You are on page 1of 32

REVIEW

Protein Delivery www.advmat.de

Rational Design of Nanocarriers for Intracellular Protein


Delivery
Xiaofei Qin, Changmin Yu, Jing Wei, Lin Li,* Chengwu Zhang, Qiong Wu, Jinhua Liu,
Shao Q. Yao,* and Wei Huang*

or abnormal in the body. For example, the


Protein/antibody therapeutics have exhibited the advantages of high speci- insulin which were approved by the US
ficity and activity even at an extremely low concentration compared to small Food and Drug Administration (FDA) in
molecule drugs. However, they are accompanied by unfavorable physico- 1982 are the most effective protein thera-
chemical properties such as fragile tertiary structure, large molecular size, pies for the treatment of diabetes mel-
litus type I and type II. In addition, the
and poor penetration of the membrane, and thus the clinical use of protein
protein therapeutics are also utilized for
drugs is hindered by inefficient delivery of proteins into the host cells. To augmenting an existing pathway, pro-
overcome the challenges associated with protein therapeutics and enhance viding a novel function or interfering
their biopharmaceutical applications, various protein-loaded nanocarriers with organisms. It is well-known that
with desired functions, such as lipid nanocapsules, polymeric nanoparticles, aberrant mitochondrial function and
protein homeostasis contribute to the
inorganic nanoparticles, and peptides, are developed. In this review, the
occurrence and development of neurode-
different strategies for intracellular delivery of proteins are comprehensively generative diseases, such as Parkinson’s
summarized. Their designed routes, mechanisms of action, and potential disease (PD) and Alzheimer’s disease
therapeutics in live cells or in vivo are discussed in detail. Furthermore, the (AD). Parkin functions as an E3 ubiquitin
perspective on the new generation of delivery systems toward the emerging ligase and participates in all stages of the
area of protein-based therapeutics is presented as well. mitochondrial life cycle (from biogenesis
to mitophagy). Previous studies showed
that parkin expression was benefit to
mitochondrial pathology in flight mus-
1. Introduction cles and dopaminergic neurons in PD Drosophila models.[2]
Another protein PINK1, an upstream protein of parkin, was
Proteins display a complex set of functions in the body, such recently reported to act as an important role in the pathology
as catalyzing of biochemical reactions, transporting molecules of AD. Gene therapy-mediated PINK1 overexpression can pro-
within living cell or from one organ to another, formation of mote the clearance of damaged mitochondria and then alle-
membrane receptors and channels, providing intracellular and viate amyloid-β peptide accumulation and cognitive loss. Also,
extracellular scaffolding support.[1] Proteins can be used in overexpression of parkin was able to rescue all the mutant
many therapeutics by replacing the proteins which are deficient phenotypes in pink1 null flies.[3] Recent studies also showed
that protein–disulfide isomerase could alleviate endoplasmic
reticulum (ER) stress and unfolded protein response signaling
Dr. X. Qin, Prof. C. Yu, J. Wei, Prof. L. Li, Prof. C. Zhang, Dr. Q. Wu, pathway, and afford neuroprotection in cellular PD models.[4]
Prof. J. Liu, Prof. W. Huang
Therefore, the regulation of many proteins is tightly associated
Key Laboratory of Flexible Electronics (KLOFE) and Institute
of Advanced Materials (IAM) with human diseases including cancer, cardiovascular diseases,
Nanjing Tech University (NanjingTech) neurological disorders, and viral infection. All these proteins
30 South Puzhu Road, Nanjing 211800, P. R. China may function as therapeutic proteins to combat disease in the
E-mail: iamlli@njtech.edu.cn; iamwhuang@njtech.edu.cn future.
Prof. S. Q. Yao Until now, the protein market grows so rapidly that many
Department of Chemistry
National University of Singapore other protein therapeutics, including Fabrazyme (agalsidase beta
Singapore 117543, Singapore for Fabry disease), PEGINTRON (pegInterferon-α2b for hepa-
E-mail: chmyaosq@nus.edu.sg titis C), and Cotazym (pancrelipase for cystic fibrosis or pan-
Prof. W. Huang creatic insufficiency) have been approved for clinical use.[5] The
Shaanxi Institute of Flexible Electronics (SIFE) market of the therapeutic proteins holds tremendous potential
Northwestern Polytechnical University (NPU)
and it is estimated that by the end of 2019, the global market for
127 West Youyi Road, Xi’an 710072, P. R. China
bioengineered protein drugs is valued at $222.7 billion, which
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/adma.201902791. include therapeutic purpose, protein vaccines, and protein-based
diagnostics.[6] Compared with small molecules, proteins possess
DOI: 10.1002/adma.201902791 unique advantage properties including less toxicity, high substrate

Adv. Mater. 2019, 31, 1902791 1902791  (1 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

specificity, low susceptibility to multidrug resistance and less


side effects to interfere with normal biological processes, and Lin Li received his B.Sc. and
last but not the least, the clinical development and FDA approval Ph.D. degrees in chemistry
time of protein therapeutics may be faster than that of small- from Anhui University in
molecule drugs.[1,5] However, there are several unfavorable 2004 and 2009, respectively.
physicochemical properties limit their usage of therapy, such He carried out postdoctoral
as entrapment into lysosomes, fragile tertiary structure which studies with Professor Shao
susceptibility to enzymatic degradation, easy aggregation when Q. Yao at National University
administered into serum, loss of activity, easy cause of an of Singapore (NUS). He
immune response, large molecular size which would be recy- obtained his current pro-
cled to the surface of the cells, poor penetration of the mem- fessor position in Nanjing
brane due to the electrostatic repulsion and poor stability in the Tech University (NanjingTech)
gastric low pH environment.[7–12] Furthermore, many proteins in 2015 after joining
(e.g., insulin, growth hormone, and oxytocin) have short half- Academician Huang’s group. His current research inter-
lives because of the fast renal clearance, and frequent injections ests are synthetic small-molecule indicators for monitoring
cause patient’s uncomfortable and inconvenience, representing biological/chemical species in physiological environments,
an immense challenge to modern proteins therapy.[13,14] especially in the mitochondrion.

2. Strategies for Intracellular Protein Delivery Shao Q. Yao is a Chinese-


born chemist currently
Direct delivery of proteins into the cells encounter many obsta- residing in Singapore. A
cles, such as serum instability and membrane impermeable graduate of Ohio State
due to electrostatic repulsions. To solve these issues, various (B.Sc. Honors, 1993) and
nanoparticles (NPs) are designed and utilized to enhance their Purdue (Ph.D., 1998), he
physical and biological stability, promote cell and tissue penetra- joined National University of
tion and effectively aide intracellular protein delivery.[15] A wide Singapore (NUS) in 2001 and
array of nanocarrier systems including lipid, polymers, protein is currently a professor at the
complex, inorganic materials have been shown to be prom- Department of Chemistry.
ising strategies.[16–19] In these strategies, different methods are His current research interests
applied to load the delivery system with the targeted proteins, broadly fall in the field of
such as genetic modification, physical adsorption, or chemical “catalomics,” in which the aim is to use tools in chemical
conjugation. In addition, these protein delivery systems based biology and proteomics, bioimaging, and materials for
on NPs could be engineered with various ligands for targeting to proteome-wide interrogation of enzymes for potential
the desired subcellular compartments like cytosol, nucleus and therapeutics.
mitochondria. Moreover, recent studies have reported that engi-
neered nanoparticles can be tailed with integration of stimuli
triggers to make protein delivery with both spatiotemporal- and
dosage-controlled manners possible, facilitating release of drug Wei Huang received his
with desirable pharmacokinetics.[20] In the last few years, there Ph.D. degree from Peking
was an increasing interest in delivering various systems of pro- University (1992). He began
teins. In this review, we will thoroughly summarize the research his postdoctoral research
progress of nanoparticle-based delivery system in native pro- in National University of
tein delivery, especially focus on the recent studies. We will Singapore (NUS, 1993);
survey representative examples of various materials and present moved to Fudan University
diverse approaches for protein delivery to complement previous where he founded the
reports. This includes the areas of polymeric nanoparticles (nat- Institute of Advanced
ural polysaccharides and synthetic polymers), lipid-based nano- Materials (IAM, 2002); was
carriers, protein-based complex, cell-penetrating peptide-based appointed as the Deputy
nanocarriers, exosome-based protein delivery system, metal- President of Nanjing
organic framework (MOF) composites and inorganic nanoparti- University of Posts and Telecommunications (NUPT,
cles (silica, gold, magnetic and carbon) (Figure 1). 2006). He assumed his duty as the President of Nanjing
Tech University (NanjingTech, 2012) and was appointed
as Deputy President and Provost of Northwestern
3. Nanoparticle-Mediated Protein Delivery Polytechnical University (NPU, 2017). His research inter-
ests include organic/flexible electronics, nanomaterials,
3.1. Polymeric Nanocarriers for Protein Delivery
and nanotechnology.
Due to the good biocompatibility and biodegradability,
many polymers can be referred as biomaterials to produce

Adv. Mater. 2019, 31, 1902791 1902791  (2 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 1.  Schematic illustration of various material-based nanocarriers for intracellular protein delivery.

nanoformulations for proteins delivery. Some of them (e.g., and ability to open up epithelial TJs, making it an excellent
chitosan (CS) and alginate) have unique feature of adhering to choice for many biomedical applications such as wound
the mucosal surface and transiently opening the tight junctions dressing, biosensor, tissue engineering application and drug/
(TJs) between epithelial cells. Moreover, polymers can be easily protein delivery.[24–28] Various approaches have been used to
modified with functional ligands, which could enhance their prepare chitosan-based nanocarriers, including emulsified sol-
physiochemical properties or improve their cellular uptake for vent diffusion, ionic gelation, microemulsion, reverse micelle
targeting to the cells.[21] To prepare polymeric nanocarriers with formation, complex coacervation, and polyelectrolyte com-
targeted proteins, various techniques are applied for designing plexation (PEC).[29] For example, by using the precipitation–
polymer-based nanoformulations, including emulsion, sur- coacervation technique, Zaharoff and co-workers reported that
factant free emulsion, interfacial polymerization.[22,23] It is well the chitosan-based nanoparticles could encapsulate various
known that polymers can be classified as natural polymers and fluorescence-labeled proteins including FITC-labeled bovine
synthetic ones. Polysaccharides including chitosan, cytodex- serum albumin (BSA), ovalbumin (OVA), concanavalin A
trins, alginate, dextran, and their derivatives are one kind of (Con A), and insulin.[30] In addition, the size and polydisper-
most important natural polymers, which have been widely used sity index (PDI) of nanocarriers, protein encapsulation effi-
for intracellular protein delivery. ciency and protein release rates have been demonstrated to
be influenced by the molecular weight and concentration of
CS, precipitant salt concentration, and proteins size. The com-
3.1.1. Polysaccharide-Based Nanoparticles position of precipitant salt containing weak anions resulted
in high PDI particles. With increasing the concentration
Chitosan and Their Derivative-Based Nanoparticles: Chitosan is and intrinsic viscosity of CS, the particle size was increased,
referred as a family of linear, cationic amino saccharides and subsequently leading to the increase of the protein loading
produced by the partial deacetylation of chitin. They are natu- efficacy and release rates. To improve the cellular up and per-
rally abundantly available polymers composed of randomly meation of protein both in vitro and in vivo, Zhang and co-
distributed β (1 → 4)-linked N-acetyl-d-glucosamine and d-glu- workers introduced arginine-rich cell-penetrating lipopeptides
cosamine units. Generally, they are isolated at commercial (SAR6EW) and synthesized SAR6EW/CS/insulin loaded NPs.
scale from waste of crustacean shells and fungi. Because of Their results demonstrated that the SAR6EW functionalized
its promising properties, such as biodegradability, low toxicity, NPs improved the internalization of insulin into Caco-2 cells
antimicrobial, nonimmunogenicity, muco-adhesive feature via adding clathrin and caveolae mediated endocytosis and

Adv. Mater. 2019, 31, 1902791 1902791  (3 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

showed much better hypoglycemic effect as compared with which is a big obstacle for its application in protein delivery. To
unmodified NPs in diabetic rats.[31] enhance its solubility in aqueous solution, various CS deriva-
As a useful tool for functional protein delivery, chitosan- tives have been designed by chemical modifications including
based PEC is formed by charge interactions between positive alkylation, thiolation, carboxylation, quaternization, PEGyla-
charged chitosan and negative charged polyelectrolytes (e.g., tion and acylation.[38–41] The side amino and hydroxyl groups of
tripolyphosphate, alginate, hyaluronic, carrageenan, pectin, CS could be modified with diverse array of ligands, functional
gellan gum, and poly-ϒ-glutamic acid). It has proven to be groups and moieties. On the one hand, this kind of modification
a simple and mild method to fabricate protein delivered NPs could enhance its aqueous solubility in different pH solution;
as it does not require organic solvents or sonication, thus can on the other hand, it could also improve stability, mucoadhe-
provide protection to the encapsulated proteins.[32,33] Hu et al. sivity, and TJs abilities by coupling with the targeted moieties.
developed a chitosan-based PEC delivery system consisting of Yao and co-workers recently reported CMCS-based multifunc-
carboxymethyl pachyman (CMP) and CS.[34] CMP is negatively tional NPs by modification of carboxymethyl chitosan (CMCS)
charged polysaccharides which could interact with CS to form with l-valine (LV) and phenylboronic acid (PBA). As shown in
CMP/CS NPs through PEC method. The diameter of CMP/CS Figure 2, PBA was served as hydrophobic and glucose-sensitive
NPs was in the range of 100–200 nm as the BSA being loaded unit, which could combine with the 1,2-diols of glucose to form
into this nanocarrier with encapsulation efficiency of 52.9%. soluble phenyl borates and result in the swelling of the NPs,
However, no further in vitro or in vivo assay was performed allowing “triggered release” of >92% of insulin with concen-
to confirm the enzymatic activity of protein. Generally, pro- tration of glucose at 20 × 10−3 m. LV was used to facilitate the
teins were encapsulated in CS-tripolyphosphate (TPP) NPs by absorption of the small intestine. Both in vitro and in vivo assay
interacting with both the positively charged CS and negatively indicated that the multifunctional NPs could internalize the
charged TPP. Furthermore, the physicochemical characteristics loaded insulin into HT-29 cells and protect insulin from deg-
of NPs were highly influenced by the deacetylation degree of radation in the simulated gastric fluid and simulated intestinal
chitosan, the ratio of TPP and chitosan, ionic strength and pH. fluid environments. Furthermore, this delivery system exhib-
Recently, Stie et al. prepared CS-TPP NPs. Two proteins BSA ited improved hypoglycemic effect after oral administration to
and p53 with different isoelectrical points could be success- the diabetic rats than that of free insulin.[42]
fully encapsulated in the NPs and effectively internalized by CS derivatives could conserve their positive charge at a neu-
the SK-Mel 28 cells.[35] Moreover, the higher positively charged tral pH value by quaternization of CS. Among them, trimethyl
NPs under mildly acidic conditions (pH 5.5) could facilitate the CS (TMC) is well studied derivative as it possesses mucoadhe-
cellular uptake of their protein-cargo. Clustered regularly inter- sive and TJs abilities. Compared with other quaternized CS-
spaced short palindromic repeats (CRISPR) and the CRISPR- based NPs, TMC-based NPs exhibited strongest effectiveness on
associated protein9 (Cas9) system associated technologies have the opening of TJs, thus could deliver insulin across Caco-2 cell
been considered as promising approach for the treatment of monolayers.[43] To prove the enhancement of the TJs opening
genetic diseases, which could be guided to specific genomic ability and the transepithelial transport of insulin-loaded TMC
locations and cleave a target DNA sequence by a short RNA NPs, Liu et al. designed a dissociable “mucus-inert” hydrophilic
search string, enabling precise genome editing and cell engi- coating of N-(2-hydroxypropyl) methacrylamide copolymer
neering for treating genetic disorders.[36] There are two delivery (pHPMA) derivative on the surface of TMC NPs. Experiments
ways of genome editing. One way is the delivery of DNA or displayed that the novel self-assembled NPs permeated effi-
mRNA that can generate Cas9 proteins alongside accompa- ciently through both epithelium and blanket of mucus gel,
nying single guide RNA (sgRNA) inside cells, and the other enhanced the cellular uptake and exhibited higher hypogly-
way is the direct delivery of CRISPR/Cas9 ribonucleoproteins cemic effect on diabetic rats than uncoated TMC NPs.[44]
(RNPs) into live cells and animals. Compared with the former, Besides the above chemical modifications of CS-based NPs,
the latter has more advantages include reduced off-target thiolation, PEGylation acylation, as well as amino acid coupling
effects, low toxicity, high editing efficacy, etc. Recently, Qiao in the construction of functional nanocarriers have been devel-
et al. utilized biodegradable materials CS and red fluorescent oped for protein delivery. Recently, Gan and co-workers reported
protein (RFP) to prepare NPs for delivering RNPs alongside CS-based deoxycholic acid modified NPs (DANPs) to overcome
DNA donors (ssDNA).[37] In order to enhance the electrostatic multiple barriers of the intestinal epithelium by exploiting the
interaction between positively charged RFP@CS and nega- bile acid pathway.[45] It is noted that DANPs were internalized
tively charged Cas9 RNPs and facilitate Cas9 enzyme to enter into the intestinal epithelium through apical sodium-dependent
the nucleus for efficient genome editing, the Cas9 enzyme bile acid transporter (ASBT)-mediated endocytosis. Due to the
was inserted twenty glutamate residues in the N terminus interaction with cytosolic ileal bile acid-binding protein, DANPs
and three repeating nuclear localization signals (NLS) at the could escape rapidly from endolysosome, thus protecting the
C terminus. Finally, the constructed Cas9 RNP-ssDNA-RFP@ insulin from degradation and facilitating the intracellular traf-
CS NPs were internalized into the cytoplasm via caveolae- and ficking. Finally, DANPs accessed the basolateral membrane and
micropinocytosis-mediated endocytosis followed by endosomal promoted the basolateral release of insulin. Further pharmaco-
escape, releasing Cas9 RNPs and ssDNA donors, and eventu- dynamic and pharmacokinetic studies in diabetic rats proved
ally entering the nucleus for efficient genome editing. that the oral administration of insulin-loaded DANPs with
Natural CS is soluble in the presence of acid solution due enteric capsules coating led to slower but prolonged reduction
to free protonatable amino groups at the glucosamine units. in blood glucose levels and exerted a stronger hypoglycemic
However, CS is not soluble in aqueous solution at neutral pH, effect.

Adv. Mater. 2019, 31, 1902791 1902791  (4 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 2.  a) Schematical illustration of the CMCS-PBA-LV-based multifunctional nanocarriers to overcome multiple barriers for oral delivery of insulin.
b) TEM images of CMCS-PBA-LV-based multifunctional nanocarriers. c) CLSM images of HT-29 cells treated with FITC-insulin loaded NPs after
incubation for 6 h. d) Insulin in vitro release profiles of insulin-loaded NPs against SGF (pH = 1.2) and SIF (pH = 6.8) in PBS solution with different
concentration of glucose. e) Blood glucose levels in diabetic rats followed oral administration of insulin-loaded nanocarriers, saline and insulin solu-
tion. INCs: insulin-loaded nanocarriers. Adapted with permission.[42] Copyright 2016, Elsevier B.V.

