1 s2.0 S0753332220312968 Main

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Biomedicine & Pharmacotherapy 134 (2021) 111103

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Nanomedicines: Redefining traditional medicine


Weijia Lu a, 1, Jing Yao b, 1, Xiao Zhu a, c, d, e, *, Yi Qi a, c, d, e, *
a
Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China
b
First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
c
Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang
524023, China
d
The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang 524023, China
e
The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China

A R T I C L E I N F O A B S T R A C T

Keywords: Nanomedicines offer nanoscale drug delivery system. They offer ways of promising drug transportation, and
Cancer address the issues of lack of targeting and permeability of traditional drugs. The physical and chemical properties
Nanoparticles in the domain of nanomedicine applications in vivo have not been sufficiently delivered. What’s more, the
Nanotechnology
metabolic of nanomedicines is not clear enough. Those factors which mentioned above determine that many
Targeting therapy
Review
nanomedicines have not yet realized clinical application due to their safety problems and in vivo efficacy. For
example, they may cause immune response and cytotoxicity, as well as the ability to clear organs in vivo, the
penetration ability of them and the lack of targeting ability may also cause poor efficacy of drugs in vivo. In this
review, the new progresses of different kinds of nanomedicines (including gold nanoparticles, nanorobots, black
phosphorus nanoparticles, brain diseases, gene editing and immunotherapy etc.) in anti-tumor, antibacterial,
ocular diseases and arteriosclerosis in recent years were summarized. Their shortcomings were pointed out, and
the new methods to improve the biosafety and efficacy were summarized.

1. Introduction Traditional small molecule drugs have no special selectivity, and


they need to pay attention to the dosage and frequency, and can lead to
The field of nanomedicine is the science and technology of diag­ serious side effects. They do not pass through some biological barriers,
nosing, treating and preventing disease and traumatic injury, of and their efficacy may also affect due to the poor solubility. In contrast,
relieving pain, and of preserving and improving human health, using nanomedicines are targeted and help to reduce drug toxicity and
molecular tools and molecular knowledge of the human body. Nano­ improve bioavailability. It can slow-release drugs and prolong the half-
medicines can be endowed with characteristics different from tradi­ life of them. More importantly, the solubility and permeability of
tional drugs by embedding drugs in carriers or modifying nanodrugs [1, insoluble drugs should be improved to make the drugs cross biological
2]. barriers [3]. It is primarily used in disease diagnosis, treatment,

Abbreviations: AIDS, acquired immune deficiency syndrome; AuNFs, gold nanoframeworks; BBB, blood-brain barrier; BP, black phosphorus; BPNS, black phos­
phorus nanosheets; BTZ, bortezomib; BV, biliverdin; CNV, choroidal neovascularization; CO, carbon monoxide; CPT, camptothecin; CRISPR, clustered regularly
interspaced short palindromic repeats; CuS, copper sulfide; DCs, dendritic cells; DDS, drug delivery system; EGFR, epidermal growth factor receptor; EPR effect,
enhanced permeability and retention effect; GSH, glutathione; H2, hydrogen; H2S, hydrogen sulfide; HIV, human immunodeficiency virus; ICD, immunogenic cell
death; IDO1, indoleamine 23-dioxygenase 1; IgM, immunoglobulin M; MFIONs, magnetosome-like ferrimagnetic iron oxide nanochains; MNPs, magnetic nano­
particles; MSCs, mesenchymal stem cells; MTOR, rapamycin; NaCl, sodium chloride; NICS, nano immunoconjugates; NIR, near-infrared; NIR-II, the second near-
infrared; NO, nitric oxide; NPs, nanoparticles; PACT, photoacoustic computed tomography; PCE, photothermal conversion efficiency; PD, Parkinson’s disease; PDT,
photodynamic therapy; PEG, polyethylene glycol; PMLA, poly(β-L-malic acid); PTT, photothermal therapy; ROS, reactive oxygenspecies; SIRT1, silent information
regulator; SnTe, tin telluride; STING, stimulator of interferon genes; TDLNs, tumor-draininglymph nodes; tFNA, tetrahedral framework nucleic acid; WAT, white
adipose tissue.
* Corresponding authors at: Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong
Medical University, Zhanjiang 524023, China.
E-mail addresses: xzhu@gdmu.edu.cn (X. Zhu), qiyi7272@gdmu.edu.cn (Y. Qi).
1
These authors have contributed equally to this work.

