nanoparticles functionalized
An environmental route of exposure affects the formation of with a cell-penetrating peptide (VG-21) from vesicular
nanoparticle coronas in blood plasma, J. Proteomics, 2016, stomatitis virus, Biomaterials, 2014, 35, 9484–9494.
137, 52–58. 70 T. Morais, et al., Effect of surface coating on the bio-
53 V. H. Nguyen and B. J. Lee, Protein corona: A new approach distribution profile of gold nanoparticles in the rat, Eur.
for nanomedicine design, Int. J. Nanomed., 2017, 12, J. Pharm. Biopharm., 2012, 80, 185–193.
3137–3151. 71 W. Poon, X. Zhang, D. Bekah, J. G. Teodoro and J. L. Nadeau,
54 P. Aggarwal, J. B. Hall, C. B. McLeland, M. A. Dobrovolskaia Targeting B16 tumors in vivo with peptide-conjugated gold
and S. E. McNeil, Nanoparticle interaction with plasma nanoparticles, Nanotechnology, 2015, 26, 285101.
proteins as it relates to particle biodistribution, biocom- 72 Y. Kojima, et al., CD47-blocking antibodies restore phagocytosis
patibility and therapeutic efficacy, Adv. Drug Delivery Rev., and prevent atherosclerosis, Nature, 2016, 536, 86–90.
2009, 61, 428–437. 73 X. Liu, H. Kwon, Z. Li and Y. Fu, Is CD47 an innate
55 C. Corbo, R. Molinaro, M. Tabatabaei, O. C. Farokhzad and immune checkpoint for tumor evasion?, J. Hematol. Oncol.,
M. Mahmoudi, Personalized protein corona on nano- 2017, 10, 12.
particles and its clinical implications, Biomater. Sci., 2017, 74 N. G. Sosale, K. R. Spinler, C. Alvey and D. E. Discher, Macro-
5, 378–387. phage engulfment of a cell or nanoparticle is regulated by
56 D. E. Owens and N. A. Peppas, Opsonization, biodistribu- unavoidable opsonization, a species-specific ‘Marker of Self’
tion, and pharmacokinetics of polymeric nanoparticles, Int. CD47, and target physical properties, Curr. Opin. Immunol.,
J. Pharm., 2006, 307, 93–102. 2015, 35, 107–112.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3605
View Article Online
75 S. B. Willingham, et al., The CD47-signal regulatory protein 93 E. J. W. Verwey, Theory of the Stability of Lyophobic
alpha (SIRPa) interaction is a therapeutic target for human Colloids, J. Phys. Chem., 1947, 51, 631–636.
solid tumors, Proc. Natl. Acad. Sci. U. S. A., 2012, 109, 94 B. Derjaguin and L. Landau, Theory of the stability of
6662–6667. strongly charged lyophobic sols and of the adhesion of
76 P. L. Rodriguez, et al., Minimal ‘Self’ Peptides That Inhibit strongly charged particles in solutions of electrolytes, Prog.
Phagocytic Clearance and Enhance Delivery of Nano- Surf. Sci., 1993, 43, 30–59.
particles, Science, 2013, 339, 971–975. 95 M. Boström, D. R. M. Williams and B. W. Ninham, Specific
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
77 Y. Qie, et al., Surface modification of nanoparticles enables ion effects: Why DLVO theory fails for biology and colloid
selective evasion of phagocytic clearance by distinct macro- systems, Phys. Rev. Lett., 2001, 87, 168103.
phage phenotypes, Sci. Rep., 2016, 6, 26269. 96 S. Kittler, et al., The influence of proteins on the dispersa-
78 P. L. Rodriguez, et al., Minimal ‘Self’ Peptides That Inhibit bility and cell-biological activity of silver nanoparticles,
Phagocytic Clearance and Enhance Delivery of Nano- J. Mater. Chem., 2010, 20, 512–518.
particles, Science, 2013, 339, 971–975. 97 J. S. Gebauer, et al., Impact of the Nanoparticle Protein
79 Q. Peng, et al., Preformed albumin corona, a protective Corona on Colloidal Stability and Protein Structure, Lang-
coating for nanoparticles based drug delivery system, muir, 2012, 28, 9673–9679.
Biomaterials, 2013, 34, 8521–8530. 98 A. Albanese and W. C. W. Chan, Effect of Gold Nanopar-
80 S. Schöttler, et al., Protein adsorption is required for stealth ticle Aggregation on Cell Uptake and Toxicity, ACS Nano,
effect of poly(ethylene glycol)- and poly(phosphoester)- 2011, 5, 5478–5489.
coated nanocarriers, Nat. Nanotechnol., 2016, 11, 372–377. 99 J. Nam, N. Won, H. Jin, H. Chung and S. Kim, pH-induced
81 J. Mariam, S. Sivakami and P. M. Dongre, Albumin corona aggregation of gold nanoparticles for photothermal cancer
on nanoparticles – a strategic approach in drug delivery, therapy, J. Am. Chem. Soc., 2009, 131, 13639–13645.
Drug Delivery, 2016, 23, 2668–2676. 100 R. Pamies, et al., Aggregation behaviour of gold nanoparticles
82 N. Bertrand, et al., Mechanistic understanding of in vivo in saline aqueous media, J. Nanopart. Res., 2014, 16, 2376.
protein corona formation on polymeric nanoparticles and 101 S. Neupane, et al., Probing the Aggregation Mechanism of
impact on pharmacokinetics, Nat. Commun., 2017, 8, 777. Gold Nanoparticles Triggered by a Globular Protein,
83 I. S. Yermolenko, et al., Origin of the nonadhesive proper- J. Phys. Chem. C, 2017, 121, 1377–1386.
ties of fibrinogen matrices probed by force spectroscopy, 102 C. A. Mirkin, R. L. Letsinger, R. C. Mucic and J. J. Storhoff, A
Langmuir, 2010, 26, 17269–17277. DNA-based method for rationally assembling nanoparticles
84 R. Safiullin, et al., Fibrinogen matrix deposited on the into macroscopic materials, Nature, 1996, 382, 607–609.
surface of biomaterials acts as a natural anti-adhesive 103 A. Gupta, et al., Ultrastable and Biofunctionalizable Gold
coating, Biomaterials, 2015, 67, 151–159. Nanoparticles, ACS Appl. Mater. Interfaces, 2016, 8, 14096–14101.
85 M. Kendall, P. Ding and K. Kendall, Particle and nanoparticle 104 R. H. Wu, et al., A facile route to tailoring peptide-stabilized
interactions with fibrinogen: the importance of aggregation gold nanoparticles using glutathione as a synthon, Mole-
in nanotoxicology, Nanotoxicology, 2011, 5, 55–65. cules, 2014, 19, 6754–6775.
86 L. Foucaud, M. R. Wilson, D. M. Brown and V. Stone, 105 R. Lévy, et al., Rational and combinatorial design of
Measurement of reactive species production by nano- peptide capping ligands for gold nanoparticles, J. Am.
particles prepared in biologically relevant media, Toxicol. Chem. Soc., 2004, 126, 10076–10084.
Lett., 2007, 174, 1–9. 106 R. Lévy, Peptide-Capped Gold Nanoparticles: Towards Arti-
87 T. M. Sager, et al., Improved method to disperse nano- ficial Proteins, ChemBioChem, 2006, 7, 1141–1145.
particles for in vitro and in vivo investigation of toxicity, 107 G. Zhou, et al., Robust aqueous quantum dots capped with
Nanotoxicology, 2007, 1, 118–129. peptide ligands as biomaterials: Facile preparation, good
88 P. Bihari, et al., Optimized dispersion of nanoparticles for stability, and multipurpose application, Part. Part. Syst.
biological in vitro and in vivo studies, Part. Fibre Toxicol., Charact., 2014, 31, 382–389.
2008, 5, 14. 108 T. Mizutaru, et al., Cysteine-containing oligopeptide b-sheets
89 A. Albanese and W. C. W. Chan, Effect of Gold Nanoparticle as redispersants for agglomerated metal nanoparticles,
Aggregation on Cell Uptake and Toxicity, ACS Nano, 2011, 5, J. Mater. Chem. A, 2015, 3, 17612–17619.
5478–5489. 109 H. Poblete, et al., New Insights into Peptide-Silver Nanoparticle
90 T. L. Moore, et al., Nanoparticle colloidal stability in cell Interaction: Deciphering the Role of Cysteine and Lysine in
culture media and impact on cellular interactions, Chem. the Peptide Sequence, Langmuir, 2016, 32, 265–273.
Soc. Rev., 2015, 44, 6287–6305. 110 N. Todorova, et al., Surface presentation of functional
91 J. Cui, et al., A Framework to Account for Sedimentation peptides in solution determines cell internalization effi-
and Diffusion in Particle-Cell Interactions, Langmuir, 2016, ciency of TAT conjugated nanoparticles, Nano Lett., 2014,
32, 12394–12402. 14, 5229–5237.
92 N. Feliu, X. Sun, R. A. Alvarez Puebla and W. J. Parak, 111 Y. Cesbron, U. Shaheen, P. Free and R. Lévy, TAT and HA2
Quantitative Particle-Cell Interaction: Some Basic Physico- facilitate cellular uptake of gold nanoparticles but do not
chemical Pitfalls, Langmuir, 2017, 33, 6639–6646. lead to cytosolic localisation, PLoS One, 2015, 10, e0121683.
3606 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
112 Z. Wang, R. Lévy, D. G. Fernig and M. Brust, The peptide 128 A. Battistoni, et al., A histidine-rich metal binding domain
route to multifunctional gold nanoparticles, Bioconjugate at the N terminus of Cu, Zn-superoxide dismutases from
Chem., 2005, 16, 497–500. pathogenic bacteria. A novel strategy for metal chaperon-
113 G. H. Woehrle, L. O. Brown and J. E. Hutchison, Thiol- ing, J. Biol. Chem., 2001, 276, 30315–30325.
functionalized, 1.5-nm gold nanoparticles through ligand 129 K. E. Sapsford, et al., Kinetics of metal-affinity driven self-
exchange reactions: Scope and mechanism of ligand assembly between proteins or peptides and CdSe-ZnS
exchange, J. Am. Chem. Soc., 2005, 127, 2172–2183. quantum dots, J. Phys. Chem. C, 2007, 111, 11528–11538.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
114 A. M. Smith, et al., Quantitative analysis of thiolated ligand 130 D. E. Prasuhn, et al., Combining Chemoselective Ligation
exchange on gold nanoparticles monitored by 1H NMR with Polyhistidine-Driven Self-Assembly for the Modular
spectroscopy, Anal. Chem., 2015, 87, 2771–2778. Display of Biomolecules on Quantum Dots, ACS Nano, 2010,
115 N. G. Bastus, et al., Homogeneous conjugation of peptides 4, 267–278.
onto gold nanoparticles enhances macrophage response, 131 E. R. Goldman, et al., Self-assembled luminescent CdSe-ZnS
ACS Nano, 2009, 3, 1335–1344. quantum dot bioconjugates prepared using engineered
116 L. Turell, R. Radi and B. Alvarez, The thiol pool in human poly-histidine terminated proteins, Anal. Chim. Acta, 2005,
plasma: The central contribution of albumin to redox 534, 63–67.
processes, Free Radical Biol. Med., 2013, 65, 244–253. 132 A. M. Dennis, et al., Surface ligand effects on metal-affinity
117 A. Kassam, G. Bremner, B. Clark, G. Ulibarri and R. B. Lennox, coordination to quantum dots: Implications for nanoprobe
Place exchange reactions of alkyl thiols on gold nanoparticles, self-assembly, Bioconjugate Chem., 2010, 21, 1160–1170.