Thiolated CSs are synthesized by chemically modification core. Due to possess smaller size and higher storage stability,
of sulfhydryl bearing agents such as glutathione, cysteine and the Arg-CS-NPs was selected for the further study. Subsequent
thioglycolic. Obviously, the major advantage of thiolated CS results showed that this codelivery nanosystem was taken-up
derivatives is the thiol functional group, which could bind into HeLa cells and exhibited synergetic effects to enhance the
with cysteine-rich subdomains of glycoproteins in the mucus apoptosis-inducing activity.
layer, thus promoting efficient mucoadhesion in the intestinal
wall. Special modification of CS derivatives with amino acids
contain various ability in anticoagulation, cell penetrating func- 3.1.2. Synthetic Polymer–Based Nanoparticles
tion and enhanced TJs opening with positive charges in neutral
conditions, thus can be used as promising materials for protein Poly(dl-lactide-co-glycolide) (PLGA): PLGA is one of the most
delivery. Recently, a series of amino acid modified CS deriva- important synthetic polymers, which has been developed as a
tives (Lys-CS, Arg-CS, and Phe-CS) were synthesized to shield promising delivery material. It is a biocompatible and biode-
nano droplet of linoleic acid for the codelivery of caspase-3 pro- gradable synthetic polymer that has been proved for safe drug
tein and paclitaxel.[46] The positively charged caspase-3 could delivery without causing tissue damage.[47,48] PLGA is a polymer
adsorb on the surface of nanocapsules to enhance the stability, ester that is composed of two α hydroxyacids lactic and glycolic
and hydrophobic drug paclitaxel was encapsulated into the oil acids. Nguyen and co-workers systematically characterized

Adv. Mater. 2019, 31, 1902791 1902791  (5 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

physiochemical properties of six polymeric NPs including gela- of 80–110 nm possessed loading efficiency of insulin as high
tion, alginate, CS, PLGA, CMCS-coated PLGA, and PLGA/PEG- as 90% and exhibited fivefold higher cellular uptake than that
based NPs.[49] By comparison, PLGA-based NPs possessed the of BSA-coated NPs. Obviously, the transferrin coated NPs could
best performance, such as the smallest size (≈160 nm), con- enhance their transepithelial transport via receptor-mediated
sistent particles sizes in PBS, water and simulated lung fluid, transcytosis and elicit a remarkable hypoglycemic response at
the most favorable profiles for effective delivery to lung tissue, the dose of 50 U k−1. It is noted that cell-penetrating peptides
as well as the highest cytocompatibility in human alveolar (CPPs) could also be utilized in PLGA-based nanocarrier for
type-1 epithelial cells. These results further demonstrated that increasing its cellular uptake and transportation. For example,
PLGA-based NPs are promising carriers for pulmonary pro- Wei and co-workers prepared poly(arginine)8 (L-R8 and D-R8)
tein/DNA delivery. Moreover, another approach was applied to functionalized PLGA NPs.[62] Due to a group of cationic amino
improve the mucoadhesion and cell uptake of PLGA-based NPs acid sequences in CPP, significant ability for membrane trans-
for mucosal delivery proteins by coating positive charged poly- location was achieved. In addition, comparing with unmodified
mers or proteins on their surface[50–53] For example, To improve NPs, the L-R8 and D-R8-modified NPs could improve hypogly-
proteins stability and their enzymatic activities, Amoozgar and cemic effects by 2.5- and 3.7-times and enhanced bioavailabili-
Goldberg synthesized a delivery system with proteins coating ties of insulin by 3.2- and 4.4-times, respectively.
on the particles surface PLGA NPs.[53] In this study, various Poly(allylamine)s: Poly(allyamine)s could be synthesized by
proteins including lysozyme, deoxyribonuclease (DNase), anti- Michael-type polyaddition of primary or bis-secondary amines
CD62E antibody, and collagenase I were immobilized on the to bis(acrylamide)s, which are water-soluble, biocompatible and
surface of NPs by Michael addition or Schiff-base formation biodegradable cationic polymers. Poly(allyamine)s have some
with polydopamine. Doxorubicin (Dox) was encapsulated into advantages such as low cytotoxicity and easy degrade into oli-
the NPs that could sustainedly release to immunosuppressive gomeric products in aqueous media within days or weeks, thus
M2 macrophages. In order to increase the transcellular uptake make them to be potential in drug delivery application. Previous
of PLGA-based NPs and enhance the protein absorption, var- studies were used to designed bioreducible poly(allyamine)s
ious ligands were conjugated for specific binding with the cel- based nanocarriers for intracellular protein delivery. By Michael-
lular receptors. For example, folic acid (FA) is often used as a type polyaddition of 4-amino-1-butanol (ABOL) to cystamine
target for many epithelial cancer cells as those cells could over- bisacrylamide (CBA), the functionalized poly(allyamine)s
express the folate receptor, which could promote endocytosis were synthesized. Next, the positively charged (CBA-ABOL)/
of FA-attached nanocarriers via the folate receptor. Recently, proteins PECs were developed via the self-assembly of negative
Jiang and co-workers fabricated the FA-CS-coated NPs based proteins with positive disulfide-containing poly(allyamine)s.
PLGA for oral delivery of insulin.[54] Experimental data showed Experimental results demonstrated that the bioreducible
that this system protected insulin against digestive enzyme poly(allyamine)s NPs showed higher mucoadhesive properties
solution and improved the cellular uptake of HT-29 cells and than poly(allyamine)s NPs without disulfide bonds. Moreover,
intracellular delivery. In vivo studies further revealed that this system exhibited fast disintegration in reductive solution,
these insulin-loaded nanocarriers could prolong the hypogly- which could promote the release of the proteins in the intracel-
cemic effect in the rats. Due to possess galactose residues in lular environment.[63,64] In order to efficiently deliver the posi-
molecule, lactose acid is the ligand for the hepatic asialoglyco tively charged proteins, Engbersen and co-workers synthesized
protein receptor, which can be conjugated with PLGA for liver poly(allyamine)s polymers with negatively charged citraconate
target. For example, Zhou et al. reported lactose acid-PLGA/ε- groups in the side chains.[65] The citraconamide carboxylate
polylysine NPs for BSA delivery.[55] In this system, lactose acid derivatives in the side chains become hydrolyzed with the for-
was selected as liver-targeted ligand. ε-Polylysine was used as mation of protonated amino moieties, which could promote
protein protectant and antiacidic agent to enhance the stability polymer–endosomal membrane interactions and enhance the
of BSA and cellular uptake in HepG2 cells. In vitro and in vivo cationic protein lysozyme delivery into the cytosol. In addition,
experiments exhibited the significant decreased initial burst the side chains were linked by disulfide, so that the intracellular
release and enhanced liver distribution in mice. In addition, cleavage of disulfide linkages of poly(allyamine)-based NPs
the same approaches were applied for synthesizing PLGA-l-va- would further improve the protein unpacking in the cytosol.
line,[56] PLGA-gambogic acid,[57] PLGA-PEG-concanavalin A,[58] Further results showed that the loading lysozyme was released
and PLGA-PEG-FA conjugates.[59] Upon emulsification, insulin- in its active form from the nanocarriers and successfully inter-
loaded NPs were formed with surface expressing functional nalized into human umbilical vein endothelial cells.
ligands for specific receptors binding and mediated transport in Cell-Penetrating Poly(disulfide)s: The structures of cell-pene-
vitro and in vivo. Moreover, the PLGA-based NPs could be also trating poly(disulfide)s (CPDs) is similar as polyarginine except
coated with functional proteins (e.g., transferrin and Con A), the polypeptides backbone is replaced with poly(disulfide)s.[66,67]
which are used to bind to the receptors for enhancing oral The CPDs were initially developed by Matile’s group in 2013,
insulin delivery.[60,61] For instance, poly(ester amide)s (PEAs) which could be grown directly on the substrates by surface-
and PLGA were selected to form PLGA-PEA NPs with trans- initiated ring-opening disulfide-exchange polymerization
ferrin coating as functional target.[61] In this system, PEAs were method.[68,69] The polymer size could be adjusted by changing
prepared with various Phe to Arg ratios to adjust the NPs with a the polymerization time, initiator and monomer concentra-
broad range of properties, thereby effecting the zeta potential of tion. Interestingly, the changes of side chain moieties would
NPs from +11.8 to 31.3 mV and insulin release kinetics. Experi- further affect their intracellular destination. In comparison
mental results showed that all PLGA-PEA NPs with small size with CPPs, CPDs could efficiently enter the cells and deliver

Adv. Mater. 2019, 31, 1902791 1902791  (6 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 3.  a) Structure of AO-SH and Tz-SH initiators. b) Schematic illustration of CPD-based intracellular delivery of native proteins with PTM-based
tagging and traceless tagging approaches. a,b) Adapted with permission.[72] Copyright 2018, Wiley-VCH. c) Schematic illustration of thiol-mediated
uptake of CPDs with no endosomal trapping. Reproduced with permission.[67] Copyright 2015, Royal Society of Chemistry.

the cargos into the cytosol via thiol-mediated pathways without (post-translation modification (PTM)-based tagging and trace-
being trapped by endocytic vesicles. In addition, CPDs could be less tagging) for delivery of “native” proteins (Figure 3b).[72] In
rapidly degraded in the cytosol by glutathione (GSH)-assisted this study, glycoproteins (including all antibodies) were chemi-
depolymerization (<5 min) and showed minimal cytotoxicity, cally or chemoenzymatically modified with biorthogonal tag
thus can be used for intracellular delivery of other thiol- and subsequently conjugated with corresponding oxime-con-
containing or modified cargos.[70] For example, in order to effi- taining CPD/TzCPD. On the other hand, nonglycosylated pro-
ciently and directly deliver proteins into cell cytosol, Yao and teins (e.g., BSA) with primary amines react with two cleavable
co-workers designed CPDs with three types of thiol-containing linkers NBL and TCOL which contain p-nitrophenylcarbonate.
initiators that comprise biotin, nitrilotriacetic acid (NTA) and Experimental data showed that “native” proteins could be suc-
tetrazine (Tz), respectively.[71] These structures of AO-SH and cessfully released in live cells undergo GSH-catalyzed disulfide
Tz-SH initiators are shown in Figure 3a. Four of recombinant cleavage followed by spontaneous cleavage of the self-immo-
proteins including avidin, trans-cyclooctyne-BSA (TCO-BSA), late linker. Furthermore, functional glycoprotein horseradish
TCO-anti-rabbit immunoglobulin G (IgG) and BRD-4, either peroxidase (HRP) and nonglycosylated protein ribonuclease A
covalently or noncovalently conjugated with CPDs were suc- (RNase A) were selected to investigate their biological activi-
cessfully delivered into different mammalian cells via endo- ties. The subsequent results displayed that the delivered HRP
cytosis-independent pathways. In addition, CPD-assisted remained activity of enzyme by catalyzing oxidization of a suit-
delivery of functional proteins recombinant (His)6-tagged cas- able substrate in the presence of H2O2 and RNase A. These
pase-3 into HeLa cells could retain its biological activity. More- results indicated that PTM-based tagging/traceless tagging
over, to avoid genetical or chemical modification of proteins CPDs conjugated delivery strategy is an effective approach for
which might affect their activities, recently, the same group intracellular delivery and release of native proteins with imme-
reported two complementary CPD-facilitated approaches diate bioavailability.

Adv. Mater. 2019, 31, 1902791 1902791  (7 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Copolymer-Based Nanoformulations: Block copolymers gener- in tumor vascular endothelia and stroma. R300 was utilized
ally embrace two or more distinct polymer chains. Until now, to deplete platelets, which could promote the diffusion of NPs
various approaches have been applied to synthesize block into solid tumors. Thus, the R300 and Dox were specifically
copolymers, such as anionic, radical, cationic, ring-opening, and selectively released into tumor tissues, subsequently sup-
photo, and group-transfer polymerization methods. These pressed tumor growth and metastasis formation in an MCF-7
block polymers could be designed with tailored functionality, mouse tumor model.
variable molecular weight (Mw), and desired chemical com- Polymersomes are polymeric vesicles made of amphi-
position. In this way, block polymers could form numerous philic block copolymers, which contain an aqueous interior
nanostructures such as NPs, micelles, polymersomes and surrounded by a bilayer membrane for loading hydrophobic
nanogels. Thus, copolymer-based nanocarriers have been and hydrophilic drugs. In comparison with liposomes, poly-
widely used for drugs delivery, wound dressing and tissue mersomes are more stable, easier adjusted with their stim-
engineering.[73,74] uli-responsiveness, membrane thickness, and size.[73] In
Micelles (polymers) are organized auto-assembly formed addition, their surface could be modified with functional
in a liquid and composed of amphiphilic macromolecules, in moieties, such as CPP for efficient cellular uptake and pro-
general amphiphilic di or tri-block copolymers made of solvo- teins delivery into different cell lines.[81] In the same way as
philic and solvophobic blocks. These micelles could be used as micelles, polymersomes could also be prepared for proteins
the platforms for cargos delivery. For example, previous studies delivery system with thermos/pH/reduction-sensitive copoly-
used cationic block polymers PEG-poly[N-{N′-(2-aminoethyl)-2- mers.[82–85] For example, using reversible addition-fragmen-
aminoethyl}aspartamide] (PEG-pAsp(DET)) to form a pH-trig- tation chain transfer polymerization (RAFT) method, Cheng
gered charge-conversion polymeric micelles.[75,76] The charged et al. successfully synthesized thermo-sensitive poly(ethylene
density of proteins cytochrome c (CytC) and IgG were tempo- glycol)-b-poly(acrylic acid)-b-poly(N-isopropylacrylamide)
rarily increased by conjugating with charge-conversional moie- (PEG-PAA-PNIPAM) triblock copolymers.[82] After incuba-
ties, citraconic acid amide (Cit) or cis-aconitic acid (Aco). Results tion with crosslinking agent cystamine, the polymersomes
showed that the micelles were stable at the normal physiological were formed with stable structure in PBS at 37 °C, but were
pH value of 7.4, however, dissociated and released the native rapid disassembled in the presence of dithiothreitol. In vitro
proteins at the endosomal pH of 5.5. In addition, the dissocia- assay showed that various proteins including BSA, OVA,
tion of micelles led to the fast endosomal escape of proteins and CytC, and lysozyme could be encapsulated into the poly-
diffused them into the cytoplasm. Gao et al. developed another mersomes and released under an intracellular mimicking
pH-sensitive positively charged polymeric micelles based on reductive environment. In addition, the CytC released from
methoxy-PEG-b-poly(β-amino ester). In this system, the pro- the poly­mersomes into the cytosol of MCF-7 cells showed
teins were bonded with piperidine and imidazole rings by elec- higher apoptosis as comparing with free CytC. Recently,
trostatic and hydrogen bonding interactions.[77] Apart from the dual-responsive polymersomes were developed by Zhong
pH-triggered release in proteins encapsulated micelles, light or and co-workers, which was based on poly(ethylene glycol)-
sugar-responsive polyion complex micelles were also fabricated SS-poly(2-(diethyl amino)ethyl methacrylate) (PEG-SS-PDEA)
for efficient protein delivery.[78,79] For example, Ji and co-workers diblock copolymers.[83] After loading model proteins BSA and
synthesized poly(N,N-dimethyl-N-(2-(methacryloyloxy)ethyl)- CytC, this dual-bioresponsive polymersomes could efficiently
N-((2-nitrobenzyl)oxy)-2-oxoethanaminium bromide)-block- release the protein under acidic and/or reductive conditions.
poly(carboxybetaine methacrylate) (PDMNBMA-b-PCBMA) Cell-based experimental results showed that the released
cationic block copolymer that could self-assemble as micelles CytC could enhance apoptosis of MCF-7 cells. In order to
with negative charged BSA.[78] In vitro assay demonstrated that achieve targeted tumor treatment with therapeutic proteins,
the micelles disassembled into zwitterionic carboxybetaine units Zhong’s group synthesized two copolymers, poly(ethylene
under UV irradiation. In addition, micelles showed fast cellular glycol)-b-poly(2,4,6-trimethoxybenzylidene-pentaerythritol
internalizations by A549 cells and controllable BSA release in carbonate)-b-poly(succinic acid carbonate) (PEG-PTMBPEC-
the presence of UV irradiation. Shi and co-workers reported PSAC) and poly(ethylene glycol)-b-poly(2,4,6- trimethoxy-
two kinds of micelles based on PEG-b-poly(glutamic acid-co-glu- benzylidene-1,1,1-tris(hydroxymethyl)ethane methacrylate)-
tamicamidophenylboronic acid) (PEG-b-P(Glu-co-GluPBA)) and b-poly(acrylic acid) (PEG-PTTMA-PAA).[84,85] The function-
PEG-b-poly(L-lysine-co-ε-3,4-dihydroxyphenylcarboxyl-l-lysine) alized polymersomes with tumor homing ligands were
(PEG-b-P(Lys-co-LysCA)) copolymers for intracellular protein fabricated by modifying PEG-PTMBPEC-PSAC with pros-
delivery, respectively.[79] Either negatively or positively charged tate cancer-targeting ligand 2-[3-[5-amino-1-carboxypentyl]-
proteins (insulin or CytC) could be encapsulated into the ureido]-pentanedioic acid (Acupa) and PEG-PTTMA-PAA
micelles under mild conditions. The micelles were quite stable with lung cancer-targeting ligand anisamide, respectively.
under physiological condition but disassembled in the pres- Decorated pH-responsive poly­ mersomes dissociated under
ence of excess fructose or endosomal pH. Recently, Nie and co- tumors environment (acidic condition), resulting in prompt
workers designed poly(etherimide)-poly(lactic-co-glycolic acid)2 protein granzume B release and potent tumor cell apop-
(PEI-(PLGA)2) based micelles with lipid-peptide shell coating. tosis. The in vivo results showed that both cancer-targeting
These micelles could codeliver both antibody R300 and anti- ligands improved the accumulation within tumor, further
tumor agent doxorubicin into tumor tissues (Figure  4).[80] In indicating the polymersomes could be served as promising
this multifunctional nanocarrier, the lipid-peptide shell could nanocarriers for intracellular delivery of protein drugs.
be cleaved by metalloproteinase 2 (MMP2) which overexpressed Recently, the same group developed smart polymersomes

Adv. Mater. 2019, 31, 1902791 1902791  (8 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 4.  a) Schematic configuration of the PEI-(PLGA)2 based Dox-loaded micelles (P-D), the antibody 300 was absorbed onto the surface to form
P-D-R. Lecithins, PEGylated phospholipids and MMP2 cleavable peptides were layered onto the surface of the P-D-R micelles to form multifunctional
PLP-D-R micelles. b) MMP2 is overexpressed on the surface of tumor endothelial and stroma cells, the encapsulated R300 is released once the shell
layer of PLP-D-R is cleaved by MMP2. R300 could bind to platelet-surface receptors and deplete platelets by inducing microaggregation of 3–5 plate-
lets. c) The Dox-loaded micelles could enter to the tumor tissues as the absence of platelets in tumor induces openings in the vessel walls. d) Release
kinetics of Dox and the MMP2 accelerated the release of Dox. PLP-D-R and other drug formulations were administered into e) MCF-7 tumor-bearing
mice, and tumor volume was measured every 3 days. Reproduced with permission.[80] Copyright 2017, Springer Nature.

dually functionalized with cRGD and fusogenic GALA system, protein-loaded NPs with uniform size and high
peptides for targeting to ανβ3-positive A549 lung cancer loading efficiency were formed by polyelectrolyte compl-
cells and enhancing cytosolic release of CytC.[86] The poly­ exation. In vivo experimental results showed that the FGF-2
mersomes were assembled with PEG-b-poly(trimethylene could be protected by PEG stealth corona and the released
carbonate-co-dithiolane trimethylene carbonate)-spermine proteins kept their biological activity in bone marrow mesen-
(PEG-b-P(TMC-co-DTC)-spermine), cRGD-PEG-b-P(TMC-co- chymal stem cells.
DTC), and maleimide-PEG-bP(TMC-co-DTC) which postmod- As one of the most promising nanocarriers for intracellular
ified with GALA-SH. Apoptosis assays showed that cRGD/ protein delivery, diverse polymeric delivery systems have been
GALA decorated polymersomes induced better apoptosis of extensively designed for protein delivery and achieved much
A549 cells than polymersomes without GALA. progress. However, owing to the complexity of polymer-based
Apart from the micelles and polymersomes, copolymers nanocarriers with multicomponent 3D structures, careful
could also bind with proteins by electrostatic interaction or designing and construction are needed to make sure reproduc-
hydrogen bond to form nanocarriers.[87,88] Due to possess ible formulations in the large-scale manufacturing processes.
specific properties, heparin sulfate proteoglycans are well Furthermore, the smart polymers with multicomponent and
studied for storage and protection of numerous growth fac- multifunction, as well as controlled release on demand and
tors in the extracellular matrix. For example, the heparin- high efficient diffusion of therapeutic protein through a stim-
block-PEG copolymer based NPs were synthesized by Silva ulus property of the disease microenvironment are urgently
et al. to load fibroblast growth factor (FGF-2).[88] In this needed.[89,90]

Adv. Mater. 2019, 31, 1902791 1902791  (9 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 5.  a) Schematic illustration of TRAIL/Dox–gel–liposome (Gelipo) for site-specific drug delivery. Using R8H3 modified liposome as the nanocar-
riers sequentially deliver TRAIL to the plasma membrane and Dox to the nuclei for combination cancer treatment. b) The diameter of TRAIL/Dox-Gelipo
measured by DLS and TEM. c) HAase degrade the m-HA shell and stimulate the extracellular release of TRAIL d) In vitro cytotoxicity of TRAIL-Gelipo,
Dox-Gelipo and TRAIL/Dox-Gelipo after 30 min of HAase pre-treatment toward MDA-MB-231 cells for 24 h. e) The MDA-MB-231 tumor growth curves
after intravenous injection of different formulations of Dox at a dose of 2 mg kg−1. f) Representative images of MDA-MB-231 xenograft tumors of the
mice after treatment with saline, the Dox solution, Dox-Gelipo and TRAIL/Dox-Gelipo respectively. MBA: a crosslinker, N,N′-methylenebisacrylamide,
EPC: Egg phosphatidylcholine. Adapted with permission.[94] Copyright 2014, Wiley-VCH.