https://doi.org/10.1016/j.biopha.2020.111103
Received 21 October 2020; Received in revised form 24 November 2020; Accepted 1 December 2020
Available online 15 December 2020
0753-3322/© 2020 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

monitoring and other drugs or biological products to improve human However, black phosphorus nanosheets (BPNS) can be used as their new
health [4,5]. In recent years, nanomedicine has great development po­ substitution of high drug loading, which has better biocompatibility and
tential and application prospect in clinical treatment of the tumor. In rapid degradation [20,21]. The organic combination of BPNS with Cu2 +
addition to tumor treatment, nanomedicine can also be used in can enhance their photothermal effect and make them possess excellent
anti-infection, ophthalmic disease, neurological disease, gene editing, rapid degradation and photothermal stability, which are not available in
immunotherapy and so on. Although nanotechnology has been proposed traditional phototherapeutic materials [22].
for decades [6], because of the great changes in physical and chemical In recent years, it has become a research hotspot to study the com­
properties of nanomaterials, the physicochemical properties and meta­ bination of multiple models and new nanomaterials for the treatment of
bolic pathways of nanotechnology still need to be studied [7]. tumors. BPNS realize the combination of chemotherapy and photo­
There is now broad consensus among medical researchers and thermal therapy for the treatment of pulmonary metastatic tumors [23].
practitioners that along with personalized medicine and regenerative The combination of photothermal therapy and photodynamic therapy is
medicine, nanomedicine is likely to revolutionize our definitions of what also realized, which improves the stability of the drug [20]. In addition
constitutes human disease and its treatment. At present, research on to cancer applications, the newly prepared black phosphorus (BP) drugs
intelligent nanorobots is also very popular. The rise of artificial intelli­ can also be used to assist in the treatment of depression [24]. BP has
gence (AI) also brings hope for the clinical transformation of nano­ potent biological activity and can be used as an antioxidant in
medicines [8]. In light of these developments, incorporation of key oxidation-reduction targeted therapy, which is a new field to be
nanomedicine concepts into the general medical curriculum ought to be explored. Hou et al. suggested that it could be effectively removed
considered. Although there have been a lot of reports on nanomedicines reactive oxygen species (ROS) in the kidney and protected the kidney
which focus on the developments of new carriers rather than clinical from ROS damage [25].
transformation. The purpose of this paper is to review the main progress
of nanomedicine and its application in different therapeutic strategies,
and to make a prospect for its future development [9]. 2.4. Gold nanoparticles

