Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

REVIEW

www.advhealthmat.de

Recent Progress in the Design and Application of


Supramolecular Peptide Hydrogels in Cancer Therapy
Ying Cai, Chao Zheng, Fengqin Xiong, Wei Ran, Yihui Zhai, Helen H. Zhu, Hao Wang,
Yaping Li,* and Pengcheng Zhang*

support, chemical, and biological cues at


Supramolecular peptide hydrogel (SPH) is a class of biomaterials a spatiotemporal precision. The ECM can
self-assembled from peptide-based gelators through non-covalent modulate cell adhesion, cell-to-cell com-
interactions. Among many of its biomedical applications, the potential of SPH munication, and differentiation, via chang-
in cancer therapy has been vastly explored in the past decade, taking ing its composition and therefore physico-
chemical and biological properties.[1] Given
advantage of its good biocompatibility, multifunctionality, and injectability.
the vital role of ECM in cell modulation,
SPHs can exert localized cancer therapy and induce systemic anticancer ECM-based materials have been widely ex-
immunity to prevent tumor recurrence, depending on the design of SPH. This plored in tissue engineering and cancer
review first gives a brief introduction to SPH and then outlines the major therapy.[2–5] Despite the functionalities of
types of peptide-based gelators that have been developed so far. The natural ECM and the development of tissue
decellularization methodology, the biomed-
methodologies to tune the physicochemical properties and biological
ical application of ECM is hampered by its
activities are summarized. The recent advances of SPH in cancer therapy as limited source.[6,7] Besides, the complexity
carriers, prodrugs, or drugs are highlighted. Finally, the clinical translation in the composition of ECM also makes fur-
potential and main challenges in this field are also discussed. ther engineering challenging.
As an alternative strategy, synthetic hy-
drogels have been explored as mimicry of
natural ECM.[8–10] Biopolymers, synthetic
1. Introduction polymers, and supramolecular hydrogelators have been uti-
lized to create such functional hydrogels.[11–18] Compared with
Extracellular matrix (ECM) is a 3D scaffold for cell adhe- hydrogels formed by covalent crosslinking, supramolecular
sion and biomolecule binding, providing cells with structural hydrogels formed by non-covalent and dynamic chemical bonds
are unique for their reversibility and simplicity in molecular
Y. Cai, C. Zheng, F. Xiong, Dr. W. Ran, Y. Zhai, Prof. Y. Li, Prof. P. Zhang design.[18–21] Non-covalent bonds (1–5 kJ mol−1 ) are several
State Key Laboratory of Drug Research and Center of Pharmaceutics orders of magnitude weaker compared with covalent bonds
Shanghai Institute of Materia Medica (600–4000 kJ mol−1 ), and thus can be tuned to respond to mild
Chinese Academy of Sciences
Shanghai 201203, China changes in the temperature, ions, and biological cues.[22,23] For
E-mail: ypli@simm.ac.cn; pzhang@simm.ac.cn example, DNA oligomers containing moieties can be easily
Y. Cai, Dr. W. Ran, Y. Zhai, Prof. Y. Li, Prof. P. Zhang co-assembled into the supramolecular hydrogel, which could
University of Chinese Academy of Sciences thus recognize and respond to endogenous nucleic acids such
Beijing 100049, China as microRNA. Supramolecular peptide hydrogels (SPHs) that
C. Zheng, F. Xiong, Prof. H. Wang are formed by self-assembly of peptide and its derivatives have
China State Institute of Pharmaceutical Industry
Shanghai 200040, China
additional advantages and broad applications in biomedical
Prof. H. H. Zhu
engineering due to their intrinsic and tunable biofunctionality
State Key Laboratory of Oncogenes and Related Genes and biocompatibility.[24–27] SPHs usually have shape-persistent
Renji-Med-X Stem Cell Research Center nanostructure with a high degree of internal order, which
Department of Urology is widely used in living systems. Importantly, short peptides
Ren Ji Hospital (e.g., cyclicRGDD FK) are able to exert significant biological
School of Medicine and School of Biomedical Engineering
Shanghai Jiao Tong University activities (such as promote cell adhesion) and can be easily
Shanghai 200127, China integrated into peptide hydrogelator design to create biofunc-
Prof. P. Zhang tional SPHs.[22,28] Moreover, small peptide hydrogelators can
Yantai Key Laboratory of Nanomedicine and Advanced Preparations be more easily degraded and excreted through kidneys. So far,
Yantai Institute of Materia Medica SPHs have been developed to mimic natural collagen,[29]
Shandong 264000, China
to enhance wound-healing,[30] to realize biochemicals-
The ORCID identification number(s) for the author(s) of this article responsive drug release,[31] to create optoelectronic biomaterials
can be found under https://doi.org/10.1002/adhm.202001239 and pressure sensors,[32,33] and to exert drug-free cancer cell
DOI: 10.1002/adhm.202001239 eradication.[34]

Adv. Healthcare Mater. 2020, 2001239 2001239 (1 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Given the unique features, SPHs are ideal for local ther- 2.1.1. Poly- and Oligo-Peptides
apy. Localized therapy is advantageous for high and prolonged
drug exposure in the lesions with low systemic side effects. Ex- Peptides with specific sequences can naturally form hydrogels.
amples are drug-eluting beads and Gliadel wafer, which have Two such examples are elastin and collagen, which are abun-
been approved for the treatment of hepatocellular carcinoma and dant in connective tissues of animals. Elastin is a highly elastic
brain tumor, respectively. Among many local drug delivery sys- protein characteristic for its repeated VPGVG sequence. Chilkoti
tems, supramolecular hydrogels including SPHs receive increas- and colleagues created an elastin-like peptide (ELP) with repeated
ing attention because of their tunable mechanical properties, VPGVG segments, and initially used ELP as a tag to facilitate
stimuli-responsive drug release, and injectability.[24,27] While ef- protein purification through a process termed as “inverse tran-
fective and safe in treating primary tumors,[35–37] conventional sition cycling,” as the ELP tag could aggregate and form hydro-
supramolecular hydrogels that deliver drugs acting on cancer gel under physiological conditions.[46] Given the unique nature
cells usually have limited efficacy against metastatic tumors of ELP, the same group recently designed a thermally respon-
in distant organs. Recently, the development of in situ pep- sive hydrophobic elastin-like polypeptide (VPGVG)120 , which was
tide gelation technology in response to kinase/phosphatase en- further fused with a death receptor agonist (a hexameric protein
ables the formation of hydrogels in metastatic sites.[38,39] In composed of oligomers of the 3rd fibronectin type III domain
addition, SPH-based localized treatments have been found to of tenascin that is engineered to bind death receptor 5 [DR5])
elicit systemic antitumor activity by priming immune responses to create an injectable in situ hydrogel for cancer therapy.[47]
against tumors.[40–43] These discoveries boost interest in develop- While ELP is an intrinsically disordered peptide, collagen has
ing SPHs for cancer therapy. well-defined secondary and tertiary structures.[48,49] Collagen is
In this progress report, we first summarize peptide hydroge- the most abundant protein in human ECM, and has characteris-
lators that have been reported so far and then highlight current tic proline-hydroxyproline-glycine (Pro-Hyp-Gly) repeating units.
strategies that have been explored to modulate the mechanical, Although as a natural component of ECM, the application of col-
chemical, and biological properties of the hydrogels. We then fo- lagen is hindered by its limited source and difficulty in qual-
cus on the recent progress in SPH-based cancer therapy. The po- ity control. To create a synthetic version of collagen, Hartgerink
tential of SPHs in clinical cancer therapy and the remaining chal- and colleagues created a class of poly-peptides composed of Pro-
lenges are briefly discussed. Hyp-Gly repeating units.[50] The most successful version of these
synthetic collagen mimics is (Pro-Lys-Gly)4 (Pro-Hyp-Gly)4 (Asp-
Hyp-Gly)4 , which could form salt-bridged hydrogen bonds be-
tween lysine and aspartate to stabilize the triple-helix structure
2. Design of Supramolecular Peptide Hydrogel in a sticky-ended assembly.[29] The peptides could replicate the
Peptide is a class of biomaterials with high diversity. Eight- self-assembly of collagen and assemble into nanofibers with char-
thousand different tri-peptides can be composed using 20 types acteristic triple-helical packing and length with a lower bound of
of natural amino acids, but only a few of them can self-assemble several hundred nanometers, which further entangled with each
and form SPHs in aqueous solution.[44] For biomedical appli- other to give a hydrogel. The protein-derived peptide hydrogela-
cations, longer peptides and further derivatization are required, tors typically have good biocompatibility. However, only a small
which make experimental optimization impossible. Thus, it is portion of natural proteins form hydrogel, and thus hydrogelators
necessary to start with known peptide hydrogelators and then with novel sequence must be explored to expand the hydrogelator
optimize the molecular design for specific purposes. In this sec- library.
tion, we summarized well-established peptide hydrogelator sys- In addition to long and protein-derived peptides, short and ra-
tems and the methodologies for SPH property control. tionally designed peptides have been explored as hydrogelators
because of ease in preparation. Since hydrogen bonding in 𝛽-
sheet is directional and can facilitate the formation of 1D fib-
rils, many 𝛽-sheet forming peptides have been designed. The
2.1. Hydrogelator Design and Hydrogel Formation ionic self-complementary peptide RADA16-I (COCH3 -(RADA)4 -
CONH2 ) designed by Zhang and colleagues is one excellent
The formation of SPHs starts with self-assembly of peptide hy- example.[51] According to the sliding diffusion model, the pep-
drogelators and requires balanced attractive and repulsive forces tide will have two distinctive sides, one hydrophobic side with
between the resulting assemblies that are dominantly filaments. an array of alanines and the other with alternating aspartic acids
In general, the self-assembly of peptide hydrogelators is driven by and arginines. Driven by hydrophobic force and inter-molecular
intermolecular attraction forces such as hydrogen bonding, hy- ionic interactions, these peptides could adopt an anti-parallel 𝛽-
drophobic interaction, chelating effect and 𝜋–𝜋 stacking, which sheet structure and self-assemble into nanofibers, which then
are mainly balanced by electrostatic and solvation forces. The en- formed hydrogel consisting of >99.5% water. Another such ex-
tanglement of filaments on the contrary mainly mediated by salt ample is the MAX1 (V(KV)4 D PL PT(KV)4 -NH2 ) peptide that can
bridge. These forces can be encoded and finely tuned by adjust- adopt 𝛽-hairpin structures with a lateral and facial association,
ing peptide sequence and auxiliary moieties.[45] According to the which can form hydrogel after the addition of Dulbecco’s Mod-
chemical structure of the building blocks, peptide-based gelators ified Eagle’s Medium (DMEM) into the peptide solution.[52,53]
can be divided into five categories including poly-/oligo-peptide, Schneider and colleagues further reported that the solution
peptide amphiphile, aromatic peptide, peptide-drug amphiphile, pH required for hydrogel formation could be finely tuned by
and other peptide derivatives (Figure 1). engineering net charge of the peptide and exact position of

Adv. Healthcare Mater. 2020, 2001239 2001239 (2 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 1. Representative sequence and structure of peptide-based hydrogelators. These hydrogelators can be mainly divided into four categories includ-
ing poly-/oligo-peptide, peptide amphiphile, aromatic peptide, and peptide-drug amphiphile.

substitution within the peptide sequence.[54] A point substitu- whole sequence space of tripeptide using coarse-grained simu-
tion of one lysine residue of amphiphilic 𝛽-hairpin peptides lation considering the aggregation propensity and hydrophilic-
MAX1 with glutamic acid (MAX8, V(KV)4 D PPTKVEV(KV)2 -NH2 ) ity of different tripeptides.[44] The simulation screened out some
was sufficient to decrease the critical pH at which the peptide new tripeptide hydrogelators such as KYF, KYY, KFF, and KYW,
forming hydrogel. Amino acids with phosphorylated side group the gelation capability of which was further confirmed experi-
could also be integrated into the sequence, which enabled enzy- mentally. Despite its powerful capability in predicting peptide hy-
matic dephosphorylation to control SPH formation.[55] In addi- drogelators, the coarse-grained simulation requires the param-
tion to 𝛽-sheet-forming peptides, those adopted 𝛼-helical struc- eters of each residue to be defined, and is thus limited by cur-
tures are also able to form hydrogel with high water content. rent understanding of the amino acid residues. Although already
First of this class was reported by Woolfson and colleagues, a huge sequence space, the peptides can be further functional-
and the hSAFAAA p1 ((KIAALKA)2 EIAALEAENAALEA) formed ized through palmitoylation, glycosylation, and phosphorylation,
purely 𝛼-helical fibrils, which entangled to give hydrogel with which are widely adopted by nature.
>99% water.[56]
The discovery that dipeptide such as diphenylalanine (FF)
could also self-assemble into hydrogels propagates researchers 2.1.2. Peptide Amphiphile
to find more short peptide hydrogelators.[57] As we mentioned
above, the huge sequence space of peptide is one major hurdle Palmitoylation is one important post-translational modification
for experimental screening. To address this limitation, Ulijn and of proteins, which regulates protein trafficking and intracellular
colleagues built a dynamic combinatorial peptide library based signaling.[59] Inspired by this process, researchers have generated
on unprotected homo- and hetero-dipeptides.[58] These dipep- a huge class of peptide derivatives called peptide amphiphiles
tides were subjected to continuous enzymatic condensation, hy- (PAs), which were created by conjugating peptides with fatty
drolysis, and sequence exchange, and self-assembling candidates acids like myristic acid (C14 ) and palmitic acid (C16 ). Given their
would be amplified as a result of free-energy change associated amphiphilic nature, the PAs could spontaneously arrange into
with the assembly process. More importantly, it was able to pur- nanosized structures in water, which process was driven by hy-
posely select different peptide sequences and nanoscale mor- drophobic interaction. Stupp and coworkers reported the first
phologies by varying environmental conditions of screening. An- PA consisting of a long alkyl tail (C16 ), four consecutive cys-
other strategy to explore large peptide sequence space is com- teine residues, a flexible linker of three glycine residues, a sin-
putational simulation. Ulijn and coworkers first explored the gle phosphorylated serine residue and an Arg-Gly-Asp (RGD)