3.2. Lipid-Based Nanoparticles cholesterol-incorporated dicetyl phosphate. Because of their


special structure, various hydrophobic or hydrophilic drugs
Lipid-based NPs are made of natural, semi-synthetic or syn- could be filled into the liposomes and then delivered into cells
thetic lipids, such as fatty acid, fatty alcohols, phospholipids, via endocytosis or liposome-cell fusion.[16,91,93] For example,
long and medium chain monoglycerides, diglycerides and tri- to enhance therapeutic efficacy, Jiang et al. designed gel–lipo-
glycerides. Based on their structures and compositions, they some complex to codeliver two kinds of anticancer drugs,
can be classified as solid lipid nanoparticles (SLNs), liposomes, namely tumor necrosis factor-related apoptosis-inducing ligand
nanoemulsions and nanocapsules. The majority of lipids (TRAIL) and Dox (Figure 5).[94] Herein, the Dox was first encap-
excipients are derived from dietary oil or fats. Due to their sulated into CPP (R8H3) modified liposome. A crosslinked gel
biocompatibility, biodegradability, low toxicity, and the ability composed of enzymatically degradable hyaluronic acid (HA)
of overcoming the multiple biological barriers (e.g., intestinal was attached on the surface of liposome. In the meantime,
fluids, mucus layer, and intestinal epithelium), lipid-based the protein TRAIL was encapsulated in the outer shell. The
nanoparticles are considered as promising protein delivery hyaluronidase (HAase) is highly overexpressed at the tumor
system. Until now, commercial success has been achieved with microenvironment, which could degrade the HA shell, thereby
some lipid-based NPs for oral administration of protein or promoting the released TRAIL to bind with the death recep-
peptide-like drugs, which encourages researchers to design tors of the plasma membrane and facilitate the tumor cellular
new developed lipid-based nanocarriers.[91] uptake of Dox-R8H3-liposomes. In vitro and in vivo experiment
demonstrated that this multifunctional codelivery gel–liposome
system showed significant inhibition of the tumor growth in
3.2.1. Liposomes the MDA-MB-231 xenograft tumor animal model.
Liguori et al. reported two different protein-loaded liposomes
Liposomes as delivery vesicles are first discovered in 1961 by by using cell-free protein synthesis technologies.[95] Both of
Bangham and Horne.[92] This kind of drug loaded system is voltage dependent anion channel protein and Bak protein were
most extensively studied. Until now, there are several liposome- successfully delivered into living cells with bioactivity. One
based products, which contain small molecules drugs, that have of the major drawbacks for liposomal delivery system is that
been approved by FDA for human use.[93] Liposomes are bilay- they are taken up by endocytosis and finally degraded in lys-
ered vesicles with an internal aqueous cavity. Their structure osomes, which could induce the side effect. To overcome this
components are composed of amphiphilic lipids or phospho- issue, Kube et al. synthesized fusogenic liposomes, which could
lipids including diplamitoylphosphatidylcholine (DPPC), dis- deliver intracellular proteins with different size ranged from
tearoyl-phosphatidylcholine (DSPC), lecithin, stearylamine, and 2.3 kDa (fluorescently tagged LifeAct) over 27 kDa (EGFP) to

Adv. Mater. 2019, 31, 1902791 1902791  (10 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 6.  Schematic illustration of bioreducible lipid-like materials and negatively supercharged proteins for effective protein delivery and genome
editing. Reproduced with permission.[102] Copyright 2016, PNAS.

240 kDa (R-phycoerythrin).[96] In general, the encapsulated effi- results suggested that cationic lipids could establish a powerful
ciency and release rate of proteins or peptides within liposomes delivery system for in vivo genome editing. Furthermore, Xu
are greatly influenced by their interaction with liposomal com- and co-workers synthesized a bioreducible lipid with disulfide
ponents such as lipids, surfactants, the polymers (e.g., chi- bond as linkage to the hydrophobic tail.[102] After complexing
tosan) or inorganic materials (e.g., silica). These components, with negatively supercharged GFP-Cre or Cas9/sgRNA, these
which are coupled on the liposomal surface, could enhance nanocarriers could be structurally breakable under the reduc-
the stability of nanocarriers, control the release time of drugs tive intracellular environment and promote the cargo proteins
and promote proteins disposition at the specific site.[91,97,98] For to escape from endosome (Figure 6).
instance, Opanasopit and co-workers synthesized methylated
N-(4-N,N-dimethylaminobenzyl) CS, which is water soluble at
physiological pH. After being coated on the surface of liposome 3.2.2. Solid Lipid Nanoparticles
by electrostatic interaction, this system showed an enhanced
loading efficiency of BSA and protein permeability across the Solid lipid nanoparticles possess excellent biodegradability and
Caco-2 cell monolayers with low cytotoxicity.[99] Recently, Sal- biocompatibility, specific function by the suitable chemical
maso and co-workers designed a pH-sensitive stearoyl-PEG- modification and rapid uptake by cells, which have been con-
poly(methacryloyl sulfadimethoxine)-decorated liposome sidered as a promising developed submicron gene/protein
system with BSA loading for bladder cancer treatment.[97] delivery system.[103,104] Like the above liposomes, SLNs are typi-
Results revealed that the BSA-loaded liposomes were colloi- cally colloidal particles with an average diameter between 10
dally stable at neural pH but showed remarkably high adhe- and 1000 nm. The difference with liposomes is, generally, SLNs
sion with MB49 cells and more effective administration to mice possess a solid lipid core matrix that can load lipophilic mole-
via bladder instillation under acidic environment. In addition, cules. To improve the stability, the lipid core is next stabilized by
Matsumura and co-workers fabricated polyampholyte-modified surfactants.[105] Thus, in comparison with liposomes, SLNs are
liposomes in conjunction with the freeze concentration strategy more stable for biological applications. Over the past 25 years,
to increase lysozyme protein internalization and the efficiency there are many techniques (e.g., water/oil/water emulsions
of endosomal escape in living cells.[98] The results showed that method and hydrophobic ion pairing technique) have been used
uptaken capacity of lysozyme was increased fourfold in com- for achieving loadings of proteins within SLN.[91,106,107] In order
parison with that of unfrozen cells. The temperature could be to improve the proteins loading efficiency, the hydrophobic ion
designed as a factor that affects the protein release rate when pairing technique is applied to enhance the lipophilic parti-
the liposomes incorporate with thermosensitive materials. tion coefficient of hydrophilic peptide leuprolide. Amphipathic
Lysolipid is a kind of widely studied materials which could molecule sodium stearate was selected to prepare hydrophobic
form the thermal sensitive liposomes. For example, Xu and ion pairing of leuprolide. This technique combined with O/O
co-workers prepared the lysolipid liposome with the diameter emulsion–evaporation that can not only increased the protein
of 145 nm.[100] This drug loaded system showed good stability entrapment, but enhanced the stability of the whole system.[107]
at 37.5 °C but rapid released albumin at 42 and 44.5 °C. The Furthermore, Yang et al. fabricated a novel Gel-Core-SLNs by
genome-editing Cas9 is natively cationic, which could not com- double emulsion and thermal sensitive gel technology.[108] In
plex with cationic lipids. To efficiently deliver those proteins this system, the loading efficiency of insulin and thymopentin
into mammalian cells by cationic lipids, Zuris et al. changed in the Gel-Core-SLNs were 57.36% and 61.97%, respectively.
the proteins to be highly anionic either by complexation with Furthermore, the functional peptide ligands are usually utilized
polyanionic single-guided RNA (Cas9:sgRNA) or by fusion to be modified with lipids to enhance cellular internalization
with the supernegatively charged (−30) green fluorescent pro- and drug absorption. Epithelium targeting peptides and CPPs
tein (GFP).[101] In this way, the stable nanocarriers were formed are two main functional peptides.[109,110] For example, Lv and co-
through electrostatic self-assembly between cationic lipids workers designed SLNs modified with stearic acid–octaarginine
and anionic protein complex. Subsequent in vitro and in vivo (SA–R8) to deliver the insulin.[109] Due to the positively charged

Adv. Mater. 2019, 31, 1902791 1902791  (11 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

SA–R8, the zeta potential of the SLNs dramatically changed that used for pharmaceuticals, therapeutics and diagnosis.[113]
from −32.13 to +29.87 mV, which could increase the Caco cells The SiNPs with different structure and morphologies could be
internalization of SLNs and bioavailability of insulin. Recently, fabricated by using sol–gel chemistry combined with different
Huang and co-worker used goblet cells affinity peptide CSKSS- structure directing agents. According to the topography, those
DYQC (CSK) and CPP IRQRRRR (IRQ) to conjugate with SiNPs could be divided into mesoporous silica nanoparticles
polyoxyethylene (40) stearate, by which the salmon calcitonin– (MSNs), dendritic silica nanoparticles, hollow silica nanoparti-
loaded SLNs are prepared and characterized.[110] In vitro and in cles (HSNs) and solid silica nanoparticles.[114] Figure  7 shows
vivo studies showed the peptide modified SLNs facilitated the silica-based NPs with different structure and morphologies that
internalization of protein drugs on Caco-2/HT29-MTX co-cul- used for protein delivery.
tured cells and permeation in excised rat duodenum mucosa. Mesoporous Silica Nanoparticles: The MCM-41 is one clas-
Besides modification of the lipids surface for improving the tar- sical type of MSNs, which is prepared using modified Stöber
geted protein delivery, the protein drug can also be modified method. In this method, cationic surfactant cetyltrimethylam-
with functional ligands to enhance the treatment of diseases. monium bromide (CTAB) is usually used as a soft template.[125]
For instance, Huang and co-workers synthesized the mem- This MSN possesses the uniform pore size of ≈2 nm, which
brane permeable sequence (MPS)-conjugated proteins CytC has been extensively studied for delivering the small drug
(MPS-CytC), which subsequently complexed with PEGylated, molecules.[113,126] However, it is difficult to encapsulate large
anisamide modified SLNs to enable specific targeting to the proteins into such small pores. In order to load the large pro-
tumor site in H460 lung carcinoma.[111] In this system, both teins, larger molecular weight template or swelling agents were
anisamide ligand and MPS could drive protein drugs to the used to expand the pore size.[114,116,127] For example, Slowing
specific tumor site in a xenograft model and exhibited effi- et al. synthesized the MCM-41 with large pore (diameter,
ciency anticancer treatment. Zwitterions are electrically neutral 5.4 nm) by using the mesitylene as a swelling agent.[127] The
compounds that consist of cation and anion group. NPs with membrane-impermeable protein CytC could be encapsulated
zwitterion lipids coating exhibit excellent mucus penetrating into the MSNs and delivered into the cytoplasm of HeLa cell.
ability as possessing neutral and superhydrophilic surface. In Gu et al. designed a facile strategy for preparing the large pore
addition, their special property that similar to the membrane of MSNs with the N,N-dimethylhexadecylamine (DMHA) as a
cells makes them easily to be uptaken by living cells.[112] pore size mediator.[116] In addition, the pore size could be tuned
In conclusion, due to their relative simplicity and safety, lipid- from 2.7 to 4.6 nm by adjusting the amount of DMHA. Obvi-
based delivery systems for protein delivery in live cells and tis- ously, the loading amount of CytC was correlated with their
sues have been extremely studied. However, the low efficient pore size. By adding isopropanol as a coswelling agent to adjust
permeability to tissue is still the main limitation for this strategy the pore size (4.6–7.6 nm), large-pore and well-dispersed ben-
in clinical trials. In addition, some issues, such as easy accumu- zene bridged mesoporous organosilica nanoparticles (MOSNs)
lation in the phagocytes, the high cost of raw material still hinder with only ≈50 nm diameter were synthesized by using biphasic
their clinical use. Thus, other alternative materials are needed to synthesis method.[128] In this system, the MOSNs with a larger
offer unique advantages, as well as for specific disease. pore size (7.6 nm) contained a higher loading amount of
RNase A and more sustained release profile of RNase A into
the cells. SBA-15 is another classical type of MSNs, which has
3.3. Inorganic Nanoparticles large pore size (5–30 nm) and could be utilized to delivery large
therapeutic proteins. However, the particle size are in range
Inorganic nanoparticles have generated some considerable of 800 nm to 2 µm, which limit their usage for hardly cel-
interest, such as easily tunable size and modified surface, which lular uptake.[126] Recently, small SBA-15 rods with 80–200 nm
enable them to efficiently enter into targeted cells, as well as in length and 18 ± 8 nm in width were synthesized.[129] This
contain minimal toxicity and long circulation time. Either by type SBA-15 rods showed better BSA and lysozyme absorption
noncovalent interaction or covalent attachment, proteins could and cellular uptake than that of conventional MCM-41. To con-
be encapsulated or stabilized by these NPs. Typically, these trol the release profiles of proteins, MSN pore surface could
NPs are taken up by cells via endocytosis or membrane fusion be engineered with citraconic amide which could control the
mechanism. The loaded proteins next escape from endosomal CytC release by adjusting the pH of the releasing medium. The
or directly go into cytosol or other organelle.[18] positive protein CytC, which was entrapped in the pore sur-
face functionalized MSNs could be released at endosomal pH
and maintain the same enzymatic activity as native CytC.[130]
3.3.1. Silica Nanoparticles Sun et al. recently fabricated pH- and glucose-sensitive nano-
carriers through coating dextran-maleic acid and 3-amidophe-
Nowadays, silica is an important element that has been broadly nylboronic acid (APBA) on the surface of MSNs.[131] In the
used as an additive in food, oral drugs, and cosmetics, which is neutral or acidic pH, the shell outside the MSNs would shrink
also recommended as “generally recognized as safe” by FDA. so that the insulin in the nanopore was blocked. However, the
Due to their impressive features, such as low cost, biocom- insulin could be released after the shell swelled at alkaline pH
patible, good physiochemical stability, versatile surface func- and exist of glucose. Furthermore, in vitro and in vivo studies
tionalization, and advanced stages of clinical development, demonstrated that the nanocarriers exhibited excellent cell
silica-based nanoparticles (SiNPs) have been applied for storage internalization without cytotoxicity and produced an obvious
and delivery of drugs including protein molecules and enzymes hypoglycemic effect. The pore size of MSNs mentioned above

Adv. Mater. 2019, 31, 1902791 1902791  (12 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 7.  Silica-based NPs with different structures for protein delivery. TEM images of a) PEGylated linker-MSNs. Adapted with permission.[115] Copyright
2011, American Chemical Society. b) MSNs using cationic surfactants as templating agent. Adapted with permission.[116] Copyright 2013, Elsevier Inc.
c) Hollow MSNs. Adapted with permission.[117] Copyright 2015, Royal Society of Chemistry. d) Silica nanorattles. Adapted with permission.[118] Copyright
2013, Wiley-VCH. e) Aldehyde-functionalized dendritic MSNs. Adapted with permission.[119] Copyright 2016, Elsevier B.V. f) GSH-responsive biodegrad-
able dendritic mesoporous organosilica nanoparticles. Adapted with permission.[120] Copyright 2016, American Chemical Society. g) Core–core-structured
MSNs. Adapted with permission.[121] Copyright 2015, Wiley-VCH. i) Amine-functionalized hollow dendritic MSNs. Adapted with permission.[122] Copyright
2016, Wiley-VCH. k) Hollow MOSNs. Adapted with permission.[123] Copyright 2016, American Chemical Society. m–p) Rough silica NPs with various
shell particles sizes. Adapted with permission.[124] Copyright 2015, Royal Society of Chemistry. Electron tomography (ET) slice of h) core–core-structured
MSNs. Reproduced with permission.[121] Copyright 2015, Wiley-VCH. j) Amine-functionalized hollow dendritic MSNs. Reproduced with permission.[122]
Copyright 2016, Wiley-VCH. l) Hollow MOSNs. Reproduced with permission.[123] Copyright 2016, American Chemical Society.

is normally below 20 nm, which could load relatively small pro- achieved with the magnetic stimuli or at acidic environment.
teins such as CytC, RNase A, and insulin but difficult to encap- This strategy has been considered as a promising design for the
sulate larger proteins (e.g., mTFP-Ferritin, ≈20 nm). Thus, the next generation of biomedical applications.
MSNs with large pore are urgently needed for large proteins Besides the proteins being encapsulated in large pore of
delivery. Recently, Omar et al. reported silica–iron oxide hybrid MSNs, another method for protein delivery is to attach the tar-
MSNs with large mesoporous from 20 to 60 nm, which not geted proteins on the surface of SiNPs through the electrostatic
only could immobilize large protein Mtfp-Ferritin, but also interaction or chemical modification. For example, Kane and co-
could be rapid biodegraded in fetal bovine serum (FBS) and/ workers prepared the SiNPs (≈20 nm) with the hydrophobic long
or at a magnetic responsiveness.[132] As showed in Figure 8, the chains on the surface. The targeted proteins could be attached
controllable release of the protein in vitro and in live cells was on the particle surface through electrostatic and hydrophobic

Adv. Mater. 2019, 31, 1902791 1902791  (13 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 8.  a) Schematic illustration of the loading of large proteins into silica–iron oxide–propylamine nanocomposites, and hypothesis interaction
between NPs and mTFP-Ferritin. b) TEM micrographs of the NPs. c) High-magnification TEM micrograph of the NPs supporting the Sherrer’s equation
finding of 4.5 nm iron oxide nanophases incorporated into the silica framework. d) The degradability of NPs in FBS. Release profile of proteins from
the NPs via e) different pH and f) magnetic actuation. CLSM images of the protein loaded NPs in HeLa cells. g) Nucleus were stained in blue with
DAPI dyes, h) mTFP-Ferritin protein appears with the green fluorescence, i) cells membranes in red with CellMask and j) merged images. Adapted
with permission.[132] Copyright 2016, Elsevier. B.V.

interaction. Subsequent biological results proved that intracel- to connect the proteins with the silica nanoparticles is another
lular delivery of specific proteins with this system could keep powerful way to deliver the proteins to the target site. In order to
the protein bioactivity and influence signaling pathways.[133,134] control the protein release at specific environment (e.g., acidic
In addition, Sun et al. designed the gold nanoparticle (AuNP)- pH and glucose) or avoid the loss of biomolecules over time,
capped MSNs with luciferase and luciferin coimmobiliza- those proteins could be modified on the surface with specific
tion.[115] The AuNPs were connected by disulfide linkage, which linkage.[138–140] To an extent, the highly sensitive proteins could
could be cleaved by intracellular disulfide-reducing antioxidant also maintain their bioactivity. For example, Lin and co-workers
and subsequent resulted in the release of the luciferase and synthesized the MSN-based nanocarriers to allow efficient intra-
luciferin. This system could be designed as tumor growth and cellular protein delivery.[138] In this system, the cyclic adeno-
metastasis monitor. Recently, Li et al. prepared the SiNPs with sine monophosphase (cAMP) was first filled in the nanopore,
ultrasmall luminescent crystals embedding for BSA protein followed by the modification of insulin with gluconic acid as a
delivery.[135] In this system, the BSA bonded with the amino linkage. In the presence of saccharides, the linkage was cleaved,
groups on the surface of SiNPs through the electrostatic interac- subsequently resulted in the release of insulin and cAMP. Tra-
tion and hydrophilic absorption. The protein releasing progress ditional way to bind the proteins on the surface of nanoparti-
was monitored by the upconversion emission intensity of lumi- cles often leads to the immediate formation of dense monolayer,
nescent crystals. To enhance the loading efficiency of proteins which would not affect the bioactivity of the proteins with the
on the surface of silica particles and promote the cellular uptake, robust tertiary structure (e.g., GFP, myoglobin, or HRP), but
a kind of novel SiNPs with controllable surface roughness was not suitable for the highly sensitive proteins.[140,141] To keep
fabricated (Figure 7m–p). The proteins could be immobilized in the bioactivity of these sensitive proteins, the zwitterion-stabi-
the void spaces of rough surface. Results showed that the hydro- lized silica nanoparticles have been synthesized by Niemeyer’s
phobic modification of the roughness surface enhanced the pro- group.[140] In this system, Schiff base was used to immobilize
tein adsorption capacity and improved the cellular uptake per- the sensitive proteins, which could effectively load the very deli-
formance.[124,136,137] Using the chemical modification method cate functional proteins to generate active biohybrid system.