2. The main types and research progress of nanomedicines Gold nanoparticles (NPs) are made of gold and are characterized by
safety. Hocevar has shown that they do not adversely affect the immune
Nanomedicines include inorganic nanomaterials (metal & metal function of B lymphocytes in vitro for the first time [26]. They could be
oxide nanoparticles, carbon nanotubes, black phosphorus nano­ used as materials of photothermal therapy and for tumor treatment [27],
particles), dendrimers, polymers, micelles, liposomes, nanocrystals, etc. weight loss therapy and so on [28,29]. Recently, tumor cells have been
Here are some of the more important nanomedicines and their research found to have the ability to synthesize gold nanoparticles (NPs). This
progress. new biogenic route of gold NPs synthesis can synthesize spherical,
anisotropic and high aspect ratio gold nanomaterials [27,30]. Molecular
2.1. Nanocrystals dynamics simulation and experimental photothermal therapy show that
these nanomaterials are proven to be excellent imaging agents and drug
Nanocrystals are colloidal dispersive systems mainly formed by carriers, improve bioavailability in vitro and in vivo, and demonstrate
drugs, which can be stabilized and dispersed by modification of a small good safety [31–34]. The molecular dynamic simulation and supported
amount of surfactant or polymer materials [10]. They have high drug with experimental photothermal therapy shown excellent application of
loading capacity, which approach 100 %. They can improve the solu­ these nanomaterials into nanomedicine for clearing biofilm and pro­
bility and bioavailability of the drugs without carrier materials [11,12]. moting the grown of fibroblast [35,36]. The gold nanoparticles loaded
with clustered regularly interspaced short palindromic repeats (CRISPR)
2.2. Liposomes could be used to edit the genes of hematopoietic stem cells, promising
cures for blood diseases and acquired immune deficiency syndrome
Due to the modifiable sites, liposome nanodrugs have excellent (AIDS) [37].
biocompatibility and dependability, which can achieve targeting, However, gold nanoparticles have the problem of aggregation. The
improve the water solubility, slow-release drugs, and can also be made stability of gold nanoparticles in biological samples can be enhanced and
into vaccine carriers [13]. So far, they are most variety of nanomedicines the aggregation in vivo can be prevented by coating gold nanoparticles
on the market. The in vivo stability of mesoporous silica nanoparticles with protective polymer and making them spherical or selenium- (Se-)
can be improved by encapsulating them with liposomes [14]. Never­ terminated polyethylene glycol (PEG) [26,38]. Zhang et al. has com­
theless, although the manufacturing process can also reduce their bined gold nanoparticles and DNA ribbon, improving the targeting
curative effect, liposomes may cause drug leakage when conditions such properties of simple DNA nanoassembly materials. It is expected to be
as the pH or temperature change [15]. Liposome hydrogel can improve used as a new carrier for tumor targeting drug delivery [39].
the stability of the liposome nanodrug. The temperature can affect the
diffusion efficiency of liposomes in biological hydrogels. Preparation of
liposome nanodrugs with different phase transition temperature (Tm) 2.5. Polymeric nanomedicines
helps the treatment of diseases with different diffusion requirements
[16]. Polymeric/polymer nanoparticles is a material composed of poly­
It has been reported that liposome drugs attracted Immunoglobulin mer, combining metal particles, inorganic nonmetal particles or organic
M (IgM) in vivo and induce immunogenicity, and the protein corona of matter at the nanoscale [40]. They could be utilized to treat neurological
plasma protein formation also affected their targeting [17]. Using the diseases and improve the drug delivery of the brain, extending the drug
apolipoprotein in the plasma as a target factor does not cause immu­ blood circulation time and improving the material utilization. Chen
nogenicity, and can be utilized to cross the blood-brain barrier (BBB) et al. developed a newly nanomedrug for Parkinson’s disease, opening
[18]. up an oral approach that can cross the gastrointestinal and blood-brain
barriers [41]. A novel polymer can reduce the off-target toxicity of
2.3. Black phosphorus (BP) chemotherapy coupled with photodynamic therapy [42].
The polymeric nanomedicine synthesized by mimicking the viral
Carbon nanotubes have recently been added to the SIN (‘Substitute It structure can control drug release, and assist macromolecular drugs to
Now’) list to their carcinogenicity and reproductive toxicity [19]. break away from vesicles and avoid being degraded by lysosomes [43].