J. Am. Chem. Soc., 2006, 128, 3476–3477. 133 S. Behzadi, et al., Cellular uptake of nanoparticles: journey
118 D.-H. Tsai, et al., Adsorption and conformation of serum inside the cell, Chem. Soc. Rev., 2017, 46, 4218–4244.
albumin protein on gold nanoparticles investigated using 134 S. Barua and S. Mitragotri, Challenges associated with
dimensional measurements and in situ spectroscopic penetration of nanoparticles across cell and tissue bar-
methods, Langmuir, 2011, 27, 2464–2477. riers: A review of current status and future prospects, Nano
119 S. Dominguez-medina, S. Mcdonough, P. Swanglap, Today, 2014, 9, 223–243.
C. F. Landes and S. Link, In situ measurement of bovine 135 J. Gilleron, et al., Image-based analysis of lipid nanoparticle-
serum albumin interaction with gold nanospheres, Lang- mediated siRNA delivery, intracellular trafficking and endo-
muir, 2012, 28, 9131–9139. somal escape, Nat. Biotechnol., 2013, 31, 638–646.
120 T. A. Larson, P. P. Joshi and K. Sokolov, Preventing protein 136 L. I. Selby, C. M. Cortez-Jugo, G. K. Such and A. P. R. Johnston,
adsorption and macrophage uptake of gold nanoparticles Nanoescapology: progress toward understanding the endo-
via a hydrophobic shield, ACS Nano, 2012, 6, 9182–9190. somal escape of polymeric nanoparticles, Wiley Interdiscip.
121 F. Pinaud, D. King, H. Moore and S. Weiss, Bioactivation Rev.: Nanomed. Nanobiotechnol., 2017, 9, e1452.
and Cell Targeting of Semiconductor CdSe/ZnS Nanocrys- 137 Y. Huang, et al., Curb challenges of the ‘Trojan Horse’
tals with Phytochelatin-Related Peptides Bioactivation and approach: Smart strategies in achieving effective yet safe
Cell Targeting of Semiconductor CdSe/ZnS Nanocrystals cell-penetrating peptide-based drug delivery, Adv. Drug
with Phytochelatin-Related Peptides, J. Am. Chem. Soc., Delivery Rev., 2013, 65, 1299–1315.
2004, 126, 6115–6123. 138 S. M. Farkhani, et al., Cell penetrating peptides: Efficient
122 J. Xu, P. Ruchala, Y. Ebenstain, J. J. Li and S. Weiss, Stable, vectors for delivery of nanoparticles, nanocarriers, thera-
compact, bright biofunctional quantum dots with peutic and diagnostic molecules, Peptides, 2014, 57, 78–94.
improved peptide coating, J. Phys. Chem. B, 2012, 116, 139 W. B. Kauffman, T. Fuselier, J. He and W. C. Wimley,
11370–11378. Mechanism Matters: A Taxonomy of Cell Penetrating
123 F. Porta, et al., Gold nanoparticles capped by peptides, Peptides, Trends Biochem. Sci., 2015, 40, 749–764.
Mater. Sci. Eng., B, 2007, 140, 187–194. 140 M. Kristensen, D. Birch and H. M. Nielsen, Applications
124 Z. Krpetić, P. Nativo, F. Porta and M. Brust, A multidentate and challenges for use of cell-penetrating peptides as
peptide for stabilization and facile bioconjugation of gold delivery vectors for peptide and protein cargos, Int. J. Mol.
Nanoparticles, Bioconjugate Chem., 2009, 20, 619–624. Sci., 2016, 17, 185.
125 F. Aldeek, M. Safi, N. Zhan, G. Palui and H. Mattoussi, 141 A. A. Keller, et al., Relationships between cargo, cell
Understanding the Self-Assembly of Proteins onto Gold penetrating peptides and cell type for uptake of non-
Nanoparticles and Quantum Dots Driven by Metal- covalent complexes into live cells, Pharmaceuticals, 2013,
Histidine Coordination, ACS Nano, 2013, 7, 10197–10210. 6, 184–203.
126 R. J. Sundberg and R. B. Martin, Interactions of histidine 142 F. Madani, S. Lindberg, Ü. Langel, S. Futaki and A. Gräslund,
and other imidazole derivatives with transition metal ions Mechanisms of cellular uptake of cell-penetrating peptides,
in chemical and biological systems, Chem. Rev., 1974, 74, J. Biophys., 2011, 2011, 414729.
471–517. 143 H. A. Rydberg, M. Matson, H. L. Åmand, E. K. Esbjörner
127 J. Schmitt, H. Hess and H. G. Stunnenberg, Affinity and B. Nordén, Effects of tryptophan content and back-
purification of histidine-tagged proteins, Mol. Biol. Rep., bone spacing on the uptake efficiency of cell-penetrating
1993, 18, 223–230. peptides, Biochemistry, 2012, 51, 5531–5539.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3607
View Article Online
144 H. D. Herce, A. E. Garcia and M. C. Cardoso, Fundamental 162 S. Futaki, et al., Arginine-rich peptides. An abundant
molecular mechanism for the cellular uptake of guanidinium- source of membrane-permeable peptides having potential
rich molecules, J. Am. Chem. Soc., 2014, 136, 17459–17467. as carriers for intracellular protein delivery, J. Biol. Chem.,
145 C. Bechara and S. Sagan, Cell-Penetrating Peptides: 20 2001, 276, 5836–5840.
Years Later, Where Do We Stand?, FEBS Lett., 2013, 587, 163 W. P. R. Verdurmen, M. Mazlami and A. Plückthun, A
1693–1702. quantitative comparison of cytosolic delivery via different
146 J. S. Wadia, R. V. Stan and S. F. Dowdy, Transducible protein uptake systems, Sci. Rep., 2017, 7, 13194.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
TAT-HA fusogenic peptide enhances escape of TAT-fusion 164 T. Jiang, et al., Tumor imaging by means of proteolytic
proteins after lipid raft macropinocytosis, Nat. Med., 2004, activation of cell-penetrating peptides, Proc. Natl. Acad. Sci.
10, 310–315. U. S. A., 2004, 101, 17867–17872.
147 K. Kettler, K. Veltman, D. van de Meent, A. van Wezel and 165 Y. Zhang, M. K. So and J. Rao, Protease-modulated cellular
A. J. Hendriks, Cellular uptake of nanoparticles as deter- uptake of quantum dots, Nano Lett., 2006, 6, 1988–1992.
mined by particle properties, experimental conditions, and 166 H. Gao, et al., Angiopep-2 and activatable cell-penetrating
cell type, Environ. Toxicol. Chem., 2014, 33, 481–492. peptide dual-functionalized nanoparticles for systemic
148 G. Guidotti, L. Brambilla and D. Rossi, Cell-Penetrating glioma-targeting delivery, Mol. Pharmaceutics, 2014, 11,
Peptides: From Basic Research to Clinics, Trends Pharmacol. 2755–2763.
Sci., 2017, 38, 406–424. 167 T. J. Harris, et al., Protease-triggered unveiling of bioactive
149 M. Zhu, et al., Physicochemical properties determine nano- nanoparticles, Small, 2008, 4, 1307–1312.
material cellular uptake, transport, and fate, Acc. Chem. 168 M. B. Hansen, et al., Constrained and UV-activatable cell-
Res., 2013, 46, 622–631. penetrating peptides for intracellular delivery of lipo-
150 A. Gautam, et al., CPPsite: A curated database of cell somes, J. Controlled Release, 2012, 164, 87–94.
penetrating peptides, Database, 2012, bas015. 169 E. Koren, A. Apte, A. Jani and V. P. Torchilin, Multi-
151 K. Saar, et al., Cell-penetrating peptides: A comparative functional PEGylated 2C5-immunoliposomes containing
membrane toxicity study, Anal. Biochem., 2005, 345, 55–65. pH-sensitive bonds and TAT peptide for enhanced tumor
152 S. R. Schwarze, A. Ho, A. Vocero-Akbani and S. F. Dowdy, cell internalization and cytotoxicity, J. Controlled Release,
In Vivo Protein Transduction: Delivery of a Biologically Active 2012, 160, 264–273.
Protein into the Mouse, Science, 1999, 285, 1569–1572. 170 A. A. Kale and V. P. Torchilin, Enhanced transfection
153 C. Rudolph, et al., Application of novel solid lipid nano- of tumor cells in vivo using ‘Smart’ pH-sensitive TAT-
particle (SLN)-gene vector formulations based on a dimeric modified pegylated liposomes, J. Drug Targeting, 2016,
HIV-1 TAT-peptide in vitro and in vivo, Pharm. Res., 2004, 15, 538–545.
21, 1662–1669. 171 J. Kalia and R. T. Raines, Hydrolytic stability of hydrazones
154 Y. Qin, et al., Comparison of four different peptides to and oximes, Angew. Chem., Int. Ed., 2008, 47, 7523–7526.
enhance accumulation of liposomes into the brain, J. Drug 172 K. E. Sapsford, et al., Functionalizing nanoparticles with
Targeting, 2012, 20, 235–245. biological molecules: developing chemistries that facilitate
155 R. R. Sawant and V. P. Torchilin, Enhanced cytotoxicity of nanotechnology, Chem. Rev., 2013, 113, 1904–2074.
TATp-bearing paclitaxel-loaded micelles in vitro and 173 A. Dirksen, S. Yegneswaran and P. E. Dawson, Bisaryl
in vivo, Int. J. Pharm., 2009, 374, 114–118. Hydrazones as Exchangeable Biocompatible Linkers, Angew.
156 J.-Y. Lee, et al., Cell-penetrating chitosan/doxorubicin/TAT Chem., Int. Ed., 2010, 49, 2023–2027.
conjugates for efficient cancer therapy, Int. J. Cancer, 2011, 174 A. Saha, S. K. Basiruddin, A. R. Maity and N. R. Jana,
128, 2470–2480. Synthesis of nanobioconjugates with a controlled average
157 J. Liu, et al., TAT-modified nanosilver for combating multidrug- number of biomolecules between 1 and 100 per nanoparticle
resistant cancer, Biomaterials, 2012, 33, 6155–6161. and observation of multivalency dependent interaction with
158 G. Sharma, et al., Cell penetrating peptide tethered bi-ligand proteins and cells, Langmuir, 2013, 29, 13917–13924.
liposomes for delivery to brain in vivo: Biodistribution and 175 M. Zhao, M. F. Kircher, L. Josephson and R. Weissleder,
transfection, J. Controlled Release, 2013, 167, 1–10. Differential conjugation of tat peptide to superpara-
159 A. Koshkaryev, A. Piroyan and V. P. Torchilin, Bleomycin in magnetic nanoparticles and its effect on cellular uptake,
octaarginine-modified fusogenic liposomes results in improved Bioconjugate Chem., 2002, 13, 840–844.
tumor growth inhibition, Cancer Lett., 2013, 334, 293–301. 176 J. Hoyer, U. Schatzschneider, M. Schulz-Siegmund and
160 H. Xia, et al., Penetratin-functionalized PEG-PLA nano- I. Neundorf, Dimerization of a cell-penetrating peptide
particles for brain drug delivery, Int. J. Pharm., 2012, 436, leads to enhanced cellular uptake and drug delivery,
840–850. Beilstein J. Org. Chem., 2012, 8, 1788–1797.
161 G. Sharma, A. Modgil, T. Zhong, C. Sun and J. Singh, 177 J. C. Breger, et al., Nanoparticle cellular uptake by dendritic
Influence of short-chain cell-penetrating peptides on wedge peptides: achieving single peptide facilitated delivery,
transport of doxorubicin encapsulating receptor-targeted Nanoscale, 2017, 9, 10447–10464.
liposomes across brain endothelial barrier, Pharm. Res., 178 M. Di Pisa, G. Chassaing and J. M. Swiecicki, Translocation
2014, 31, 1194–1209. mechanism(s) of cell-penetrating peptides: Biophysical
3608 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
studies using artificial membrane bilayers, Biochemistry, 196 K. Ulbrich, T. Hekmatara, E. Herbert and J. Kreuter,
2015, 54, 194–207. Transferrin- and transferrin-receptor-antibody-modified
179 C. Dalal, A. Saha and N. R. Jana, Nanoparticle Multivalency nanoparticles enable drug delivery across the blood-brain
Directed Shifting of Cellular Uptake Mechanism, J. Phys. barrier (BBB), Eur. J. Pharm. Biopharm., 2009, 71, 251–256.