Adv. Mater. 2019, 31, 1902791 1902791  (14 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 9. a) Schematic illustration of the preparation of bisorthosilicate-containing azo monomer based; CPD coated; fluorescent dye and black
hole quencher doped; antibody encapsulated biodegradable HSNs for intracellular delivery. b) The CPD-Cetuximab@BS-qNP were incubated under
normoxic and hypoxic conditions and protein was released under hypoxic condition with fluorescence turn on imaging. c) XTT assay of different cells
treated with CPD-Cetuximab@BS-NP under normoxic and hypoxic conditions. Reproduced with permission.[148] Copyright 2017, Wiley-VCH.

Dendritic Silica Nanoparticles: Owning to the large degradation.[143,144] For example, Mou and co-workers have
mesoporous channels and special dendritic superstructures, successfully confined HRP in the cavity of HSNs.[145] In vitro
the dendritic mesoporous silica nanoparticles (DMSNs) have studies showed that this system could keep the enzyme stability
recently attracted a growing interest in protein delivery applica- in the presence of the denaturant and urea. Furthermore, the
tion.[142] Comparing with conventional MSNs, this kind of novel catalytic activity was also preserved as the prodrug indole-3-
porous materials possess larger pore size from ≈10 to ≈45 nm acetic acid was oxidized by HRP to produce toxic free radicals
that can enhance large protein loading capacity.[121,123] For for killing HeLa cells. Another study showed that the mag-
example, Yu and co-workers reported a anion assisted approach nesium HSN, which was prepared using a modified classical
to synthesize DMSNs with the pore size of 29.5 nm. By further Stöber method, displayed highly amount of hemoglobin and
alkaline etching with Na2CO3 solution, the dendritic hollow anticancer drugs absorption.[146] Next cell-based experiments
mesoporous silica nanoparticles (DHMSNs) could be fabri- exhibited that this codelivery of HSNs system had efficient
cated.[123] Results demonstrated that the DHMSNs possessed anticancer activity. In order to improve the loading efficiency of
higher loading capacity of BSA and lower hemolytic activity proteins, a new kind of HSNs, hierarchically structured HSNs
than DMSNs, showing great potential in delivering large bio- which possess shell-in-shell structure, was reported by Cao
molecules. In addition, Zhu and co-workers prepared aldehyde et al.[147] In this system, glucose oxidase could be immobilized
functionalized DMSNs which could form the pH labile imine not only onto the external surface, but also inside the inner
bonds between aldehyde groups on the DMSNs surface and channel. Comparing with normal HSNs, hierarchically HSNs
primary amines of BSA.[119] Due to the linkage of labile imine exhibited higher mechanical stability and enzyme loading
bonds, it could control the BSA release by changing the pH capacity, lower enzyme leaching. By adding breakable orga-
value. By using bis[3-(triethoxysilyl)propyl]tetrasulfide (BTES) nosilica matrices, such as bispropyldisulfide derivatives in the
as silica source, Yang et al. synthesized the disulfide bond- preparation of hierarchically HSNs, the native proteins CytC,
linked biodegradable DMSNs with different pore sizes.[120] TRAIL Apo2 ligand, and onconase could be encapsulated in
Results revealed that the DMSNs could load a mass of RNase A. the HSNs and released in their functional conformations once
Furthermore, the degradation of the DMSNs at high intracel- against external stimuli.[144] Recently, Yuan et al. successfully
lular GSH level resulted in the fast release of loading RNase A, designed biodegradable HSNs for RNaseA or native antibody
indicating this system could deliver protein effectively in living Cetuximab intracellular delivering.[148] Schematic of prepa-
cells but reduce the cytotoxicity. ration of HSNs and the intracellular delivery of delivery are
Hollow Silica Nanoparticles: Hollow silica nanoparticles pos- shown in Figure  9a. In this nanosystem, the out shell of bio-
sess a series of advantages including large surface area, low degradable HSNs are synthesized by bisorthosilicate-containing
density and tunable void volume that could offer more storage azo monomer, which could be reduced by flavoproteins under
spaces for loading proteins. Moreover, due to the external hypoxic conditions. In addition, the surface of HSNs were
shell protection, the proteins encapsulated into the internal coated with CPD that promoted the HSNs transported into
spaces could preserve the bioactivity from the enzymatic cells via endocytosis-independent uptake. Moreover, the HSNs

Adv. Mater. 2019, 31, 1902791 1902791  (15 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

were doped with fluorescent dye and black hole quencher, thus showing significantly uptake in mice and growth inhibition of
could simultaneous image and monitor the intracellular pro- xenograft tumors without detectable systemic toxicity.[157] In
tein release and cell condition under hypoxic conditions. The order to promote the proteins release from gold nanoparticles,
results showed that Cetuximab encapsulated biodegradable Reich and co-workers designed hollow gold nanoshells that
nanocapsules (CPD-Cetuximab@BS-NP) successfully uptaken coated by a DNA layer.[159] The DNA layer provided terminal
and released in hypoxic A431 cells. Importantly, the released amine groups used to conjugate the Ni2+ and protein chelator.
Cetuximab inhibited pEGFR expression, and subsequently Under the irradiation of NIR laser pulses, this system could
leading to time-dependent cell death (Figure 9b,c). be thermalized and release the loading proteins GFP into the
Over the years, many antibodies/proteins loaded nanocar- cytosol. Recently, AuNPs were functionalized with arginate
riers with desired functions have been reported for intracel- that could bind oligo-glutamic acid tagged protein.[160] This
lular delivery. The delivery of native antibody/proteins into coengineering system successfully transported proteins with
subcellular targets is regarded as promising strategy for poten- different size, charge and function into the cytosol or nucleus.
tial clinical application but rarely reported. In this regard, In addition, the engineered proteins Cre recombinase and
Yao and co-workers first designed a TPP-modified biodegrad- granzyme A could reserve bioactivity after cytosolic delivery.
able HSNs for intracellular mitochondria delivery of different The drug proteins could be trapped and degraded inside
kinds of proteins and antibodies, such as BSA, Cetuximab, endosomes and lysosomes, which has been considered as one
monoamine oxidase A (MAO-A), and anticytochrome c oxi- of the main obstacles in protein delivery. To improve cellular
dase subunit 2 (anti-MTCO2).[149] In this system, two types of uptaken efficiency and avoid endosomal sequestration, Rotello
monomers, a disulfide monomer bis[3(triethoxysilyl)propyl]- and co-workers designed a serious AuNP-stabilized capsule for
disulfide (BTEPDS) and tetraethylorthosilicate (TEOS), were effective protein delivery.[161,162] The AuNPs were first bonded
used to make HSNs, which could slowly degraded around with a tetrapeptide, His-Lys-Arg-Lys (HKRK), which interacted
24–48 h under the intracellular GSH. CPDs were conjugated with carboxylates of the oil to pin the AuNPs on the capsule
on the surface of HSNs via a two-step biorthogonal reaction. surface. After that, the negative proteins were attached on the
The TPP/CPDs facilitated the HSNs rapid internalization into AuNPs through the electrostatic interaction. In this system,
the mitochondria before premature degradation by cytosolic the capsule surface could fuse with cell membrane and then
GSH. Finally, a mitochondria enzyme MAO-A and functional release the proteins directly into the cytosol. By using caspase-3
native antibody anti-MTCO2 were successfully delivered into as model, the delivered proteins effectively induced apoptosis
mitochondria and led to detectable antibody-target binding, of HeLa cells, indicating it is a powerful strategy for efficient
thus providing novel methods of subcellular antibodies/pro- delivery of functional proteins in their native forms.[163,164] In
teins delivery for improving protein-based therapies. order to further optimize the AuNPs stabilized capsules system
for delivering large proteins into the cytosol, the same group
designed AuNPs with 1-guanidino-2-(4-imidazole)propionic
3.3.2. Metal-Based Nanoparticles acid (GIPA) as the terminal group. the guanidine group could
interact with both protein payload and the oil core to stabilize
Metal-based nanoparticles have been considered as promising the capsules structure. Final results showed that the large pro-
delivery systems due to their small size, nondeformable shape teins dsRed and β-gal were effectively transduced into cytosol
and diversity, etc. In addition, the intrinsic physiochemical and remained its enzymatic activity by GIPA AuNPs stabilized
properties, such as photoacoustic and photothermal effect capsules (Figure 10).[165]
make them as excellent candidates in the cancer treatment. Recently, AuNPs have been utilized as Cas9 RNP loaded
Among them, the gold nanoparticles have been well studied for nanocarriers for direct cytosolic delivery and efficient gene
protein carrier construction. editing. The AuNPs were conjugated with arginine or DNA,
Gold Nanoparticles: The AuNPs contain highly tunable sur- which could complex with donor DNA and Cas9 RNP to pre-
face area that can be engineered with various biomolecules pare stabilized nanocarriers.[166,167] The arginine AuNPs can
such as DNA/RNA (e.g., aptamers),[150] proteins (e.g., TAT self-assemble with glutamate residues/NLS modified Cas9 RNP
peptide, aspartic acid, and human transferrin),[151–154] carbo- to form nanoassemblies through the charge interaction.[166]
hydrate, polymer (e.g., chitosan)[155] and lipid.[156] Fabrication In addition, the DNA could also bind with Cas9 RNP-loaded
of AuNPs surfaces with functional biomolecules could make AuNPs to form stable nanocarriers. After conjugation with
the particles high binding affinity and long-term stability, and polymer PAsp(DET), this system could trigger endosomal dis-
promote drugs efficient transport into the cells or organs. ruption.[167] Both kinds of AuNPs could successfully deliver
Recently, Lee and co-workers developed a multiple proteins Cas9 RNP into the cytoplasm and induce gene editing in vitro
delivery system based on AuNPs functionalized with His- and or in vivo.
GST-tag aptamers.[157,158] With this system, any recombinant Magnetic Nanoparticles (MNPs): The magnetic nanoparticles
protein labelled a commonly used tag (e.g., His and GST) could have been developed for several technological applications such
be purified and loaded onto the aptamer-conjugated AuNPs. as medical application, biosensing, magnetic storage media
It is noted that the specific apoptosis-inducing BIM protein and contrast agents in magnetic resonance imaging (MRI).
was largely localized in the mitochondria of HeLa cells and In order to stabilize the MNPs, their surface could be coated
exhibited efficient antitumor activity. Furthermore, epidermal with polymer (e.g., alginate and chitosan), inorganic materials
growth factor (EGF), a target protein could be loaded onto the (e.g., silica and gold) or modified with chemical compounds
AuNP–(His-GST)–aptamer system with BIM protein together, for binding drugs (e.g., proteins, antibodies, enzymes, or

Adv. Mater. 2019, 31, 1902791 1902791  (16 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 10.  a) Schematic illustration of GIPA AuNPs stabilized capsules are utilized for dsRed and β-galactosidase delivery. LSCM images showing
b) dsRed and c) FITC-β-gal delivery into HeLa cells by GIPA AuNPs stabilized capsules. Reproduced with permission.[165] Copyright 2016, Royal Society
of Chemistry.

nucleotides). In addition, the external magnetic field is often Metal Organic Framework–Based Nanoparticles: Metal-organic
used to promote MNPs to target to the specific organ, tissue frameworks, which were first discovered in 1989 by Robson,[173]
or tumor.[168,169] For instance, Yang and co-workers constructed are class of porous crystalline materials that are synthesized
long-circulating heparin functionalized MNPs for protamine with metal containing nodes and organic ligands.[174,175] Their
protein delivering.[170] The MNPs were coated with starch and structure, crystal morphology, pore shape and size, physico-
subsequently modified with a mount of 20 kDa PEG chains to chemical functionality (e.g., stability, hydrophobicity, and sur-
favor MNPs with passive target and long circulation. Moreover, face charge) can be determined by monodentate ligands.[176]
this system also showed the high capability for MRI and effi- Moreover, the surface modification of MOFs can enhance
ciency for subcutaneous tumor magnetic targeting. Recently, their biomedical properties like improving their stability under
Lee and co-workers fabricated a MNPs system for proteins physiological conditions, introducing additional biological func-
transporting and trigger release with redox response as stimuli tionalities and providing the MOFs with bioadhesive or tar-
(Figure 11).[171] In this system, the Cy5.5-labeled human serum geting properties.[177] Due to their mutable properties, MOFs
albumin (Cy5.5-HSA) was complexed with MNPs through the show great promise in many potential applications such as
electrostatic interaction, which was served as MRI and NIR catalysis, separation, chemical sensing and drug delivery.[178–181]
imaging probe in animal and tumor models (Figure 11c,d). MOFs are of interest as promising drug delivery carriers
The disulfide-linked methoxy-poly-(ethylenelycol)-block- mainly due to their high drug loading capability. However,
poly(dopamine-diethylenetriamine-l-glutamate) ligands were most studies are focus on delivering small molecular drugs,
synthesized and the functional groups (dopamine-diethylen- indeed, many poor solubility and/or stability drugs (e.g.,
etriamine) facilitated the self-assembly of MNPs. In the intra- busulfan, Dox, cidofovir, nimesulide, cisplatin, and siRNAs)
cellular environment, the Cy5.5-HSA could be rapidly released have been successfully loaded into MOFs which not only pro-
under high GSH conditions, and remained the bioactivity in tect drugs from biodegradation but also enhance their thera-
the living cells. By combining different nanoparticles, Chung peutic efficacy.[178,182] On the other hand, MOFs can also be
and co-workers developed a multifunctional gold coated MNPs/ utilized as carriers for enzyme immobilization and delivering
engineered protein G system to deliver antibodies into cells.[172] because of their high surface area, large pore volume, easily
The protein G was first engineered with an affinity tag which pore size tuning and mild fabricated conditions.[183] Generally,
could bind with NTA-Ni(II) on the surface of gold-coated MNPs. the preparation strategies of MOF–enzyme composites can
Next, this system was constructed by fusing the cell penetration be categorized into for main types: surface attachment, cova-
peptides with the N- and C-terminals of protein G. Cell-based lent linkage, pore entrapment, and coprecipitation.[184] Those
experiments demonstrated that this system could deliver the enzymes could interact with MOFs either by electrostatic attrac-
antibodies into host cells with targeting, imaging, and manipu- tion, hydrogen bonding interaction or covalent conjugations.
lation of mitochondria. It is noteworthy that MOF-immobilized enzymes offer many

Adv. Mater. 2019, 31, 1902791 1902791  (17 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 11.  a) Schematic illustration of the Cy5.5-HSA-loaded MNP for intracellular protein delivery. b) In vitro release of Cy5.5-HSA from redox sensi-
tive RMNs in PBS. c) In vivo NIR imaging and d) in vivo T2-weighted MR imaging of breast cancer tumor-bearing mice after intravenous injection of
Cy5.5-HSA-loaded MNP. Reproduced with permission.[171] Copyright 2017, American Chemical Society.

advantages such as improving the reusability of immobilized the enzymes from proteolysis and chelating.[188] Li et al. suc-
enzymes, enhancing the catalytic activities and stability under cessfully encapsulated a nerve agent detoxifying enzyme,
denaturing conditions.[185] For example, Zhou and co-workers organophosphorus acid anhydrolase into a water stable zirco-
fabricated a series of highly stable MOFs and found that one nium MOF PCN-128y (4.4 nm) with a high loading. Moreover,
of the MOFs, PCN-333(Al), exhibits largest cage (5.5 nm) and the immobilized enzyme into PCN-128y displayed long-term
highest void volumes (3.84 cm3 g−1), and could encapsulate stabilities at high temperature (70 °C) and in dry form.[189]
high loading amounts of HRP, CytC and microperoxidase-11 Recently, some novel methods are developed to synthesize
(MP-11). Further experiments proved that those immobilized protein-loaded MOFs and their enzymatic activity are evaluated
enzymes showed better performance under harsh conditions in vitro and/or in vivo experiments.[190–192] Zhang et al. used
and exhibited smaller Km than free enzymes.[186] The conven- facile synthesis protocols to prepare protein embedded MOFs
tional fabrication methods of enzyme–MOF composites were vaccines. Herein, OVA was selected as model antigen that was
used organic solvent, which would easily denature enzymes. It encapsulated into ZIF-8. In addition, the unmethylated cyto-
is urgent to pursue a facile and mild reaction conditions for the sine–phosphate–guanine oligodeoxynucleotides (CpG ODNs)
synthesis of composites to preserve enzymes’ activities. Shieh which used as vaccine adjuvants were attached on the surface of
et al. developed a de novo approach under aqueous conditions to the OVA@ZIF-8 nanoparticles to improve the biocompatibility
embed catalase into zeolitic imidazolate framework 90 (ZIF-90) and immunogenicity of the nanoparticle. Both in vitro and in
supports of pore sizes smaller than the size of the enzyme. vivo experiments proved that MOF-based vaccine platform had
The composites not only enhanced the stability and recycla- good biocompatibility and pH stimulated release properties to
bility of the enzyme, but also retained the peroxidase activity delivery antigens and CpG ODNs into targeted cells, inducing
of catalase.[187] In order to improve the therapeutic effect, Ge strong cellular and humoral immune response.[190] Lian et al.
and co-workers prepared a multiple enzyme-embedded ZIF-8 recently developed antioxidant enzymes (SOD) and catalase-
within 30 min at 25 °C by simply mixing aqueous solutions loaded MOFs and found those nano-MOFs could maintain
containing zinc ions, 2-methylimidazole, glucose oxidase and the enzymatic activity and protect human cells from toxic reac-
HRP. The confinement of the MOF scaffolds greatly shielded tive oxygen species for up to a week. Interestingly, although

Adv. Mater. 2019, 31, 1902791 1902791  (18 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 12.  a) Schematic illustration of MOF NPs for intracellular protein delivery. b) TEM image of caspase 3/HSA@ZIF-8. c) Long term activity of
β-Gal and β-Gal in ZIF-8. d) Fluorescent images of Hela cells treated with caspase 3/HSA@ZIF-8 for 4h. e) Cell viability after treatment with HSA@
ZIF-8 or caspase 3/HSA@ZIF-8 of different concentration for 4h. Reproduced with permission.[194] Copyright 2018, American Chemical Society.

the nano-MOFs were localized inside lysosomes, they still keep pathway and preserve the activity of proteins throughout the
their chemical stability and enzymatic activity against the pro- intracellular delivery process (Figure 12).
tease degradation and low pH environment.[191] As mentioned MOFs are also an emerging class of platform for Cas9 RNP
above, surface modification of MOFs could enhance their cytosolic delivery and CRISPR/Cas9 genome editing. The
biomedical abilities, previous studies proved that MOF con- ZIF-90 or ZIF-8/Cas9 RNP nanocarriers could be fabricated by
jugated with DNA could stabilize them in high concentration self-assembling with Zn2+, proteins and imidazole-2-carboxal-
NaCl (0.4 m) media and ensure facile cellular entry.[193] Based dehyde (2-ICA) or 2-methylimidazole.[195,196] Yang et al. found
on previous studies, Wang et al. fabricated nucleic acid-MOF that the ZIF-90 could be disintegrated by high concentration of
based delivery carriers, the results showed that the insulin ATP (10 × 10−3 m) and released the protein inside cells due to
was encapsulated into MOF with high loading and enzymatic the competitive coordination between Zn2+ and ATP that dis-
activity and a tenfold enhancement of cellular uptake were assembles ZIF-90. In the ZIF-90 system, various proteins with
achieved.[192] Using the biomimetic mineralization method, different molecular weights, including BSA, SOD, GFP, RNase
Chen et al. synthesized a biocompatible polyvinylpylrolidoe A-NBC and Cas9 RNP are encapsulated into the ZIF-90. In
(PVP) coated with protein-encapsulating ZIF-8 NPs for effi- addition, the functional proteins RNase A-NBC and Cas9 RNP
cient intracellular delivery.[194] In this systems, not only single were successfully delivered into cells for enhancing cytotoxicity
protein such as BSA, ferritin, EGFP, or β-Gal, but also multiple against HeLa cells or inducing genome editing.[195]
proteins caspase 3/HSA or BSA/RFP/β-Gal could be success-
fully coencapsulated into single ZIF-8 NPs with good monodis-
persity. The coated PVP enhanced the stability of NPs and the 3.3.3. Carbon-Based Nanomaterials
encapsulated proteins retained their activities throughout the
encapsulation and release. Finally, the protein-encapsulated Due to possess various attractive properties, carbon-based
NPs could enter cells through a lipid-raft-mediated endocytosis nanomaterials have been shown to extremely useful in energy