Adv. Healthcare Mater. 2020, 2001239 2001239 (3 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

sequence.[60] The resulting PA could form a nanostructured fi- realized.[74] In the above cases, the aromatic groups only con-
brous scaffold-like ECM which could be used for tissue regen- tributed to self-assembly. Ulijn and coworkers conjugated an or-
eration. The directional packing of PA molecules was guided by ganic chromophore diketopyrrolopyrrole (DPP) with two amino
𝛽-sheets orienting parallel to the long axis of nanofibers, which acid (Phe, Tyr, Leu) ethyl esters (DPP-(XOMe)2 ), which could
could be promoted by using a less branched peptide. The peptide form hydrogel at the presence of 𝛼-chymotrypsin.[75] The result-
sequence was also important, but the first amino acid residue ing hydrogel could produce 1 O2 as a result of DPP-mediated pho-
following the alkyl segment plays a vital role in promoting 𝛽- todynamic reaction. Li’s group set up a machine learning sys-
sheet formation and axial elongation into fibers.[61,62] These find- tem and trained it with a structurally diverse hydrogel library
ings suggest that the morphology, surface chemistry, and poten- with over 2000 peptides with characterized chemical features,
tial bioactivity can be engineered more freely by changing the including topological and physicochemical properties.[76] They
amino acid sequences. Indeed, the morphological of PA-based then utilized this trained system to predict the self-assembly be-
fibrils could be tuned from short twisted ribbon, to long twisted havior of new peptide-like molecules, and successfully obtained
ribbons and to helical ribbons via engineering three phenylala- 81 peptide-like hydrogelators, expanding the library of aromatic
nine residues.[63] The PAs could also be tailored to respond to peptide hydrogelator.
pH change and divalent ions,[64] and to display bioactive lig-
ands such as 𝛽-alanine-histidine carnosine dipeptide motif and
YEALRVANEVTLN.[65,66] In addition to the increased versatility 2.1.4. Peptide-Drug Amphiphile
of peptide sequence, PAs also provide an additional hydropho-
bic space for potential drug encapsulation, which makes them Modifications of peptide backbone with fatty acids and aro-
extremely useful in drug delivery. matic moieties mainly affect their self-assembly. By conjugating
functional molecules such as drugs to peptide could introduce
additional bioactivities while modulating self-assembly. Camp-
2.1.3. Aromatic Peptide tothecin (CPT) is one drug that has been widely investigated
for peptide-drug amphiphile development because of its potent
The study on FF peptide suggested that aromatic interaction con- antitumor activity and planar structure facilitating 1D molecu-
tributes to the free energy of self-assembly and also guides di- lar packing. Cui and coworkers first tested this concept by con-
rectional molecular stacking. Given the vital role of 𝜋–𝜋 stack- jugating CPT to a 𝛽-sheet-forming peptide sequence (VQIVY)
ing during self-assembly, many aromatic peptide hydrogelators derived from the Tau protein through a reducible disulfylbu-
have been created. The most widely used aromatic moieties tyrate (buSS) linker.[77] Since the successful proof-of-concept
for peptide modification are 9-fluorenylmethoxycarbonyl (Fmoc) study, the group dedicated to proving the versatility and tun-
and naphthalene (Nap). Fmoc-FF is an aromatic peptide that ability of this particular platform. They demonstrated that dif-
has been vastly studied. Gazit and colleagues first synthesized ferent numbers of CPT molecules could be conjugated with one
Fmoc-FF and found that this molecule could form a rigid ma- peptide to modulate drug loading and morphology without af-
terial with macroscopic characteristics of a gel at a concentra- fecting 1D molecular packing.[77] Since different cancer cells
tion of <1 wt%.[67] The observation was confirmed by Ulijn and may express distinct receptors, the formation of hydrogel should
colleagues, who first suggested that the hydrogel was formed be ideally independent of peptide sequence. Indeed, peptide of
through 𝜋–𝜋 interlocked 𝛽-sheets.[68] The Fmoc-FF contains various sequences such as hydrophilic 𝛽-sheet-forming peptide
a carboxyl group, and thus requires an alkaline environment GV2 Q2 HKD-OH and hydrophilic and cyclic peptide iRGD could
for self-assembly, which may not be physiologically relevant. be used instead of hydrophobic 𝛽-sheet-forming peptide VQIVY
Nilsson et al. thus created a set of C-terminal cation-modified to realize pH-responsive hydrogel formation and enhanced tis-
Fmoc-F derivatives that were positively charged across a broad sue penetration of both peptide-drug amphiphiles and encapsu-
range of pH values, which could form sheet-based nanotubes lated drugs.[78,79] In addition, linkers that could respond to dif-
and hydrogel at physiological pH.[69] In addition to Fmoc-FF, ferent stimuli could also be easily integrated into the molecule
Ulijn et al. also designed a series of Fmoc-dipeptides and found design to enable controlled drug release at the presence of glu-
that both the sequence and position of dipeptide would affect tathione (disulfylbutyrate or disulfanyl-ethyl carbonate linker) or
self-assembly.[70] For tyrosine-containing peptide hydrogelators matrix metalloproteinase-2 (MMP-2 cleavable PLGLAG).[78–80] In
(Fmoc-YX-OH), alkaline phosphatase (ALP) responsive hydro- these examples, the hydrophobic interaction and 𝜋–𝜋 stacking
gel formation could be realized by design a hydrogelator precur- among CPT molecules are critical for peptide-CPT amphiphile
sor (Fmoc-pYX-OH).[71] Very recently, a novel aromatic dipeptide self-assembly into nanosized filaments such as fibers and nan-
(Fmoc-Lys(Fmoc)-Asp) was reported by Gazit and coworkers.[72] otubes, and the salt bridge and depleted electrostatic repulsion
Due to the strong intramolecular and intermolecular aromatic in- mediated by multivalent ions in the solution are key for hydrogel
teraction, the reported molecule had an ultra-low critical gelation formation.
concentration (0.002 wt%, 28.3 × 10−6 m). More importantly, it In addition to CPT, other drug molecules have also been ex-
showed a novel two-step assembly process, starting from sphere- plored. Parquette and coworkers created a 5-fluorouracil (5-Fu)
based opaque hydrogel to filament-based transparent hydrogel. based peptide-drug amphiphile by conjugating 5-Fu to the 𝜖-
In addition to Fmoc, other aromatic molecules such as naph- amine of the C-terminal lysine residue of Fmoc-KK via succinate
thalene have also been used to create aromatic dipeptide and or acetamide linkage.[81] Driven by the hydrophobic 𝜋–𝜋 associ-
oligopeptide.[73] When phosphorylated tyrosine was introduced ation between uracil and Fmoc groups, the molecules could self-
into the molecule, ALP-responsive hydrogelation could also be assemble into nanofibers or nanotubes, which further entangled

Adv. Healthcare Mater. 2020, 2001239 2001239 (4 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

to form self-supporting hydrogels. The self-assembly of a chlo- 2.2. Hydrogel Characterization and Property Control
rambucil (CRB)-based peptide-drug amphiphile has been inves-
tigated by Yang and colleagues. By conjugating CRB to a phos- Hydrogels formed by different hydrogelators may have very dis-
phorylated peptide GD FD FpD Y, the resulting conjugate, CRB- tinct properties. Besides, different functional molecules could be
GD FD FpD Y, only self-assembled into stable nanofibers and hy- encapsulated either covalently or non-covalently. Given that the
drogels after the removal of phosphate group by ALP due to ECMs at different organs are distinct from each other in prop-
weakened electrostatic repulsion between the molecules.[82] Non- erties such as mechanical strength, stimuli-responsive decom-
steroidal anti-inflammatory drugs have also been used to cre- position, and controlled release of active molecules, hydrogels
ate peptide-drug amphiphile-based hydrogels. Xu and colleagues that can be finely tuned in these properties are highly required
conjugated naproxen to the N-terminus or 𝜖-amine of lysines (Figure 2).
of a set of peptides (D FD F, D FD FD Y, D FD FD K, or D FD FD KD Y).[83]
All the peptide-naproxen amphiphiles self-assembled into fila-
ments and formed self-standing hydrogels due to the strong 𝜋–𝜋 2.2.1. Morphology
stacking between the drug molecules and side chains of pheny-
lalanines. Zhong and coworkers conjugated naproxen to short The morphology of a nanoparticle (e.g., size and shape) has
peptide dendrons (E3 FID and E3 FNP), and the resulting conju- great influences on its interaction with biomacromolecules and
gates could self-assemble and form hydrogel at the assistance cells.[90,91] Though most hydrogels are formed by the entangle-
of zinc ions.[84] Similarly, Chen et al. successfully created an ment of 1D filaments, they can be in the form of nanofiber, nan-
(S)-ketoprofen (Ket) based hydrogel by conjugating Ket to either otape, or nanotube, which are distinct in their surface property
L- or D-VEVE peptide.[85] In these cases, all the drugs were of and molecular organization. In addition, the filaments are un-
small molecular weight and planar structure, which would fa- der dynamic changes, which is characteristic of supramolecule
cilitate molecular packing through 𝜋–𝜋 stacking. Cui and col- structures. An effective strategy to control the morphology of the
leagues recently conjugated a bulky anticancer drug paclitaxel filaments is varying the chemical structure of peptide hydrogela-
to the GGRGDRPLGVRVVV peptide to obtain a peptide-drug tor. For instance, C16 -VEVE self-assembled into nanobelts, while
amphiphile, which could also form a self-supporting reverse C16 -VVEE, C16 -EVEV, and C16 -EEVV tended to form singular
bolaamphiphile (RBA) hydrogel at the presence of MMP-2 in nanofibers, ribbons, and interwound nanofibers, respectively.[92]
PBS.[86] This result demonstrated that drug of different chemical The addition of GRGD sequence at the C-terminus of the pep-
properties can all potentially be engineered to become a hydroge- tide amphiphile did not affect the morphology of the filaments
lator through proper peptide conjugation. and further hydrogel formation. Non-peptidic segments could
also influence the morphology of the filaments. Cui and col-
leagues conjugated one, two, or four CPT molecules to one
VQIVY peptide, and the drug amphiphile with four CPT seg-
2.1.5. Other Peptide Derivatives ments self-assembled into nanotubes, while the rest two formed
nanofibers.[77] Aside from tuning the molecular structure of pep-
In the above cases, amide bond of peptide contributes to the tide hydrogelators, drug loading was also able to affect the mor-
self-assembly of the hydrogelators. For instance, Hartgerink phology of the self-assemblies. Cui and colleagues reported that
and coworkers showed that hydrogen bond among the first peptide-paclitaxel conjugate (PTX-buSS-CGN2 Q2 (KOEG )4 , Spax)
amino acid residues following hydrocarbon tail is critical for could form filaments only at the presence of free PTX.[93] Li and
self-assembly of peptide amphiphiles.[87] Peptidomimetic hydro- colleagues showed that the morphology of the filament could also
gelators with various backbone amide modifications have been be modulated by the kinetic control of self-assembly pathway.[94]
investigated, giving rise to hydrogels of diverse properties and The Fmoc-GF2 Q2 NYKD-OH self-assembled into nanofibers and
increased stability.[88] Given the important role of interpeptide hy- hydrogel with low mechanical strength at room temperature, but
drogen bonding, Gazit and colleagues replaced the amide bond formed twisted ribbons and self-supporting hydrogels after being
in Fmoc-FF peptide with the urea moiety, which increased hy- heated for 2 h at 70 °C. The capability of modulating morphology
drogen bonding capabilities between the molecules. The urei- of the filaments will provide a feasible way to adjust the molecular
dopeptide, Fmoc-F-NHCONH-F-OH (Fmoc-FuF), formed hydro- display of ligand and anchor points on their surfaces.
gels with improved mechanical properties compared with Fmoc-
FF, with additional benefits such as anti-fouling and controlled
release of urea stemmed from the urea moiety.[88] Another cate- 2.2.2. Mechanical Property
gory of peptide-mimetic materials is peptoid, the side chain of
which is appended to the nitrogen atom of the peptide back- Hydrogels are distinct from solutions in their mechanical prop-
bone instead of the 𝛼-carbons. Peptoid is more flexible than pep- erty that is important for their biomedical applications. As natural
tides since intramolecular CO–HN hydrogen bonds are removed. hydrogels, ECMs in different parts of human body show different
After specific engineering, tripeptoids could also form hydrogel mechanical properties. The storage modulus of the soft tissues
with the assistance of organic solvents.[89] In addition to the nat- range from 103 Pa (in the brain, lung, and kidney) to 105 Pa (in the
ural peptides, ureidopeptides and peptoids greatly expanded the bladder).[95] The stiffness of the ECMs is mainly determined by
territory of peptide-derived materials, providing enormous pos- the concentrations of ECM fibers including elastic fibers and col-
sibilities to obtain functional hydrogels for specific biomedical lagen fibers and is under dynamic change over time in response
applications. to physiological and pathological stimulus.[96] These properties

Adv. Healthcare Mater. 2020, 2001239 2001239 (5 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 2. Characterization and major properties of SPH. Some methods to modulate the properties are mentioned. The hydrogelators can self-assemble
into different filamentous structures, and form SPH of different and controllable mechanical, chemical, and biological properties.