Chem. C, 2016, 120, 6778–6786. 197 X. Ying, et al., Dual-targeting daunorubicin liposomes
180 C. Dalal and N. R. Jana, Multivalency Effect of TAT-Peptide- improve the therapeutic efficacy of brain glioma in animals,
Functionalized Nanoparticle in Cellular Endocytosis J. Controlled Release, 2010, 141, 183–192.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
and Subcellular Trafficking, J. Phys. Chem. B, 2017, 121, 198 J.-Q. Gao, et al., Glioma targeting and blood-brain barrier
2942–2951. penetration bydual-targeting doxorubincin liposomes,
181 I. Nakase, S. Kobayashi and S. Futaki, Endosome- Biomaterials, 2013, 34, 5628–5639.
disruptive peptides for improving cytosolic delivery of 199 Z. Pang, et al., Brain delivery and cellular internalization
bioactive macromolecules, Biopolymers, 2010, 94, 763–770. mechanisms for transferrin conjugated biodegradable
182 C. Plank, B. Oberhauser, K. Mechtler, C. Koch and polymersomes, Int. J. Pharm., 2011, 415, 284–292.
E. Wagner, The influence of endosome-disruptive peptides 200 Y. Li, et al., A dual-targeting nanocarrier based on
on gene transfer using synthetic virus like gene transfer poly(amidoamine) dendrimers conjugated with transferrin
systems, J. Biol. Chem., 1994, 269, 12918–12924. and tamoxifen for treating brain gliomas, Biomaterials,
183 T. Kakudo, et al., Transferrin-Modified Liposomes Equipped 2012, 33, 3899–3908.
with a pH-Sensitive Fusogenic Peptide: An Artificial Viral- 201 M. J. Gomes, P. J. Kennedy, S. Martins and B. Sarmento,
like Delivery System, Biochemistry, 2004, 43, 5618–5628. Delivery of siRNA silencing P-gp in peptide- functionalized
184 K. Takayama, et al., Enhanced intracellular delivery using nanoparticles causes efflux modulation at the blood –
arginine-rich peptides by the addition of penetration accel- brain barrier, Nanomedicine, 2017, 12, 1385–1399.
erating sequences (Pas), J. Controlled Release, 2009, 138, 202 J. Huwyler, D. Wu and W. M. Pardridge, Brain drug delivery
128–133. of small molecules using immunoliposomes, Proc. Natl.
185 B. R. Liu, et al., Endocytic Trafficking of Nanoparticles Acad. Sci. U. S. A., 1996, 93, 14164–14169.
Delivered by Cell-penetrating Peptides Comprised of Nona- 203 Y. Zhang, F. Calon, C. Zhu, R. J. Boado and W. M. Pardridge,
arginine and a Penetration Accelerating Sequence, PLoS Intravenous Nonviral Gene Therapy Causes Normalization
One, 2013, 8, e67100. of Striatal Tyrosine Hydroxylase and Reversal of Motor
186 R. A. Morshed, et al., Cell-Penetrating Peptide-Modified Impairment in Experimental Parkinsonism, Hum. Gene
Gold Nanoparticles for the Delivery of Doxorubicin to Ther., 2003, 14, 1–12.
Brain Metastatic Breast Cancer, Mol. Pharmaceutics, 2016, 204 Z. Pang, et al., Preparation and brain delivery property of
13, 1843–1854. biodegradable polymersomes conjugated with OX26,
187 H. Gao, Progress and perspectives on targeting nanoparticles J. Controlled Release, 2008, 128, 120–127.
for brain drug delivery, Acta Pharm. Sin. B, 2016, 6, 268–286. 205 S.-S. Kim, et al., A nanoparticle carrying the p53 gene
188 B. Oller-Salvia, M. Sánchez-Navarro, E. Giralt and M. Teixidó, targets tumors including cancer stem cells, sensitizes
Blood-brain barrier shuttle peptides: an emerging paradigm glioblastoma to chemotherapy and improves survival,
for brain delivery, Chem. Soc. Rev., 2016, 45, 4690–4707. ACS Nano, 2014, 8, 5494–5514.
189 W. A. Banks, From blood–brain barrier to blood–brain 206 W. M. Pardridge, Blood–brain barrier drug delivery of IgG
interface: new opportunities for CNS drug delivery, Nat. fusion proteins with a transferrin receptor monoclonal
Rev. Drug Discovery, 2016, 15, 275–292. antibody, Expert Opin. Drug Delivery, 2015, 12, 207–222.
190 A. M. Grabrucker, et al., Nanoparticle transport across the 207 P. Yue, et al., OX26/CTX-conjugated PEGylated liposome as
blood brain barrier, Tissue Barriers, 2016, 4, e1153568. a dual-targeting gene delivery system for brain glioma,
191 L. G. Dubois, et al., Gliomas and the vascular fragility of Mol. Cancer, 2014, 13, 191.
the blood brain barrier, Front. Cell. Neurosci., 2014, 8, 417. 208 R. Huang, W. Ke, Y. Liu, C. Jiang and Y. Pei, The use of
192 S. Santra, et al., Rapid and effective labeling of brain tissue lactoferrin as a ligand for targeting the polyamidoamine-
using TAT-conjugated CdS:Mn/ZnS quantum dots, Chem. based gene delivery system to the brain, Biomaterials, 2008,
Commun., 2005, 3144–3146. 29, 238–246.
193 L. Liu, et al., Self-assembled cationic peptide nanoparticles 209 Y. Yu, et al., The proton permeability of self-assembled
as an efficient antimicrobial agent, Nat. Nanotechnol., polymersomes and their neuroprotection by enhancing a
2009, 4, 457–463. neuroprotective peptide across the blood-brain barrier
194 Y. Cheng, et al., Blood-brain barrier permeable gold nano- after modification with lactoferrin, Nanoscale, 2014, 6,
particles: An efficient delivery platform for enhanced 3250–3258.
malignant glioma therapy and imaging, Small, 2014, 10, 210 K. Hu, et al., Lactoferrin-conjugated PEG-PLA nano-
5137–5150. particles with improved brain delivery: In vitro and
195 Y. Liu, et al., Paclitaxel loaded liposomes decorated with in vivo evaluations, J. Controlled Release, 2009, 134, 55–61.
a multifunctional tandem peptide for glioma targeting, 211 K. Hu, et al., Lactoferrin conjugated PEG-PLGA nano-
Biomaterials, 2014, 35, 4835–4847. particles for brain delivery: Preparation, characterization
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3609
View Article Online
and efficacy in Parkinsons disease, Int. J. Pharm., 2011, decorated gold nanoparticles, Biomaterials, 2015, 37,
415, 273–283. 425–435.
212 Y. Ye, et al., A novel lactoferrin-modified b-cyclodextrin 229 Y. Endo-Takahashi, et al., Preparation of Angiopep-2
nanocarrier for brain-targeting drug delivery, Int. J. Pharm., Peptide-Modified Bubble Liposomes for Delivery to the
2013, 458, 110–117. Brain, Biol. Pharm. Bull., 2016, 39, 977–983.
213 Z. Su, et al., Lactoferrin-modified poly(ethylene glycol)- 230 P. J. Gaillard, et al., Enhanced brain delivery of liposomal
grafted BSA nanoparticles as a dual-targeting carrier for methylprednisolone improved therapeutic efficacy in a
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
treating brain gliomas, Mol. Pharmaceutics, 2014, 11, model of neuroinflammation, J. Controlled Release, 2012,
1823–1834. 164, 364–369.
214 Y.-C. Kuo and Y.-C. Chen, Targeting delivery of etoposide 231 J. Rip, et al., Glutathione PEGylated liposomes: pharmaco-
to inhibit the growth of human glioblastoma multiforme kinetics and delivery of cargo across the blood–brain
using lactoferrin- and folic acid-grafted poly(lactide-co- barrier in rats, J. Drug Targeting, 2014, 22, 460–467.
glycolide) nanoparticles, Int. J. Pharm., 2015, 479, 138–149. 232 M. Rotman, et al., Enhanced glutathione PEGylated lipo-
215 J. H. Lee, J. A. Engler, J. F. Collawn and B. A. Moore, somal brain delivery of an anti-amyloid single domain
Receptor mediated uptake of peptides that bind the human antibody fragment in a mouse model for Alzheimer’s
transferrin receptor, Eur. J. Biochem., 2001, 268, 2004–2012. disease, J. Controlled Release, 2015, 203, 40–50.
216 R. Prades, et al., Delivery of gold nanoparticles to the brain 233 C. Zhan, et al., Micelle-based brain-targeted drug delivery
by conjugation with a peptide that recognizes the trans- enabled by a nicotine acetylcholine receptor ligand, Angew.
ferrin receptor, Biomaterials, 2012, 33, 7194–7205. Chem., Int. Ed., 2011, 50, 5482–5485.
217 R. Prades, et al., Applying the retro-enantio approach to 234 C. Fang, et al., Temozolomide nanoparticles for targeted
obtain a peptide capable of overcoming the blood-brain glioblastoma therapy, ACS Appl. Mater. Interfaces, 2015, 7,
barrier, Angew. Chem., Int. Ed., 2015, 54, 3967–3972. 6674–6682.
218 P. J. Gaillard, C. C. Visser, C. C. M. Appeldoorn and J. Rip, 235 S. Chen, M. Ahmadiantehrani, N. G. Publicover, K. W. J.
Enhanced brain drug delivery: Safely crossing the blood-brain Hunter and X. Zhu, Thermal Decomposition Based Synthesis
barrier, Drug Discovery Today: Technol., 2012, 9, e155–e160. of Ag-In-S/ZnS Quantum Dots and Their Chlorotoxin-
219 M. Demeule, et al., Identification and design of peptides as Modified Micelles for Brain Tumor Cell Targeting, RSC
a new drug delivery system for the brain, J. Pharmacol. Exp. Adv., 2015, 74, 60612–60620.
Ther., 2008, 324, 1064–1072. 236 B. Oller-Salvia, et al., MiniAp-4: A Venom-Inspired Pepti-
220 W. Ke, et al., Gene delivery targeted to the brain using an domimetic for Brain Delivery, Angew. Chem., Int. Ed., 2016,
Angiopep-conjugated polyethyleneglycol-modified polyamido- 55, 572–575.
amine dendrimer, Biomaterials, 2009, 30, 6976–6985. 237 G. Tosi, et al., NIR-labeled nanoparticles engineered for
221 H. Xin, et al., Angiopep-conjugated poly(ethylene glycol)- brain targeting: In vivo optical imaging application and
co-poly(e-caprolactone) nanoparticles as dual-targeting fluorescent microscopy evidences, J. Neural Transm., 2011,
drug delivery system for brain glioma, Biomaterials, 2011, 118, 145–153.
32, 4293–4305. 238 A. Vilella, et al., Insight on the fate of CNS-targeted
222 H. Xin, et al., Anti-glioblastoma efficacy and safety of nanoparticles. Part I: Rab5-dependent cell-specific uptake
paclitaxel-loading Angiopep-conjugated dual targeting and distribution, J. Controlled Release, 2014, 174, 195–201.
PEG-PCL nanoparticles, Biomaterials, 2012, 33, 8167–8176. 239 Y. Liu, et al., Brain-targeting gene delivery and cellular
223 H. Yan, et al., Two-order targeted brain tumor imaging by internalization mechanisms for modified rabies virus gly-
using an optical/paramagnetic nanoprobe across the blood coprotein RVG29 nanoparticles, Biomaterials, 2009, 30,
brain barrier, ACS Nano, 2012, 6, 410–420. 4195–4202.
224 X. Sun, et al., Co-delivery of pEGFP-hTRAIL and paclitaxel 240 T.-E. Park, et al., Enhanced BBB permeability of osmoti-
to brain glioma mediated by an angiopep-conjugated lipo- cally active poly(mannitol-co-PEI) modified with rabies
some, Biomaterials, 2012, 33, 916–924. virus glycoprotein via selective stimulation of caveolar
225 R. Huang, et al., Angiopep-conjugated nanoparticles for endocytosis for RNAi therapeutics in Alzheimer’s disease,
targeted long-term gene therapy of parkinson’s disease, Biomaterials, 2015, 38, 61–71.