Adv. Mater. 2019, 31, 1902791 1902791  (19 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 13.  Schematic illustration of nuclear protein LB1-loaded SWCNTs enter into HeLa cells and quantification of SWCNTs localize to the nucleus of
cells by using fluorescence lifetime imaging microscopy. Reproduced with permission.[204] Copyright 2016, American Chemical Society.

storage, electricity, renewable energy, biosensing, and biomedi- and proteins, the laccase could be immobilized on the CNTs and
cine. Based on their different structures, carbon nanomaterials showed high electrocatalytic activity.[206] Another important and
have been divided into different forms, including carbon nano- feasible route to enhance the proteins immobilization is the
tubes (CNTs), nanodiamonds (NDs), fullerenes, carbon dots, modification of CNTs with polymers. Until now, many poly­
and carbon nanospheres (CNs). merization techniques (e.g., anionic polymerization and radical
Carbon Nanotubes: Carbon nanotubes can be designed as a polymerization) have been used to prepare polymer function-
graphite sheet rolled up into a nanoscale tube. There are two alized CNTs, their physicochemical and biological properties
kinds of CNTs according to the number of walls around the could be affected after polymer-functionalization.[198,207] The
nanotubes, namely, single-wall carbon nanotubes (SWCNTs) most studies of polymer functionalization CNTs are focus on
and multiwall CNTs. Due to the excellent electrical, mechanical, biocatalysts, gas detector, and biosensor; however, few studies
electromechanical, and chemical properties, CNTs have been are applied for protein delivery and disease therapy. Thus, this
used for a number of potential applications in various research research area will attract an increasing interest in future.
area, including reinforced materials, hydrogen storage, elec- Nanodiamonds: Nanodiamonds are a type of carbon nano-
tronic devices, field-emission materials, biomedical imaging, particles that possess a truncated octahedral architecture with
and drug delivery.[197] CNTs are receiving a great deal of atten- diameter of 2–10 nm. Beside the advantages of the superior
tion for protein immobilization and delivery. CNTs could be properties of bulk diamond, including high thermal conduc-
modified to generate functional groups (e.g., –COOH, –OH) tivity, electrical resistivity, optical properties, chemical stability,
for proteins interaction via either noncovalent (e.g., electrostatic superior hardness and Young’s modulus, NDs also contain
or hydrophobic interaction) or covalent conjugations.[198–201] more advantages than bulk diamond, such as larger surface
For instance, Dai and co-workers reported SWCNT-based area, smaller size, better biocompatibility, and higher adsorp-
nanocarriers, which could complex with protein through elec- tion capacity.[208,209] In comparison with other carbon nanopar-
trostatic interaction. In this study, they uncovered that the ticles, NDs showed less toxic to a variety of cell types and did
CNTs could transport proteins, such as streptavidin, protein not produce significant reactive oxygen species.[210] Due to easy
A (SpA), BSA, and CytC, into living cells by energy-dependent surface modification of functional groups, NDs display various
endocytosis.[202,203] In addition, Boyer et al. recent developed medical application in drug delivery, tissue scaffolds, protein
a noncovalent recombinantly engineered lamin B1 (LB1)- mimics, and surgical implants.[208] For example, Lora Huang
based SWCNTs.[204] In this system, the LB1 contains a nuclear and Chang designed carboxylation/oxidization NDs that coated
localization sequence which could promote the functionalized poly-l-lysine for covalent attachment of proteins.[211] The protein
SWCNTs to localize in the nucleus. Moreover, the uptaken CytC was successfully absorbed and immobilized on NDs. How-
process of SWCNTs in cells can be precisely determined and ever, the biological activity was not investigated in this study.
directly visualized by using confocal Raman and fluorescence Another study showed that hormone insulin could be nonco-
lifetime imaging microscopes (Figure 13). valently bound to detonated NDs and released by changing pH
In order to stabilize protein, linkages are normally designed value.[212] Studies showed that the adsorbed insulin remained
for covalent immobilization of proteins onto CNTs. For example, largely inactive but preserved function followed by desorp-
acid oxidized SWCNTs was incubated with 1-ethyl-3-(3-dimeth- tion. Recently, Hu et al. prepared ND–protein complex for
ylaminopropyl)carbodiimide (EDC) and biotin-LC-PEO-amine oral delivery of proteins.[213] The complex could facilitate pro-
to afford biotin functionalized SWCNTs.[205] The SWCNTs tein desorption in the cytosol by the functionalization of poly-
could bind with the specific protein streptavidin and enter into d-lysine (PDL) on the surface of NDs (Figure  14a). With this
human promyelocytic leukemia cells and human T cells via the system, RNase protein could be delivered into intestinal cells of
endocytosis pathway. In addition, by a typical glutaraldehyde Drosophila, and consequently triggered regenerative divisions of
crosslinking reaction between 1-aminopyrene-modified CNTs intestinal stem cells for tissue repair (Figure 14b,c).

Adv. Mater. 2019, 31, 1902791 1902791  (20 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 14.  a) Adsorption of RFP to NDs or NDs-PDL in PBS and desorption of RFP from NDs or NDs-PDL in cell lysate. b) Viability assays of HeLa
cells after incubation with RNase, NDs-PDL, or NDs-PDL-RNase. c) CLSM imaging of oral delivery of RFP by NDs-PDL into Drosophila midgut. RFP:
Red fluorescent protein. Modified with permission.[213] Copyright 2017, American Chemical Society.

Other Carbon-Based Nanomaterials: Graphene oxide (GOx) Carbon nanospheres are another promising platform that
is derived from graphene upon the oxidative treatment. It has been applied for delivering proteins. Scaffold matrix attach-
has been widely used as nanocarrier based on its promising ment region binding protein 1 (SMAR1) have been found to
features, such as hydrophilic property, high surface area and downregulate in advanced stages of human breast cancer.
destabilization of lipid bilayer membranes. For example, Geest Recently, Paknikar and co-workers prepared SMAR1 loaded
and co-workers prepared OVA-modified GOx nanosheets and CNs to control breast tumor in mice model.[219] Interestingly,
demonstrated the nanosheets could be efficiently internalized this nanocomplex could internalize and localize in the nucleus
by dendritic cells.[214] In addition, Zhang and co-workers fabri- of MCF-7 cells. Furthermore, the delivered SMAR1 exhibited
cated the PEGylated GOx nanovector for protein delivery.[215] In apoptotic, antiproliferative and antiangiogenic activities and
this study, RNase A, BSA and Kinase A could be loaded on the significantly reduced the tumor volumes in metastatic breast
nanovector via π–π stacking and other noncovalent interactions. tumor bearing Balb/c mice.
Cell-based experiments showed that the intracellular delivery Until now, various inorganic nanoparticle-mediated delivery
of RNase A kept activity and could lead to cell death. To pro- systems have been studied for intracellular delivery of protein.
tect Cas9/sgRNA against enzymatic cleavage, a new function- These inorganic nanoparticles with unique physical/chemical
alized GOx with PEG and PEI modification was fabricated by properties display great potential for development of a non-
Yue et al.[216] The results showed that GOx-based nanomaterials toxic, biocompatible and nonimmunogenic delivery system. In
provided an effective platform for Cas9/sgRNA delivery and addition, unlike the lipid-based nanoparticles, these inorganic
protection. In this system, the loaded Cas9/sgRNA adsorb on nanoparticles are considered to be easier to scale up manu-
the Gox-PEG-PEI via physical adsorption and π-stacking inter- facturing. The stability of these inorganic nanoparticles ena-
actions. The nanocarriers could deliver Cas9/sgRNA in human bles them with long-term circulation in the body, however, in
cells through endocytosis and release based on the proton another respect, makes them difficulty to be decomposed and
sponge effect of PEI, resulting in efficient genome editing. cleared from body, thus finally leading to the accumulation in
Carbon nanodots (C-dots) contain numbers of stacked gra- the liver, lung and kidney.[105] Therefore, development of the
phene layers with the size of diameter from 1 to 10 nm. Their biodegradable inorganic nanoparticle-based system is an effec-
properties are similar as that of graphene and have been tive way to solve this issue.
designed for protein delivery. For instance, Liu and co-workers
discovered that the hydrophobic and van der Waals forces inter-
actions between two serum proteins (HSA and ϒ-globulins) 3.4. Cell-Penetrating Peptides
and C-dots by a series of photophysical measurements, which
can be used for proteins delivery.[217] Furthermore, Zhang et al. Besides the modification with nanoparticles/polymer, cell-pen-
mixed C-dots with EGFP to prepare C-dots/EGFP nanocomplex etrating peptide itself could also be used for proteins delivery.
with a size of around 200 nm. This nanosystem exhibited effec- The first CPP, TAT peptide, has been discovered from the
tively EGFP delivery into the cytoplasm of HeLa cells.[218] human immunodeficiency virus (HIV) by Frankel and Pabo in

Adv. Mater. 2019, 31, 1902791 1902791  (21 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

1988, which arouse scientists intense interest as transporting enter into cells. Based on discovering the annexin family binds
cargos for intracellular uptake.[220] During the past three dec- to phosphatidylethanolamine, an annexin derived CPP that
ades, numerous CPPs have been developed and identified as riches in hydrophobic residues was screened and synthesized
delivering the bioactive cargos in vitro and in vivo. Those CPPs from twelve isoforms of annexin.[223] Results showed that this
can be derived from proteins of microorganisms, plasmid dis- CPP could be served as a potential candidate for the successful
play screenings, phage, antimicrobial peptides, heparin-, DNA-, delivery of functional β-gal and Cre recombinase in vitro and in
RNA-binding proteins and signal peptides.[221–223] For example, vivo. Due to possess minimal toxicity to mammalian cells, anti-
Fu et al. proposed a brain-targeting peptide RDP derived from microbial peptides (AMPs) have drawn an increasing attraction
the rabies virus glycoprotein, which is consisting of 39 amino to the scientists. For instance, Liu and co-workers screened and
acids residues, for efficient target delivery of biological active discovered a 13-residue peptide, aurein 1.2 (GLFDIIKKIAESF),
proteins, such as luciferase, β-Gal, brain derived neurotrophic which could also promote the endosomal escape of a variety of
factor and glial cell-derived neurotrophic factor, into the cen- cargos fused to +36 GFP.[229] Recently, a cationic, amphiphilic
tral nervous system.[224,225] The in vivo results showed that peptide with a strong membrane lytic activity was selected as
the conjugate of RDP and proteins exhibited significant neu- template for the endosomolytic peptides by Akishiba et al.[230]
roprotective effect. Although CPPs can be used to effectively This endosomolytic peptides (L17E) were developed by intro-
deliver proteins into the host cells, however, due to the limita- ducing one or two negatively charged glutamic acid residues
tion of instability in poor serum, CPPs can be trapped in the into the hydrophobic face. In this system, the positive charges
endosomes and lysosomal degradation, induce cytotoxic at of peptide were retained to facilitate the cell surface adsorption.
high concentrations once transduced into cells, thus resulting By the induction of micropinocytosis, this system could enable
in poor achievement in their biological outcomes. To overcome the cytosolic release of endocytosed proteins (Cre recombi-
this issue, numerous strategies, such as introduction of the nase and IgG) in terms of physicochemical peptide-membrane
endosomolytic agents used to disrupt endosomal membrane, interactions. Furthermore, the L17E could also mediate cyto-
have been developed to improve the stability and transporting solic delivery of exosome-encapsulated proteins in living cells
efficiency.[226] For instance, Erazo-Oliveras et al. fabricated (Figure 15a,b). The cell-based result showed that saporin-encap-
tetramethylrhodamine-labeled dimer of the TAT and discovered sulated exosomes in the presence of L17E led to significant cell
that this kind of TAT could deliver the EGFP into the cytosol death. (Figure 15c).
and nucleus in several cell lines.[227] Nischan and co-workers The length of peptides is much shorter than antibodies,
synthesized another cyclic CPP to promote the GFP delivery and thus show a less expensive option for protein delivery. In
in the cytosol and nucleus.[228] Specifically, the chemoselec- addition, peptides have the advantage of being nonimmuno-
tive copper-catalyzed azide–alkyne cycloaddition was applied genic in comparison to the antibodies. These features make
to access a cyclic arginine rich TAT–GFP conjugate. In com- the peptides as promising delivery platform for a wide range
paration with linear TAT–GFP, cyclic TAT–GFP could directly of protein-based therapeutics.[231] However, some limitations,

Figure 15.  a) Schematic illustration of L17E enhances the efficacy of exosome-mediated intracellular delivery. b) CLSM imaging of the ability of exog-
enous antibody to target cellular protein in the presence or absence of L17E. c) HeLa cells were treated with exosomes that contained saporin with or
without L17E. Reproduced with permission.[230] Copyright 2017, Springer Nature.

Adv. Mater. 2019, 31, 1902791 1902791  (22 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

such as the lack of cell selectivity of CPPs, are still needed to be to prepare the proteins nanoparticles, including collagen, gel-
solved. For instance, it is essential to develop protein delivery atin, albumin, casein, elastin, whey proteins, soy proteins, silk
systems with the features of tumor targeting and these mem- proteins, gliadin, and lectins.[234,235]
brane permeable CPPs to make the site-specific delivery and
intracellular trafficking. Such smart CPP-based delivery system
could be designed with either tumor-targeted peptides or com- 3.5.1. Protein Cages
bined features in multicomponent delivery systems to enhance
the delivery/release capability of proteins. Additionally, further Protein cages, such as ferritins, viral capsids, and heat shock
development and research, particularly in vivo studies will be proteins, are attractive nanoscale cargo delivery vehicles which
essential for the novel peptide-based protein delivery systems. have been used for protein delivery. The diameter of cages is
usually less than 30 nm which can exit the vasculature, per-
meate tissue, and enter the lymphatic system by passive dif-
3.5. Protein-Based Nanoparticles fusion. In the protein cage-based NPs, most protein drugs
could be localized on the surface of cages via either covalent
Comparing with other nanocarriers, such as polymer, lipid, bioconjugation or genetic fusion.[17] For example, Cao and co-
and inorganic nanoparticles, protein-based nanoparticles have workers applied genetic method to generate human ferritin
more advantages including biocompatibility, biofunctionality, H-chain protein (FTH1)-based NPs that fused with epidermal
molecular recognition in the biological environment, and abun- growth factor receptor (EGF-FTH1).[236] Cell-based results
dant renewable sources and biodegradability into small amino demonstrated that this EGF-FTH1 NPs could specifically bind
acid. More importantly, the protein nanoparticles could be pre- to breast cancer MCF-7 cells and MDA-MB-231 cells but not
cise genetic engineered with functional sites of distinct loca- normal breast epithelial MCF-10A cells. Kang and co-workers
tions for specific cells targeting and protein delivering.[232,233] genetically introduced the fragment crystallizable region (Fc)-
The extraordinary binding capacity of various drugs could be binding peptide (FcBP) into the loop of the ferritin cage. In this
achieved by different interactions between multifunctional cage, both human and rabbit IgGs could noncovalent bind with
groups of polypeptides and therapeutic compounds.[232] Two FcBP-presenting ferritin to form antibody-based nanosystem
main approaches can be used to prepare the protein- for breast cancer treatment (Figure  16a–d).[237] In addition,
based nano­ particles, namely self-assembly and desolvation the trastuzumab or rabbit antibody could be mixed with the
methods.[232] Until now, there have been many kinds of proteins FcBP-representing ferritin cage without any chemical conjuga-
extracted from animals or plants to be served as the substrates tion to form the complex. This complex could specific bind to

Figure 16.  a) Surface and ribbon diagram representation of ferritin from the position of the fourfold axis of symmetry and b) the subunits with the
inserted FcBP and extra glycine residues shown as connected spheres. c) Schematic illustration of the construction of a noncovalent antibody/fluo-
rescent labeled protein cage complex. d) TEM images of FcBP-ferritin. e) Fluorescent microscopic images of SKBR3 breast cancer cells treated with
Cy3-trastuzumab/FcBP-ferritin complexes. Modified with permission.[237] Copyright 2012, Elsevier Ltd.

Adv. Mater. 2019, 31, 1902791 1902791  (23 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 17.  Schematic illustration of the genetic and enzymatic manipulation of prostate cancer cell-penetrating M13 bacteriophage to generate nano-
carriers for the intracellular delivery of functional horseradish peroxidase to a human prostate cancer cell line. Adapted with permission.[239] Copyright
2014, American Chemical Society.

the SKBR3 breast cancer cell line or attach to the KB cell line, whey proteins, casein, silk proteins, gliadin, elastin, soy pro-
which overexpresses target HER2 or folate receptor respectively teins, and lectins.[234] For instance, Chan and co-workers
(Figure 16e). discovered a novel platform for protein delivery based on
Cry3Aa protein crystals that from the bacterium Bacillus thur-
3.5.2. Virus-Like Nanocarriers ingiensis.[241] The reporter proteins mCherry and GFP could
be efficiently delivered into mammalian cells and animals by
Inspired by the development of commercial vaccines for human Cry3Aa fusion protein platform. Regarding the noncytotoxic
papilloma and hepatitis B viruses, a broad multidisciplinary and customizable properties, the silk proteins from Bombyx
approaches have been used to design the virus-like particles mori and recombinant spider silk proteins eADF4 are con-
(VLPs) for protein delivery.[238] Those VLPs can be morpholog- sidered as promising candidates for proteins delivery.[242]
ical diversity without the viral genome and derived from micro- Herein, lysozyme was selected as a model protein and loaded
bial, mammalian, plant and insect viruses. The protein drugs on eADF4 particles surface by electrostatic interactions. In
can be loaded on the surface or inside of VLPs and efficiently this system, lysozyme could be released as decreasing pH or
internalized by cells.[238] M13 filamentous bacteriophage, which increasing ionic strength environment to weaken the electro-
consists of 5 coat proteins (p3, p6, p7, p8, and p9) is a well- static interactions. Among these protein-based nanocarriers,
studied nanocarrier with a length of ≈930 nm and width of the natural fibrous protein silk fibroin shows several advan-
≈6 nm. It has been used to deliver biotinylated HRP or GFP tages of high loading capacity and controllable drug release
intracellularly through genetic and enzymatic manipulation kinetics. However, the supernegative charge of fibroin parti-
(Figure 17).[239] The specific VLP was generated from the pros- cles limited their usage in the intracellular delivery. In order
tate cancer CPP (Ypep 2) equipped on p3, which could selec- to achieve cellular uptake and maintain proteins activities in
tively penetrate into PC-3 cell lines. Studies showed that the the cytoplasm, fibroin particles have been fabricated by coating
functional protein HRP delivered into the host cells could pre- with cationic lipid layers, showing high stability and low tox-
serve its biological activity. Recently, Chatterjee and co-workers icity in vitro assay. Furthermore, tyrosinase, luciferase, GFP,
created Gag fusion based VLP using a truncated form of influ- and HRP could be effectively encapsulated into the particles
enza neuraminidase or hemagglutinin to fuse with murine and led to hyper-pigmentation and tumor regression in mice,
IFN-γ and human TNF-related apoptosis-inducing ligand.[240] suggesting the possibility of fibroin based particles for deliv-
With this VLP, various proteins, including Cre recombinase, ering proteins in clinical applications.[243] BSA is a nonim-
GFP, cytotoxic enzymes Fcy::Fur or human Caspase 8 could be munogenic, nontoxic, and biodegradable protein material that
delivered into the inside of cells. has been used as proteins carrier owning to their high binding
capacity of protein drugs. For example, Sun and co-workers
3.5.3. Protein-Based Complex synthesized chitosan-stabilized BSA NPs with NEL-like mole­
cule- 1 protein (NELL-1) loading.[244] This NPs system showed
Until now, numbers of proteins extracted from animals or high entrapment efficiency up to 89.30% and maintained sus-
plants have been developed as the substrates to prepare the tained release during 8 days. Recently, a poly(oligo (ethylene
protein-based nanoparticles, such as collagen, gelatin, albumin, glycol) methyl ether methacrylate)-albumin conjugate was