are associated with cell differentiation and functions in particu- in the middle of the RAD16 peptide ((RADA)4 ) increased the stiff-
lar tissues.[97,98] Therefore, it is critical to adjust the mechanical ness of the hydrogel (G′RADA16 = 265 Pa, G″RADA16 = 42 Pa; G′A8G
properties for the purposed biomedical applications. To quantify = 469 Pa, G″A8G = 56 Pa).[101] The enhanced stiffness of A8G
the mechanical strength of hydrogels, storage modulus G′ and probably resulted from the formation of twisted 𝛽-sheet struc-
loss modulus G″ are the most widely used parameters. G′ reflects tures and nanofiber bundles due to a transition in the molecu-
the rigidity of hydrogels, and is closely related to its injectabil- lar packing. These results suggested that a balanced rigidity and
ity, tissue retention capacity, and cell interaction. It is sensitive to flexibility of filament should be reached to achieve high stiff-
changes in micro- or nano-structure of viscoelastic systems and ness. In addition to the peptide sequence, Hamley and cowork-
thus can be modulated at both scales. G″ describes the energy ers also found that terminal modification of a peptide hydrogela-
transformed into heat when a material is deforming and reflects tor could also significantly affect the G′ of the hydrogel.[102] They
the viscosity. The mechanical property of a hydrogel can be mod- showed that N-acylation and C-termini histidine of peptide H2 N-
ulated via changing hydrogelator structure and combination, as VEQLTEEQKNEFKAAFDIFVLGA-OH increased the G′ by three-
well as components in the solution environment (Figure 3). fold to a value of ≈13 kPa. Lin et al. explored the effect of iso-
Engineering chemical structure of hydrogelator is one strat- meric hydrocarbons (C7 ) on the stiffness of hydrogel, and found
egy to modulate the mechanical property of a hydrogel. Since the that the G′ decreased with an increase in the ring size of the tail
hydrogels were formed by 1D filaments, Stupp and colleagues through regulating molecular packing at the interface between
explored the effect of strength of 𝛽-sheet on the stiffness of the the peptide domain and the hydrophobic core.[103] In addition to
resulting hydrogel.[99] They created a set of peptide amphiphiles the composition of the hydrogelator, Schneider and colleagues
with various number of valines and alanines at different positions further demonstrated that the chirality of a peptide hydrogelator
relative to the hydrocarbon tails, and found that valine positioned could be used as a design tool to control the mechanical rigidity
at proximity to the hydrocarbon tail could increase the stiffness of peptide-based hydrogels.[104] An enantiomeric peptide mixture
and mechanical property (e.g., two alanine replaced with valines of MAX1 and DMAX1 showed threefold greater rigidity than that
in the hydrocarbon tail increased the G′ by tenfold). Similarly, Xu of hydrogel prepared from either of pure enantiomer because of
and colleagues substituted the valines of MAX1 sequence with energetically favorable interactions between enantiomers.
isoleucines that had a higher propensity for 𝛽-sheet hydrogen Engineering structure of hydrogelator, while effective in mod-
bonding to obtain MAX1I8 ((IK)4 VD PPT(KI)4 -NH2 ).[100] MAX1I8 ulating the stiffness of the hydrogel, may also affect other proper-
showed a faster gelation process than MAX1 (30 s vs 75 min to ties of the hydrogel. Since the interaction between the filaments
reach a G′ of 100 Pa), and formed hydrogel with a G′ as high as is another important factor that determines the mechanical prop-
1320 Pa after 2 h. However, Muraoka and coworkers showed that erty of the hydrogel, a simple yet effective way to modulate hy-
substitution of alanine with glycine (A8G, RADARADA(RADA)2 ) drogel stiffness is manipulating ionic strength that can affect

Adv. Healthcare Mater. 2020, 2001239 2001239 (6 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 3. Peptide design for modulation of mechanical property of SPHs.

electrostatic interactions. Indeed, Williams and coworkers found surfactant-like Fmoc-S in different concentrations to produce hy-
that G′ of the Fmoc-FRGDF hydrogel increased from 10 to 3200, drogels with tunable stiffness (1, 13, and 32 kPa).[110] Fmoc-R,
7500, and 11 000 Pa in PBS solutions of 0.05, 0.25, 0.5, and 0.75 another surfactant-like molecule, has also been used to increase
m, respectively.[105] Given that the concentration of salts must be the Fmoc-FF-based hydrogels.[111] The above strategies used non-
limited within a tolerable range for biomedical application, Stupp covalent crosslinkers, and Guler and colleagues explored the ef-
and coworkers used oligo-L-lysines of different molecular lengths fect of glutaraldehyde, a covalent crosslinker, on the stiffness of
to precisely tune anionic gel stiffness and found that G′ increased lauryl-VVAGKK-Am (K2 -PA) hydrogel.[112] The G′ of the hydro-
by 10.5 Pa for each additional lysine monomer in the oligo- gel was significantly increased after the addition of glutaralde-
L-lysine chain.[106] In addition to these small molecular cross- hyde, which could exceed 105 Pa. More importantly, the viscoelas-
linkers, macromolecules and even nanoparticles could be used tic properties of the hydrogel could further be tuned by changing
to modulate the mechanical property of the hydrogels through ratios of glutaraldehyde or addition other dialdehydes of differ-
non-specific or specific intermolecular interactions. Huang and ent molecular length. Pyridoxal-5-phosphate (P5P) has also been
coworkers demonstrated that mixing ovalbumin (OVA) protein investigated by Xu and colleagues to modulate the mechanical
with Nap-HHFF peptides can enhance the mechanical property property of the SPH.[113]
up to 15-fold.[107] Nilsson and colleagues utilized specific molec-
ular recognition between mannose-modified fibrils with con-
canavalin A (ConA) to modulate hydrogel stiffness, and found 2.2.3. Chemical Property
that the G′ of mannose-functionalized Ac-(FKFE)2 -NH2 peptide
hydrogels increased from 148 ± 9 to 466 ± 61 Pa after the ad- Natural ECMs are under constant chemical modification to ex-
dition of ConA.[108] Ulijn and coworkers further created a spe- ert desired functions. Therefore, SPHs need to exert stimuli-
cific and multivalent nano-crosslinker by decorating magnetic responsive chemical property to maximize their activities in
nanoparticles with thermolysin, which could generate hydroge- biomedical applications. A feasible strategy to control the
lator Fmoc-TF-NH2 in situ.[109] This resulted in up to a tenfold chemical properties of a hydrogel is to introduce stimuli-
increase (from 1.9 × 103 to 15 × 103 Pa) in G′ of the hydrogel responsive moieties into the molecular design (Figure 4). Light-
compared to a conventional soluble enzyme system. Crosslinkers responsive groups are those under broad investigation, as chem-
could also be introduced into filaments by co-assembly strategy. ical reactions could be easily triggered by irradiation.[114–116]
For instance, Ulijn and coworkers co-assembled Fmoc-FF with Schneider and colleagues reported a self-assembly hydrogel

Adv. Healthcare Mater. 2020, 2001239 2001239 (7 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 4. Peptide design for modulation of chemical property of SPHs. The letter (X) represents GSH, NO, or H2 S.

composed of 𝛽-sheet-forming peptides, the lysyl sorbamide side colleagues, and the hydrogel filaments could rapidly break into
chains of which could polymerize after exposure to ultraviolet fragments prior to reassembling into spherical micelles upon
(UV) radiation.[117] Instead of lysyl sorbamide, flavin mononu- MMP-2 exposure.[86]
cleotide (FMN) and [3-(3-aminomethylphenylazo)phenyl] acetic
acid (AMPP) have also been used to create peptide hydrogel with
light responsiveness.[118,119] 2.2.4. Bioactivity
For biomedical applications, it is ideal to create hydrogels that
can respond to biological cues such as reducing agents, enzymes, Hydrogel such as ECM is not merely a scaffold providing
or other biomolecules.[120] Nitric oxide (NO) and hydrogen sul- space for cells, but more importantly, provides biological sig-
fide (H2 S) are molecules that have important biological func- nals to tune the behavior of cells. Natural ECM fulfills this task
tions. To create peptide hydrogels that could respond to these through two strategies. First, ECM incorporates functional lig-
molecules, Yang and coworkers designed a short peptide, GFFY, ands such as RGD-containing domain into the filaments. Sec-
with self-immolative aromatic groups to yield supramolecular hy- ond, ECM recruits cells to functionalize the matrix. Similar
drogelators that could form hydrogels. The self-immolative cap- strategies have been employed to create biomimetic hydrogels
ping group could be removed in the presence of NO, H2 S, or (Figure 5). For instance, Stupp and coworkers incorporated a
GSH, causing gel–sol phase transition.[121] Tumor microenviron- laminin-mimetic IKVAV sequence into their peptide amphiphile
ment is characteristic for enhanced expression of protease such design (C16 V2 A2 E4 GIKVAV), creating a supramolecular hydro-
as matrix metalloproteinases (MMPs). MMP-responsive hydro- gel for neural transdifferentiation.[123] Yang and colleagues mod-
gels could thus be potentially used in cell behavior control and ified their peptide hydrogelator with folic acid (FA) to generate
tumor-specific drug delivery.[20] Xu and colleagues designed an FA-functionalized hydrogel, which significantly improved the re-
MMP-2 responsive peptide hydrogelator (Ac-I3 SLKG-NH2 ) by in- tention and survival of pluripotent stem (iPS) cells.[124] Li and
corporating MMP-2 substrate sequence into molecule design.[122] colleagues reported a new peptide gelator (Nap-FFRGD) to en-
Upon exposure to MMP-2 overexpressing HeLa cells, the self- capsulate vascular endothelial growth factor (VEGF), and the re-
assembled nanofibers and the entangled hydrogel network can be sulting hydrogel enhanced blood capillary density in mice.[125]
significantly disrupted. A similar design was reported by Cui and In addition to a direct chemical modification to the chemical

Adv. Healthcare Mater. 2020, 2001239 2001239 (8 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 5. Peptide design for modulation of bioactivity of SPHs.

structures,[126] co-assembly strategy was also explored to func- modulated at different levels from molecular design to hydrogel
tionalize SPHs. For instance, Ulijn and coworkers reported the composition, which facilitates their biomedical applications.
co-assembly of Fmoc-FF and Fmoc-RGD to construct small-
molecule-based bioactive hydrogels through 𝜋–𝜋 stacking of
Fmoc groups.[127] The same group also co-assembled Fmoc-FF 2.3. Control of Cargo Loading and Release
with aromatic carbohydrate to generate minimalistic proteogly-
can mimics, which could prolong the stability of growth factors As we discussed above, peptide hydrogel has intrinsic biological
and preserve the viability of cells in the SPH.[128] By the modi- activities. These activities could be further enhanced by incorpo-
fication of fatty acids and aromatic rings, the range of gelators rating additional biologically active components. These compo-
has been extended. Interestingly, Gazit et al. combined Fmoc-FF nents can be small-molecule drugs, protein therapeutics, gene
peptide with a conductive polymer polyaniline (PAni) to form a therapeutics, or even cells, and the resulting hydrogels have
two-component and conductive hydrogel, which could be used been used in cancer therapy, regenerative medicine, and immune
for dynamic range pressure sensing and as a conductive inter- engineering.[62,132,133] Functional components can be introduced
face for electrogenic cardiac cells.[32] into the hydrogels through either physical entrapment or chemi-
In contrast to the above strategies, Hartgerink and colleagues cal conjugation (Figure 6). Effective cargo loading and its control-
investigated the infiltrated cell-mediated K2 (SL)6 K2 peptide hy- lable release are key factors to maximize their efficacy.
drogel remodeling.[129] The K2 (SL)6 K2 hydrogel could induce
rapid cell infiltration, and then be enriched with natural collagen-
containing ECM and a wide range of cytokines and growth fac- 2.3.1. Physical Entrapment
tors produced by the infiltrating cells, which could support cells
present in the wound healing process.[130] To study the effect SPHs are characteristic of their non-covalent nature, mesh-like
of ionic charged domains at the N- and C-termini, they further structures, and capability of immobilizing liquid. Therefore, the
designed a series of multidomain peptides (MDPs) including cargoes can be easily entrapped within the filament network
K2 (SL)6 K2 , R2 (SL)6 R2 , E2 (SL)6 E2 , and D2 (SL)6 D2 . All the studied through mixing, and their release from the SPHs could be ad-
MDPs showed similar nanostructures and physical properties, justed via tuning interactions between the cargoes and filament
but they trigger markedly different inflammatory responses. The network. Schneider and colleagues revealed that the size of the
negatively-charged peptide hydrogels elicited minimal inflam- cargoes and their electrostatic interactions with hydrogel net-
mation characterized by tissue-resident macrophage infiltration, works are two important factors influencing release. They devel-
fast remodeling, and no collagen deposition or blood vessel for- oped a multi-compartment hydrogel composed of EGFR kinase
mation within the implants. In contrast, the positively-charged inhibitor Erlotinib (ERL) and Doxorubicin (DOX) vesicles, and
peptides were highly infiltrated by immune cells, were remod- found that the ERL was released in early time due to its small
eled at a slower rate, would promote angiogenesis, and resulted molecular size while the larger DOX vesicles showed a much
in a high degree of collagen deposition.[131] These studies showed slower release. The sequential treatment (first ERL and then
that the biological activity of the peptide-based hydrogels could be DOX) first inhibited EGFR signaling, which sensitized LN229

Adv. Healthcare Mater. 2020, 2001239 2001239 (9 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 6. Methods of loading cargos into SPHs. Cargoes including small-molecule drugs, biomacromolecules, nanoparticles, and even cells can be
easily entrapped within the filament network through physical entrapment and chemical conjugation.

glioblastoma cells to the subsequent DOX.[134] Similar result was Another strategy to control the release of entrapped car-
also observed when Zhang and colleagues investigated the ef- goes is to disrupt mesh structure. Given the large size of the
fect of protein size on their diffusion out of the SPH using mesh in the SPHs, it is ideal for entrapment and controlled re-
single-molecule fluorescence correlation spectroscopy (FCS).[135] lease of biomacromolecules and nanoparticles. Hamachi’s group
To understand the effect of electrostatic interaction on drug re- used a H2 O2 -responsive supramolecular hydrogel as the uni-
lease, the same group created a negatively charged SPH (Ac- versal matrix, coupling with enzymatic reactions with the gel
VEVSVSVEVDPLPTEVSVEVEV-NH2 , AcVES3) and a set of en- response.[31] The hydrogel could response to input stimuli like
hanced green fluorescent protein (EGFP) analogues with dif- H2 O2 molecules and various disease-related biomarkers, includ-
ferent positively charged interaction domains (IDs) at their N- ing glucose, lactose, and uric acid, which could be converted
termini.[136] These IDs exerted weak, medium, or strong affin- into H2 O2 by the hydrogel-enzyme hybrids. They combined two
ity for the fibrils, realizing controlled release of EGFP from the different gelators BPmoc-F3 and NPmoc-F2 that could respond
AcVES3 SPH. Wang et al. even showed that a strong electro- to oxidation and reduction, respectively, and built Boolean logic
static interaction between the cargoes and dense hydrogel net- gates (OR and AND) into materials. The addition of both NADH
work would even retain cargo enzyme chloroperoxidase (CPO) and glucose (AND logic-regulated) induced a gel–sol transition
in the Fmoc-Y-OH-based SPHs for more than two weeks without and release about 90% of the initially encapsulated drugs. In com-
significant release.[137] parison with hydrogel network limited or hydrophobic interac-
In contrast with biomacromolecules and nanoparticles that tion controlled drug release mechanism, gel–sol transition me-
are large and multi-valent, small molecular drugs must have a diated drug release is more suitable for stimuli-responsive bolus
strong affinity for hydrogelators in order to be retained in and drug release.
sustainedly released from SPHs. Nilsson and coworkers cre-
ated an SPH self-assembled from Fmoc-F5 -Phe-DAP (DAP, di-
aminopropane) for localized delivery of diclofenac.[138] Due to 2.3.2. Co-Assembly
the 𝜋–𝜋 interaction between the altered quadrupole in the aro-
matic benzyl side chain of the gelator and aromatic rings of Co-assembly is another important strategy to realize supramolec-
the diclofenac, sustained release of the drug from SPH over ular encapsulation and controlled release. This strategy has
two weeks in vivo after a single injection was achieved. Li been utilized to combine B and T epitopes by conjugating the
and colleagues explored the influence of hydrogelators’ struc- epitopes to Q11 (Ac-QQKFQFQFEQQ-Am) peptide domain re-
ture on drug encapsulation efficiency.[94] They created a set spectively, achieving a high-titer antibodies and a modular T-
of peptide hydrogelators with different peptide sequences and cell response.[140] Similar strategy has been reported by Yang
hydrophobic tails (C16 -GN2 Q2 NYKD-OH, Fmoc-GN2 Q2 NYKD- and colleagues for delivery of phosphorylated antigens such as
OH, and Fmoc-GF2 Q2 NYKD-OH) for encapsulation of losar- TpSN, CpTW, and KpTL.[141] The hydrogel slowly released the
tan. They showed that the drug loading was the highest in C16 - co-assembled antigens and protect them from early dephos-
GN2 Q2 NYKD-OH, the self-assembly of which contained a large phorylation. In addition to peptides, the same group also ex-
hydrophobic cavity for losartan. However, Pochan et al. showed plored the co-assembly of proteins with SPH. In one such exam-
that vincristine, a hydrophobic chemotherapeutics, could also be ple, they modified antiHER2 affibody with hydrophobic drugs,
encapsulated within MAX8-based SPH with constant drug re- which co-assembled with NBD-𝛽A-GD FD FpD Y (NBD: 4-Chloro-
lease over the course of one month.[139] Since MAX8 did not con- 7-nitrobenzo-2-oxa-1,3-diazole) to realize high affibody loading
tain hydrophobic tail, their findings suggested that the side chain and prolonged release.[142] In another example, they demon-
of MAX8 may contribute to shielding vincristine from surround- strated the co-assembly of flurbiprofen (Fbp)-modified peptide
ing environment. Fbp-GD FD FD YD K(𝛾E)2 -NH2 with protein antigens such as OVA