Pharm. Res., 2013, 30, 2549–2559. 241 W. Lu, et al., Cationic albumin-conjugated pegylated nano-
226 X. Ying, et al., Angiopep-conjugated electro-responsive particles as novel drug carrier for brain delivery,
hydrogel nanoparticles: Therapeutic potential for epilepsy, J. Controlled Release, 2005, 107, 428–448.
Angew. Chem., Int. Ed., 2014, 53, 12436–12440. 242 Y.-L. Xie, W. Lu and X.-G. Jiang, Improvement of cationic
227 D. Ni, et al., Dual-targeting upconversion nanoprobes albumin conjugated pegylated nanoparticles holding
across the blood-brain barrier for magnetic resonance/ NC-1900, a vasopressin fragment analog, in memory deficits
fluorescence imaging of intracranial glioblastoma, ACS induced by scopolamine in mice, Behav. Brain Res., 2006,
Nano, 2014, 8, 1231–1242. 173, 76–84.
228 S. Ruan, et al., Tumor microenvironment sensitive doxor- 243 W. Lu, J. Wan, Q. Zhang, Z. She and X. Jiang, Aclarubicin-
ubicin delivery and release to glioma using angiopep-2 loaded cationic albumin-conjugated pegylated nanoparticle
3610 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
for glioma chemotherapy in rats, Int. J. Cancer, 2007, 120, 261 H. Gao, et al., In vitro and in vivo intracellular distribution
420–431. and anti-glioblastoma effects of docetaxel-loaded nano-
244 H. J. Byeon, et al., Doxorubicin-loaded nanoparticles consisted particles functioned with IL-13 peptide, Int. J. Pharm.,
of cationic- and mannose-modified-albumins for dual-targeting 2014, 466, 8–17.
in brain tumors, J. Controlled Release, 2016, 225, 301–313. 262 H. Gao, et al., Tumor cells and neovasculature dual target-
245 P. Carmeliet and R. K. Jain, Angiogenesis in cancer and ing delivery for glioblastoma treatment, Biomaterials, 2014,
other diseases, Nature, 2000, 407, 249–257. 35, 2374–2382.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
246 R. K. Jain and T. Stylianopoulos, Delivering nanomedicine 263 B. Wang, et al., Nanoparticles functionalized with Pep-1 as
to solid tumors, Nat. Rev. Clin. Oncol., 2010, 7, 653–664. potential glioma targeting delivery system via interleukin
247 Y. H. Bae and K. Park, Targeted drug delivery to tumors: 13 receptor a2-mediated endocytosis, Biomaterials, 2014,
Myths, reality and possibility, J. Controlled Release, 2011, 35, 5897–5907.
153, 198–205. 264 P. Laakkonen, K. Porkka, J. A. Hoffman and E. Ruoslahti, A
248 U. Prabhakar, et al., Challenges and key considerations tumor-homing peptide with a targeting specificity related
of the enhanced permeability and retention effect for to lymphatic vessels, Nat. Med., 2002, 8, 751–755.
nanomedicine drug delivery in oncology, Cancer Res., 265 P. P. Karmali, et al., Targeting of albumin-embedded paclitaxel
2013, 73, 2412–2417. nanoparticles to tumors, Nanomedicine, 2009, 5, 73–82.
249 J. W. Nichols and Y. H. Bae, EPR: Evidence and fallacy, 266 L. Roth, et al., Transtumoral targeting enabled by a novel
J. Controlled Release, 2014, 190, 451–464. neuropilin-binding peptide, Oncogene, 2012, 31, 3754–3763.
250 F. Danhier, To exploit the tumor microenvironment: Since 267 Z.-Z. Yang, J.-Q. Li, Z.-Z. Wang, D.-W. Dong and X.-R. Qi,
the EPR effect fails in the clinic, what is the future of Tumor-targeting dual peptides-modified cationic lipo-
nanomedicine?, J. Controlled Release, 2016, 244, 108–121. somes for delivery of siRNA and docetaxel to gliomas,
251 T. J. Anchordoquy, et al., Mechanisms and Barriers in Biomaterials, 2014, 35, 5226–5239.
Cancer Nanomedicine: Addressing Challenges, Looking 268 S. M. Weis, Vascular permeability in cardiovascular disease
for Solutions, ACS Nano, 2017, 11, 12–18. and cancer, Curr. Opin. Hematol., 2008, 15, 243–249.
252 M. Björnmalm, K. J. Thurecht, M. Michael, A. M. Scott and 269 Z. Panagi, et al., Effect of dose on the biodistribution and
F. Caruso, Bridging Bio-Nano Science and Cancer Nano- pharmacokinetics of PLGA and PLGA-mPEG nanoparticles,
medicine, ACS Nano, 2017, 11, 9594–9613. Int. J. Pharm., 2001, 221, 143–152.
253 T. Teesalu, K. N. Sugahara, V. R. Kotamraju and E. Ruoslahti, 270 D. Dehaini, R. H. Fang and L. Zhang, Biomimetic strate-
C-end rule peptides mediate neuropilin-1-dependent cell, gies for targeted nanoparticle delivery, Bioeng. Transl.
vascular, and tissue penetration, Proc. Natl. Acad. Sci. Med., 2016, 1, 30–46.
U. S. A., 2009, 106, 16157–16162. 271 L. D. Field, J. B. Delehanty, Y. Chen and I. L. Medintz,
254 K. N. Sugahara, et al., Tissue-Penetrating Delivery of Peptides for Specifically Targeting Nanoparticles to Cellular
Compounds and Nanoparticles into Tumors, Cancer Cell, Organelles: Quo Vadis?, Acc. Chem. Res., 2015, 48, 1380–1390.
2009, 16, 510–520. 272 P. Valadon, et al., Screening phage display libraries for
255 K. N. Sugahara, et al., Coadministration of a Tumor- organ-specific vascular immunotargeting in vivo, Proc.
Penetrating Peptide Enhances the Efficacy of Cancer Natl. Acad. Sci. U. S. A., 2006, 103, 407–412.
Drugs, Science, 2010, 328, 1031–1036. 273 J. Wang, et al., Selection of phage-displayed peptides on
256 J. Wang, et al., Targeted gene delivery to glioblastoma live adherent cells in microfluidic channels, Proc. Natl.
using a C-end rule RGERPPR peptide-functionalised poly- Acad. Sci. U. S. A., 2011, 108, 6909–6914.
ethylenimine complex, Int. J. Pharm., 2013, 458, 48–56. 274 C.-H. Wu, I.-J. Liu, R.-M. Lu and H.-C. Wu, Advancement
257 A. M. Hamilton, et al., Nanoparticles coated with the and applications of peptide phage display technology in
tumor-penetrating peptide iRGD reduce experimental biomedical science, J. Biomed. Sci., 2016, 23, 8.
breast cancer metastasis in the brain, J. Mol. Med., 2015, 275 V. J. Yao, et al., Ligand-targeted theranostic nanomedicines
93, 991–1001. against cancer, J. Controlled Release, 2016, 240, 267–286.
258 X. Cun, et al., A Novel Strategy through Combining iRGD 276 G. P. Smith, Filamentous fusion phage: novel expression
Peptide with Tumor-Microenvironment-Responsive and vectors that display cloned antigens on the virion surface,
Multistage Nanoparticles for Deep Tumor Penetration, Science, 1985, 228, 1315–1317.
ACS Appl. Mater. Interfaces, 2015, 7, 27458–27466. 277 K. Omidfar and M. Daneshpour, Advances in phage display
259 H. Gao, et al., Glioma-homing peptide with a cell- technology for drug discovery, Expert Opin. Drug Discovery,
penetrating effect for targeting delivery with enhanced 2015, 10, 651–669.
glioma localization, penetration and suppression of 278 M. J. Mcguire, S. Li and K. C. Brown, Biopanning of Phage
glioma growth, J. Controlled Release, 2013, 172, 921–928. Displayed Peptide Libraries for the Isolation of Cell-
260 Y. Wang, et al., Multifunctional mesoporous silica-coated Specific Ligands, Methods Mol. Biol., 2009, 504, 291–321.
graphene nanosheet used for chemo-photothermal synergistic 279 K. C. Brown, Peptidic tumor targeting agents: the road
targeted therapy of glioma, J. Am. Chem. Soc., 2013, 135, from phage display peptide selections to clinical applica-
4799–4804. tions, Curr. Pharm. Des., 2010, 16, 1040–1054.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3611
View Article Online
280 K. Deramchia, et al., New human antibody fragments 297 H. Ding, et al., HER2-positive breast cancer targeting and
homing to atherosclerotic endothelial and subendothelial treatment by a peptide-conjugated mini nanodrug, Nano-
tissues: An in vivo phage display targeting human anti- medicine, 2017, 13, 631–639.
bodies homing to atherosclerotic tissues, Am. J. Pathol., 298 Q. Mu, et al., Anti-Her2/neu Peptide/Conjugated Iron Oxide
2012, 180, 2576–2589. Nanoparticles for Targeted Delivery of Paclitaxel to Breast
281 D. Rajotte, et al., Molecular heterogeneity of the vascular Cancer Cells, Nanoscale, 2015, 7, 18010–18014.
endothelium revealed by in vivo phage display, J. Clin. 299 W. Arap, et al., Targeting the prostate for destruction
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
Invest., 1998, 102, 430–437. through a vascular address, Proc. Natl. Acad. Sci. U. S. A.,
282 K. Porkka, P. Laakkonen, J. A. Hoffman, M. Bernasconi and 2002, 99, 1527–1531.
E. Ruoslahti, A fragment of the HMGN2 protein homes to 300 C.-Y. Yeh, J.-K. Hsiao, Y.-P. Wang, C.-H. Lan and H.-C. Wu,
the nuclei of tumor cells and tumor endothelial cells Peptide-conjugated nanoparticles for targeted imaging and
in vivo, Proc. Natl. Acad. Sci. U. S. A., 2002, 99, 7444–7449. therapy of prostate cancer, Biomaterials, 2016, 99, 1–15.
283 J. A. Hoffman, et al., Progressive vascular changes in a 301 M. G. Kolonin, P. K. Saha, L. Chan, R. Pasqualini and
transgenic mouse model of squamous cell carcinoma, W. Arap, Reversal of obesity by targeted ablation of adipose
Cancer Cell, 2003, 4, 383–391. tissue, Nat. Med., 2004, 10, 625–632.
284 M. E. Åkerman, W. C. Chan, P. Laakkonen, S. N. Bhatia 302 A. Azhdarinia, et al., A peptide probe for targeted brown
and E. Ruoslahti, Nanocrystal targeting in vivo, Proc. Natl. adipose tissue imaging, Nat. Commun., 2013, 4, 2472.
Acad. Sci. U. S. A., 2002, 99, 12617–12621. 303 M. N. Hossen, K. Kajimoto, H. Akita, M. Hyodo and
285 D. Simberg, et al., Biomimetic amplification of nanoparti- H. Harashima, A comparative study between nanoparticle-
cle homing to tumors, Proc. Natl. Acad. Sci. U. S. A., 2007, targeted therapeutics and bioconjugates as obesity medica-
104, 932–936. tion, J. Controlled Release, 2013, 171, 104–112.
286 H. F. Dvorak, D. R. Senger, A. M. Dvorak, V. S. Harvey and 304 Y. Xue, X. Xu, X.-Q. Zhang, O. C. Farokhzad and R. Langer,
J. McDonagh, Regulation of extravascular coagulation by Preventing diet-induced obesity in mice by adipose tissue
microvascular permeability, Science, 1985, 227, 1059–1061. transformation and angiogenesis using targeted nano-
287 W. J. M. Mulder, et al., Quantum dots with a paramagnetic particles, Proc. Natl. Acad. Sci. U. S. A., 2016, 113, 5552–5557.
coating as a bimodal molecular imaging probe, Nano Lett., 305 T. Dvir, et al., Nanoparticles Targeting the Infarcted Heart,
2006, 6, 1–6. Nano Lett., 2011, 11, 4411–4414.