Adv. Mater. 2019, 31, 1902791 1902791  (24 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

generated to encapsulate the anticancer proteins Sprouty 1.[245] conditions. Final results showed the antitumor proteins that
In contrast to BSA/protein complex, the PEGylated BSA/protein escaped from endosome could exhibit enhanced apoptotic
complex micelle was more stable and had higher anticancer activity to MCF-7 cells. Due to the positive charge of some pro-
efficacy by inhibiting the growth of 3D MCF-7 multicellular teins, they could bind to anionic cell surface proteoglycans and
tumor spheroids. Elastin is another protein material that has exhibit cell-permeability. With those positively charged proteins
been well studied for protein delivery. Actually, elastin and or peptides, the genetically fused protein drugs could possess
elastin derivative-based NPs possess many unique properties, high levels of cytosolic delivery.[250,251] For instance, superposi-
such as biocompatibility, elasticity, dermal regeneration ability tively charged GFP was selected and fused to mCherry or Cre
and structure tunability in response to temperature change. recombinase. With this system, significantly higher levels of
Elastin-like peptides (ELPs) are produced recombinantly from internalized protein and greater efficiencies of Cre-induced
synthetic genes and can be soluble at the inverse transition recombination were achieved by comparing with transduc-
temperatures (Tt), but self-assemble as coacervates when the tion domains protein delivering system.[251] In fact, therapeutic
temperature above the Tt.[246] According to this property, ELPs peptides are one of important classes of pharmaceutical, how-
can be genetically fused to high keratinocyte growth factor ever, many peptides exhibit poor stability and oral bioavail-
(KGF) and self-assembled into NPs at physiological tempera- ability, short half-life in the circulatory system and the lack of
tures.[247] Subsequent results demonstrated that this KGF-ELPs tissue specificity. Those disadvantages would further limit their
NPs could enhance granulation and re-epithelialization, as clinical usage. Recent study showed that through embedding
well as improve would healing in diabetic mice. Recently, Cho the therapeutic peptides in the polypeptides backbone, the
and co-workers designed PEGylated human α-elastin nano- therapeutic peptides/proteins could be delivered to the target
particles which could load significant amounts of insulin and site without the above issues.[252] As shown in Figure  18a,b,
BSA, It is noted that sustained protein release for 72 h could three independent polypeptides consisting of functional pep-
be achieved in this nanosystem.[248] GY(EIAALEK)3GC (E3) tides (Tat-NR2B9c: a neuroprotective peptide fused with CPP;
and GY-(KIAALKE)3GC (K3) peptides which can be formed as melittin (Mel): a cytolytic peptide for treatment of stroke and
coiled-coil motifs have drawn remarkable attention for their cancer) self-assemble as activatable protein nanoparticles
pH-triggered unfolding behaviors at late endosomal environ- (APNPs). The APNPs contain enzyme-responsive sequences
ment. For example, Zhong and co-workers fabricated E3/K3 and tumor targeting peptide (RTIGPSV) that deliver the thera-
coiled-coil peptide-crosslinked hyaluronic acid nanogels.[249] peutic peptides to targeted tissues and stimulated release active
Various antitumor proteins including saporin, CytC could be peptides at proteases enriched microenvironment, enhancing
encapsulated into the nanogel and released under mild acidic the antitumor effect of Mel (Figure 18c,d).

Figure 18.  a) Schematic illustration of polypeptides containing functional modular motifs and the formation of self-assembled NPs through pairwise
coiled-coil dimerization. b) TEM images of Melittin-NPs (scale bar: 20 nm). c) Cytotoxicity of Mel-APNPs with and without MMP-2 treatment and
free melittin peptide on MDA-MB-231 cells. d) Antitumor effect of Mel-APNPs evaluated in mice bearing MDA-MB-231 tumors. Adapted with permis-
sion.[252] Copyright 2018, Wiley-VCH.

Adv. Mater. 2019, 31, 1902791 1902791  (25 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

Figure 19.  Schematic illustration of development of an exosome-based drug delivery system. The exosomes can be modified with the targeting pep-
tide by the transfection method, then the exosomes are secreted by a various of cells which containing other functional cellular components such as
siRNA, microRNA, small molecules and proteins, the protein drugs could be encapsulated into the exosomes by various methods, then the functional
exosomes can be targeted on the specific receptors of cells and deliver the protein drugs into the cells.

Protein-based delivery platforms show exciting ways to sig- Figure 19 Schematic illustration of development of an exosome-
nificantly improve protein delivery and therapeutic effects. based drug delivery system.
While the progress is still needed to develop the potential in The method of exosome-producing cells transfection is
the improvement and mechanistic understanding. One of generally very effective for loading of functional proteins into
the potential strategies is suggested to increase the efficacy of exosomes. In this way, the transfection of donor cells is gen-
protein-based nanoparticles through the combination of these erated to overexpressed specific proteins on the surface mem-
nanoparticles with other FDA-approved treatments. In addition, brane of the exosome. After that, the engineered exosomes are
future work will also indicate the feasibility of using protein- isolated from the donor cells and used for disease treatments.
based nanoparticles and their derivative in combination therapy Saydam and co-workers first developed genetic cells which
in the clinical field of tumor treatment in vivo. overexpressed the suicide therapeutic mRNA/protein for cyto-
sine deaminase (CDE) fused in-frame with uracil phosphoribo-
syltransferase (UPRT).[254] The CDE-UPRT can convert prodrug
3.6. Exosomes 5-fluorocytosine to 5-fluoro-deoxyuridine monophosphate
(5-FdUMP), which could cause cells to go under apoptosis.
Exosomes are lipoproteic nanosized vesicles which are secreted Based on the conversion of CDE-UPRT to 5-fluorocytosine, the
from a variety of cells, particularly from dendritic cells, T cells, isolated exosomes carried high amounts of CDE-UPRT mes-
B cells and macrophages. They can interact with cellular mem- sage and protein were injected into schwannomas and sub-
branes as abundance of adhesive proteins attach on their nat- sequently resulted in regression of the tumor in combination
ural lipid bilayers. Exosomes play functional roles in removing with prodrug of 5-fluorocytosine. Own to its wider tropism
obsolete proteins. In addition, they also facilitate the transfer and high effective membrane fusion without adverse effect,
of proteins, siRNA, mRNAs, microRNAs and other functional vascular stomatitis virus protein (VSVG) is routine for viral
cellular components in the cell or cell-to-cell communications. pseudotyping to enhance the target range and transduction
Moreover, exosomes deliver their payload (small molecular efficiency. By expressing gene-encoding VSVG fusion proteins
drugs, proteins, nucleic acid) to target cells as they could con- in the mother cells, the exosomes that encapsulated VSVG
tain various surface proteins and specific vector ligands such were isolated, which could be developed as the protein delivery
as heat shock proteins, tetraspanins, lysosomal proteins and nanosystem for enhancing the uptaken efficiency by recipient
fusion proteins. Until now, various approaches have been used cells.[255,256] Another two studies have shown that VSVG based
to develop exosomes based nanocarriers, such as chemical- fusogenic exosomes could efficiently load proteins and mediate
based transfection, electroporation, cell activation, transfec- the delivery of therapeutic membrane proteins. One example is
tion of exosome-producing cells, and incubation method.[253] the generation of fusion proteins, VSVG that integrated GFP,

Adv. Mater. 2019, 31, 1902791 1902791  (26 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

YFP or luciferase in the exosomal membrane, which could applications, especially for materials which are not biode-
preserve their function in the targeted cells.[255] The other one gradable.
is, Kim and co-workers fabricated fusogenic exosomes by har- 2) Many obstacles still need to be overcome during the prepa-
boring VSVG and GFP-fused tetraspanin CD63 (CD63-GFP as ration of proteins loaded nanocarriers, either covalent modi-
a reporter).[256] The membrane of fusogenic exosomes could fications to protein surface or noncovalent strategies may
fuse with recipient cell membranes at acidic pH. In this system, cause the protein structure changes and subsequent loss of
the model protein GLUT4 was incorporated into the exosomal function, the development nanocarriers in organic solvents
surface and finally retained their functionality after intracellular or interaction between proteins and materials could also alter
delivery. the protein structures.
Catalase is one of the most efficient antioxidants that is 3) After entering the body, nanocarriers elimination via mecha-
often used to treat PD. In order to effectively encapsulate the nism or excretion processes should be considered serious-
catalase into exosomes, various approaches including soni- ly as many of them are resistant to elimination routes, for
cation, extrusion, freeze–thaw cycles, and permeabilization instance, due to the large size and higher chemical stability,
with saponin have been developed. Studies showed that the injected semiconductor quantum dots in mice remained
catalase encapsulated into exosomes could preserve enzymatic intact for more than 2 years in mouse tissues.[260]
activity. Particularly, the methods of sonication and extrusion 4) There are still many nanocarriers entrapped into the en-
showed the highest catalytic activity.[257] Recently, Kabanov dosomes and difficult to escape into cytosol. Highly efficient
and co-workers uncovered that naïve exosomes released from for the intracellular transduction is still a challenge for nano-
macrophage (M-exosomes) could penetrate the blood-brain bar- carriers.
rier (BBB).[258] M-exosomes contained the integrin lymphocyte 5) Even though many proteins were successfully delivered into
function-associated antigen 1 (LFA-1) and intercellular adhe- cells and animal model, showing promising biological activ-
sion molecule 1 (ICAM-1), both of which play important role ity. However, most proteins are selected as model proteins
in the uptake of M-exosomes in brain endothelial cells. Studies which may contain strong structure and keep stability in the
showed that the inflammation processes could promote the biological environment, whether all protein drugs showed
exosomes uptake in the BBB cells because of the overexpres- biological activities once delivered into targets need to further
sion of ICAM-1 receptors. The model protein brain derived confirm.
neurotrophic factor (BDNF) could bind with exosomes by the 6) Small mouse model is frequently used to test the biological
electrostatic and polysaccharide interactions. High amount of activities of proteins loaded NPs, whether the positive results
BDNF was successfully delivered to brain under inflammation, could be reproduced in clinical trials that need to be further
indicating M-exosomes are promising nanocarriers for brain verified. Thus, it is necessary to build up novel models that
delivery of therapeutic proteins. represent the better human conditions.
Despite the numerous advantages of using exosomes as drug 7) Last but not least, every single person is truly unique with
carriers, several challenges are still remained. For instance, the clinical, genomic and environmental information, and this
composition of exosomes and their functions are considered uniqueness should be used to guide the disease manage-
to be the major challenge.[259] It is known that exosomes are ment, personalized nanomedicine contain multifunctional
involved in cellular communication from the host to the recip- abilities for early detection, combination of diagnostics with
ient cell. However, the transported biomolecules, their roles, therapeutics, and delivering proteins to the specific target,
and the associated heterogeneity of the exosomes are unclear however, due to the lack of sufficient patient’s information
exactly. Exosomes also suffer from the heterogeneity of the and the communication among cells are changing all the
biomolecules inside the aqueous lumen and on the surface. time, functionalized nanocarriers conjugate specific ligands
In addition, the scale-up process to produce a large amount of should be changed judiciously, which is still at the early stage
the vesicles is difficult to achieve. Further research is preferred of development and meets various challenges.
to require into the composition of exosomes and their delivery
efficacy in vivo.
To date, several strategies to develop protein-loaded nano-
carriers have been utilized, those nanocarriers are built with
4. Summary and Outlook various materials for enhancing drug encapsulation and
improving the protein delivery to the cytosol by endosomal
Various nanocarriers have been developed for intracellular pro- escape. Moreover, the symbiosis of different materials opens
teins delivery and shown great promise for enabling the cur- up new opportunities for building novel multifunctional nano-
rent and next generations of therapeutics. However, there still carriers, polymers within protein cages offer the possibility of
remain many challenges that include the following points: increasing the loading density of drug molecules and proteins.
Combination therapy provide the potential for enhanced effi-
1) The choice of materials is used to be platform must be con- cacy through synergies between therapeutics. Using nanocar-
sidered carefully. That is because those materials have been riers for codelivery small molecules and proteins is regarding
screened in vitro and approved biocompatibility and nontox- as a promising therapeutic strategy which has been applied in
icity to the cells, may not accurately predict the in vivo toxic- cancer therapy.[261] Moreover, to improve protein intracellular
ity. Hence, it is necessary to investigate the in vivo results delivery and active targeting, especially for the tumor cells, the
exposure to nanomaterials before any potential therapeutic surface of nanocarriers could be incorporated with cell receptor

Adv. Mater. 2019, 31, 1902791 1902791  (27 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

targeting ligands (e.g., folic acid and hyaluronic acid), cell pen- Conflict of Interest
etrating peptide, or antibody (e.g., Herceptin and transferrin)
The authors declare no conflict of interest.
to specifically bind with a receptor of the cancer cell mem-
brane. Clearly, many NP-based drug delivery systems have
been focused on targeted drug delivery, especially the surface
modification of targeted ligands for tumor-specific targeting Keywords
and enhancing the anticancer treatment. In recent years, orga- inorganic nanoparticles, lipids, polymeric nanoparticles, protein delivery
nelle and subcellular drug targeting has gained increasingly
acceptance. For example, targeting the nuclear is a direct way to Received: May 2, 2019
interfere DNA transcription and damage the genetic informa- Revised: July 4, 2019
tion. Thus, the subcellular targeting strategy could achieve max- Published online: September 8, 2019
imum therapeutic responsive as well as avoiding cytotoxic side
effects of the drugs. However, the protein-loaded NPs for the
subcellular targeted and disease treatment are rarely reported.
[1] B. Leader, Q. J. Baca, D. E. Golan, Nat. Rev. Drug Discovery 2008,
The development of protein delivered NPs for targeting spe-
7, 21.
cific subcellular could be regarded as the new generation drug [2] C. Ng, M. S. H. Guan, C. Koh, X. Ouyang, F. Yu, E. Tan,
delivery technology for the precise disease treatment. S. P. O’Neill, X. Zhang, J. Chung, K. Lim, J. Neurosci. 2012, 32,
The first nanotherapeutic Doxil (PEGylated liposomal Dox) 14311.
has been approved by FDA in 1995 and used to treat ovarian [3] L. Hang, J. Thundyil, K. Lim, Ann. N. Y. Acad. Sci. 2015, 1350, 37.
cancer and multiple myeloma.[262] Abraxane, a paclitaxel loaded [4] F. Du, Q. Yu, S. Yan, G. Hu, L. Lue, D. G. Walker, L. Wu, S. F. Yan,
albumin-bound nanoparticles, was available in the market in K. Tieu, S. S. Yan, Brain 2017, 140, 3233.
2005 and achieved great commercial success for the treatment [5] M. Yu, J. Wu, J. Shi, O. C. Farokhzad, J. Controlled Release 2016,
of recurrent breast cancer.[263] Nowadays, at least 15 liposome 240, 24.
[6] M. M. Pakulska, S. Miersch, M. S. Shoichet, Science 2016, 351,
drugs or nanomedical based therapies have been approved by
aac4750.
FDA for commercial use. Global markets are projected to reach
[7] R. T. Dean, Nature 1975, 257, 414.
177.6 billion $US by 2019.[264,265] Those successful applications [8] W. Wang, Int. J. Pharm. 1999, 185, 129.
confirmed the high potential in the development of protein- [9] M. C. Manning, K. Patel, R. T. Borchardt, Pharm. Res. 1989, 06,
loaded nanomedicines. However, the clinical development of 903.
protein loaded nanocarriers is still in their early stages. Most [10] M. C. Manning, D. K. Chou, B. M. Murphy, R. W. Payne,
protein drugs contain fragile tertiary structure that are sensitive D. S. Katayama, Pharm. Res. 2010, 27, 544.
to the unfavored physicochemical conditions. The development [11] A. S. De Groot, W. Martin, Clin. Immunol. 2009, 131, 189.
of multifunctional NPs to achieve the long-term stability of pro- [12] S. Frokjaer, D. E. Otzen, Nat. Rev. Drug Discovery 2005, 4, 298.
tein drugs for commercialization is a great challenge. Among [13] A. Szlachcic, M. Zakrzewska, J. Otlewski, Biotechnol. Adv. 2011, 29,
436.
the numerous materials that applied for drugs delivery systems,
[14] T. Vermonden, R. Censi, W. E. Hennink, Chem. Rev. 2012, 112,
various types of VLPs could be manufacture at a large scale
2853.
by a large assortment of expression systems (bacterial, insect, [15] C. D. Spicer, C. Jumeaux, B. Gupta, M. M. Stevens, Chem. Soc.
yeast, mammalian, and plant), and therapeutic protein could be Rev. 2018, 47, 3574.
genetic produced with VLPs as a protein loaded nanocarriers [16] Z. Gu, A. Biswas, M. Zhao, Y. Tang, Chem. Soc. Rev. 2011, 40,
by expression systems. Moreover, some VLPs are exceptionally 3638.
stable biological materials that can withstand high temperature, [17] M. Rother, M. G. Nussbaumer, K. Renggli, N. Bruns, Chem. Soc.
thus VLPs can be regarded as promising vehicles for potential Rev. 2016, 45, 6213.
commercialization. [18] F. Scaletti, J. Hardie, Y.-W. Lee, D. C. Luther, M. Ray, V. M. Rotello,
In conclusion, the potential for protein delivery with nano- Chem. Soc. Rev. 2018, 47, 3421.
[19] J. de A. Pachioni-Vasconcelos, A. M. Lopes, A. C. Apolinário,
carriers in this field is tremendous, and more and more novel
J. K. Valenzuela-Oses, J. S. R. Costa, L. de O. Nascimento,
material or delivery vehicles emerge and can be uncovered by
A. P. Jr., L. R. S. Barbosa, C. de O. Rangel-Yagui, Biomater. Sci. 2016,
further scientific research. 4, 205.
[20] Y. Lu, W. Sun, Z. Gu, J. Controlled Release 2014, 194, 1.
[21] Y. Yun, Y. W. Cho, K. Park, Adv. Drug Delivery Rev. 2013, 65, 822.
[22] K. M. El-Say, H. S. El-Sawy, Int. J. Pharm. 2017, 528, 675.
Acknowledgements [23] S. Gupta, A. Jain, M. Chakraborty, J. K. Sahni, J. Ali, S. Dang, Drug
X.Q. and C.Y. contributed equally to this work. This study was Delivery. 2013, 20, 237.
financially supported by the National Natural Science Foundation [24] S. K. Samal, M. Dash, S. Van Vlierberghe, D. L. Kaplan, E. Chiellini,
of China (81672508 and 61601218), Jiangsu Provincial Foundation C. van Blitterswijk, L. Moroni, P. Dubruel, Chem. Soc. Rev. 2012,
for Distinguished Young Scholars (BK20170041), Natural Science 41, 7147.
Foundation of Shaanxi Province (2019JM-016), GSK-EDB Trust Fund [25] S. S. Silva, J. F. Mano, R. L. Reis, Green Chem. 2017, 19, 1208.
(R-143-000-688-592), the Synthetic Biology Research & Development [26] H. Liu, C. Wang, C. Li, Y. Qin, Z. Wang, F. Yang, Z. Li, J. Wang,
Programme (SBP) of National Research Foundation (SBP-P4 and SBP-P8) RSC Adv. 2018, 8, 7533.
of Singapore, China-Sweden Joint Mobility Project (51811530018), [27] G. Rassu, E. Soddu, M. Cossu, E. Gavini, P. Giunchedi, A. Dalpiaz,
and Fundamental Research Funds for the Central Universities and the J. Drug Delivery Sci. Technol. 2016, 32, 77.
Natural Science Foundation of Guangdong Province (2017A030313299). [28] L. Hu, Y. Sun, Y. Wu, Nanoscale 2013, 5, 3103.