2
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

2.6. Medical nanorobots antibodies) [62,63]. In 2018, Li et al. developed an independent DNA
nanorobot system, which can recognize tumor-associated endothelial
2.6.1. Functions of medical nanorobots cells, induced tumor vascular embolism. Thus the tumor blood supply
Nanorobots refer to the size of nanoscale robot, which can accurately was blocked without affecting the vascular system in healthy tissues
build and manipulate objects on the nanoscale. Their advantages lie in [64] (Fig. 1).
their targeting and controllable movement, which contributes the ability However, aptamers are also unstable. To solve this problem, Ma et al.
of drugs to cross biological barriers [44,45]. At present, there are increased the biological stability and anticancer activity of aptamer by
numerous researches on the treatment of tumors. For example, Wu et.al adding tetrahedral framework nucleic acid (tFNA) to the anti-HER2
used photoacoustic computed tomography (PACT) to realize real-time aptamer [65].
imaging and control the nanorobots tract [46].
2.6.2.3. Micro/nanomachines based on biological cells. Connected with
2.6.2. Classification of medical nanorobots cells or bacteria and other organisms, micro/nanomachines based on
Nanorobots can be divided into two types: outfield drive and self- biological cells are endowed with autonomous movement ability [50,
drive. The driving force of outfield drives generally comes from 66]. There are built by coupling the motile microorganism (responsible
outfield, such as an optical drive, the sound field drive, magnetic field for movement) with the synthetic structure (providing other functions)
drive and so on [47,48]. Self-drive nanorobots can obtain power from [67]. Cells used to build their units include bacteria, sperm cells, heart
the fluid environment through chemical reactions or enzymes [49]. In muscle cells, and white blood cells [68,69]. These nanorobots enable the
recent years, the research on biological hybrid nanobots that use biology active and targeted delivery of drugs in vivo, avoiding over-dependence
to promote nanobots has also increased [50]. on EPR effects, and can simultaneously possess biological characteris­
tics. For example, microalgae tend to light, while biological cells such as
2.6.2.1. Magnetic nanorobots. The nanomedicines driven by the mag­ sperm and bacteria have an intrinsic system for responding to hypoxia
netic field were called magnetic nanorobots, or magnetic nanoparticles stimuli, as well as a directional environment (temperature, magnetic
(MNPs). One of the advantages of magnetic nanorobots is that they can field, pH, glucose) [66].
enhance the penetration of drugs in solid tumors. However, it only re­ The latest trend of research on biological hybrid nanorobots focuses
flected in superficial tumors in the past. In recent years, some new on their application in precision medicine for clinical transition, such as
progress has been made in further deepening the permeability of mag­ how to reduce the size of nanorobots [45,70].
netic nanoparticles. Liu developed a system of two oppositely polarized
external magnets could increase the drug permeability in solid tumors
by five times [51]. A nanospinners made of magnetic nanoparticles 2.7. Polymeric micelles
could also enhance the penetration depth of drugs by applying me­
chanical force to the rotating magnetic field and targeting the mito­ Polymeric micelles have core-shell structure, which can encapsulate
chondria of deep cancer cells [52]. Zhou et al. used a two-magnet hydrophobic drugs, provide modification points and reduce drug
configuration to introduce a constant magnetic field gradient, which toxicity [71]. Micellar docetaxel regulates tumor immune microenvi­