288 S. Shen, et al., Targeting mesoporous silica-encapsulated 306 M. M. Nguyen, et al., Enzyme-Responsive Nanoparticles for
gold nanorods for chemo-photothermal therapy with near- Targeted Accumulation and Prolonged Retention in Heart
infrared radiation, Biomaterials, 2013, 34, 3150–3158. Tissue after Myocardial Infarction, Adv. Mater., 2015, 27,
289 G. Colombo, et al., Structure-activity relationships of linear 5547–5552.
and cyclic peptides containing the NGR tumor-homing 307 A. P. Mann, et al., A peptide for targeted, systemic delivery
motif, J. Biol. Chem., 2002, 277, 47891–47897. of imaging and therapeutic compounds into acute brain
290 M. Gupta, et al., Dual targeted polymeric nanoparticles injuries, Nat. Commun., 2016, 7, 11980.
based on tumor endothelium and tumor cells for 308 L. Agemy, et al., Targeted nanoparticle enhanced proapop-
enhanced antitumor drug delivery, Mol. Pharmaceutics, totic peptide as potential therapy for glioblastoma, Proc.
2014, 11, 697–715. Natl. Acad. Sci. U. S. A., 2014, 111, 11906.
291 S. Kunjachan, et al., Passive versus active tumor targeting 309 A. Hayashi, et al., A strategy for efficient cross-presentation
using RGD- and NGR-modified polymeric nanomedicines, of CTL-epitope peptides leading to enhanced induction of
Nano Lett., 2014, 14, 972–981. in vivo tumor immunity, J. Controlled Release, 2007, 117,
292 F. Curnis, et al., NGR-tagged nano-gold: A new CD13- 11–19.
selective carrier for cytokine delivery to tumors, Nano 310 K. Matsuo, et al., Efficient generation of antigen-specific cellular
Res., 2016, 9, 1393–1408. immunity by vaccination with poly(g-glutamic acid) nano-
293 X. Wang, B. Wang and Q. Zhang, Anti-tumor targeted drug particles entrapping endoplasmic reticulum-targeted peptides,
delivery systems mediated by aminopeptidase N/CD13, Biochem. Biophys. Res. Commun., 2007, 362, 1069–1072.
Acta Pharm. Sin. B, 2011, 1, 80–83. 311 B. Alberts, et al., Molecular biology of the cell, Garland
294 S.-M. Lee, et al., Targeting Bladder Tumor Cells In vivo and Science, 2008.
in the Urine with a Peptide Identified by Phage Display, 312 P. Ponka and C. N. Lok, The transferrin receptor: role in
Mol. Cancer Res., 2007, 5, 11–19. health and disease, Int. J. Biochem. Cell Biol., 1999, 31,
295 Y. Wei, et al., Polydopamine and peptide decorated 1111–1137.
doxorubicin-loaded mesoporous silica nanoparticles as a 313 T. R. Daniels, et al., The transferrin receptor and the
targeted drug delivery system for bladder cancer therapy, targeted delivery of therapeutic agents against cancer,
Drug Delivery, 2017, 24, 681–691. Biochim. Biophys. Acta, 2012, 291–317.
296 L. Geng, et al., HER2 targeting peptides screening 314 D. W. Bartlett, H. Su, I. J. Hildebrandt, W. A. Weber and
and applications in tumor imaging and drug delivery, M. E. Davis, Impact of tumor-specific targeting on the
Theranostics, 2016, 6, 1261–1273. biodistribution and efficacy of siRNA nanoparticles
3612 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
measured by multimodality in vivo imaging, Proc. Natl. 331 N. Surti and A. Misra, Wheat germ agglutinin-conjugated
Acad. Sci. U. S. A., 2007, 104, 15549–15554. nanoparticles for sustained cellular and lung delivery of
315 C. H. J. Choi, C. A. Alabi, P. Webster and M. E. Davis, budesonide, Drug Delivery, 2008, 15, 81–86.
Mechanism of active targeting in solid tumors with 332 X. Gao, et al., Brain delivery of vasoactive intestinal peptide
transferrin-containing gold nanoparticles, Proc. Natl. Acad. enhanced with the nanoparticles conjugated with wheat
Sci. U. S. A., 2010, 107, 1235–1240. germ agglutinin following intranasal administration,
316 J. Wang, S. Tian, R. A. Petros, M. E. Napier and J. M. J. Controlled Release, 2007, 121, 156–167.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
DeSimone, The Complex Role of Multivalency in Nano- 333 Q. Liu, et al., Nose-to-brain transport pathways of wheat
particles Targeting the Transferrin Receptor for Cancer germ agglutinin conjugated PEG-PLA nanoparticles,
Therapies, J. Am. Chem. Soc., 2010, 132, 11306–11313. Pharm. Res., 2012, 29, 546–558.
317 A. Salvati, et al., Transferrin-functionalized nanoparticles 334 Y. Zhang, et al., Transporter protein and drug-conjugated
lose their targeting capabilities when a biomolecule corona gold nanoparticles capable of bypassing the blood-brain
adsorbs on the surface, Nat. Nanotechnol., 2013, 8, 137–143. barrier, Sci. Rep., 2016, 6, 25794.
318 J. Y. Yhee, et al., Tumor-targeting transferrin nanoparticles 335 N. Foster, M. A. Clark, M. A. Jepson and B. H. Hirst, Ulex
for systemic polymerized siRNA delivery in tumor-bearing europaeus 1 lectin targets microspheres to mouse Peyer’s
mice, Bioconjugate Chem., 2013, 24, 1850–1860. patch M-cells in vivo, Vaccine, 1998, 16, 536–541.
319 J. C. Kraft, J. P. Freeling, Z. Wang and R. J. Y. Ho, Emerging 336 P. N. Gupta, K. Khatri, A. K. Goyal, N. Mishra and S. P. Vyas,
Research and Clinical Development Trends of Liposome M-cell targeted biodegradable PLGA nanoparticles for oral
and Lipid Nanoparticle Drug Delivery Systems, J. Pharm. immunization against hepatitis B, J. Drug Targeting, 2007,
Sci., 2014, 103, 29–52. 15, 701–713.
320 K. Liu, et al., Self-assembled targeted nanoparticles based 337 B. Malik, et al., Microfold-cell targeted surface engineered
on transferrin-modified eight-arm-polyethylene glycol– polymeric nanoparticles for oral immunization, J. Drug
dihydroartemisinin conjugate, Sci. Rep., 2016, 6, 29461. Targeting, 2012, 20, 76–84.
321 K. Wang, et al., Self-assembled IR780-loaded transferrin 338 K. Misstear, et al., Targeted nasal vaccination provides
nanoparticles as an imaging, targeting and PDT/PTT agent antibody-independent protection against Staphylococcus
for cancer therapy, Sci. Rep., 2016, 6, 27421. aureus, J. Infect. Dis., 2014, 209, 1479–1484.
322 A. M. Master and A. Sen Gupta, EGF receptor-targeted 339 P. Sapra and T. M. Allen, Ligand-targeted liposomal anti-
nanocarriers for enhanced cancer treatment, Nanomedicine, cancer drugs, Prog. Lipid Res., 2003, 42, 439–462.
2012, 7, 1895–1906. 340 M. M. Cardoso, I. N. Peca and A. C. A. Roque, Antibody-
323 C.-L. Tseng, et al., Targeting efficiency and biodistribution Conjugated Nanoparticles for Therapeutic Applications,
of biotinylated-EGF-conjugated gelatin nanoparticles Curr. Med. Chem., 2012, 19, 3103–3127.
administered via aerosol delivery in nude mice with lung 341 V. H. Shargh, H. Hondermarck and M. Liang, Antibody-
cancer, Biomaterials, 2008, 29, 3014–3022. targeted biodegradable nanoparticles for cancer therapy,
324 T. Shimada, et al., Development of targeted therapy with Nanomedicine, 2016, 11, 63–79.
paclitaxel incorporated into EGF-conjugated nanoparticles, 342 D. A. Richards, A. Maruani and V. Chudasama, Antibody
Anticancer Res., 2009, 29, 1009–1014. fragments as nanoparticle targeting ligands: a step in the
325 H. Jin, et al., Investigating the specific uptake of EGF- right direction, Chem. Sci., 2017, 8, 63–77.
conjugated nanoparticles in lung cancer cells using 343 F. Mack, M. Ritchie and P. Sapra, The next generation of
fluorescence imaging, Cancer Nanotechnol., 2010, 1, 71–78. antibody drug conjugates, Semin. Oncol., 2014, 41, 637–652.
326 R. Matsumoto, R. Hara, T. Andou, M. Mie and E. Kobatake, 344 A. Thomas, B. A. Teicher and R. Hassan, Antibody–drug
Targeting of EGF-displayed protein nanoparticles with conjugates for cancer therapy, Lancet Oncol., 2016, 17,
anticancer drugs, J. Biomed. Mater. Res., Part B, 2014, e254–e262.
102, 1792–1798. 345 R. Huang, M. M. Kiss, M. Batonick, M. P. Weiner and B. K.
327 Y. Assal, Y. Mizuguchi, M. Mie and E. Kobatake, Growth Kay, Generating Recombinant Antibodies to Membrane
Factor Tethering to Protein Nanoparticles via Coiled-Coil Proteins through Phage Display, Antibodies, 2016, 5, 11.
Formation for Targeted Drug Delivery, Bioconjugate Chem., 346 J. M. Reichert, Antibodies to watch in 2017, mAbs, 2017, 9,
2015, 26, 1672–1677. 167–181.
328 X. Chen, et al., Inflamed leukocyte-mimetic nanoparticles 347 H. R. Hoogenboom, Selecting and screening recombinant
for molecular imaging of inflammation, Biomaterials, antibody libraries, Nat. Biotechnol., 2005, 23, 1105–1116.
2011, 32, 7651–7661. 348 J. K. H. Liu, The history of monoclonal antibody develop-
329 S. Yang, et al., Biomimetic, synthetic HDL nanostructures for ment - Progress, remaining challenges and future innova-
lymphoma, Proc. Natl. Acad. Sci. U. S. A., 2013, 110, 2511–2516. tions, Ann. Med. Surg., 2014, 3, 113–116.
330 H. Lu, H. Zhang, D. Zhang, H. Lu and D. Ma, A Biocompa- 349 C. F. C. Fernandes, et al., Camelid single-domain anti-
tible Reconstituted High-Density Lipoprotein Nano-System bodies as an alternative to overcome challenges related to
as a Probe for Lung Cancer Detection, Med. Sci. Monit., the prevention, detection, and control of neglected tropical
2015, 21, 2726–2733. diseases, Front. Immunol., 2017, 8, 653.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3613
View Article Online
350 U. Krishnamurti and J. F. Silverman, HER2 in Breast Cancer: 367 D. E. Lopes de Menezes, M. J. Kirchmeier, J.-F. Gagne,
A Review and Update, Adv. Anat. Pathol., 2014, 21, 100–107. L. M. Pilarski and T. M. Allen, Cellular Trafficking and
351 J. W. Park, et al., Anti-HER2 immunoliposomes for targeted Cytotoxicity of Anti-Cd19- Targeted Liposomal Doxorubicin
therapy of human tumors, Cancer Lett., 1997, 118, 153–160. in B Lymphoma Cells, J. Liposome Res., 1999, 9, 199–228.
352 J. W. Park, et al., Tumor targeting using anti-her2 immuno- 368 J. Guo, et al., Image-guided and tumor-targeted drug delivery
liposomes, J. Controlled Release, 2001, 74, 95–113. with radiolabeled unimolecular micelles, Biomaterials, 2013,
353 J. W. Park, et al., Anti-HER2 Immunoliposomes: Enhanced 34, 8323–8332.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
Efficacy Attributable to Targeted Delivery, Clin. Cancer Res., 369 F. Chen, et al., In vivo tumor targeting and image-guided
2002, 8, 1172–1181. drug delivery with antibody-conjugated, radiolabeled meso-
354 T. Kikumori, T. Kobayashi, M. Sawaki and T. Imai, Anti- porous silica nanoparticles, ACS Nano, 2013, 7, 9027–9039.
cancer effect of hyperthermia on breast cancer by magnetite 370 L. Karmani, et al., 89Zr-labeled anti-endoglin antibody-
nanoparticle-loaded anti-HER2 immunoliposomes, Breast targeted gold nanoparticles for imaging cancer: implications
Cancer Res. Treat., 2009, 113, 435–441. for future cancer therapy, Nanomedicine, 2014, 9, 1923–1937.