Adv. Healthcare Mater. 2020, 2001239 2001239 (10 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

and hepatitis B surface antigen (HBsAg).[143] During the co- PTX. However, the location of the protease-cleavable peptide in
assembly between peptide and protein antigens, OVA or HB- the filament is important. Enzymatic cleavage events could be
sAg served as coacervation stimulators to induce hydrogelation inhibited if the peptide substrate was hidden in the assembled
and colocalized onto peptide nanofibers. Compared with pro- structures.[149] In the above cases, the drug molecules mostly
teins that were physically encapsulated in hydrogels, antigen re- located at the hydrophobic domain of the filaments. However,
lease from co-assembled nanofibers was much slower. In addi- functional peptide and amino acid could be integrated into the
tion to biomacromolecules, small molecules could also be loaded peptidic segment of the hydrogelator. For instance, Hartgerink
through the co-assembly strategy, in which the co-assembled and coworkers integrated a hydrophilic drug N6-(1-iminoethyl)-
drugs also played essential roles in hydrogel formation. For L-lysine (L -NIL) (an inhibitor of inducible nitric oxide synthase,
instance, Zhong and colleagues reported that DOX could co- iNOS) into K2 (SL)6 K2 MDP to give L -NIL-MDP, which showed
assembled with peptide Nap-GFFYGRGDH by electrostatic in- comparable activity in inhibiting inducible nitric oxide syn-
teraction to form hydrogel which would release the DOX under thase (iNOS) in vitro and in vivo.[150] Yang et al. used tuftsin
weakly acidic conditions.[144] The same group also found that cis- (TKPR) with Nap-GD FD FD Y to form a hybrid peptide compound
platin (CDDP) could co-assemble with Nap-FFYERGD peptide Nap-GD FD FD YTKPR, which formed hydrogel and enhanced the
to form a rigid hydrogel.[145] In the above two cases, the two aro- maturation of DCs and phagocytosis of macrophages.[151] Sim-
matic peptides themselves were not able to form rigid hydrogels. ilarly, Ruokolainen et al. conjugated carnosine dipeptide (𝛽-
alanine-histidine, 𝛽AH) onto a KTT sequence from “Matrixyl”
lipopeptides to a lipopeptide C16KTT𝛽AH, which showed dose-
2.3.3. Chemical Conjugation dependent cytotoxicity to MCF-7 cancer cells.[65] It is also possible
to conjugate proteins on the surfaces of filaments, which strategy
Physical entrapment, while a feasible strategy, usually shows could be used to realize stimuli-responsive and sustained drug
a fast drug release. Chemical conjugation provides a precisely release.
controlled method to achieve high drug loading and prolonged
drug release. In the peptide-drug amphiphile section, the re-
3. Supramolecular Peptide Hydrogel in Cancer
quirement for drug and peptide has been discussed in detail.
In this section, we mainly focus on the design of crosslinker,
Therapy
which must be reversible and stimuli-responsive, as most drugs As we have discussed above, SPHs are advantageous for tun-
must be released as their parental forms in order to be active. able physicochemical properties and biological activity, good bio-
Hydrolysable ester bond is a widely used bond for drug conju- compatibility, and multiple spaces for both hydrophobic and hy-
gation. 1-naphthaleneacetic acid (NAA) that was conjugated to drophilic molecules. Moreover, they could be further function-
peptide G′FFY (NAA-G′FFY) through an ester bond could slowly alized to exert stimuli-responsive (dis)-assembly and drug re-
release NAA in aqueous solution, which is in sharp contrast with lease, thus making it a perfect candidate for drug delivery.[152]
amide bond.[146] In addition to spontaneous hydrolysis, drugs co- Given their unique advantages, SPHs have been explored as both
valently loaded into SPH through an ester bond could also be supramolecular drug and biocompatible vector for localized and
specifically released at the presence of carboxylic-ester hydrolase systemic cancer therapy.[153]
(CES) in the cells.[147] In these cases, the focus was given to the
release of free drug from the conjugate. Cui and colleagues de-
veloped a set of GSH-responsive peptide-drug amphiphiles, and 3.1. Small Molecular Drug Delivery
found that drug release from SPHs could be divided into two
steps, filament dissociation and free drug release. They first cre- Small molecular weight drugs are widely used in cancer therapy
ated a peptide-paclitaxel conjugate with buSS linker (PTX-buSS- due to their well-defined efficacy against a broad spectrum of can-
G2 V3 RGDR), which could form self-supporting hydrogels.[148] cers. However, most of them suffer from shortcomings such as
They showed that the release of hydrogelator monomers from limited anticancer efficacy and serious systematic toxicity, which
the SPH was controlled by their critical aggregation concentra- are resulted from non-targeted distribution and short half-life of
tion (CAC). Since the CAC was constant, a linear release pro- the drugs in the tumors. Localized small molecular weigh drug
file could be achieved rather than burst release. The monomers delivery with injectable SPHs could realize prolonged drug expo-
were converted into free PTX after their internalization into sure in the tumors, which can potentially increase efficacy and
cells. Given the low CAC of the monomer and limited release lower systemic toxicity.
medium, the hydrogel maintained a sustainable drug release
over the period of several months. This observation was further 3.1.1. Delivery of Chemotherapeutics with SPH
confirmed on another peptide-drug amphiphile (camptothecin-
buSS-GV2 Q2 HKD, CPT-HKD) based SPH, which also showed The filament network of the SPHs can hinder the diffusion of
a steady and linear release of CPT.[78] The dis-assembly of fil- the entrapped molecules out of the depot, the capability of which
aments could also be engineered to respond to extracellular can be experimentally quantified by measuring the diffusion con-
biomolecules. The same group further introduced an MMP-2 stant of a model fluorophore through fluorescence recovery after
cleavable sequence into their molecular design.[86] MMP-2 could the photobleaching (FRAP) method.[154] Aside from the physi-
accelerate filament fragmentation into spheres and short rods, cal barrier, the rate of drug diffusion is also influenced by non-
which could also be internalized by cells. This design signifi- covalent interaction between the encapsulated drugs and the net-
cantly enhanced the conversion of peptide-drug amphiphiles into work. Nie and colleagues designed a hexapeptide hydrogelator

Adv. Healthcare Mater. 2020, 2001239 2001239 (11 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

(FEF3 K), which could form nanofibers with antiparallel 𝛽-sheet tors. Photosensitizers such as Chlorin e6 (Ce6) have also been
structure and hydrogel.[155] Due to the strong 𝜋–𝜋 interaction be- delivered with (FHFD)2 -based hydrogel for photodynamic ther-
tween the filaments and DOX, a low but effective dose of DOX apy of cancer.[161] Liu and colleagues took this work a step fur-
could be maintained in the tumor, which significantly reduced ther by conjugating recombinant elastin-like polypeptide (ELP)
the side-effect of DOX in the treatment of 4T1 breast cancer, and with Ce6 to function as a convection-enhanced delivery (CED)
enhanced the tumor growth inhibition efficacy at the primary tu- driven photoradiation-controlled intratumoral depot (PRCITD).
mor site, and prolonged the survival to over 40 days. This design They showed that the penetration of ELP could be enhanced by
requires direct intratumoral injection of the SPH. Ulijn et al. CED under a 660 nm laser, and the generated reactive oxygen
took one step further and created an matrix metalloproteinase- species (ROS) catalyzed periodic cysteine residues of ELP and in-
9 (MMP-9) responsive mixed peptide micelle for DOX delivery duced their crosslink into a therapeutic stable depot. Compared
using aromatic moiety-containing PhAc-FFAGLDD and MMP-9 with control groups without crosslinking, the PRCITD showed
degradable GFFLGLDD and their analogues.[156] These micelles high stability and significantly developed the retention in tumors,
could convert into fibrous nanostructure after reaching MDA- improving the anti-tumor efficiency in a Ce6 dose-dependent
MB-231 tumor site with high MMP-9 expression and form DOX way.[162] Since the activity of Ce6 must be triggered with localized
depot for localized therapy of cancer. In addition to DOX, other irradiation, this strategy could further decrease off-tumor side ef-
chemotherapeutics such as vincristine,[139] tanshinone IIA,[157] fects.
and triptolide[158] have also been physically encapsulated in SPHs Jeffrey et al. validated the concept of delivering immune
for localized cancer therapy. These hydrogels maintained the con- modulators with SPH (Figure 7b).[41] They encapsulated cyclic
centrations of drugs within a suitable range, which greatly en- dinucleotides dithio-(RP,RP)-[cyclic[A(2′,5′)pA(3′,5′)p]] (CDNs),
hanced the efficacy and safety of chemotherapy. a stimulator of interferon genes (STING) agonist, within MDP
Physical entrapment provides a feasible way to load to form the “STINGel” through the favorable electrostatic inter-
chemotherapeutics into SPH. However, it is difficult to achieve actions between the CDN’s negative thiophosphate linkages and
high drug loading, and the drug release and hydrogel de- the positive lysine residues at the peptide termini. The STINGel
composition are not synchronized. SPH self-assembled from was injected subcutaneously in MOC2-E6E7 murine oral cancer
peptide-drug amphiphile could be one possible strategy to tumor, realizing localized and sustained release of CDNs, which
address this issue. For instance, Cui et al. developed a camp- was eightfold slower compared with standard collagen hydrogel
tothecin (CPT)-based self-assembling prodrug (SAPD) hydrogel group. This local treatment induced tumor rejection and pro-
for the treatment of glioblastoma multiforme (GBM) after max- longed overall survival of model mice.
imal tumor resection. The covalent linkage yielded a fixed drug In the above examples, the small molecular-weight drugs de-
loading as high as 23% drug loading (in weight), and ensured the livered by SPHs mainly targeted cancer cells. Li and coworkers
synchronous kinetical behaviors of CPT and the HKD hydrogel. aimed to create an SPH for sustained delivery of losartan, an in-
The SAPD hydrogel showed a steady and linear drug release hibitor of cancer-associated fibroblast (CAF).[94] CAF is one of
of more than 30 days in vitro. After intracranial implantation the major components of tumor stromal cells and contributes
into the cavity left over by surgical resection, the SAPD hydrogel to ECM remodeling and cancer metastasis. Prolonged losartan
penetrated deeply into the brain parenchyma, and inhibited the treatment is necessary to block the function of CAF, and thus
recurrence of GBM (Figure 7a).[78] To target inoperable tumors, a high drug loading and prolonged drug retention are necessary.
Zhong et al. designed a hydrogel precursor (HCPT-SA-FFEssEE) To achieve this goal, they designed and screened a set of hydorge-
by conjugating HCPT-SA-NHS with FFEssEE. When exposed lators and found that C16 -GN2 Q2 NYKD-OH (C16-N) showed the
to GSH, HCPT-SA-FFEssEE would be cleaved and converted to highest drug loading because the long hydrocarbon tail formed
HCPT-SA-FFE, which could form a supramolecular hydrogel in a flexible and large hydrophobic cavity for losartan. The losartan-
the tumors. The hydrogel at a concentration of 5.0% w/v HCPT- loaded nanofibers could form hydrogel under physiological con-
peptide could release drugs sustainedly for at least 72 h at the ditions, and retained in the orthotopic 4T1 tumor for over nine
presence of MD-MBA-231 cells.[159] In addition, SPHs have been days after intratumoral injection, which was in sharp contrast
reported to deliver small weight molecules loaded nanoparticles. with losartan-loaded spherical micelles formed by DSPE-PEG.
Mo and colleagues created PTX-CT by encapsulating PTX into The SPH significantly inhibited the incidence of CAF and the
a cell-penetrating-peptide (CPP) R8 H3 -modified transfersomes. synthesis of ECM, and therefore enhanced intratumoral accumu-
Then PTX-CTs were entrapped into Fmoc-FFF-Dopa-based lation and penetration of DOX-liposomes, as well as their antitu-
SPH.[160] The hydrogel could be applied as a patch on the skin mor efficacy.
for transdermal administration of PTX. Due to its network struc-
ture, the SPH design significantly prolonged PTX-CT retention
and allow a much longer interaction time for PTX-CT to squeeze 3.1.3. Co-Deliver Chemotherapeutics and Other Small Molecular
through the skin barrier compared with PTX-CT solution. Drugs with SPH

Combination treatment is widely adopted in clinical cancer ther-


3.1.2. Delivery of Other Small Molecular Weight Drugs with SPH apy to maximize efficacy. SPH is a convenient platform for co-
delivery of multiple drugs through mixing.[163,164] Besides, SPH
In addition to chemotherapeutics, other small molecular weight could be formed by self-assembling prodrugs, which offers a
drugs have also been explored in cancer therapy, including pho- new method for combinational therapy. For example, Wang et al.
tosensitizers, molecule-targeted drugs, and immune modula- first created a self-assembling prodrug via conjugating two CPT

Adv. Healthcare Mater. 2020, 2001239 2001239 (12 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 7. Schematic illustrations of SPHs loaded with small molecular weight drugs. a) The design and mechanism of action of self-assembling prodrug
(SAPD) hydrogels. Reproduced with permission.[78] Copyright 2020, Elsevier B.V. b) The design of “STINGel” formed by CDN-loaded MDP hydrogel.
Reproduced with permission.[41] Copyright 2018 Elsevier Ltd.

molecules to a tumor homing and penetrating peptide iRGD signal at the periphery of the tumor. The CDA-diCPT-iRGD also
(cyclic-CRGDKGPDC)[165] to obtain diCPT-iRGD. This peptide- induced a higher percentage of intratumoral CD8+ T cells and
drug amphiphile could assemble into SPH, which could entrap elicited tumor regression, which was in sharp contrast with CDA-
STING agonist cyclic di-AMP (c-di-AMP (CDA)) for co-delivery diC8-iRGD (Figure 8b).[79] Recently, they developed diCPT-iRGD
of CPT and CDA into the GL-261 brain tumor. The resulting to serve as a universal dispersing agent for low-molecular-weight
SPH showed much better tumor penetration throughout the tu- hydrophobic drugs. A second therapeutic agent like DOX or cur-
mor and the tumor periphery compared with the control hydro- cumin (Cur) could be co-delivered for combination therapy.[166]
gel composed of diCPT-iRGRD (Figure 8a), which manifested no In these examples, the SPH was injected directly into the tumor.