greatly enhances the accumulation and penetration of MNPs in solid ronment and increases the proportion between M1 and M2-like mac­
tumor models [53]. rophages, which can be used in combination chemotherapy and
It is a research hotspot to make magnetic nanorobots overcome the immunotherapy [72]. It should not be ignored that photodynamic
complex structure of the human body to reach specific sites. In 2019, therapy combined with chemotherapy is toxic, and a polymeric micelle
Huang et al. developed a magnetic nanorobot that can change its shape can control drug release and reduce off-target toxicity [42]. Li et al.
according to pass through narrow, curved blood vessels. It can move found that the synergistic effect of cisplatin crosslinking and polymeric
freely in the body with the fluid flows, depending on changes in the prodrug could be used to prepare polymer prodrug micelle in order to
surrounding electric field [54]. In the same year, Zhong et al. developed further improve its retention time in blood circulation [3].
a new drug to study the nucleus of tumor cells by using optical tweezers,
without destroying the cell membrane or cytoskeleton. It can use to 3. Advances in the application of nanomedicines in different
study the characteristics of cancer cells, and may point out the direction fields
for strengthening diagnosis and treatment [55].
In order to make the magnetic nanorobots effective, it is necessary to 3.1. Applications of nanomedicines in cancer
monitor their track [56]. Yan et al. replaced fluorescence based imaging
with MR imaging, which has a stronger ability to penetrate tissues [57]. The dense tumor tissues make it difficult for traditional drugs to
The nanorobots can be precisely concentrated in a small area of the penetrate. However, nanomedicine can increase drug penetration, pro­
retina, by using clinical optical coherence tomography [58]. Currently, longing circulation time, and reduce the accumulation of drugs in non-
some magnetic nanoparticles have begun to be used for in vitro diag­ target tissues, thus having unique advantages in the treatment of tumors.
nosis and treatment, but there is still a long way to go before they are The vascular endothelial cell gap of the tumor is large, so nanomedicine
widely used for in vivo diagnosis and treatment due to the limited can enter into the tumor tissue through the vascular endothelial cell gap
cytotoxicity of nanomaterials [4]. and enrich, which is called enhanced permeability and retention effect
(EPR effect) of the tumor [73]. Mitochondria of tumor cells can be used
2.6.2.2. DNA nanorobots. DNA can connect other molecules with its as targets for nanomedicine because they produce ROS [20,74].
negative potential phosphate groups after damaging the highly struc­ Increasing the osmotic and promoting cell lysis through nanoparticles
tured water surface on its surface, which plays an important role in its containing sodium chloride (NaCl) nanoparticles is also a new way to
stability [59]. DNA origami can be used for self-assembly of nano­ kill tumors [75]. In addition, Wang et al. proposed the development of
particles or as a vehicle to guide drug delivery. The former reduces nano-gas drugs (that is the gas-releasing nanomedicines), which can be
immunogenicity by assembling new systems from the bottom-up to used to release hydrogen (H2), carbon monoxide (CO), nitric oxide (NO),
reduce the size of nanomedicines [60]. The latter can achieve voluntary and hydrogen sulfide (H2S) to inhibit mitochondrial respiration of tumor
movement to the target in vivo by connecting the aptamers [61]. Their cells, so as to treat cancer and other diseases [76].
activities are actually triggered by environmental stimuli (such as pH or
temperature) or by chemicals (such as DNA strands, small molecules, 3.1.1. Immunotherapy of tumor with nanomedicines
Targeting the mechanism of tumor immune escape, tumor