355 F. Corsi, et al., HER2 expression in breast cancer cells is 371 T. A. Elbayoumi and V. P. Torchilin, Tumor-Specific
downregulated upon active targeting by antibody-engineered Antibody-Mediated Targeted Delivery of Doxils Reduces
multifunctional nanoparticles in mice, ACS Nano, 2011, 5, the Manifestation of Auricular Erythema Side Effect in
6383–6393. Mice, Int. J. Pharm., 2008, 357, 272–279.
356 F. M. Kievit, et al., Targeting of primary breast cancers and 372 F. Fay, et al., Conatumumab (AMG 655) coated nano-
metastases in a transgenic mouse model using rationally particles for targeted pro-apoptotic drug delivery,
designed multifunctional SPIONs, ACS Nano, 2012, 6, Biomaterials, 2011, 32, 8645–8653.
2591–2601. 373 G. Morral-Ruı́z, P. Melgar-Lesmes, C. Solans and M. J.
357 L. Fiandra, et al., Assessing the in vivo targeting efficiency Garcı́a-Celma, Multifunctional polyurethane-urea nano-
of multifunctional nanoconstructs bearing antibody- particles to target and arrest inflamed vascular environ-
derived ligands, ACS Nano, 2013, 7, 6092–6102. ment: A potential tool for cancer therapy and diagnosis,
358 M. Jang, et al., Trastuzumab-conjugated liposome-coated J. Controlled Release, 2013, 171, 163–171.
fluorescent magnetic nanoparticles to target breast cancer, 374 L. M. Herda, D. R. Hristov, M. Lo Giudice, E. Polo and
Korean J. Radiol., 2014, 15, 411–422. K. A. Dawson, Mapping of molecular structure of the
359 C. NDong, et al., Tumor cell targeting by iron oxide nanoscale surface in bionanoparticles, J. Am. Chem. Soc.,
nanoparticles is dominated by different factors in vitro 2017, 139, 111–114.
versus in vivo, PLoS One, 2015, 10, e0115636. 375 C. Parolo, et al., Design, preparation, and evaluation of a
360 J. Hamzehalipour Almaki, et al., Trastuzumab-decorated fixed-orientation antibody/gold-nanoparticle conjugate as
nanoparticles for in vitro and in vivo tumor-targeting an immunosensing label, ACS Appl. Mater. Interfaces, 2013,
hyperthermia of HER2+ breast cancer, J. Mater. Chem. B, 5, 10753–10759.
2017, 5, 7369–7383. 376 M. K. Greene, et al., Generating Next-Generation Antibody-
361 C. Mamot, et al., Epidermal growth factor receptor- Nanoparticle Conjugates through the Oriented Installation
targeted immunoliposomes significantly enhance the effi- of Non-Engineered Antibody Fragments, Chem. Sci., 2018,
cacy of multiple anticancer drugs in vivo, Cancer Res., 2005, 9, 79–87.
65, 11631–11638. 377 W. Jiang, B. Y. S. Kim, J. T. Rutka and W. C. W. Chan,
362 C. Mamot, et al., Tolerability, safety, pharmacokinetics, Nanoparticle-mediated cellular response is size-dependent,
and efficacy of doxorubicin-loaded anti-EGFR immuno- Nat. Nanotechnol., 2008, 3, 145–150.
liposomes in advanced solid tumours: A phase 1 dose- 378 H. Han and M. E. Davis, Single-antibody, targeted nano-
escalation study, Lancet Oncol., 2012, 13, 1234–1241. particle delivery of camptothecin, Mol. Pharmaceutics,
363 R. Marega, et al., Antibody-functionalized polymer-coated 2013, 10, 2558–2567.
gold nanoparticles targeting cancer cells: an in vitro and 379 M. Colombo, et al., Tumour homing and therapeutic effect
in vivo study, J. Mater. Chem., 2012, 22, 21305–21312. of colloidal nanoparticles depend on the number of
364 R. R. Sawant, A. M. Jhaveri, A. Koshkaryev, F. Qureshi and attached antibodies, Nat. Commun., 2016, 7, 13818.
V. P. Torchilin, The effect of dual ligand-targeted micelles 380 A. L. Schwartz, Receptor cell biology: receptor-mediated
on the delivery and efficacy of poorly soluble drug for endocytosis, Pediatr. Res., 1995, 38, 835–843.
cancer therapy, J. Drug Targeting, 2013, 21, 630–638. 381 D. K. Cureton, C. E. Harbison, E. Cocucci, C. R. Parrish and
365 N. Senzer, et al., Phase I study of a systemically delivered T. Kirchhausen, Limited Transferrin Receptor Clustering
p53 nanoparticle in advanced solid tumors, Mol. Ther., Allows Rapid Diffusion of Canine Parvovirus into Clathrin
2013, 21, 1096–1103. Endocytic Structures, J. Virol., 2012, 86, 5330–5340.
366 D. E. Lopes de Menezes, L. M. Pilarski and T. M. Allen, 382 L. Zhao, et al., Nanoparticle vaccines, Vaccine, 2014, 32, 327–337.
In vitro and in vivo targeting of immunoliposomal 383 J. D. Smith, L. D. Morton and B. D. Ulery, Nanoparticles
doxorubicin to human B-cell lymphoma, Cancer Res., as synthetic vaccines, Curr. Opin. Biotechnol., 2015, 34,
1998, 58, 3320–3330. 217–224.
3614 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
384 M. Luo, L. Z. Samandi, Z. Wang, Z. J. Chen and J. Gao, 401 W. Tao, K. S. Ziemer and H. S. Gill, Gold nanoparticle–M2e
Synthetic nanovaccines for immunotherapy, J. Controlled conjugate coformulated with CpG induces protective
Release, 2017, 263, 200–210. immunity against influenza A virus, Nanomedicine, 2014,
385 L. J. Cruz, et al., Controlled release of antigen and Toll-like 9, 237–251.
receptor ligands from PLGA nanoparticles enhances 402 D. Safari, et al., Gold nanoparticles as carriers for a
immunogenicity, Nanomedicine, 2017, 12, 491–510. synthetic Streptococcus pneumoniae type 14 conjugate
386 S. T. Reddy, et al., Exploiting lymphatic transport and vaccine, Nanomedicine, 2012, 7, 651–662.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
complement activation in nanoparticle vaccines, Nat. 403 J. W. Stone, et al., Gold nanorod vaccine for respiratory
Biotechnol., 2007, 25, 1159–1164. syncytial virus, Nanotechnology, 2013, 24, 295102.
387 K. T. Gause, et al., Immunological Principles Guiding the 404 Z. Zhao, T. Wakita and K. Yasui, Inoculation of plasmids
Rational Design of Particles for Vaccine Delivery, ACS encoding Japanese encephalitis virus PrM-E proteins with
Nano, 2017, 11, 54–68. colloidal gold elicits a protective immune response in
388 K. Lee and Y. Yu, Janus nanoparticles for T cell activation: BALB/c mice, J. Virol., 2003, 77, 4248–4260.
clustering ligands to enhance stimulation, J. Mater. Chem. 405 P. Di Gianvincenzo, et al., Negatively charged glyconano-
B, 2017, 5, 4410–4415. particles modulate and stabilize the secondary structures
389 B. Chen, et al., Janus Particles as Artificial Antigen- of a gp120 V3 loop peptide: Toward fully synthetic HIV
Presenting Cells for T Cell Activation, ACS Appl. Mater. vaccine candidates, Bioconjugate Chem., 2015, 26, 755–765.
Interfaces, 2014, 6, 18435–18439. 406 M. Lamberti, et al., Carbon nanotubes: Properties, biome-
390 H. De La Peña, et al., Artificial exosomes as tools for basic dical applications, advantages and risks in patients and
and clinical immunology, J. Immunol. Methods, 2009, 344, occupationally-exposed workers, Int. J. Immunopathol.
121–132. Pharmacol., 2015, 28, 4–13.
391 F. Giannoni, et al., Clustering of T Cell Ligands on 407 D. Pantarotto, et al., Synthesis, structural characterization,
Artificial APC Membranes Influences T Cell Activation and immunological properties of carbon nanotubes
and Protein Kinase C y Translocation to the T Cell Plasma functionalized with peptides, J. Am. Chem. Soc., 2003,
Membrane, J. Immunol., 2005, 174, 3204–3211. 125, 6160–6164.
392 S. A. C. Carabineiro, Applications of gold nanoparticles in 408 J. Meng, et al., Carbon nanotubes conjugated to tumor lysate
nanomedicine: Recent advances in vaccines, Molecules, protein enhance the efficacy of an antitumor immuno-
2017, 22, 857. therapy, Small, 2008, 4, 1364–1370.
393 K. Niikura, et al., Gold nanoparticles as a vaccine platform: 409 N. Yandar, et al., Immunological profile of a Plasmodium
Influence of size and shape on immunological responses vivax AMA-1 N-terminus peptide-carbon nanotube conju-
in vitro and in vivo, ACS Nano, 2013, 7, 3926–3938. gate in an infected Plasmodium berghei mouse model,
394 L. A. Dykman and N. G. Khlebtsov, Immunological proper- Vaccine, 2008, 26, 5864–5873.
ties of gold nanoparticles, Chem. Sci., 2017, 8, 1719–1735. 410 M. Zeinali, M. Jammalan, S. K. Ardestani and N. Mosaveri,
395 R. Kumar, et al., Nanovaccines for malaria using Plasmo- Immunological and cytotoxicological characterization of tuber-
dium falciparum antigen Pfs25 attached gold nano- culin purified protein derivative (PPD) conjugated to single-
particles, Vaccine, 2015, 33, 5064–5071. walled carbon nanotubes, Immunol. Lett., 2009, 126, 48–53.
396 Y.-S. Chen, Y.-C. Hung, W.-H. Lin and G. S. Huang, Assess- 411 C. H. Villa, et al., Single-walled carbon nanotubes deliver
ment of gold nanoparticles as a size-dependent vaccine peptide antigen into dendritic cells and enhance IgG
carrier for enhancing the antibody response against synthetic responses to tumor-associated antigens, ACS Nano, 2011,
foot-and-mouth disease virus peptide, Nanotechnology, 2010, 5, 5300–5311.
21, 195101. 412 D. Pantarotto, et al., Immunization with Peptide-Functionalized
397 L. A. Dykman, et al., Use of a synthetic foot-and-mouth Carbon nanotubes Enhances Virus-Specific Neutralizing Anti-
disease virus peptide conjugated to gold nanoparticles for body Response, Chem. Biol., 2003, 10, 961–966.
enhancing immunological response, Gold Bull., 2015, 48, 413 M. Kalkanidis, et al., Methods for nano-particle based
93–101. vaccine formulation and evaluation of their immunogeni-
398 A. E. Gregory, et al., Conjugation of Y. pestis F1-antigen to city, Methods, 2006, 40, 20–29.
gold nanoparticles improves immunogenicity, Vaccine, 414 D. L. V. Greenwood, et al., Vaccination against foot-and-
2012, 30, 6777–6782. mouth disease virus using peptides conjugated to nano-
399 T. Mocan, et al., In vitro administration of gold nanoparticles beads, Vaccine, 2008, 26, 2706–2713.
functionalized with MUC-1 protein fragment generates anti- 415 M. Skwarczynski, et al., Polyacrylate dendrimer nano-
cancer vaccine response via macrophage activation and particles: A self-adjuvanting vaccine delivery system,
polarization mechanism, J. Cancer, 2015, 6, 583–592. Angew. Chem., Int. Ed., 2010, 49, 5742–5745.