Adv. Healthcare Mater. 2020, 2001239 2001239 (13 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 8. The design and mechanism of action of SPH loaded with CPT and CDA. a) The chemical structure of diCPT-iRGD. b) The diCPT-iRGD could self-
assemble into nanotubes (NT) with positive surface charge and thus adsorb negatively charged CDA. The resulting CDA-loaded NT solution could form
hydrogel after intratumoral injection, and release CPT and CDA for combinational chemotherapy and immunotherapy. Reproduced with permission.[79]
Copyright 2020, Springer Nature.

In another example, Liu and colleagues developed a novel pro- 3.2. Biomacromolecule Delivery
drug, namely Taxol-EYSV, which could from SPH in the tumor af-
ter intravenous injection.[167] The Taxol-EYSV molecule was syn- Cancer therapy using biomacromolecular drugs such as anti-
thesized by conjugating Taxol to a tripeptide tyroservatide (YSV, gens, proteins, antibodies and nucleic acids is becoming pop-
histone deacetylase and P-glycoprotein inhibitor) through an es- ular. Most of these drugs, while of high potency, suffer from
ter bond that could be cleaved under hydrolysis. The YSV peptide short half-life due to fast clearance or degradation. Given that
then formed hydrogel for Taxol deposition, and inhibited histone SPH can entrap molecules in a hydrophilic network, many SPHs
deacetylase and P-glycoprotein. Therefore, the Taxol-EYSV-based have been created and used to protect biomacromolecular drugs
hydrogel overcame Taxol resistance and enhanced its anti-tumor from deactivation and clearance.[107,168,169] For instance, anti-
efficiency. These studies highlight the versatility of SPH in tumor cancer peptide G(IIKK)3 I-NH2 (G3) would be eliminated from
targeted delivery of small molecules, demonstrating that one or tumors quickly after systemic administration. To improve its effi-
multiple drugs could be feasibly introduced into SPH either co- ciency, Xu and colleagues screened out Ac-I3 SLKG-NH2 through
valently or non-covalently to exert synergistic anticancer effects. rational design to form an MMP-2 degradable SPH for G3

Adv. Healthcare Mater. 2020, 2001239 2001239 (14 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 9. The design and mechanism of action of fibrin-based SPH. A solution containing fibrinogen and aCD47@CaCO3 nanoparticles was applied into
the post-surgery tumor bed with a solution of thrombin, which together formed hydrogel in situ. Under mild acidic tumor microenvironment, CaCO3 was
degraded to elevated pH and release aCD47, which re-polarized TAM and inhibited CD47-mediated immune evasion. Reproduced with permission.[177]
Copyright 2018, Springer Nature.

delivery. The SPH could retain in the tumor for a prolonged pe- juvant to evoke the immune response. The peptide could form
riod, and release G3 in a “cell-demanded” way due to the MMP-2 a hydrogel by simple heating-cooling process and the antigens
responsiveness of SPH.[122] In contrast to this study, Mo and col- like OVA could be mixed into the hydrogel through a vortex.[172]
leagues used enzyme WQ9-2 to initiate SPH formation.[170] The The Nap group could be replaced with flurbiprofen or carprofen,
enzyme-catalyzed a condensation reaction between two precur- nonsteroidal anti-inflammatory drugs (NSAID), and the result-
sors Fmoc-F and FF-Dopa to form hydrogelator Fmoc-FFF-Dopa, ing peptide hydrogels had both antitumor immunity and anti-
which entrapped hirudin (an antiangiogenic protein), and TRAIL inflammatory properties.[173]
(an apoptosis-inducing cytokine) in the hydrogel. This hydrogel Antibodies have long half-lives in circulation and show a high
showed great protein-loading capacity without bioactivity damage affinity for corresponding antigens. However, anticancer antibod-
and provided sustained release in the MDA-MB-231 tumor. ies that can recognize antigens universally expressed through the
In addition to the anticancer peptides and proteins, SPHs have body should be targeted delivered to the tumor. For instance,
also been used to deliver antigens. Premature degradation of off-tumor accumulation of anti-PD-L1 and anti-PD1 antibodies
antigen peptides is one challenge that limits immunization ef- cause severe side effects in the lungs. Another potential tar-
ficiency, which is critical for phosphorylated antigens. Yang et al. get for cancer immunotherapy is the integrin-associated protein
thus created a Ca2+ -induced gelator Nap-GD FD FpD Y to protect (IAP)—CD47 known as a “don’t eat me” signal.[174–176] Although
phosphorylated antigens (TpSN, CpTW, and KpTL) from dephos- CD47 was upregulated in many tumors, it is also regularly ex-
phorylation by in vivo phosphatase.[141] While protecting the anti- pressed on other normal tissue. To limit anti-CD47 antibody in
gens, the D-tetrapeptide-based hydrogel itself also helped to pro- the B16F10 murine melanoma, Gu’ group loaded the anti-CD47
duce antibodies and inducing the immune response. With the antibody in the calcium carbonate nanoparticles and then encap-
help of SPH, the ratio of generated antibodies against phospho- sulated them in the fibrous gel, which formed quickly upon the
rylated peptides increased. Fast clearance is another issue pre- mixture of fibrinogen and thrombin (Figure 9).[177] This treat-
venting effective immunization, as prolonged stimulation is nec- ment changed the tumor microenvironment (pH and immune
essary to maximize antigen presentation. Liu et al. thus created checkpoint), and induced the M1-oriented polarization of tumor-
a peptide hydrogelator (Fbp-GD FD FD YD K(𝛾E)2 -NH2 ), which self- associated macrophages (TAM). High antitumor immunity was
assembled upon the mixture of the gelator with protein anti- realized using this SPH design, without systemic side effects.
gens ovalbumin (OVA) and hepatitis B surface antigen (HBsAg). Nucleic acid-based vaccination has unique advantages because
The formed SPH significantly delayed antigen release and fa- specific antigens can be easily encoded into the sequence with-
cilitated their delivery to antigen-presenting cells, allowing an out tedious protein expression and purification. However, nucleic
effective immune response and repression of the, E.G.,7-OVA acid-based vaccine suffers from enzymatic degradation and lim-
tumor growth.[143] Recently, many peptides are found to be po- ited expression efficiency. As a response to this challenge, Op-
tent adjuvants.[171] Nap-GFFY peptide designed by Yang et al. penheim et al. designed a set of cationic peptide hydrogelators
was one such example and reported to serve as a peptide ad- and found that the HLT2 peptide (net charge = +5 under neutral

Adv. Healthcare Mater. 2020, 2001239 2001239 (15 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 10. Schematic illustration of the mechanism of action of alkaline phosphatase (ALP). The Nap-D FD FD Yp D EMe2 was cleaved by ALP to generate
Nap-D FD FD YD EMe2 , which self-assembled in the cancer cells to induce cell stress and apoptosis. Reproduced with permission.[180] Copyright 2019,
Elsevier Inc.

pH) could efficiently encapsulate a plasmid DNA (TA) encoding with the function of mitochondria. Yang and colleagues created
a melanoma-specific gp100 antigen and adjuvant HMGB1 (high- a dual-responsive hydrogelator (NBD-GFFpY-ss-ERGD), which
mobility group binding protein 1). This hydrogel protected the could transform into NBD-GFFY-ss-ERGD and NBD-GFFY-thiol
DNA from swift enzymatic degradation and fragmentation and subsequently after exposure to extracellular ALP and intracellu-
improved DNA retention and expression. The SPH-based vac- lar GSH.[39] The intracellular hydrogel caused the death of tumor
cine induced an acute inflammatory response, improved infiltra- cells selectively.
tion of macrophages and polymorphonuclear cells, and enhanced
the immune response mediated by CD4+ /IFN𝛾 + expressing Th1
cells and gp100-specific antibodies.[178] 3.4. Combination Delivery

Combination therapy of cancer with small molecular drugs and


3.3. Drug-Free Anticancer SPH biomacromolecules is becoming popular in recent years. This is
because small molecular weight chemotherapeutics are potent
The self-assembly of peptide and its derivatives, not only creates immunogenic cell death (ICD) inducers and biomacromolecules
hydrogels but in some cases endows the molecules with self- are usually effective immune modulators, which together could
assembly-based anticancer activity. Xu and colleagues linked an L - induce prolonged tumor regression. As we discussed above,
amino acid to the C-terminal of a D -tripeptide (Nap-D FD FD YR) to SPHs can provide both hydrophilic network and hydropho-
create a peptide hydrogelator that could assemble into a crescent- bic cavity to accommodate small molecular weight drugs and
shaped supramolecular structure at the presence of ALP.[179] biomacromolecules either covalently or non-covalently, which
They showed that the formed supramolecular structure would make SPH an ideal platform for combination therapy of
damage cell membrane of HeLa and HS-5 cells and interact with cancer.
the endoplasmic reticulum (ER) to induce cell death, which pro-
cess depended on the formation of the crescent-shaped mor-
phology. A similar phenomenon was observed on another ALP- 3.4.1. Combinational Delivery of Small Molecular and
responsive peptide hydrogelator (Nap-D FD FD Yp D EMe2 ), which im- Biomacromolecular Drugs
proved the survival of mice bearing osteosarcoma tumor (Fig-
ure 10).[180] SPH could also be engineered to target mitochon- As we discussed above, many peptides can serve as po-
dria other than ER to kill tumor cells. A feasible strategy to tent adjuvants to potentiate antitumor immune response.
achieve this is conjugating mitochondria-targeting moiety such Inspired by this, immune active peptides were engineered
as triphenyl phosphonium (TPP) to peptide hydrogelators. For onto peptide hydrogelators for co-delivery of small molecu-
instance, Xu et al. modified a pair of enantiomeric tripeptides lar weight ICD inducers. Melittin, a 26-amino-acid polypep-
with TPP to produce phosphorylated tetrapeptides (L-1P or D- tide (GIGAVLKVLTTGLPALISWIKRKRQQ-NH2 ) with the po-
1P).[181] They found that the oligomers could self-assemble on tential to perforate on cell membranes and modulate the
the surface of human osteosarcoma cells at the presence of ALP, innate immune response,[182,183] was the one most investi-
and the hydrogel was internalized by the cancer cells to interfere gated. For instance, Jin et al. created a melittin-containing

Adv. Healthcare Mater. 2020, 2001239 2001239 (16 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