3
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

Fig. 1. DNA nanorobots block tumor blood vessels.


DNA nanobots loaded thrombins recognize the nuclear protein and change their shape, releasing thrombins to cause the tumor blood vessel obstruction. Thus the
tumor is a lack of nutrition.

immunotherapy can control and kill tumor cells by activating and Therefore, it is necessary to improve PTT or develop novel nano­
strengthening the immune system, or by cutting off the inhibitory im­ materials with clear metabolism and good bio-safety, better photo­
mune regulation of the immune checkpoint with checkpoint inhibitors stability, and high PCE [85,86].
[77]. Nevertheless, the efficacy of immunotherapy has individual dif­ Endogenous materials have the advantage of having a clear meta­
ferences, which is usually merely used as an adjuvant therapy combined bolic mechanism. Biliverdin (BV), the recently developed near-infrared
with surgery, chemotherapy, radiotherapy, and so on [78]. (NIR) absorption pigment, has no obvious toxic and side effects [85].
A mounting number of studies have been conducted on the appli­ The development of nanomaterials with high absorption capacity in the
cation of nanomedicine in combination with immunotherapy. The ad­ second near-infrared (NIR-II) is helpful to improve the photothermal
vantages are as follows: first, the synergistic effect of multiple therapies therapy of deep tumors. The newly developed materials include new
can make up for the deficiencies and avoiding tumor metastasis, such as gold nanoframeworks (AuNFs), the Gd- and copper sulfide (CuS)-inte­
improving the application rate of tumor immune drugs in different pa­ grated nanogel [87,88]. The nanomedicine developed by Zhang et al.
tients [79,80]. Second, the delivery capacity is strong enough to carry a was wrapped with MnO2 and soybean phospholipid, which has high PCE
variety of immune checkpoint inhibitors. Third, anti-cancer immunity (NIR-I is 38.2 %, NIR-II is 43.9 %). It is also mediated by NIR-II mediated
can be reinforced by inducing immunogenic cell death (ICD) [75]. The and has good biodegradability [89].
indoleamine 2,3-dioxygenase 1 (IDO1) siRNA was delivered to In the aspect of improving PCE, there are also some new nanodrugs.
tumor-draining lymph nodes (TDLNs) and tumor tissues simultaneously For example, NIR - absorbing quaterrylenediimide chromophore has
by cationic lipid-assisted nanoparticles, which could interfere with the high stability and high photothermal conversion efficiency (PCE) [90].
IDO1 pathway and enhance the immune response induced by ICD [81]. In addition, a nanomedicine that can be degraded by enzymes in vivo
The tumor microenvironment can affect the effect of immuno­ has the PCE compared with the other PTT agents previously reported
therapy. The delivery of NO to the tumor by nanosystem can improve [84] (Table 1). Generally speaking, the PCE of polymer nanoparticles is
the tumor microenvironment. However, it used to rely too much on
external excitation, but current research shows that nanomedicines
combine radioisotope to stimulate the continuous release of NO by Table 1
endogenous light, which can help activate the immune system to elim­ Comparison of photothermal conversion efficiency of various photothermal
inate metastatic tumors [82]. Semiconducting Polymer Nanoreporters agents.
can be used for immunoactivated near-infrared chemiluminescence Usage Nanoparticles PCE References
imaging during cancer immunotherapy. It can sensitively distinguish Tumor BP@Cu nanostructure 35.4% Hu et al.(2020)
immune cells from other cells (including cancer and normal cells) and is [17]
used to track T cell activation [83]. Tumor Biliverdin (BV) nanoagents 34.3 % Xing et al.(2019)
[61]
Tumor water-soluble quaterrylenediimide 64.7 ± 4 % Liu et al.(2018)
3.1.2. Photothermal therapy of tumor with nanomedicines (QDI) [62]
Photothermal therapy (PTT) has the advantages of noninvasive, Tumor Gd-/CuS-Loaded Functional 26.7 % Zhang et al.(2020)
precise manipulation, and good therapeutic effect. The principle is to Nanogels [64]
make the high PCE of the material gathered near the tumor tissue. And Tumor SnTe@MnO2-SP NSs 43.9 %(NIR Zhang et al.(2019)
II) [65]
when exposed to an external light source (usually near-infrared), the
Tumor π-conjugated oligomer 82 % Li et al.(2019) [60]
local high temperature of tumor tissue is caused to kill cancer cells. At nanoparticles
present, the nanomaterials that have been studied for PPT have their
BP, black phosphorus; NIR-II, the second near-infrared; PCE, photothermal
own shortcomings and are slightly inadequate in clinical practice [84].
conversion efficiency; SnTe, tin telluride; SP, soybean phospholipid.