400 H. Cai, et al., Glycopeptide-functionalized gold nano- 416 D. Chiu, et al., Biomineralization and size control of stable
particles for antibody induction against the tumor asso- calcium phosphate core-protein shell nanoparticles:
ciated mucin-1 glycoprotein, Bioorg. Med. Chem., 2016, 24, Potential for vaccine applications, Bioconjugate Chem.,
1132–1135. 2012, 23, 610–617.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3615
View Article Online
417 W. Zhou, A. O. Moguche, D. Chiu, K. Murali-Krishna and 434 A. J. van Hell, et al., Self-Assembly of Recombinant Amphi-
F. Baneyx, Just-in-time vaccines: Biomineralized calcium philic Oligopeptides into Vesicles, Biomacromolecules,
phosphate core-immunogen shell nanoparticles induce 2007, 8, 2753–2761.
long-lasting CD8+ T cell responses in mice, Nanomedicine, 435 T. Shimada, S. Lee, F. S. Bates, A. Hotta and M. Tirrell,
2014, 10, 571–578. Wormlike Micelle Formation in Peptide-Lipid Conjugates
418 R. A. Schwendener, Liposomes as vaccine delivery systems: Driven by Secondary Structure Transformation of the
a review of the recent advances, Ther. Adv. Vaccines, 2014, Headgroups, J. Phys. Chem. B, 2009, 113, 13711–13714.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
3616 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
cancer cell imaging and real-time monitoring of drug between sequence and structure, J. Struct. Biol., 2008,
release, Nat. Nanotechnol., 2016, 11, 388–394. 163, 258–269.
452 S. Vauthey, S. Santoso, H. Gong, N. Watson and S. Zhang, 469 D. N. Woolfson, Building fibrous biomaterials from alpha-
Molecular self-assembly of surfactant-like peptides to form helical and collagen-like coiled-coil peptides, Biopolymers,
nanotubes and nonovesicles, Proc. Natl. Acad. Sci. U. S. A., 2010, 94, 118–127.
2002, 99, 5355–5360. 470 B. Apostolovic, M. Danial and H.-A. Klok, Coiled coils:
453 S. Santoso, W. Hwang, H. Hartman and S. Zhang, Self- attractive protein folding motifs for the fabrication of self-
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
assembly of Surfactant-like Peptides with Variable Glycine assembled, responsive and bioactive materials, Chem. Soc.
Tails to Form Nanotubes and Nanovesicles, Nano Lett., Rev., 2010, 39, 3541.
2002, 2, 687–691. 471 M. M. Stevens, N. T. Flynn, C. Wang, D. A. Tirrell and
454 M. Rad-Malekshahi, et al., The Supramolecular Organiza- R. Langer, Coiled-coil peptide-based assembly of gold
tion of a Peptide-Based Nanocarrier at High Molecular nanoparticles, Adv. Mater., 2004, 16, 915–918.
Detail, J. Am. Chem. Soc., 2015, 137, 7775–7784. 472 V. N. Malashkevich, R. A. Kammerer, V. P. Efimov,
455 S. Gudlur, et al., Peptide Nanovesicles Formed by the Self- T. Schulthess and J. Engel, The Crystal Structure of a
Assembly of Branched Amphiphilic Peptides, PLoS One, Five-Stranded Coiled Coil in COMP: A Prototype Ion Chan-
2012, 7, e45374. nel?, Science, 1996, 274, 761–765.
456 A. J. van Hell, D. J. A. Crommelin, W. E. Hennink and 473 S. Raman, G. Machaidze, A. Lustig, U. Aebi and
E. Mastrobattista, Stabilization of peptide vesicles by intro- P. Burkhard, Structure-based design of peptides that self-
ducing inter-peptide disulfide bonds, Pharm. Res., 2009, assemble into regular polyhedral nanoparticles, Nanome-
26, 2186–2193. dicine, 2006, 2, 95–102.
457 A. J. van Hell, M. M. Fretz, D. J. A. Crommelin, W. E. 474 T. A. P. F. Pimentel, et al., Peptide nanoparticles as novel
Hennink and E. Mastrobattista, Peptide nanocarriers for immunogens: Design and analysis of a prototypic severe
intracellular delivery of photosensitizers, J. Controlled acute respiratory syndrome vaccine, Chem. Biol. Drug Des.,
Release, 2010, 141, 347–353. 2009, 73, 53–61.
458 H. Tanisaka, et al., Near-infrared fluorescent labeled pep- 475 S. A. Kaba, et al., A nonadjuvanted polypeptide nanoparti-
tosome for application to cancer imaging, Bioconjugate cle vaccine confers long-lasting protection against rodent
Chem., 2008, 19, 109–117. malaria, J. Immunol., 2009, 183, 7268–7277.
459 I. W. Hamley, Lipopeptides: from self-assembly to bioac- 476 N. Wahome, et al., Conformation-specific Display of 4E10
tivity, Chem. Commun., 2015, 51, 8574–8583. and 2F5 Epitopes on Self-assembling Protein Nano-
460 J. A. Hutchinson, S. Burholt and I. W. Hamley, Peptide particles as a Potential HIV Vaccine, Chem. Biol. Drug
hormones and lipopeptides: from self-assembly to thera- Des., 2012, 80, 349–357.
peutic applications, J. Pept. Sci., 2017, 23, 82–94. 477 F. Boato, et al., Synthetic Virus-Like Particles from Self-
461 S. Lv, et al., Doxorubicin-loaded amphiphilic polypeptide- Assembling Coiled-Coil Lipopeptides and Their Use in
based nanoparticles as an efficient drug delivery system for Antigen Display to the Immune System, Angew. Chem.,
cancer therapy, Acta Biomater., 2013, 9, 9330–9342. Int. Ed., 2007, 46, 9015–9018.
462 X. Xu, H. Yuan, J. Chang, B. He and Z. Gu, Cooperative 478 R. Sharma, A. Ghasparian, J. A. Robinson and K. C.
hierarchical self-assembly of peptide dendrimers and Mccullough, Synthetic Virus-Like Particles Target Dendritic
linear polypeptides into nanoarchitectures mimicking Cell Lipid Rafts for Rapid Endocytosis Primarily but Not
viral capsids, Angew. Chem., Int. Ed., 2012, 51, 3130–3133. Exclusively by Macropinocytosis, PLoS One, 2012, 7, e43248.
463 X. Xu, et al., Smart Nanovehicles based on ph-triggered 479 J. S. Rudra, Y. F. Tian, J. P. Jung and J. H. Collier, A self-
disassembly of supramolecular peptide-amphiphiles for effi- assembling peptide acting as an immune adjuvant, Proc.
cient intracellular drug delivery, Small, 2014, 10, 1133–1140. Natl. Acad. Sci. U. S. A., 2010, 107, 622–627.
464 Y. Li, et al., Capsid-like supramolecular dendritic systems 480 C. V. Pecot, G. A. Calin, R. L. Coleman, G. Lopez-Berestein
as pH-responsive nanocarriers for drug penetration and and A. K. Sood, RNA interference in the clinic: challenges
site-specific delivery, Nanomedicine, 2016, 12, 355–364. and future directions, Nat. Rev. Cancer, 2011, 11, 59–67.
465 Z. Zhang, et al., Virus-Inspired Mimics Based on Dendritic 481 J. Burnett and J. Rossi, RNA-based therapeutics: current
Lipopeptides for Efficient Tumor-Specific Infection and Sys- progress and future prospects, Chem. Biol., 2012, 19,
temic Drug Delivery, Adv. Funct. Mater., 2015, 25, 5250–5260. 60–71.
466 Y. Wu and J. H. Collier, a-Helical coiled-coil peptide 482 L. W. Seymour and A. J. Thrasher, Gene therapy matures in
materials for biomedical applications, Wiley Interdiscip. the clinic, Nat. Biotechnol., 2012, 30, 588–593.
Rev.: Nanomed. Nanobiotechnol., 2017, 9, 1–17. 483 C. Chakraborty, A. R. Sharma, G. Sharma, C. G. P. Doss and
467 P. Burkhard, M. Meier and A. Lustig, Design of a minimal S.-S. Lee, Therapeutic miRNA and siRNA: Moving from
protein oligomerization domain by a structural approach, Bench to Clinic as Next Generation Medicine, Mol.
Protein Sci., 2000, 9, 2294–2301. Ther.–Nucleic Acids, 2017, 15, 132–143.
468 D. A. D. Parry, R. D. B. Fraser and J. M. Squire, Fifty years of 484 M. E. Martin and K. G. Rice, Peptide-guided Gene Delivery,
coiled-coils and a-helical bundles: A close relationship AAPS J., 2007, 9, 18–29.
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3617
View Article Online
485 G. Y. Wu and C. H. Wu, Receptor-mediated in vitro gene 501 A. Chilkoti, M. R. Dreher and D. E. Meyer, Design of thermally
transformation by a soluble DNA carrier system, J. Biol. responsive, recombinant polypeptide carriers for targeted
Chem., 1987, 262, 4429–4432. drug delivery, Adv. Drug Delivery Rev., 2002, 54, 1093–1111.
486 G. Y. Wu and C. H. Wu, Evidence for Targeted Gene 502 M. R. Dreher, et al., Evaluation of an elastin-like polypeptide-
Delivery to Hep G2 Hepatoma Cells in Vitro, Biochemistry, doxorubicin conjugate for cancer therapy, J. Controlled
1988, 27, 887–892. Release, 2003, 91, 31–43.
487 R. C. Adami and K. G. Rice, Metabolic stability of glutar- 503 M. R. Dreher, et al., Temperature triggered self-assembly of
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
aldehyde cross-linked peptide DNA condensates, J. Pharm. polypeptides into multivalent spherical micelles, J. Am.
Sci., 1999, 88, 739–746. Chem. Soc., 2008, 130, 687–694.
488 D. L. McKenzie, K. Y. Kwok and K. G. Rice, A potent new 504 J. A. Mackay, et al., Self-assembling chimeric polypeptide-
class of reductively activated peptide gene delivery agents, doxorubicin conjugate nanoparticles that abolish tumours
J. Biol. Chem., 2000, 275, 9970–9977. after a single injection, Nat. Mater., 2009, 8, 993–999.
489 Tarwadi, J. Jazayeri, R. J. Prankerd and C. W. Pouton, 505 J. R. Mcdaniel, S. R. Macewan, M. Dewhirst and A. Chilkoti,
Preparation and in vitro evaluation of novel lipopeptide Doxorubicin-conjugated chimeric polypeptide nanoparticles
transfection agents for efficient gene delivery, Bioconjugate that respond to mild hyperthermia, J. Controlled Release,
Chem., 2008, 19, 940–950. 2012, 159, 362–367.
490 K. Rittner, et al., New Basic Membrane-Destabilizing Pep- 506 W. Kim, J. Thévenot, E. Ibarboure, S. Lecommandoux and
tides for Plasmid-Based Gene Delivery in Vitro and in Vivo, E. L. Chaikof, Self-assembly of thermally responsive
Mol. Ther., 2002, 5, 104–114. amphiphilic diblock copolypeptides into spherical
491 L. Crombez, et al., Targeting cyclin B1 through peptide- micellar nanoparticles, Angew. Chem., Int. Ed., 2010, 49,
based delivery of siRNA prevents tumour growth, Nucleic 4257–4260.