Adv. Mater. 2019, 31, 1902791 1902791  (28 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[29] P. Mukhopadhyay, R. Mishra, D. Rana, P. P. Kundu, Prog. Polym. [64] S. Cohen, G. Coué, D. Beno, R. Korenstein, J. F. J. Engbersen,
Sci. 2012, 37, 1457. Biomaterials 2012, 33, 614.
[30] B. prasanth Koppolu, S. G. Smith, S. Ravindranathan, S. Jayanthi, [65] G. Coué, C. Freese, R. E. Unger, C. James Kirkpatrick,
T. K. Suresh Kumar, D. A. Zaharoff, Biomaterials 2014, 35, 4382. J. F. J. Engbersen, Acta Biomater. 2013, 9, 6062.
[31] L. Li, L. Yang, M. Li, L. Zhang, Carbohydr. Polym. 2017, 174, 182. [66] N. Chuard, G. Gasparini, A. Roux, N. Sakai, S. Matile, Org. Biomol.
[32] Y. Luo, Q. Wang, Int. J. Biol. Macromol. 2014, 64, 353. Chem. 2015, 13, 64.
[33] V. Bourganis, T. Karamanidou, O. Kammona, C. Kiparissides, [67] G. Gasparini, E.-K. Bang, J. Montenegro, S. Matile, Chem.
Eur. J. Pharm. Biopharm. 2017, 111, 44. Commun. 2015, 51, 10389.
[34] Y. Hu, T. Yang, X. Hu, Polym. Bull. 2012, 68, 1183. [68] E.-K. Bang, G. Gasparini, G. Molinard, A. Roux, N. Sakai, S. Matile,
[35] M. B. Stie, H. S. Thoke, O. G. Issinger, J. Hochscherf, B. Guerra, J. Am. Chem. Soc. 2013, 135, 2088.
L. F. Olsen, Colloids Surf., B 2019, 174, 216. [69] G. Gasparini, G. Sargsyan, E. K. Bang, N. Sakai, S. Matile, Angew.
[36] P. D. Hsu, E. S. Lander, F. Zhang, Cell 2014, 157, 1262. Chem., Int. Ed. 2015, 54, 7328.
[37] J. Qiao, W. Sun, S. Lin, R. Jin, L. Ma, Y. Liu, Chem. Commun. 2019, [70] G. Gasparini, E. K. Bang, G. Molinard, D. V. Tulumello, S. Ward,
55, 4707. S. O. Kelley, A. Roux, N. Sakai, S. Matile, J. Am. Chem. Soc. 2014,
[38] M. Amidi, E. Mastrobattista, W. Jiskoot, W. E. Hennink, Adv. Drug 136, 6069.
Delivery Rev. 2010, 62, 59. [71] J. Fu, C. Yu, L. Li, S. Q. Yao, J. Am. Chem. Soc. 2015, 137, 12153.
[39] M.-C. Chen, F.-L. Mi, Z.-X. Liao, C.-W. Hsiao, K. Sonaje, [72] L. Qian, J. Fu, P. Yuan, S. Du, W. Huang, L. Li, S. Q. Yao, Angew.
M.-F. Chung, L.-W. Hsu, H.-W. Sung, Adv. Drug Delivery Rev. 2013, Chem. 2018, 130, 1548.
65, 865. [73] V. Agrahari, V. Agrahari, Drug Discovery Today 2018, 23, 1139.
[40] C. Y. Wong, H. Al-Salami, C. R. Dass, J. Controlled Release 2017, [74] J. Nicolas, S. Mura, D. Brambilla, N. Mackiewicz, P. Couvreur,
264, 247. Chem. Soc. Rev. 2013, 42, 1147.
[41] Y. Yang, S. Wang, Y. Wang, X. Wang, Q. Wang, M. Chen, Biotechnol. [75] Y. Lee, T. Ishii, H. Cabral, H. J. Kim, J. H. Seo, N. Nishiyama,
Adv. 2014, 32, 1301. H. Oshima, K. Osada, K. Kataoka, Angew. Chem. 2009, 121, 5413.
[42] L. Li, G. Jiang, W. Yu, D. Liu, H. Chen, Y. Liu, Z. Tong, X. Kong, [76] Y. Lee, T. Ishii, H. J. Kim, N. Nishiyama, Y. Hayakawa, K. Itaka,
J. Yao, Mater. Sci. Eng., C 2017, 70, 278. K. Kataoka, Angew. Chem. 2010, 122, 2606.
[43] A. M. M. Sadeghi, F. A. Dorkoosh, M. R. Avadi, M. Weinhold, [77] G. H. Gao, M. J. Park, Y. Li, G. H. Im, J. H. Kim, H. N. Kim,
A. Bayat, F. Delie, R. Gurny, B. Larijani, M. Rafiee-Tehrani, J. W. Lee, P. Jeon, O. Y. Bang, J. H. Lee, D. S. Lee, Biomaterials
H. E. Junginger, Eur. J. Pharm. Biopharm. 2008, 70, 270. 2012, 33, 9157.
[44] M. Liu, J. Zhang, X. Zhu, W. Shan, L. Li, J. Zhong, Z. Zhang, [78] Q. Jin, T. Cai, Y. Wang, H. Wang, J. Ji, ACS Macro Lett. 2014, 3, 679.
Y. Huang, J. Controlled Release 2016, 222, 67. [79] J. Ren, Y. Zhang, J. Zhang, H. Gao, G. Liu, R. Ma, Y. An, D. Kong,
[45] W. Fan, D. Xia, Q. Zhu, X. Li, S. He, C. Zhu, S. Guo, L. Hovgaard, L. Shi, Biomacromolecules 2013, 14, 3434.
M. Yang, Y. Gan, Biomaterials 2018, 151, 13. [80] S. Li, Y. Zhang, J. Wang, Y. Zhao, T. Ji, X. Zhao, Y. Ding, X. Zhao,
[46] D.-Y. Wu, Y. Ma, X.-S. Hou, W. Zhang, P. Wang, H. Chen, B. Li, R. Zhao, F. Li, X. Yang, S. Liu, Z. Liu, J. Lai, A. K. Whittaker,
C. Zhang, Y. Ding, Carbohydr. Polym. 2017, 157, 1470. G. J. Anderson, J. Wei, G. Nie, Nat. Biomed. Eng. 2017, 1, 667.
[47] M. Allahyari, E. Mohit, Hum. Vaccines Immunother. 2016, 12, 806. [81] S. F. M. Van Dongen, W. P. R. Verdurmen, R. J. R. W. Peters,
[48] F. Cui, K. Shi, L. Zhang, A. Tao, Y. Kawashima, J. Controlled Release R. J. M. Nolte, R. Brock, J. C. M. Van Hest, Angew. Chem., Int. Ed.
2006, 114, 242. 2010, 49, 7213.
[49] J. U. Menon, P. Ravikumar, A. Pise, D. Gyawali, C. C. W. Hsia, [82] R. Cheng, F. Meng, S. Ma, H. Xu, H. Liu, X. Jing, Z. Zhong,
K. T. Nguyen, Acta Biomater. 2014, 10, 2643. J. Mater. Chem. 2011, 21, 19013.
[50] X. Zhang, M. Sun, A. Zheng, D. Cao, Y. Bi, J. Sun, Eur. J. Pharm. [83] J. Zhang, L. Wu, F. Meng, Z. Wang, C. Deng, H. Liu, Z. Zhong,
Sci. 2012, 45, 632. Langmuir 2012, 28, 2056.
[51] S. Dyawanapelly, U. Koli, V. Dharamdasani, R. Jain, P. Dandekar, [84] X. Li, W. Yang, Y. Zou, F. Meng, C. Deng, Z. Zhong, J. Controlled
Drug Delivery Transl. Res. 2016, 6, 365. Release 2015, 220, 704.
[52] J. Sheng, L. Han, J. Qin, G. Ru, R. Li, L. Wu, D. Cui, P. Yang, Y. He, [85] L. Lu, Y. Zou, W. Yang, F. Meng, C. Deng, R. Cheng, Z. Zhong,
J. Wang, ACS Appl. Mater. Interfaces 2015, 7, 15430. Biomacromolecules 2015, 16, 1726.
[53] Z. Amoozgar, M. S. Goldberg, Nanomedicine 2017, 12, 729. [86] P. Yao, Y. Zhang, H. Meng, H. Sun, Z. Zhong, Biomacromolecules
[54] B. Xu, G. Jiang, W. Yu, D. Liu, Y. Liu, X. Kong, J. Yao, Mater. Sci. 2019, 20, 184.
Eng., C 2017, 78, 420. [87] R. E. Whitmire, D. Scott Wilson, A. Singh, M. E. Levenston,
[55] P. Zhou, T. An, C. Zhao, Y. Li, R. Li, R. Yang, Y. Wang, X. Gao, Int. J. N. Murthy, A. J. García, Biomaterials 2012, 33, 7665.
Pharm. 2015, 478, 633. [88] C. Silva, A. Carretero, D. Soares da Costa, R. L. Reis,
[56] A. Jain, S. K. Jain, Acta Diabetol. 2015, 52, 663. R. Novoa-Carballal, I. Pashkuleva, Acta Biomater. 2017, 63, 283.
[57] R. Ganugula, M. Arora, M. Guada, P. Saini, M. N. V. R. Kumar, [89] R. Rupaimoole, F. J. Slack, Nat. Rev. Drug Discovery 2017, 16, 203.
ACS Macro Lett. 2017, 6, 161. [90] Y. Chen, D.-Y. Gao, L. Huang, Adv. Drug Delivery Rev. 2015, 81, 128.
[58] R. Sharma, U. Gupta, N. K. Garg, R. K. Tyagi, N. K. Jain, Colloids [91] Z. Niu, I. Conejos-Sánchez, B. T. Griffin, C. M. O’Driscoll,
Surf., B 2015, 127, 172. M. J. Alonso, Adv. Drug Delivery Rev. 2016, 106, 337.
[59] S. Jain, V. V Rathi, A. K. Jain, M. Das, C. Godugu, Nanomedicine [92] A. D. Bangham, R. W. Horne, J. Mol. Biol. 1964, 8, 660.
2012, 7, 1311. [93] U. Bulbake, S. Doppalapudi, N. Kommineni, W. Khan, Pharmaceu-
[60] P. Hurkat, A. Jain, A. Jain, S. Shilpi, A. Gulbake, S. K. Jain, J. Nano- tics 2017, 9, 12.
part. Res. 2012, 14, 1219. [94] T. Jiang, R. Mo, A. Bellotti, J. Zhou, Z. Gu, Adv. Funct. Mater. 2014,
[61] X. Zhu, J. Wu, W. Shan, W. Tao, L. Zhao, J. M. Lim, M. D’Ortenzio, 24, 2295.
R. Karnik, Y. Huang, J. Shi, O. C. Farokhzad, Angew. Chem., Int. Ed. [95] L. Liguori, B. Marques, A. Villegas-Mendez, R. Rothe,
2016, 55, 3309. J.-L. Lenormand, J. Controlled Release 2008, 126, 217.
[62] X. Liu, C. Liu, W. Zhang, C. Xie, G. Wei, W. Lu, Int. J. Pharm. 2013, [96] S. Kube, N. Hersch, E. Naumovska, T. Gensch, J. Hendriks,
448, 159. A. Franzen, L. Landvogt, J. P. Siebrasse, U. Kubitscheck,
[63] G. Coué, J. F. J. Engbersen, J. Controlled Release 2011, 152, 90. B. Hoffmann, R. Merkel, A. Csiszár, Langmuir 2017, 33, 1051.

Adv. Mater. 2019, 31, 1902791 1902791  (29 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[97] M. Vila-Caballer, G. Codolo, F. Munari, A. Malfanti, M. Fassan, [128] Y. Yang, Y. Niu, J. Zhang, A. K. Meka, H. Zhang, C. Xu, C. X. C. Lin,
M. Rugge, A. Balasso, M. de Bernard, S. Salmaso, J. Controlled M. Yu, C. Yu, Small 2015, 11, 2743.
Release 2016, 238, 31. [129] Y. N. Yang, S. Karmakar, J. Zhang, M. H. Yu, N. Mitter, C. Z. Yu,
[98] S. Ahmed, S. Fujita, K. Matsumura, Nanoscale 2016, 8, 15888. J. Mater. Chem. B 2014, 2, 4929.
[99] J. Kowapradit, A. Apirakaramwong, T. Ngawhirunpat, [130] H. S. Park, C. W. Kim, H. J. Lee, J. H. Choi, S. G. Lee, Y. P. Yun,
T. Rojanarata, W. Sajomsang, P. Opanasopit, Eur. J. Pharm. Sci. I. C. Kwon, S. J. Lee, S. Y. Jeong, S. C. Lee, Nanotechnology 2010,
2012, 47, 359. 21, 225101.
[100] X. Zhang, P. F. Luckham, A. D. Hughes, S. Thom, X. Y. Xu, Int. J. [131] L. Sun, X. Zhang, Z. Wu, C. Zheng, C. Li, Polym. Chem. 2014, 5,
Pharm. 2011, 421, 291. 1999.
[101] J. A. Zuris, D. B. Thompson, Y. Shu, J. P. Guilinger, J. L. Bessen, [132] H. Omar, J. G. Croissant, K. Alamoudi, S. Alsaiari, I. Alradwan,
J. H. Hu, M. L. Maeder, J. K. Joung, Z.-Y. Chen, D. R. Liu, Nat. Bio- M. A. Majrashi, D. H. Anjum, P. Martins, R. Laamarti, J. Eppinger,
technol. 2015, 33, 73. B. Moosa, A. Almalik, N. M. Khashab, J. Controlled Release 2017,
[102] M. Wang, J. A. Zuris, F. Meng, H. Rees, S. Sun, P. Deng, Y. Han, 259, 187.
X. Gao, D. Pouli, Q. Wu, I. Georgakoudi, D. R. Liu, Q. Xu, Proc. [133] S. S. Bale, S. J. Kwon, D. a Shah, A. Banerjee, J. S. Dordick,
Natl. Acad. Sci. USA 2016, 113, 2868. R. S. Kane, ACS Nano 2010, 4, 1493.
[103] C. Botto, N. Mauro, E. Amore, E. Martorana, G. Giammona, [134] D. A. Shah, S. J. Kwon, S. S. Bale, A. Banerjee, J. S. Dordick,
M. L. Bondì, Int. J. Pharm. 2017, 516, 334. R. S. Kane, Biomaterials 2011, 32, 3210.
[104] S. A. Wissing, O. Kayser, R. H. Müller, Adv. Drug Delivery Rev. 2004, [135] Y. Li, X. Chen, H. Liu, X. Mou, Z. Ren, Z. Ahmad, X. Li, G. Han,
56, 1257. Chem. Eng. J. 2017, 330, 166.
[105] Z. Bai, J. Wei, C. Yu, X. Han, X. Qin, C. Zhang, W. Liao, L. Li, [136] Y. Niu, M. Yu, S. B. Hartono, J. Yang, H. Xu, H. Zhang, J. Zhang,
W. Huang, J. Mater. Chem. B 2019, 7, 1209. J. Zou, A. Dexter, W. Gu, C. Yu, Adv. Mater. 2013, 25, 6233.
[106] M. García-Fuentes, D. Torres, M. J. Alonso, Colloids Surf., B 2003, [137] Y. Niu, M. Yu, A. Meka, Y. Liu, J. Zhang, Y. Yang, C. Yu, J. Mater.
27, 159. Chem. B 2016, 4, 212.
[107] H. Yuan, S.-P. Jiang, Y.-Z. Du, J. Miao, X.-G. Zhang, F.-Q. Hu, [138] Y. Zhao, B. G. Trewyn, I. I. Slowing, V. S. Lin, J. Am. Chem. Soc.
Colloids Surf., B 2009, 70, 248. 2009, 131, 8398.
[108] R. Yang, R.-C. Gao, C.-F. Cai, H. Xu, F. Li, H.-B. He, X. Tang, Chem. [139] X. Wu, S. Wu, L. Yang, J. Han, S. Han, J. Mater. Chem. 2012, 22,
Pharm. Bull. 2010, 58, 1195. 17121.
[109] Z.-H. Zhang, Y.-L. Zhang, J.-P. Zhou, H.-X. Lv, Int. J. Nanomed. [140] X.-D. Wang, K. S. Rabe, I. Ahmed, C. M. Niemeyer, Adv. Mater.
2012, 7, 3333. 2015, 27, 7945.
[110] T. Fan, C. Chen, H. Guo, J. Xu, J. Zhang, X. Zhu, Y. Yang, [141] P. Aggarwal, J. B. Hall, C. B. Mcleland, M. A. Dobrovolskaia,
Z. Zhou, L. Li, Y. Huang, Eur. J. Pharm. Biopharm. 2014, 88, S. E. Mcneil, Adv. Drug Delivery Rev. 2009, 61, 428.
518. [142] X. Du, S. Zhang Qiao, Small 2015, 11, 392.
[111] S. K. Kim, M. B. Foote, L. Huang, Biomaterials 2012, 33, 3959. [143] J. Zhang, S. Karmakar, M. Yu, N. Mitter, J. Zou, C. Yu, Small 2014,
[112] W. Shan, X. Zhu, W. Tao, Y. Cui, M. Liu, L. Wu, L. Li, Y. Zheng, 10, 5068.
Y. Huang, ACS Appl. Mater. Interfaces 2016, 8, 25444. [144] E. A. Prasetyanto, A. Bertucci, D. Septiadi, R. Corradini,
[113] Z. Li, J. C. Barnes, A. Bosoy, J. F. Stoddart, J. I. Zink, Chem. Soc. P. Castro-Hartmann, L. De Cola, Angew. Chem., Int. Ed. 2016, 55,
Rev. 2012, 41, 2590. 3323.
[114] M. Yu, Z. Gu, T. Ottewell, C. Yu, J. Mater. Chem. B 2017, 5, 3241. [145] F. P. Chang, Y. Hung, J. H. Chang, C. H. Lin, C. Y. Mou, ACS Appl.
[115] X. Sun, Y. Zhao, V. S. Y. Lin, I. I. Slowing, B. G. Trewyn, J. Am. Mater. Interfaces 2014, 6, 6883.
Chem. Soc. 2011, 133, 18554. [146] T.-W. Sun, Y.-J. Zhu, C. Qi, F. Chen, Y.-Y. Jiang, Y.-G. Zhang, J. Wu,
[116] J. Gu, K. Huang, X. Zhu, Y. Li, J. Wei, W. Zhao, C. Liu, J. Shi, J. Col- C. Wu, J. Mater. Chem. B 2016, 4, 3257.
loid Interface Sci. 2013, 407, 236. [147] S. Cao, L. Fang, Z. Zhao, Y. Ge, S. Piletsky, A. P. F. Turner, Adv.
[117] H. Zhang, H. Xu, M. Wu, Y. Zhong, D. Wang, Z. Jiao, J. Mater. Funct. Mater. 2013, 23, 2162.
Chem. B 2015, 3, 6480. [148] P. Yuan, H. Zhang, L. Qian, X. Mao, S. Du, C. Yu, B. Peng,
[118] F. Gao, L. Li, C. Fu, L. Nie, D. Chen, F. Tang, Adv. Mater. 2013, 25, S. Q. Yao, Angew. Chem., Int. Ed. 2017, 56, 12481.
5508. [149] P. Yuan, X. Mao, X. Wu, S. S. Liew, L. Li, S. Q. Yao, Angew. Chem.,
[119] Z. Tian, Y. Xu, Y. Zhu, Mater. Sci. Eng., C 2017, 71, 452. Int. Ed. 2019, 58, 7657.
[120] Y. Yang, J. Wan, Y. Niu, Z. Gu, J. Zhang, M. Yu, C. Yu, Chem. Mater. [150] D. A. Giljohann, D. S. Seferos, P. C. Patel, J. E. Millstone,
2016, 28, 9008. N. L. Rosi, C. A. Mirkin, Nano Lett. 2007, 7, 3818.
[121] C. Xu, M. Yu, O. Noonan, J. Zhang, H. Song, H. Zhang, C. Lei, [151] P. Ghosh, X. Yang, R. Arvizo, Z.-J. Zhu, S. S. Agasti, Z. Mo,
Y. Niu, X. Huang, Y. Yang, C. Yu, Small 2015, 11, 5949. V. M. Rotello, J. Am. Chem. Soc. 2010, 132, 2642.
[122] A. K. Meka, P. L. Abbaraju, H. Song, C. Xu, J. Zhang, Small 2016, [152] C. H. J. Choi, C. A. Alabi, P. Webster, M. E. Davis, Proc. Natl. Acad.
12, 5169. Sci. USA 2010, 107, 1235.
[123] Y. Yang, S. Bernardi, H. Song, J. Zhang, M. Yu, J. C. Reid, [153] P. Wang, L. Zhang, Y. Xie, N. Wang, R. Tang, W. Zheng, X. Jiang,
E. Strounina, D. J. Searles, C. Yu, Chem. Mater. 2016, 28, 704. Adv. Sci. 2017, 4, 1700175.
[124] Y. Niu, M. Yu, J. Zhang, Y. Yang, C. Xu, M. Yeh, E. Taran, [154] H. M. Joshi, D. R. Bhumkar, K. Joshi, V. Pokharkar, M. Sastry, Lang-
J. J. C. Hou, P. P. Gray, C. Yu, J. Mater. Chem. B 2015, 3, 8477. muir 2006, 22, 300.
[125] J. S. Beck, J. C. Vartuli, W. J. Roth, M. E. Leonowicz, C. T. Kresge, [155] D. R. Bhumkar, H. M. Joshi, M. Sastry, V. B. Pokharkar, Pharm. Res.
K. D. Schmitt, C. T. W. Chu, D. H. Olson, E. W. Sheppard, 2007, 24, 1415.
S. B. McCullen, J. B. Higgins, J. L. Schlenker, J. Am. Chem. Soc. [156] S. Rana, A. Bajaj, R. Mout, V. M. Rotello, Adv. Drug Delivery Rev.
1992, 114, 10834. 2012, 64, 200.
[126] F. Tang, L. Li, D. Chen, Adv. Mater. 2012, 24, 1504. [157] S. M. Ryou, J. H. Yeom, H. J. Kang, M. Won, J. S. Kim, B. Lee,
[127] I. I. Slowing, B. G. Trewyn, V. S. Y. Lin, J. Am. Chem. Soc. 2007, 129, M. J. Seong, N. C. Ha, J. Bae, K. Lee, J. Controlled Release 2014,
8845. 196, 287.