peptide hydrogelator namely melittin-RADA32 ((RADA)8 -GG- for live DC delivery.[42] The SPH was prepared by encapsulating
GIGAVLKVLTTGLPALISWIKRKRQQ-NH2 ) for localized deliv- live dendritic cells (DCs), anti-PD-1 antibodies, and tumor anti-
ery of DOX.[40] The resulting hydrogel showed controlled re- gen ovalbumin (OVA) into RADA16 (Ac-(RADA)4 -CONH2 ) (Fig-
lease of DOX that suppressed the growth of primary B16F10 ure 12a) based scaffold. The SPH served as a mild reservoir for ec-
tumor. In the meanwhile, it also activated DC maturation in togenous DCs where they retained its bioactivity, interacted with
the draining lymph nodes, eliminated alternatively activated OVA, and finally migrated out to tumor site sustainably com-
macrophage, and activated cytotoxic T cells to kill metastatic tu- pared with OVA-pulsed DCs. It also helped to recruit endogenous
mors in vivo. The same group then optimized their molecule de- DCs and activated them by sustained OVA release, thus inducing
sign, and used melittin-(RADA)6 for KN93 (a Ca2+ /calmodulin- much stronger activation of antigen-specific effector T cells to kill
dependent protein kinase II (CAMKII) inhibitor) delivery. In tumor cells (Figure 12b). Therefore, the SPH induced strong pro-
B16F10 mouse melanoma model, melittin-(RADA)6 facilitated tective and therapeutic antitumor response.
ICD by improving the uptake of KN93 through creating tunnels
on cell membranes and induced stronger cell-mediated antitu- 4. Conclusions and Perspectives
mor immunity compared with melittin-(RADA)4 and melittin-
(RADA)8 hydrogels.[184] SPH is a class of biomaterial that closely mimics ECM both in its
Co-delivery of small molecular weight ICD inducer and structure and biological functions. It is mainly composed of short
chemokines is another widely adopted strategy in hydrogel- peptides that can be degraded into natural amino acids, which
based local cancer therapy. For instance, Chen and colleagues nature endows it with good biocompatibility and controllable im-
have developed poly(ethylene glycol)-poly(𝛾-ethyl-l-glutamate) di- munogenicity. Although a short peptide, a decapeptide, for ex-
block copolymers (mPEG-b-PELG) and PELG-PEG-PELG tri- ample, can have a sequence space with >1013 possibilities, sur-
block copolymers for codelivery of ICD inducers (cisplatin or passing most synthetic materials. Compared to the natural ECM,
DOX) and immune modulators (interleukin-15, interleukin-2, or it is self-assembled from simple building blocks through non-
interferon-𝛾).[185,186] The resulting hydrogels showed potent anti- covalent interactions such as 𝜋–𝜋 interaction, hydrogen bonding,
tumor activity via a mechanism involving CD8+ T cells and nat- hydrophobic force, and electrostatic interactions. Thus, different
ural killer (NK) cells. functional moieties can be easily integrated into SPH through a
Enhanced cytotoxic T lymphocyte infiltration triggered by ICD co-assembly strategy to create novel SPHs. With these unique ad-
inducers usually resulted in upregulation of immune check- vantages, SPH has shown great potential in cancer therapy. SPH
points. Thus, combination delivery of ICD inducers and immune has been used as localized delivery vehicles for small molecular
checkpoint inhibitors may be an effective way to enhance anti- weight drugs, biomacromolecules, nanoparticles, and even cells
cancer immunity. To test this concept, Cui and colleagues created through non-covalent or covalent encapsulation mechanisms.
an MMP-2 responsive hydrogelator diCPT-PLGLAG-iRGD (Fig- SPH itself can also directly modulate the function and viabil-
ure 11a), which could self-assemble into supramolecular nan- ity of cancer cells and stromal cells in the TME. With increas-
otubes (P-NTs) and then formed hydrogel with anti-PD1 antibod- ing advances in cancer biology and oncoimmunology, SPH has
ies under physiological conditions (Figure 11b).[80] After intra- also demonstrated its great potential in cancer immunotherapy.
tumoral injection, the diCPT-PLGLAG-iRGD showed better tu- As a result, SPH can selectively eradicate primary tumors, but at
mor penetration compared with the hydrogel formed by diCPT- the same time induce systemic anticancer immunity. Neverthe-
PLGLAG-iRGRD. At the presence of MMP-2 and GSH, free CPT less, some issues must be addressed before the clinical transla-
was released to boost ICD of cancer cells, while at the same time tion of SPH in cancer therapy. First, the key features that deter-
liberate anti-PD1 antibodies. As a result, the SPH boosted T-cell mine the biological activities of SPH mush be understood. Sim-
mediated immune response, leading to the 100% repression of ilar to ECM that can exert both protumoral and antitumoral ef-
tumor growth for all treated mice with either CT 26 colon can- fects, SPH can also exert biological activities of opposite effects.
cer or GL-261 brain cancer and repressed the tumor recurrence Recent studies by Hartgerink and Elisseeff and their colleagues
significantly. showed that the composition of hydrogels has a dramatic effect
on the types of recruited immune cells into the hydrogels and
their biological functions,[131,188] revealing that the charge and ori-
3.4.2. Combinational Delivery of Cells/Cell Debris and Other gin of hydrogels significantly affected their biological functions.
Ingredients However, more function-related features must be identified for
rational SPH design. Second, the dynamic feature of the SPH
SPHs have been widely used in 3D cell culture and tissue engi- must be controllable. SPH is characteristic of its dynamics be-
neering. Recently, SPHs have also been used for local delivery of cause of its supramolecular nature. However, the dynamic tran-
cell debris or live DC for cancer immunotherapy. Li and cowork- sition of SPH is mainly driven by physical and chemical cues,
ers designed an Fmoc-KCRGDK (FK) based SPH for encapsula- which is in sharp contrast with ECM the transitions of which
tion of JQ1 (a BRD4 inhibitor) and indocyanine green (ICG, a are mainly biological cue driven. Strategies that introduce bio-
photosensitizer) loaded dead tumor cells.[187] The resulting per- logical stimuli-responsive linkers into SPH design has been ex-
sonalized cancer vaccine, namely PVAX, could kill cancer cells tensively explored. Recently, Stupp and colleagues explored other
upon NIR irradiation to enhance DC maturation and enhance strategies such as simultaneous covalent and non-covalent hy-
tumor infiltration of CD8+ T lymphocytes, the activity of which brid polymerization and peptide-DNA hybrid building blocks,
could be further improved by the released JQ1. Instead of deliv- which can be potentially used in the design of more intelligent
ering cell debris, Wang and colleagues explored the use of SPH SPH. Third, the optimized combination treatment regime shall

Adv. Healthcare Mater. 2020, 2001239 2001239 (17 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 11. Schematic illustration of the design and mechanism of action of P-NT–aPD1 hydrogel. a) The chemical structure of diCPT-PLGLAG-iRGD.
b) PD1 antibodies could be efficiently entrapped within the network form by diCPT-PLGLAG-iRGD solution. After intratumoral injection, the solution
turned into hydrogel, which released CPT and aPD1 in response to MMP-2 and GSH, exerting combination chemotherapy and immunotherapy against
tumor. Reproduced with permission.[80] Copyright 2020, American Association for the Advancement of Science.

be further pursued. SPH has great potential in co-delivery of ternal organs, clinic-compatible and imaging-guided drug deliv-
multiple cargoes of different chemical and biological properties. ery platform must be established. Lastly, peptides may be rela-
The strategies to realize controlled release of a single therapeu- tively more expensive compared with polymers. The requirement
tic component have been vastly explored. However, the drug re- of protected amino acids during peptide synthesis and the dif-
lease kinetics of the co-delivered drugs in an SPH were usu- ficulty in peptide purification are two major challenges to ob-
ally not optimized. Since combination therapy is a routine prac- tain peptide hydrogelators. Recently, a water/dichloromethane
tice in cancer therapy, it is critical to optimize drug combina- biphasic system with macroinitiators anchored at the interface
tion, dosage, and release profiles to achieve maximized efficacy. was developed by Cheng and colleagues to simplify the impurity
Forth, imaging-guided hydrogel delivery is required to expand treatment process.[189] Method to synthesize polypeptides directly
the application of SPH. Currently, the SPH is mainly applied from amino acids has also been reported by the same group.[190]
in surgical beds, subcutaneous tissues, and tumors that can be With these new and more upcoming technologies, a cost reduc-
easily accessed (e.g., breast cancer). To treat tumors of the in- tion in peptide hydrogelator synthesis is foreseeable in the near

Adv. Healthcare Mater. 2020, 2001239 2001239 (18 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Figure 12. Schematic illustration of the design and mechanism of action of SPH-based vaccine. a) The chemical structure of RADA16 peptide. b) RADA16
peptide could form hydrogel and entrap antigens, anti-PD-1 antibodies, and DC within its network. After subcutaneous injection, the mature DCs would
migrate to the lymph nodes, while the hydrogel could further recruit naïve DC to further improve antigen presentation and CD8 T cell proliferation.
Reproduced with permission.[42] Copyright 2018, American Chemical Society.

future. Despite these challenges, clinical translation of SPH is Received: July 15, 2020
expected in the near future, given its advantages such as tumor Revised: September 4, 2020
targeted drug delivery, low systemic drug exposure, the capability Published online:
to induce systemic antitumor immunity, and realize combination
therapy, as well as good biocompatibility.

Acknowledgements [1] R. O. Hynes, Science 2009, 326, 1216.


[2] V. Z. Beachley, M. T. Wolf, K. Sadtler, S. S. Manda, H. Jacobs, M.
Y.C. and C.Z. contributed equally to this work. The authors thank the R. Blatchley, J. S. Bader, A. Pandey, D. Pardoll, J. H. Elisseeff, Nat.
National Natural Science Foundation of China (31870995, 81690265, Methods 2015, 12, 1197.
81671808, 81630052), the Youth Innovation Promotion Association of CAS [3] S. Hinderer, S. L. Layland, K. Schenke-Layland, Adv. Drug Delivery
(2017335), and the SA-SIBS Scholarship Program for financial support. Rev. 2016, 97, 260.
[4] G. S. Hussey, J. L. Dziki, S. F. Badylak, Nat. Rev. Mater. 2018, 3, 159.
Conflict of Interest [5] M. D. Davidson, J. A. Burdick, R. G. Wells, Adv. Healthcare Mater.
2020, 9, 1901682.
The authors declare no conflict of interest. [6] T. W. Gilbert, T. L. Sellaro, S. F. Badylak, Biomaterials 2006, 27, 3675.
[7] J. P. Guyette, S. E. Gilpin, J. M. Charest, L. F. Tapias, X. Ren, H. C.
Keywords Ott, Nat. Protoc. 2014, 9, 1451.
[8] M. P. Lutolf, J. A. Hubbell, Nat. Biotechnol. 2005, 23, 47.
cancer, hydrogels, local therapy, peptides, supramolecular self-assembly [9] D. Seliktar, Science 2012, 336, 1124.

Adv. Healthcare Mater. 2020, 2001239 2001239 (19 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