Acids Res., 2009, 37, 4559–4569. 507 W. Kim, C. Brady and E. L. Chaikof, Amphiphilic protein
492 H. O. Mccarthy, et al., Development and characterization micelles for targeted in vivo imaging, Acta Biomater., 2012,
of self-assembling nanoparticles using a bio-inspired 8, 2476–2482.
amphipathic peptide for gene delivery, J. Controlled 508 S. M. Janib, et al., Kinetic quantification of protein polymer
Release, 2014, 189, 141–149. nanoparticles using non-invasive imaging, Integr. Biol.,
493 W. Y. Seow and Y. Y. Yang, A class of cationic triblock 2013, 5, 183–194.
amphiphilic oligopeptides as efficient gene-delivery vec- 509 C. Garcı́a-Arévalo, et al., Immunomodulatory nanoparticles
tors, Adv. Mater., 2009, 21, 86–90. from elastin-like recombinamers: Single-molecules for
494 K. Numata, M. R. Reagan, R. H. Goldstein, M. Rosenblatt and tuberculosis vaccine development, Mol. Pharmaceutics,
D. L. Kaplan, Spider silk-based gene carriers for tumor cell- 2013, 10, 586–597.
specific delivery, Bioconjugate Chem., 2011, 22, 1605–1610. 510 A. J. Simnick, et al., In vivo tumor targeting by a NGR-
495 H. Wang, et al., Biocompatible, chimeric peptide- decorated micelle of a recombinant diblock copolypeptide,
condensed supramolecular nanoparticles for tumor cell- J. Controlled Release, 2011, 155, 144–151.
specific siRNA delivery and gene silencing, Chem. Com- 511 L. Walker, E. Perkins, F. Kratz and D. Raucher, Cell
mun., 2014, 50, 7806–7809. penetrating peptides fused to a thermally targeted bio-
496 J. R. McDaniel, D. J. Callahan and A. Chilkoti, Drug polymer drug carrier improve the delivery and antitumor
delivery to solid tumors by elastin-like polypeptides, Adv. efficacy of an acid-sensitive doxorubicin derivative, Int.
Drug Delivery Rev., 2010, 62, 1456–1467. J. Pharm., 2012, 436, 825–832.
497 F. C. M. Smits, B. C. Buddingh, M. B. Van Eldijk and 512 S. Moktan, E. Perkins, F. Kratz and D. Raucher, Thermal
J. C. M. Van Hest, Elastin-Like Polypeptide Based Nano- Targeting of an Acid-Sensitive Doxorubicin Conjugate of
particles: Design Rationale Toward Nanomedicine, Macro- Elastin-like Polypeptide Enhances the Therapeutic Efficacy
mol. Biosci., 2015, 15, 36–51. Compared with the Parent Compound In Vivo, Mol. Cancer
498 D. E. Meyer, G. A. Kong, M. W. Dewhirst, M. R. Zalutsky Ther., 2012, 11, 1547–1557.
and A. Chilkoti, Targeting a Genetically Engineered 513 D. J. Callahan, et al., Triple Stimulus-Responsive Poly-
Elastin-like Polypeptide to Solid Tumors by Local peptide Nanoparticles That Enhance Intratumoral Spatial
Hyperthermia, Cancer Res., 2001, 61, 1548–1554. Distribution, Nano Lett., 2012, 12, 2165–2170.
499 D. E. Meyer, B. C. Shin, G. A. Kong, M. W. Dewhirst and 514 D. S. Horne, Casein structure, self-assembly and gelation,
A. Chilkoti, Drug targeting using thermally responsive Curr. Opin. Colloid Interface Sci., 2002, 7, 456–461.
polymers and local hyperthermia, J. Controlled Release, 515 M. Bachar, et al., Development and characterization of a
2001, 74, 213–224. novel drug nanocarrier for oral delivery, based on self-
500 D. Raucher and A. Chilkoti, Enhanced Uptake of a Ther- assembled
mally Responsive Polypeptide by Tumor Cells in Response b-casein micelles, J. Controlled Release, 2012, 160, 164–171.
to Its Hyperthermia-mediated Phase Transition Enhanced 516 M. Haham, et al., Stability and bioavailability of vitamin D
Uptake of a Thermally Responsive Polypeptide by Tumor nanoencapsulated in casein micelles, Food Funct., 2012, 3,
Cells in., Cancer Res., 2001, 61, 7163–7170. 737–744.
3618 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018
View Article Online
517 A. Shapira, Y. G. Assaraf and Y. D. Livney, Beta-casein synthetic urinary biomarkers of thrombosis, ACS Nano,
nanovehicles for oral delivery of chemotherapeutic drugs, 2013, 7, 9001–9009.
Nanomedicine, 2010, 6, 119–126. 534 A. D. Warren, G. A. Kwong, D. K. Wood, K. Y. Lin and
518 A. Shapira, I. Davidson, N. Avni, Y. G. Assaraf and S. N. Bhatia, Point-of-care diagnostics for noncommunicable
Y. D. Livney, b-Casein nanoparticle-based oral drug deliv- diseases using synthetic urinary biomarkers and paper micro-
ery system for potential treatment of gastric carcinoma: fluidics, Proc. Natl. Acad. Sci. U. S. A., 2014, 111, 3671–3676.
Stability, target-activated release and cytotoxicity, Eur. 535 J. S. Dudani, C. G. Buss, R. T. K. Akana, G. A. Kwong and
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
J. Pharm. Biopharm., 2012, 80, 298–305. S. N. Bhatia, Sustained-Release Synthetic Biomarkers for
519 M. Bar-Zeev, Y. G. Assaraf and Y. D. Livney, b-casein Monitoring Thrombosis and Inflammation Using Point-of-
nanovehicles for oral delivery of chemotherapeutic drug Care Compatible Readouts, Adv. Funct. Mater., 2016, 26,
combinations overcoming P-glycoprotein-mediated multi- 2919–2928.
drug resistance in human gastric cancer cells, Oncotarget, 536 J.-C. Leroux, Drug Delivery: Too Much Complexity, Not Enough
2016, 7, 23322–23334. Reproducibility?, Angew. Chem., Int. Ed., 2017, 56, 15170–15171.
520 X. Zhen, X. Wang, C. Xie, W. Wu and X. Jiang, Cellular 537 L. Di, Strategic Approaches to Optimizing Peptide ADME
uptake, antitumor response and tumor penetration of Properties, AAPS J., 2015, 17, 134–143.
cisplatin-loaded milk protein nanoparticles, Biomaterials, 538 D. Mathur, et al., PEPlife: A Repository of the Half-life of
2013, 34, 1372–1382. Peptides, Sci. Rep., 2016, 6, 36617.
521 C. N. Loynachan, et al., Platinum Nanocatalyst Amplifica- 539 K. Lang and J. W. Chin, Cellular incorporation of unna-
tion: Redefining the Gold Standard for Lateral Flow Immu- tural amino acids and bioorthogonal labeling of proteins,
noassays with Ultra-Broad Dynamic Range, ACS Nano, Chem. Rev., 2014, 114, 4764–4806.
2018, 12, 279–288. 540 A. Dumas, L. Lercher, C. D. Spicer and B. G. Davis, Design-
522 P. Brangel, et al., A Serological Point-of-Care Test for the ing logical reassignment - expanding the chemistry in
Detection of IgG Antibodies against Ebola Virus in Human biology, Chem. Sci., 2015, 6, 50–69.
Survivors, ACS Nano, 2018, 12, 63–73. 541 A. J. Keefe and S. Jiang, Poly(zwitterionic)protein conju-
523 M. Cordeiro, F. Ferreira Carlos, P. Pedrosa, A. Lopez and gates offer increased stability without sacrificing binding
P. Baptista, Gold Nanoparticles for Diagnostics: Advances affinity or bioactivity, Nat. Chem., 2011, 4, 59–63.
towards Points of Care, Diagnostics, 2016, 6, 43. 542 E. M. Pelegri-Oday, E.-W. Lin and H. D. Maynard, Ther-
524 D. Quesada-González and A. Merkoçi, Nanoparticle-based apeutic protein-polymer conjugates: Advancing beyond
lateral flow biosensors, Biosens. Bioelectron., 2015, 73, pegylation, J. Am. Chem. Soc., 2014, 136, 14323–14332.
47–63. 543 S. P. Brown and A. B. Smith, Peptide/protein stapling and
525 S. Vigneshvar, C. C. Sudhakumari, B. Senthilkumaran and unstapling: Introduction of s-tetrazine, photochemical
H. Prakash, Recent Advances in Biosensor Technology for release, and regeneration of the peptide/protein, J. Am.
Potential Applications – An Overview, Front. Bioeng. Bio- Chem. Soc., 2015, 137, 4034–4037.
technol., 2016, 4, 11. 544 D. P. Fairlie and A. Dantas de Araujo, Review stapling
526 Z. Farka, T. Juřı́k, D. Kovář, L. Trnková and P. Skládal, peptides using cysteine crosslinking, Biopolymers, 2016,
Nanoparticle-Based Immunochemical Biosensors and 106, 843–852.
Assays: Recent Advances and Challenges, Chem. Rev., 545 S. Ståhl, et al., Affibody Molecules in Biotechnological and
2017, 117, 9973–10042. Medical Applications, Trends Biotechnol., 2017, 35, 691–712.
527 B. A. Kairdolf, X. Qian and S. Nie, Bioconjugated nano- 546 C. Heinis and G. Winter, Encoded libraries of chemically
particles for biosensing, in vivo imaging, and medical modified peptides, Curr. Opin. Chem. Biol., 2015, 26, 89–98.
diagnostics, Anal. Chem., 2017, 89, 1015–1031. 547 C. Heinis, T. Rutherford, S. Freund and G. Winter, Phage-
528 M. A. Eckert, et al., Novel molecular and nanosensors for encoded combinatorial chemical libraries based on bicyc-
in vivo sensing, Theranostics, 2013, 3, 583–594. lic peptides, Nat. Chem. Biol., 2009, 5, 502–507.
529 G. Rong, S. R. Corrie and H. A. Clark, In Vivo Biosensing: 548 S. Chen, et al., Dithiol amino acids can structurally shape
Progress and Perspectives, ACS Sens., 2017, 2, 327–338. and enhance the ligand-binding properties of polypep-
530 K. J. Cash and H. A. Clark, Phosphorescent nanosensors tides, Nat. Chem., 2014, 6, 1009–1016.
for in vivo tracking of histamine levels, Anal. Chem., 2013, 549 S. Chen, D. Bertoldo, A. Angelini, F. Pojer and C. Heinis,
85, 6312–6318. Peptide ligands stabilized by small molecules, Angew.
531 K. Sun, et al., In Vivo Dynamic Monitoring of Small Chem., Int. Ed., 2014, 53, 1602–1606.
Molecules with Implantable Polymer-Dot Transducer, 550 C. D. Spicer and B. G. Davis, Selective chemical protein
ACS Nano, 2016, 10, 6769–6781. modification, Nat. Commun., 2014, V5, 4740.
532 G. A. Kwong, et al., Mass-encoded synthetic biomarkers for 551 N. Krall, F. P. da Cruz, O. Boutureira and G. J. L. Bernardes,
multiplexed urinary monitoring of disease, Nat. Biotech- Site-selective protein-modification chemistry for basic biology
nol., 2013, 31, 63–70. and drug development, Nat. Chem., 2015, 8, 103–113.
533 K. Y. Lin, G. A. Kwong, A. D. Warren, D. K. Wood and 552 K. E. Sapsford, K. M. Tyner, B. J. Dair, J. R. Deschamps and
S. N. Bhatia, Nanoparticles that sense thrombin activity as I. L. Medintz, Analyzing nanomaterial bioconjugates: A review
This journal is © The Royal Society of Chemistry 2018 Chem. Soc. Rev., 2018, 47, 3574--3620 | 3619
View Article Online
of current and emerging purification and characterization 555 A. López-Serrano, R. M. Olivas, J. S. Landaluze and
techniques, Anal. Chem., 2011, 83, 4453–4488. C. Cámara, Nanoparticles: a global vision. Characteriza-
553 E. J. Cho, et al., Nanoparticle characterization: State of tion, separation, and quantification methods. Potential
the art, challenges, and emerging technologies, Mol. environmental and health impact, Anal. Methods, 2014, 6,
Pharmaceutics, 2013, 10, 2093–2110. 38–56.
554 European Technology Plattform on Nanomedicine. 556 A. Ali, et al., Synthesis, characterization, applications, and
NANOMEDICINE 2020 Contribution of Nanomedicine to challenges of iron oxide nanoparticles, Nanotechnol., Sci.
Published on 26 February 2018. Downloaded by Washington University in St. Louis on 22/05/2018 09:43:48.
3620 | Chem. Soc. Rev., 2018, 47, 3574--3620 This journal is © The Royal Society of Chemistry 2018