Adv. Mater. 2019, 31, 1902791 1902791  (30 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[158] J.-H. Yeom, M. Joo, B. Lee, K. P. Kim, N.-C. Ha, Y. Park, J. Bae, [188] X. Wu, J. Ge, C. Yang, M. Hou, Z. Liu, Chem. Commun. 2015, 51,
K. Lee, J. Ind. Eng. Chem. 2017, 45, 5. 13408.
[159] D. P. Morales, G. B. Braun, A. Pallaoro, R. Chen, X. Huang, [189] P. Li, S. Moon, M. A. Guelta, S. P. Harvey, J. T. Hupp, O. K. Farha,
J. A. Zasadzinski, N. O. Reich, Mol. Pharmaceutics 2015, 12, 600. J. Am. Chem. Soc. 2016, 138, 8052.
[160] R. Mout, M. Ray, T. Tay, K. Sasaki, G. Yesilbag Tonga, V. M. Rotello, [190] Y. Zhang, F. Wang, E. Ju, Z. Liu, Z. Chen, J. Ren, X. Qu, Adv. Funct.
ACS Nano 2017, 11, 6416. Mater. 2016, 26, 6454.
[161] C. S. Kim, R. Mout, Y. Zhao, Y. C. Yeh, R. Tang, Y. Jeong, B. Duncan, [191] X. Lian, A. Erazo-oliveras, J. Pellois, H. Zhou, Nat. Commun. 2017,
J. A. Hardy, V. M. Rotello, Bioconjugate Chem. 2015, 26, 950. 8, 2075.
[162] Y.-C. Yeh, R. Tang, R. Mout, Y. Jeong, V. M. Rotello, Angew. Chem., [192] S. Wang, Y. Chen, S. Wang, P. Li, C. A. Mirkin, O. K. Farha, J. Am.
Int. Ed. 2014, 53, 5137. Chem. Soc. 2019, 141, 2215.
[163] R. Tang, C. S. Kim, D. J. Solfiell, S. Rana, R. Mout, [193] W. Morris, W. E. Briley, E. Auyeung, M. D. Cabezas, C. A. Mirkin,
E. M. Velázquez-Delgado, A. Chompoosor, Y. Jeong, B. Yan, J. Am. Chem. Soc. 2014, 136, 7261.
Z. J. Zhu, C. Kim, J. A. Hardy, V. M. Rotello, ACS Nano 2013, 7, [194] T. T. Chen, J. T. Yi, Y. Y. Zhao, X. Chu, J. Am. Chem. Soc. 2018, 140,
6667. 9912.
[164] M. Ray, R. Tang, Z. Jiang, V. M. Rotello, Bioconjugate Chem. 2015, [195] X. Yang, Q. Tang, Y. Jiang, M. Zhang, M. Wang, L. Mao, J. Am.
26, 1004. Chem. Soc. 2019, 141, 3782.
[165] R. Tang, Z. Jiang, M. Ray, S. Hou, V. M. Rotello, Nanoscale 2016, 8, [196] S. K. Alsaiari, S. Patil, M. Alyami, K. O. Alamoudi, F. A. Aleisa,
18038. J. S. Merzaban, M. Li, N. M. Khashab, J. Am. Chem. Soc. 2018,
[166] R. Mout, M. Ray, G. Yesilbag Tonga, Y. W. Lee, T. Tay, K. Sasaki, 140, 143.
V. M. Rotello, ACS Nano 2017, 11, 2452. [197] M. Trojanowicz, TrAC, Trends Anal. Chem. 2006, 25, 480.
[167] K. Lee, M. Conboy, H. M. Park, F. Jiang, H. J. Kim, M. A. Dewitt, [198] N. Saifuddin, A. Z. Raziah, A. R. Junizah, J. Chem. 2013, 2013, 1.
V. A. Mackley, K. Chang, A. Rao, C. Skinner, T. Shobha, [199] J. Liu, A. G. Rinzler, H. Dai, J. H. Hafner, R. Kelley Bradley,
M. Mehdipour, H. Liu, W. C. Huang, F. Lan, N. L. Bray, S. Li, P. J. Boul, A. Lu, T. Iverson, K. Shelimov, C. B. Huffman,
J. E. Corn, K. Kataoka, J. A. Doudna, I. Conboy, N. Murthy, Nat. F. Rodriguez-Macias, Y. S. Shon, T. R. Lee, D. T. Colbert,
Biomed. Eng. 2017, 1, 889. R. E. Smalley, Science 1998, 280, 1253.
[168] J. Long, X. Yu, E. Xu, Z. Wu, X. Xu, Z. Jin, A. Jiao, Carbohydr. Polym. [200] Z. Guo, P. J. Sadler, S. C. Tsang, Adv. Mater. 1998, 10, 701.
2015, 131, 98. [201] B. R. Azamian, J. J. Davis, K. S. Coleman, C. B. Bagshaw,
[169] S. Laurent, D. Forge, M. Port, A. Roch, C. Robic, L. Vander Elst, M. L. H. Green, J. Am. Chem. Soc. 2002, 124, 12664.
R. N. Muller, Chem. Rev. 2008, 108, 2064. [202] N. W. S. Kam, H. Dai, J. Am. Chem. Soc. 2005, 127, 6021.
[170] J. Zhang, M. C. Shin, A. E. David, J. Zhou, K. Lee, H. He, [203] N. W. S. Kam, Z. Liu, H. Dai, Angew. Chem. 2006, 118, 591.
V. C. Yang, Mol. Pharmaceutics 2013, 10, 3892. [204] P. D. Boyer, S. Ganesh, Z. Qin, B. D. Holt, M. J. Buehler,
[171] H. Y. Yang, M. S. Jang, Y. Li, J. H. Lee, D. S. Lee, ACS Appl. Mater. M. F. Islam, K. N. Dahl, ACS Appl. Mater. Interfaces 2016, 8, 3524.
Interfaces 2017, 9, 19184. [205] N. W. S. Kam, T. C. Jessop, P. A. Wender, H. Dai, J. Am. Chem. Soc.
[172] Y. T. Lim, M. Y. Cho, J. M. Lee, S. J. Chung, B. H. Chung, Biomate- 2004, 126, 6850.
rials 2009, 30, 1197. [206] H. L. Pang, J. Liu, D. Hu, X. H. Zhang, J. H. Chen, Electrochim.
[173] B. F. Hoskins, R. Robson, J. Am. Chem. Soc. 1989, 111, 5962. Acta 2010, 55, 6611.
[174] H. Cai, Y. Huang, D. Li, Coord. Chem. Rev. 2019, 378, 207. [207] M. Adeli, R. Soleyman, Z. Beiranvand, F. Madani, Chem. Soc. Rev.
[175] C. Doonan, R. Ricco, K. Liang, D. Bradshaw, P. Falcaro, Acc. Chem. 2013, 42, 5231.
Res. 2017, 50, 1423. [208] V. N. Mochalin, O. Shenderova, D. Ho, Y. Gogotsi, Nat. Nano-
[176] M. Sindoro, N. Yanai, A.-Y. Jee, S. Granick, Acc. Chem. Res. 2014, technol. 2012, 7, 11.
47, 459. [209] Y. Zhu, J. Li, W. Li, Y. Zhang, X. Yang, N. Chen, Y. Sun, Y. Zhao,
[177] M. Giménez-Marqués, T. Hidalgo, C. Serre, P. Horcajada, Coord. C. Fan, Q. Huang, Theranostics 2012, 2, 302.
Chem. Rev. 2016, 307, 342. [210] A. M. Schrand, H. Huang, C. Carlson, J. J. Schlager, E. Osawa,
[178] P. Horcajada, R. Gref, T. Baati, P. K. Allan, G. Maurin, P. Couvreur, S. M. Hussain, L. Dai, J. Phys. Chem. B 2007, 111, 2.
G. Ferey, R. E. Morris, C. Serre, Chem. Rev. 2012, 112, 1232. [211] L.-C. Lora Huang, H.-C. Chang, Langmuir 2004, 20, 5879.
[179] V. Lykourinou, Y. Chen, X. Wang, L. Meng, T. Hoang, L. Ming, [212] R. A. Shimkunas, E. Robinson, R. Lam, S. Lu, X. Xu, X. Q. Zhang,
R. L. Musselman, S. Ma, J. Am. Chem. Soc. 2011, 133, H. Huang, E. Osawa, D. Ho, Biomaterials 2009, 30, 5720.
10382. [213] X. Hu, X. Li, M. Yin, P. Li, P. Huang, L. Wang, Y. Jiang, H. Wang,
[180] W. Liu, S. Lo, B. Singco, C. Yang, H. Huang, C. Lin, J. Mater. Chem. N. Chen, C. Fan, H. Song, ACS Appl. Mater. Interfaces 2017, 9,
B 2013, 1, 928. 18575.
[181] Y. Cao, Z. Wu, T. Wang, Y. Xiao, Q. Huo, Y. Liu, Dalton Trans. 2016, [214] H. Li, K. Fierens, Z. Zhang, N. Vanparijs, M. J. Schuijs,
45, 6998. K. Van Steendam, N. Feiner Gracia, R. De Rycke, T. De Beer,
[182] C. He, K. Lu, D. Liu, W. Lin, J. Am. Chem. Soc. 2014, 136, 5181. A. De Beuckelaer, S. De Koker, D. Deforce, L. Albertazzi,
[183] E. Gkaniatsou, C. Sicard, R. Ricoux, J.-P. Mahy, N. Steunou, J. Grooten, B. N. Lambrecht, B. G. De Geest, ACS Appl. Mater.
C. Serre, Mater. Horiz. 2017, 4, 55. Interfaces 2016, 8, 1147.
[184] X. Lian, Y. Fang, E. Joseph, Q. Wang, J. Li, S. Banerjee, C. Lollar, [215] H. Shen, M. Liu, H. He, L. Zhang, J. Huang, Y. Chong, J. Dai,
X. Wang, H. Zhou, Chem. Soc. Rev. 2017, 46, 3386. Z. Zhang, ACS Appl. Mater. Interfaces 2012, 4, 6317.
[185] F. Lyu, Y. Zhang, R. N. Zare, J. Ge, Z. Liu, Nano Lett. 2014, 14, [216] H. Yue, X. Zhou, M. Cheng, D. Xing, Nanoscale 2018, 10, 1063.
5761. [217] Z.-Q. Xu, Q.-Q. Yang, J.-Y. Lan, J.-Q. Zhang, W. Peng, J.-C. Jin,
[186] D. Feng, T. Liu, J. Su, M. Bosch, Z. Wei, W. Wan, D. Yuan, Y. Chen, F.-L. Jiang, Y. Liu, J. Hazard. Mater. 2016, 301, 242.
X. Wang, K. Wang, X. Lian, Z. Gu, J. Park, X. Zou, H. Zhou, Nat. [218] J. Zhang, M. Zheng, Z. Xie, J. Mater. Chem. B 2016, 4, 5659.
Commun. 2015, 6, 5979. [219] P. N. Bhagat, S. H. Jadhav, S. Chattopadhyay, K. M. Paknikar,
[187] F.-K. Shieh, S.-C. Wang, C.-I. Yen, C.-C. Wu, S. Dutta, L.-Y. Chou, Nanomedicine 2018, 13, 353.
J. V. Morabito, P. Hu, M. H. Hsu, K. C.-W. Wu, C.-K. Tsung, J. Am. [220] A. D. Frankel, C. O. Pabo, Cell 1988, 55, 1189.
Chem. Soc. 2015, 137, 4276. [221] F. Heitz, M. C. Morris, G. Divita, Br. J. Pharmacol. 2009, 157, 195.

Adv. Mater. 2019, 31, 1902791 1902791  (31 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.advancedsciencenews.com www.advmat.de

[222] C. Bechara, S. Sagan, FEBS Lett. 2013, 587, 1693. [246] S. R. Macewan, A. Chilkoti, J. Controlled Release 2014, 190, 314.
[223] H. Y. Kim, S. Y. Yum, G. Jang, D.-R. Ahn, Sci. Rep. 2015, 5, 11719. [247] P. Koria, H. Yagi, Y. Kitagawa, Z. Megeed, Y. Nahmias, R. Sheridan,
[224] A. Fu, Y. Wang, L. Zhan, R. Zhou, Pharm. Res. 2012, 29, 1562. M. L. Yarmush, Proc. Natl. Acad. Sci. USA 2011, 108, 1034.
[225] A. Fu, Z. Zhao, F. Gao, M. Zhang, Pharm. Res. 2013, 30, 2108. [248] J. D. Kim, Y. J. Jung, C. H. Woo, Y. C. Choi, J. S. Choi, Y. W. Cho,
[226] A. Erazo-Oliveras, N. Muthukrishnan, R. Baker, T.-Y. Wang, Colloids Surf., B 2017, 149, 122.
J.-P. Pellois, Pharmaceuticals 2012, 5, 1177. [249] L. Ding, Y. Jiang, J. Zhang, H. A. Klok, Z. Zhong, Biomacromol-
[227] A. Erazo-Oliveras, K. Najjar, L. Dayani, T.-Y. Wang, G. A. Johnson, ecules 2018, 19, 555.
J.-P. Pellois, Nat. Methods 2014, 11, 861. [250] T. Gaj, J. Liu, K. E. Anderson, S. J. Sirk, I. Carlos, F. Barbas, ACS
[228] N. Nischan, H. D. Herce, F. Natale, N. Bohlke, N. Budisa, Chem. Biol. 2014, 9, 1662.
M. C. Cardoso, C. P. R. Hackenberger, Angew. Chem., Int. Ed. 2015, [251] J. J. Cronican, D. B. Thompson, K. T. Beier, B. R. McNaughton,
54, 1950. C. L. Cepko, D. R. Liu, ACS Chem. Biol. 2010, 5, 747.
[229] M. Li, Y. Tao, Y. Shu, J. R. LaRochelle, A. Steinauer, D. Thompson, [252] X. Yu, X. Gou, P. Wu, L. Han, D. Tian, F. Du, Z. Chen, F. Liu,
A. Schepartz, Z. Y. Chen, D. R. Liu, J. Am. Chem. Soc. 2015, 137, G. Deng, A. T. Chen, C. Ma, J. Liu, S. M. Hashmi, X. Guo, X. Wang,
14084. H. Zhao, X. Liu, X. Zhu, K. Sheth, Q. Chen, L. Fan, J. Zhou, Adv.
[230] M. Akishiba, T. Takeuchi, Y. Kawaguchi, K. Sakamoto, H.-H. Yu, Mater. 2018, 30, 1705383.
I. Nakase, T. Takatani-Nakase, F. Madani, A. Gräslund, S. Futaki, [253] K. B. Johnsen, J. M. Gudbergsson, M. N. Skov, L. Pilgaard,
Nat. Chem. 2017, 9, 751. T. Moos, M. Duroux, Biochim. Biophys. Acta 2014, 1846, 75.
[231] S. Dissanayake, W. A. Denny, S. Gamage, V. Sarojini, J. Controlled [254] A. Mizrak, M. F. Bolukbasi, G. B. Ozdener, G. J. Brenner,
Release 2017, 250, 62. S. Madlener, E. P. Erkan, T. Ströbel, X. O. Breakefield, O. Saydam,
[232] L. P. Herrera Estrada, J. A. Champion, Biomater. Sci. 2015, 3, 787. Mol. Ther. 2013, 21, 101.
[233] N. M. Molino, S.-W. Wang, Curr. Opin. Biotechnol. 2014, 28, 75. [255] C. Meyer, J. Losacco, Z. Stickney, L. Li, G. Marriott, B. Lu, Int. J.
[234] A. O. Elzoghby, W. M. Samy, N. A. Elgindy, J. Controlled Release Nanomed. 2017, 12, 3153.
2012, 161, 38. [256] Y. Yang, Y. Hong, G. H. Nam, J. H. Chung, E. Koh, I.-S. Kim, Adv.
[235] L. Yin, C. Yuvienco, J. K. Montclare, Biomaterials 2017, 134, 91. Mater. 2017, 29, 1605604.
[236] X. Li, L. Qiu, P. Zhu, X. Tao, T. Imanaka, J. Zhao, Y. Huang, Y. Tu, [257] M. J. Haney, N. L. Klyachko, Y. Zhao, R. Gupta, E. G. Plotnikova,
X. Cao, Small 2012, 8, 2505. Z. He, T. Patel, A. Piroyan, M. Sokolsky, A. V. Kabanov,
[237] H. J. Kang, Y. J. Kang, Y.-M. Lee, H.-H. Shin, S. J. Chung, S. Kang, E. V. Batrakova, J. Controlled Release 2015, 207, 18.
Biomaterials 2012, 33, 5423. [258] D. Yuan, Y. Zhao, W. A. Banks, K. M. Bullock, M. Haney,
[238] A. Zeltins, Mol. Biotechnol. 2013, 53, 92. E. Batrakova, A. V Kabanov, Biomaterials 2017, 142, 1.
[239] S. M. DePorter, B. R. McNaughton, Bioconjugate Chem. 2014, 25, [259] J. E. Pullan, M. I. Confeld, J. K. Osborn, J. Kim, K. Sarkar, S. Mallik,
1620. Mol. Pharmaceutics 2019, 16, 1789.
[240] S. J. Kaczmarczyk, K. Sitaraman, H. a Young, S. H. Hughes, [260] H. Soo Choi, W. Liu, P. Misra, E. Tanaka, J. P. Zimmer, B. Itty
D. K. Chatterjee, Proc. Natl. Acad. Sci. USA 2011, 108, 16998. Ipe, M. G. Bawendi, J. V. Frangioni, Nat. Biotechnol. 2007, 25,
[241] M. S. Nair, M. M. Lee, A. Bonnegarde-Bernard, J. A. Wallace, 1165.
D. H. Dean, M. C. Ostrowski, R. W. Burry, P. N. Boyaka, [261] C. He, Z. Tang, H. Tian, X. Chen, Adv. Drug Delivery Rev. 2016, 98,
M. K. Chan, PLoS One 2015, 10, e0127669. 64.
[242] M. Hofer, G. Winter, J. Myschik, Biomaterials 2012, 33, 1554. [262] Y. Barenholz, J. Controlled Release 2012, 160, 117.
[243] W.-J. Kim, B.-S. Kim, Y.-D. Cho, W.-J. Yoon, J.-H. Baek, K.-M. Woo, [263] E. Miele, G. P. Spinelli, E. Miele, F. Tomao, S. Tomao, Int. J.
H.-M. Ryoo, Biomaterials 2017, 122, 154. Nanomed. 2009, 4, 99.
[244] Y. Li, H. Song, S. Xiong, T. Tian, T. Liu, Y. Sun, Int. J. Biol. Mac- [264] H. Zhong, G. Chan, Y. Hu, H. Hu, D. Ouyang, Pharmaceutics 2018,
romol. 2018, 109, 672. 10, 263.
[245] Y. Jiang, H. Lu, F. Chen, M. Callari, M. Pourgholami, D. L. Morris, [265] R. M. Kannan, E. Nance, S. Kannan, D. A. Tomalia, J. Intern. Med.
M. H. Stenzel, Biomacromolecules 2016, 17, 808. 2014, 276, 579.

Adv. Mater. 2019, 31, 1902791 1902791  (32 of 32) © 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like