[10] N. Annabi, A. Tamayol, J. A. Uquillas, M. Akbari, L. E. Bertassoni, C. [45] N. Habibi, N. Kamaly, A. Memic, H. Shafiee, Nano Today 2016, 11,
Cha, G. Camci-Unal, M. R. Dokmeci, N. A. Peppas, A. Khademhos- 41.
seini, Adv. Mater. 2014, 26, 85. [46] D. E. Meyer, A. Chilkoti, Nat. Biotechnol. 1999, 17, 1112.
[11] S. J. Buwalda, K. W. M. Boere, P. J. Dijkstra, J. Feijen, T. Vermonden, [47] M. T. Manzari, G. R. Anderson, K. H. Lin, R. S. Soderquist, M. Çakir,
W. E. Hennink, J. Controlled Release 2014, 190, 254. M. Zhang, C. E. Moore, R. N. Skelton, M. Fèvre, X. Li, J. J. Bellucci,
[12] H. R. Culver, J. R. Clegg, N. A. Peppas, Acc. Chem. Res. 2017, 50, S. E. Wardell, S. A. Costa, K. C. Wood, A. Chilkoti, Sci. Adv. 2019, 5,
170. eaaw9162.
[13] O. J. G. M. Goor, S. I. S. Hendrikse, P. Y. W. Dankers, E. W. Meijer, [48] S. Roberts, M. Dzuricky, A. Chilkoti, FEBS Lett. 2015, 589, 2477.
Chem. Soc. Rev. 2017, 46, 6621. [49] M. D. Shoulders, R. T. Raines, Annu. Rev. Biochem. 2009, 78, 929.
[14] J. J. Green, J. H. Elisseeff, Nature 2016, 540, 386. [50] J. A. Fallas, L. E. O’Leary, J. D. Hartgerink, Chem. Soc. Rev. 2010, 39,
[15] J. Li, L. T. Mo, C. H. Lu, T. Fu, H. H. Yang, W. H. Tan, Chem. Soc. Rev. 3510.
2016, 45, 1410. [51] H. Yokoi, T. Kinoshita, S. Zhang, Proc. Natl. Acad. Sci. U. S. A. 2005,
[16] Y. L. Li, J. Rodrigues, H. Tomas, Chem. Soc. Rev. 2012, 41, 2193. 102, 8414.
[17] F. Zhang, M. W. King, Adv. Healthcare Mater. 2020, 9, 1901358. [52] J. K. Kretsinger, L. A. Haines, B. Ozbas, D. J. Pochan, J. P. Schneider,
[18] M. J. Webber, E. A. Appel, E. W. Meijer, R. Langer, Nat. Mater. 2016, Biomaterials 2005, 26, 5177.
15, 13. [53] L. Haines-Butterick, K. Rajagopal, M. Branco, D. Salick, R. Rughani,
[19] A. Harada, Y. Takashima, M. Nakahata, Acc. Chem. Res. 2014, 47, M. Pilarz, M. S. Lamm, D. J. Pochan, J. P. Schneider, Proc. Natl. Acad.
2128. Sci. U. S. A. 2007, 104, 7791.
[20] A. M. Rosales, K. S. Anseth, Nat. Rev. Mater. 2016, 1, 15012. [54] K. Rajagopal, M. S. Lamm, L. A. Haines-Butterick, D. J. Pochan, J. P.
[21] Y. Shao, H. Y. Jia, T. Y. Cao, D. S. Liu, Acc. Chem. Res. 2017, 50, 659. Schneider, Biomacromolecules 2009, 10, 2619.
[22] T. Aida, E. W. Meijer, S. I. Stupp, Science 2012, 335, 813. [55] J. Shi, G. Fichman, J. P. Schneider, Angew. Chem., Int. Ed. 2018, 57,
[23] X. Yan, F. Wang, B. Zheng, F. Huang, Chem. Soc. Rev. 2012, 41, 6042. 11188.
[24] S. Eskandari, T. Guerin, I. Toth, R. J. Stephenson, Adv. Drug Delivery [56] E. F. Banwell, E. S. Abelardo, D. J. Adams, M. A. Birchall, A. Corrigan,
Rev. 2017, 110, 169. A. M. Donald, M. Kirkland, L. C. Serpell, M. F. Butler, D. N. Woolfson,
[25] J. Li, X. Li, Y. Kuang, Y. Gao, X. Du, J. Shi, B. Xu, Adv. Healthcare Nat. Mater. 2009, 8, 596.
Mater. 2013, 2, 1586. [57] M. Reches, E. Gazit, Science 2003, 300, 625.
[26] Y. Li, F. H. Wang, H. G. Cui, Bioeng. Transl. Med. 2016, 1, 306. [58] C. G. Pappas, R. Shafi, I. R. Sasselli, H. Siccardi, T. Wang, V. Narang,
[27] K. Sato, M. P. Hendricks, L. C. Palmer, S. I. Stupp, Chem. Soc. Rev. R. Abzalimov, N. Wijerathne, R. V. Ulijn, Nat. Nanotechnol. 2016, 11,
2018, 47, 7539. 960.
[28] R. J. Wojtecki, M. A. Meador, S. J. Rowan, Nat. Mater. 2011, 10, 14. [59] J. E. Smotrys, M. E. Linder, Annu. Rev. Biochem. 2004, 73, 559.
[29] L. E. R. O’Leary, J. A. Fallas, E. L. Bakota, M. K. Kang, J. D. Hartgerink, [60] J. D. Hartgerink, E. Beniash, S. I. Stupp, Science 2001, 294, 1684.
Nat. Chem. 2011, 3, 821. [61] H. Jiang, M. O. Guler, S. I. Stupp, Soft Matter 2007, 3, 454.
[30] V. A. Kumar, N. L. Taylor, S. Y. Shi, B. K. Wang, A. A. Jalan, M. K. [62] M. J. Sis, M. J. Webber, Trends Pharmacol. Sci. 2019, 40, 747.
Kang, N. C. Wickremasinghe, J. D. Hartgerink, ACS Nano 2015, 9, [63] E. T. Pashuck, S. I. Stupp, J. Am. Chem. Soc. 2010, 132, 8819.
860. [64] J. D. Hartgerink, E. Beniash, S. I. Stupp, Proc. Natl. Acad. Sci. U. S.
[31] M. Ikeda, T. Tanida, T. Yoshii, K. Kurotani, S. Onogi, K. Urayama, I. A. 2002, 99, 5133.
Hamachi, Nat. Chem. 2014, 6, 511. [65] V. Castelletto, C. J. C. Edwards-Gayle, F. Greco, I. W. Hamley, J.
[32] P. Chakraborty, T. Guterman, N. Adadi, M. Yadid, T. Brosh, L. Adler- Seitsonen, J. Ruokolainen, ACS Appl. Mater. Interfaces 2019, 11,
Abramovich, T. Dvir, E. Gazit, ACS Nano 2019, 13, 163. 33573.
[33] J. D. Tovar, Acc. Chem. Res. 2013, 46, 1527. [66] V. Castelletto, A. Kaur, R. M. Kowalczyk, I. W. Hamley, M. Reza, J.
[34] Y. Kuang, J. F. Shi, J. Li, D. Yuan, K. A. Alberti, Q. B. Xu, B. Xu, Angew. Ruokolainen, Biomacromolecules 2017, 18, 2013.
Chem., Int. Ed. 2014, 53, 8104. [67] A. Mahler, M. Reches, M. Rechter, S. Cohen, E. Gazit, Adv. Mater.
[35] X. Liu, X. H. Chen, M. X. Chua, Z. B. Li, X. J. Loh, Y. L. Wu, Adv. 2006, 18, 1365.
Healthcare Mater. 2017, 6, 1700159. [68] A. M. Smith, R. J. Williams, C. Tang, P. Coppo, R. F. Collins, M. L.
[36] Y. H. Wu, H. B. Wang, F. Gao, Z. Y. Xu, F. Y. Dai, W. G. Liu, Adv. Funct. Turner, A. Saiani, R. V. Ulijn, Adv. Mater. 2008, 20, 37.
Mater. 2018, 28, 1801000. [69] A. Rajbhandary, D. M. Raymond, B. L. Nilsson, Langmuir 2017, 33,
[37] H. E. Xu, T. T. Wang, C. B. Yang, X. L. Li, G. Liu, Z. M. Yang, P. K. 5803.
Singh, S. Krishnan, D. Ding, Adv. Funct. Mater. 2018, 28, 1707140. [70] V. Jayawarna, M. Ali, T. A. Jowitt, A. F. Miller, A. Saiani, J. E. Gough,
[38] Z. M. Yang, G. L. Liang, L. Wang, B. Xu, J. Am. Chem. Soc. 2006, 128, R. V. Ulijn, Adv. Mater. 2006, 18, 611.
3038. [71] M. Hughes, S. Debnath, C. W. Knapp, R. V. Ulijn, Biomater. Sci. 2013,
[39] J. Zhan, Y. B. Cai, S. S. He, L. Wang, Z. M. Yang, Angew. Chem., Int. 1, 1138.
Ed. 2018, 57, 1813. [72] P. Chakraborty, Y. Tang, T. Yamamoto, Y. Yao, T. Guterman, S.
[40] H. L. Jin, C. Wan, Z. W. Zou, G. F. Zhao, L. L. Zhang, Y. Y. Geng, T. Zilberzwige-Tal, N. Adadi, W. Ji, T. Dvir, A. Ramamoorthy, G. Wei,
Chen, A. Huang, F. G. Jiang, J. P. Feng, J. F. Lovell, J. Chen, G. Wu, E. Gazit, Adv. Mater. 2020, 32, 1906043.
K. Y. Yang, ACS Nano 2018, 12, 3295. [73] Z. Yang, G. Liang, M. Ma, Y. Gao, B. Xu, J. Mater. Chem. 2007, 17,
[41] D. G. Leach, N. Dharmaraj, S. L. Piotrowski, T. L. Lopez-Silva, Y. L. 850.
Lei, A. G. Sikora, S. Young, J. D. Hartgerink, Biomaterials 2018, 163, [74] H. Wang, Z. Luo, Y. Wang, T. He, C. Yang, C. Ren, L. Ma, C. Gong, X.
67. Li, Z. Yang, Adv. Funct. Mater. 2016, 26, 1822.
[42] P. X. Yang, H. J. Song, Y. B. Qin, P. S. Huang, C. N. A. Zhang, D. L. [75] S. Biswas, M. Kumar, A. M. Levine, I. Jimenez, R. V. Ulijn, A. B. Braun-
Kong, W. W. Wang, Nano Lett. 2018, 18, 4377. schweig, Chem. Sci. 2020, 11, 4239.
[43] Q. Chen, M. Chen, Z. Liu, Chem. Soc. Rev. 2019, 48, 5506. [76] F. Li, J. Han, T. Cao, W. Lam, B. Fan, W. Tang, S. Chen, K. L. Fok, L.
[44] P. W. Frederix, G. G. Scott, Y. M. Abul-Haija, D. Kalafatovic, C. G. Li, Proc. Natl. Acad. Sci. U. S. A. 2019, 116, 11259.
Pappas, N. Javid, N. T. Hunt, R. V. Ulijn, T. Tuttle, Nat. Chem. 2015, [77] A. G. Cheetham, P. Zhang, Y. A. Lin, L. L. Lock, H. Cui, J. Am. Chem.
7, 30. Soc. 2013, 135, 2907.

Adv. Healthcare Mater. 2020, 2001239 2001239 (20 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

[78] P. Schiapparelli, P. Zhang, M. Lara-Velazquez, H. Guerrero-Cazares, [109] M. P. Conte, J. K. Sahoo, Y. M. Abul-Haija, K. H. A. Lau, R. V. Ulijn,
R. Lin, H. Su, R. W. Chakroun, M. Tusa, A. Quinones-Hinojosa, H. ACS Appl. Mater. Interfaces 2018, 10, 3069.
Cui, J. Controlled Release 2020, 319, 311. [110] E. V. Alakpa, V. Jayawarna, A. Lampel, K. V. Burgess, C. C. West, S.
[79] F. Wang, H. Su, D. Xu, W. Dai, W. Zhang, Z. Wang, C. F. Anderson, C. J. Bakker, S. Roy, N. Javid, S. Fleming, D. A. Lamprou, J. Yang, A.
M. Zheng, R. Oh, F. Wan, H. Cui, Nat. Biomed. Eng. 2020, https: Miller, A. J. Urquhart, P. W. J. M. Frederix, N. T. Hunt, B. Péault, R.
//doi.org/10.1038/s41551-020-0597-7. V. Ulijn, M. J. Dalby, Chem 2016, 1, 298.
[80] F. Wang, D. Xu, H. Su, W. Zhang, X. Sun, M. K. Monroe, R. W. [111] M. Ghosh, M. Halperin-Sternfeld, I. Grigoriants, J. Lee, K. T. Nam,
Chakroun, Z. Wang, W. Dai, R. Oh, H. Wang, Q. Fan, F. Wan, H. L. Adler-Abramovich, Biomacromolecules 2017, 18, 3541.
Cui, Sci. Adv. 2020, 6, eaaz8985. [112] M. A. Khalily, M. Goktas, M. O. Guler, Org. Biomol. Chem. 2015, 13,
[81] Y. Sun, J. A. Kaplan, A. Shieh, H. L. Sun, C. M. Croce, M. W. Grinstaff, 1983.
J. R. Parquette, Chem. Commun. 2016, 52, 5254. [113] H. Wang, Z. Feng, A. Lu, Y. Jiang, H. Wu, B. Xu, Angew. Chem., Int.
[82] C. Liang, D. Zheng, F. Shi, T. Xu, C. Yang, J. Liu, L. Wang, Z. Yang, Ed. 2017, 56, 7579.
Nanoscale 2017, 9, 11987. [114] M. T. He, J. B. Li, S. Tan, R. Z. Wang, Y. Zhang, J. Am. Chem. Soc.
[83] J. Li, Y. Kuang, Y. Gao, X. Du, J. Shi, B. Xu, J. Am. Chem. Soc. 2013, 2013, 135, 18718.
135, 542. [115] Z. J. Qiu, H. T. Yu, J. B. Li, Y. Wang, Y. Zhang, Chem. Commun. 2009,
[84] M. Tao, S. He, J. Liu, H. Li, L. Mei, C. Wu, K. Xu, W. Zhong, J. Mater. 3342.
Chem. B 2019, 7, 469. [116] Y. Qiao, X. Liu, X. Zhou, H. Zhang, W. Zhang, W. Xiao, G. Pan, W.
[85] Z. Chen, L. Xing, Q. Fan, A. G. Cheetham, R. Lin, B. Holt, L. Chen, Cui, H. A. Santos, Q. Shi, Adv. Healthcare Mater. 2020, 9, 1901239.
Y. Xiao, H. Cui, Theranostics 2017, 7, 2003. [117] R. V. Rughani, M. C. Branco, D. J. Pochan, J. P. Schneider, Macro-
[86] R. W. Chakroun, A. Sneider, C. F. Anderson, F. Wang, P. H. Wu, D. molecules 2010, 43, 7924.
Wirtz, H. Cui, Angew. Chem., Int. Ed. 2020, 59, 4434. [118] H. Y. Liu, H. D. Nguyen, C. C. Lin, Adv. Healthcare Mater. 2018, 7,
[87] S. E. Paramonov, H. W. Jun, J. D. Hartgerink, J. Am. Chem. Soc. 2006, 1800954.
128, 7291. [119] T. M. Doran, D. M. Ryan, B. L. Nilsson, Polym. Chem. 2014, 5, 241.
[88] V. Basavalingappa, T. Guterman, Y. Tang, S. Nir, J. Lei, P. [120] H. Shigemitsu, T. Fujisaku, S. Onogi, T. Yoshii, M. Ikeda, I. Hamachi,
Chakraborty, L. Schnaider, M. Reches, G. Wei, E. Gazit, Adv. Sci. Nat. Protoc. 2016, 11, 1744.
2019, 6, 1900218. [121] D. Zheng, Z. Gao, T. Xu, C. Liang, Y. Shi, L. Wang, Z. Yang, Nanoscale
[89] H. P. Mangunuru, H. Yang, G. Wang, Chem. Commun. 2013, 49, 2018, 10, 21459.
4489. [122] C. Chen, Y. Zhang, Z. Hou, X. Cui, Y. Zhao, H. Xu, Biomacromolecules
[90] R. A. Petros, J. M. DeSimone, Nat. Rev. Drug Discovery 2010, 9, 615. 2017, 18, 3563.
[91] A. E. Nel, L. Madler, D. Velegol, T. Xia, E. M. Hoek, P. Somasundaran, [123] W. Ji, Z. Alvarez, A. N. Edelbrock, K. Sato, S. I. Stupp, ACS Appl.
F. Klaessig, V. Castranova, M. Thompson, Nat. Mater. 2009, 8, 543. Mater. Interfaces 2018, 10, 41046.
[92] H. Cui, A. G. Cheetham, E. T. Pashuck, S. I. Stupp, J. Am. Chem. Soc. [124] H. Li, J. Gao, Y. Shang, Y. Hua, M. Ye, Z. Yang, C. Ou, M. Chen, ACS
2014, 136, 12461. Appl. Mater. Interfaces 2018, 10, 24459.
[93] H. Su, W. Zhang, H. Wang, F. Wang, H. Cui, J. Am. Chem. Soc. 2019, [125] B. Cheng, Y. Yan, J. Qi, L. Deng, Z. W. Shao, K. Q. Zhang, B. Li, Z.
141, 11997. Sun, X. Li, ACS Appl. Mater. Interfaces 2018, 10, 12474.
[94] C. Hu, X. Liu, W. Ran, J. Meng, Y. Zhai, P. Zhang, Q. Yin, H. Yu, Z. [126] H. Tao, Y. Wu, H. Li, C. Wang, Y. Zhang, C. Li, T. Wen, X. Wang, Q.
Zhang, Y. Li, Biomaterials 2017, 144, 60. He, D. Wang, D. Ruan, ACS Appl. Mater. Interfaces 2015, 7, 17076.
[95] O. Chaudhuri, J. Cooper-White, P. A. Janmey, D. J. Mooney, V. B. [127] M. Zhou, A. M. Smith, A. K. Das, N. W. Hodson, R. F. Collins, R. V.
Shenoy, Nature 2020, 584, 535. Ulijn, J. E. Gough, Biomaterials 2009, 30, 2523.
[96] E. Prince, E. Kumacheva, Nat. Rev. Mater. 2019, 4, 99. [128] A. Brito, Y. M. Abul-Haija, D. S. da Costa, R. Novoa-Carballal, R.
[97] O. Chaudhuri, L. Gu, D. Klumpers, M. Darnell, S. A. Bencherif, J. L. Reis, R. V. Ulijn, R. A. Pires, I. Pashkuleva, Chem. Sci. 2019, 10,
C. Weaver, N. Huebsch, H. P. Lee, E. Lippens, G. N. Duda, D. J. 2385.
Mooney, Nat. Mater. 2016, 15, 326. [129] A. N. Moore, T. L. Lopez Silva, N. C. Carrejo, C. A. Origel Marmolejo,
[98] Z. H. Yang, H. Y. Xu, X. J. Zhao, Adv. Sci. 2020, 7, 1903718. I. C. Li, J. D. Hartgerink, Biomaterials 2018, 161, 154.
[99] E. T. Pashuck, H. Cui, S. I. Stupp, J. Am. Chem. Soc. 2010, 132, 6041. [130] N. C. Carrejo, A. N. Moore, T. L. Lopez Silva, D. G. Leach, I. C. Li, D.
[100] C. Chen, Y. Gu, L. Deng, S. Han, X. Sun, Y. Chen, J. R. Lu, H. Xu, ACS R. Walker, J. D. Hartgerink, ACS Biomater. Sci. Eng. 2018, 4, 1386.
Appl. Mater. Interfaces 2014, 6, 14360. [131] T. L. Lopez-Silva, D. G. Leach, A. Azares, I. C. Li, D. G. Woodside, J.
[101] A. Ishida, G. Watanabe, M. Oshikawa, I. Ajioka, T. Muraoka, Chem- D. Hartgerink, Biomaterials 2020, 231, 119667.
istry 2019, 25, 13523. [132] R. Huang, W. Qi, L. Feng, R. Su, Z. He, Soft Matter 2011, 7, 6222.
[102] N. Hauptstein, L. M. de Leon-Rodriguez, A. K. Mitra, Y. Hemar, I. [133] X.-D. Xu, L. Liang, C.-S. Chen, B. Lu, N.-l. Wang, F.-G. Jiang, X.-Z.
Kavianinia, N. Li, V. Castelletto, I. W. Hamley, M. A. Brimble, ACS Zhang, R.-X. Zhuo, ACS Appl. Mater. Interfaces 2010, 2, 2663.
Biomater. Sci. Eng. 2018, 4, 2733. [134] P. Majumder, U. Baxa, S. T. R. Walsh, J. P. Schneider, Angew. Chem.,
[103] Y.-A. Lin, M. Kang, W.-C. Chen, Y.-C. Ou, A. G. Cheetham, P.-H. Wu, Int. Ed. 2018, 57, 15040.
D. Wirtz, S. M. Loverde, H. Cui, Biomater. Sci. 2018, 6, 216. [135] S. Koutsopoulos, L. D. Unsworth, Y. Nagai, S. Zhang, Proc. Natl.
[104] K. J. Nagy, M. C. Giano, A. Jin, D. J. Pochan, J. P. Schneider, J. Am. Acad. Sci. U. S. A. 2009, 106, 4623.
Chem. Soc. 2011, 133, 14975. [136] S. E. Miller, Y. Yamada, N. Patel, E. Suarez, C. Andrews, S. Tau, B. T.
[105] R. Li, C. C. Horgan, B. Long, A. L. Rodriguez, L. Mather, C. J. Barrow, Luke, R. E. Cachau, J. P. Schneider, ACS Cent. Sci. 2019, 5, 1750.
D. R. Nisbet, R. J. Williams, RSC Adv. 2015, 5, 301. [137] Q. Zhang, J. Wu, J. Wang, X. Wang, C. Wu, M. Chen, Q. Wu, M. S.
[106] J. M. Godbe, R. Freeman, L. F. Burbulla, J. Lewis, D. Krainc, S. I. Lesniak, Y. Mi, Y. Cheng, Q. Wang, Angew. Chem., Int. Ed. 2020, 59,
Stupp, ACS Biomater. Sci. Eng. 2020, 6, 1196. 3732.
[107] Y. Zhao, Z. Wang, C. Mei, Z. Jiang, Y. Feng, R. Gao, Q. Wang, J. [138] D. M. Raymond, B. L. Abraham, T. Fujita, M. J. Watrous, E. S. Toriki,
Huang, Bioconjugate Chem. 2018, 29, 1519. T. Takano, B. L. Nilsson, ACS Appl. Bio Mater. 2019, 2, 2116.
[108] J. T. M. DiMaio, T. M. Doran, D. M. Ryan, D. M. Raymond, B. L. [139] J. E. Sun, B. Stewart, A. Litan, S. J. Lee, J. P. Schneider, S. A. Lang-
Nilsson, Biomacromolecules 2017, 18, 3591. hans, D. J. Pochan, Biomater. Sci. 2016, 4, 839.

Adv. Healthcare Mater. 2020, 2001239 2001239 (21 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

[140] R. R. Pompano, J. Chen, E. A. Verbus, H. Han, A. Fridman, T. Mc- [167] C. Ren, Y. Gao, Y. Guan, Z. Wang, L. Yang, J. Gao, H. Fan, J. Liu, ACS
Neely, J. H. Collier, A. S. Chong, Adv. Healthcare Mater. 2014, 3, Appl. Mater. Interfaces 2019, 11, 33706.
1898. [168] M. C. Branco, D. J. Pochan, N. J. Wagner, J. P. Schneider, Biomaterials
[141] Y. Wang, X. Li, Y. Zhang, L. Wang, Z. Yang, Chem. Commun. 2019, 2010, 31, 9527.
55, 12388. [169] V. A. Kumar, N. L. Taylor, S. Shi, N. C. Wickremasinghe, R. N.
[142] C. Liang, L. Zhang, W. Zhao, L. Xu, Y. Chen, J. Long, F. Wang, L. D’Souza, J. D. Hartgerink, Biomaterials 2015, 52, 71.
Wang, Z. Yang, Adv. Healthcare Mater. 2018, 7, 1800899. [170] T. Jiang, S. Shen, T. Wang, M. Li, B. He, R. Mo, Nano Lett. 2017, 17,
[143] Z. Wang, Y. Shang, Z. Tan, X. Li, G. Li, C. Ren, F. Wang, Z. Yang, J. 7447.
Liu, Theranostics 2020, 10, 657. [171] Y. Cai, W. Ran, Y. Zhai, J. Wang, C. Zheng, Y. Li, P. Zhang, Biomater.
[144] L. Mei, K. Xu, Z. Zhai, S. He, T. Zhu, W. Zhong, Org. Biomol. Chem. Sci. 2020, 8, 1045.
2019, 17, 3853. [172] Z. Luo, Q. Wu, C. Yang, H. Wang, T. He, Y. Wang, Z. Wang, H. Chen,
[145] C. Wu, J. Liu, Z. Zhai, L. Yang, X. Tang, L. Zhao, K. Xu, W. Zhong, X. Li, C. Gong, Z. Yang, Adv. Mater. 2017, 29, 1601776.
Acta Biomater. 2020, 106, 278. [173] Z. Wang, C. Liang, F. Shi, T. He, C. Gong, L. Wang, Z. Yang, Nanoscale
[146] Y. Chen, X. Li, J. Bai, F. Shi, T. Xu, Q. Gong, Z. Yang, Chem. Commun. 2017, 9, 14058.
2018, 54, 11721. [174] S. J. Gardai, K. A. McPhillips, S. C. Frasch, W. J. Janssen, A. Starefeldt,
[147] S. He, L. Mei, C. Wu, M. Tao, Z. Zhai, K. Xu, W. Zhong, Nanoscale J. E. Murphy-Ullrich, D. L. Bratton, P.-A. Oldenborg, M. Michalak, P.
2019, 11, 5030. M. Henson, Cell 2005, 123, 321.
[148] R. W. Chakroun, F. Wang, R. Lin, Y. Wang, H. Su, D. Pompa, H. Cui, [175] A. D. Michaels, T. E. Newhook, S. J. Adair, S. Morioka, B. J.
ACS Nano 2019, 13, 7780. Goudreau, S. Nagdas, M. G. Mullen, J. B. Persily, T. N. J. Bullock,
[149] L. L. Lock, A. G. Cheetham, P. C. Zhang, H. G. Cui, ACS Nano 2013, C. L. Slingluff, K. S. Ravichandran, J. T. Parsons, T. W. Bauer, Clin.
7, 4924. Cancer Res. 2018, 24, 1415.
[150] D. G. Leach, J. M. Newton, M. A. Florez, T. L. Lopez-Silva, A. A. Jones, [176] N. G. Ring, D. Herndler-Brandstetter, K. Weiskopf, L. Shan, J.-P. Volk-
S. Young, A. G. Sikora, J. D. Hartgerink, ACS Biomater. Sci. Eng. 2019, mer, B. M. George, M. Lietzenmayer, K. M. McKenna, T. J. Naik, A.
5, 6755. McCarty, Y. Zheng, A. M. Ring, R. A. Flavell, I. L. Weissman, Proc.
[151] X. Li, Y. Wang, S. Wang, C. Liang, G. Pu, Y. Chen, L. Wang, H. Xu, Y. Natl. Acad. Sci. U. S. A. 2017, 114, E10578.
Shi, Z. Yang, Nanoscale 2020, 12, 2111. [177] Q. Chen, C. Wang, X. Zhang, G. Chen, Q. Hu, H. Li, J. Wang, D.
[152] K. Lei, L. Tang, Biomater. Sci. 2019, 7, 733. Wen, Y. Zhang, Y. Lu, G. Yang, C. Jiang, J. Wang, G. Dotti, Z. Gu,
[153] J. L. Li, R. R. Xing, S. Bai, X. H. Yan, Soft Matter 2019, 15, 1704. Nat. Nanotechnol. 2019, 14, 89.
[154] G. Cinar, A. Ozdemir, S. Hamsici, G. Gunay, A. Dana, A. B. Tekinay, [178] S. H. Medina, S. Li, O. M. Z. Howard, M. Dunlap, A. Trivett, J. P.
M. O. Guler, Biomater. Sci. 2016, 5, 67. Schneider, J. J. Oppenheim, Biomaterials 2015, 53, 545.
[155] Y. Qi, H. Min, A. Mujeeb, Y. Zhang, X. Han, X. Zhao, G. J. Anderson, [179] Z. Feng, H. Wang, S. Wang, Q. Zhang, X. Zhang, A. A. Rodal, B. Xu,
Y. Zhao, G. Nie, ACS Appl. Mater. Interfaces 2018, 10, 6972. J. Am. Chem. Soc. 2018, 140, 9566.
[156] D. Kalafatovic, M. Nobis, J. Son, K. I. Anderson, R. V. Ulijn, Bioma- [180] Z. Q. Q. Feng, X. G. Han, H. M. Wang, T. T. Tang, B. Xu, Chem 2019,
terials 2016, 98, 192. 5, 2442.
[157] Y. Yin, C. Wu, J. Wang, F. Song, W. Yue, W. Zhong, Chem. Commun. [181] H. Wang, Z. Feng, Y. Wang, R. Zhou, Z. Yang, B. Xu, J. Am. Chem.
2017, 53, 529. Soc. 2016, 138, 16046.
[158] X. Zhao, X. Liu, P. Zhang, Y. Liu, W. Ran, Y. Cai, J. Wang, Y. Zhai, G. [182] G. Lee, H. Bae, Molecules 2016, 21, 616.
Wang, Y. Ding, Y. Li, Acta Pharm. Sin. B 2019, 9, 1050. [183] H. Kubo, D. A. Loegering, Y. Tohda, J. Bankers-Fulbright, C. R. Weiler,
[159] C. Wu, R. Li, Y. Yin, J. Wang, L. Zhang, W. Zhong, Mater. Sci. Eng., C H. Nakajima, L. L. Thomas, C. R. Adolphson, G. J. Gleich, J. Im-
2017, 76, 196. munol. Methods 1999, 227, 1.
[160] T. Jiang, T. Wang, T. Li, Y. Ma, S. Shen, B. He, R. Mo, ACS Nano 2018, [184] X. Dai, J. Meng, S. Deng, L. Zhang, C. Wan, L. Lu, J. Huang, Y. Hu,
12, 9693. Z. Zhang, Y. Li, J. F. Lovell, G. Wu, K. Yang, H. Jin, Theranostics 2020,
[161] M. Abbas, R. R. Xing, N. Zhang, Q. L. Zou, X. H. Yan, ACS Biomater. 10, 3049.
Sci. Eng. 2018, 4, 2046. [185] X. Wu, Y. Wu, H. Ye, S. Yu, C. He, X. Chen, J. Controlled Release 2017,
[162] R. Mukerji, J. Schaal, X. Li, J. Bhattacharyya, D. Asai, M. R. Zalutsky, 255, 81.
A. Chilkoti, W. Liu, Biomaterials 2016, 79, 79. [186] Q. Lv, C. He, F. Quan, S. Yu, X. Chen, Bioact. Mater. 2018, 3, 118.
[163] C. Karavasili, D. A. Andreadis, O. L. Katsamenis, E. Panteris, P. [187] T. Wang, D. Wang, H. Yu, B. Feng, F. Zhou, H. Zhang, L. Zhou, S.
Anastasiadou, Z. Kakazanis, V. Zoumpourlis, C. K. Markopoulou, S. Jiao, Y. Li, Nat. Commun. 2018, 9, 1532.
Koutsopoulos, I. S. Vizirianakis, D. G. Fatouros, Mol. Pharmaceutics [188] K. Sadtler, M. T. Wolf, S. Ganguly, C. A. Moad, L. Chung, S. Majum-
2019, 16, 2326. dar, F. Housseau, D. M. Pardoll, J. H. Elisseeff, Biomaterials 2019,
[164] C. Karavasili, E. Panteris, I. S. Vizirianakis, S. Koutsopoulos, D. G. 192, 405.
Fatouros, Pharm. Res. 2018, 35, 166. [189] Z. Song, H. Fu, J. Wang, J. Hui, T. Xue, L. A. Pacheco, H. Yan, R.
[165] K. N. Sugahara, T. Teesalu, P. P. Karmali, V. R. Kotamraju, L. Agemy, Baumgartner, Z. Wang, Y. Xia, X. Wang, L. Yin, C. Chen, J. Rodríguez-
O. M. Girard, D. Hanahan, R. F. Mattrey, E. Ruoslahti, Cancer Cell López, A. L. Ferguson, Y. Lin, J. Cheng, Proc. Natl. Acad. Sci. U. S. A.
2009, 16, 510. 2019, 116, 10658.
[166] F. Wang, H. Su, R. Lin, R. W. Chakroun, M. K. Monroe, Z. Wang, M. [190] T. Xue, Z. Song, Y. Wang, B. Zhu, Z. Zhao, Z. Tan, X. Wang, Y. Xia, J.
Porter, H. Cui, ACS Nano 2020, 14, 10083. Cheng, Macromolecules 2020, 53, 6589.

Adv. Healthcare Mater. 2020, 2001239 2001239 (22 of 23) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.advhealthmat.de

Yaping Li is a principal investigator and director in the Center of Pharmaceutics, Shanghai Institute
of Materia Medica, Chinese Academy of Sciences. He has made a series of contributions to the basic
research and clinic translation of targeted drug delivery systems, including the design, construction,
and mechanistic study of novel nanosized drug delivery systems with higher efficacy and less toxicity
for the treatment of metastatic and multi-drug resistant cancer.

Pengcheng Zhang is a professor at the Shanghai Institute of Materia Medica, Chinese Academy of
Sciences. His research interest mainly focuses on the development of nanomedicines using self-
assembling and biomimetic strategies.

Adv. Healthcare Mater. 2020, 2001239 2001239 (23 of 23) © 2020 Wiley-VCH GmbH

You might also like