4
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

higher than that of inorganic nanoparticles [40]. Phototherapy alone antibiotic resistant-bacteria. Because of its targeting, it can reduce the
cannot avoid tumor metastasis, thus it is often combined with other effective concentration of drugs and enhance the effectiveness of exist­
therapies (including chemotherapy, immunotherapy, magnetic nano­ ing antibacterial drugs [103]. According to the principle of PTT, a sul­
particles, etc.) [91,92]. fide (covellite) nanodots developed by Qiao et al. have a strong killing
effect on drug-resistant bacteria and can release Cu2 + which promotes
3.1.3. Photodynamic therapy of tumor with nanomedicines healing. It has the potential to be used in chronic, refractory wounds due
Compared with chemotherapy and radiotherapy, photodynamic to infection [104]. What’s more, hydrothermal therapy had also been
therapy (PDT) has the characteristics of non-invasive, low side effects, shown to have good antibacterial properties. This technology has
and high selectivity [93]. The principle of PDT is that photosensitizer developed a new antimicrobial strategy [105]. The strategy of killing
excited by light of specific wavelength transmits energy to the sur­ pathogenic bacteria by using reactive oxygen species produced by the
rounding oxygen to produce singlet oxygen and other reactive oxygen photocatalytic effect is considered to be one of the most promising
species (ROS), leading to tumor cell necrosis and apoptosis [94,95]. antibacterial treatments. Black phosphorous nanocrystals, which
The mitochondria of tumor cells produce too much ROS and gluta­ combine silver nanoparticles, are excellent materials [106].
thione, so it has become a promising direction of phototherapy to
transport anti-tumor drugs by targeting mitochondria, which can solve 3.4. Applications of nanomedicines in diagnosis and treatment of obesity
the problem that exogenous light has permeability limitation [20,52,94, and related metabolic diseases
96]. Beside, X-ray has better penetration than ordinary laser, which is
also a strategy for PDT of the deep tissues [55,97]. The advantage of nanomedicines is to increase the bioavailability of
However, the disadvantage lies in the short life and limited action drugs to reduce the tremendous toxicity of traditional weight-loss drugs.
range of ROS. The nanoreactor developed by Zhang et al. directly Nanomedicines for the treatment of obesity and related metabolic dis­
released camptothecin (CPT) after being activated by ROS in mito­ eases are mainly through early diagnosis, targeted delivery of drugs,
chondria, which could solve this problem [98]. Also, photodynamic reducing blood lipid concentration, controlling appetite, inhibiting fat
therapy for oxygen dependence has found a solution. For example, the absorption, transforming white adipose tissue (WAT) into brown-like
oxygen production capacity of the organism can be used to solve it by adipose tissue, etc [107–109].
coating nanoparticles on the surface of spirulina platensis [99]. What’s Using gold nanoparticles can directly eliminate adipose tissue
more, a semiconductor polymer NPs prodrug was synthesized, which through PTT, it can improve the appearance of uneven liposuction [29,
could realize the high-efficiency synergistic therapy of PDT and 110] (Fig. 2). Prevention and treatment of atherosclerosis can be ach­
chemotherapy in a hypoxic environment [93]. Cherenkov light can be ieved by inhibiting macrophage cell proliferation with nanodrugs
used as an in vivo light source for photodynamic therapy. Its low light loaded with simvastatin, thus achieving anti-inflammatory effects and
efficiency can be solved by combining nuclides with nanoparticles [82]. halting the progression of atherosclerosis [111]. Besides, increasing the
expression of silent information regulator 1 (SIRT1) in plaque can di­
3.2. Applications of nanomedicines in ocular diseases agnose atherosclerosis and inhibit its growth [109]. Nanomedicines can
avoid the side effects of vascular restenosis and thrombosis caused by
3.2.1. Deficiency of common drugs in the treatment of ocular diseases surgery (such as stent implantation). For example, an atherosclerotic
Due to the existence of physical and physiological multi-layer drug, rapamycin (MTOR), could be wrapped in a modified red blood cell
physiological barrier in the eyes, existing administration methods (eye membrane and injected into the blood to deliver the drug to the plaque
drops or intraocular injections) not only have a low utilization rate, but without surgery [12].
also bring discomfort to patients.
3.5. Applications of nanomedicines in brain diseases
3.2.2. Advances in research on the treatment of ocular diseases by
nanoscale drugs Compared with traditional drugs, nanomedicines have the advantage
Nanodrugs can increase the physiological activity of drugs, effec­ of crossing the blood-brain barrier. Recently, some studies have applied
tively increase the retention time of drugs in the eye. They solve the high it to Parkinson’s disease, ischemic stroke and so on [112]. The nano­
effective concentration problem that common drugs have, and also assist medicine used in brain diseases is mainly polymer. For example, Chen
drugs to penetrate the physiological barrier of eyes [100]. Wu et al. et al. have developed an orally available drug for Parkinson’s disease
coated the liquid coating liquid layer on a spiral-shaped magnetic (PD), which can cross the gastrointestinal barrier and BBB [41]. Nano­
nanorobot and made it get rid of the bondage of biological macromol­ drugs can be used in mesenchymal stem cells (MSCs) - based therapy,
ecules through the control of an external magnetic field, so as to break which can improve the secretion level of natural stem cells and cross the
through the blood-ocular barrier and accurately concentrate in the le­ blood-brain barrier. It can be used in the recovery treatment of ischemic
sions of the eyes [44]. stroke [113].
The antibacterial function and penetrating ability of nanomedicine Using poly (β-L-malic acid) (PMLA), as a carrier to construct immune
provides new ideas for the treatment of ocular diseases. Ye et al. checkpoint inhibitors CTLA-4 and PD-1 antibody nano immunoconju­
developed the AuAgCu2O-bromfenac sodium nanoparticles, which are gates (NICS) can effectively cross BBB and induce a local immune
based on the principle of PTT, combined with the high-efficiency anti­ response of gliomas. It is expected to be used in the treatment of brain
bacterial properties of silver nanoparticles and the advantages of copper gliomas [114]. Nanodrugs used in the treatment of depression can in­
ions in promoting healing. It has the potential to be used for the infection crease drug loading, improve the biocompatibility of fluoxetine. More
of drug-resistant bacteria in the eyes [101]. Wang et al. developed a importantly, it can greatly speed up the treatment of depression [24].
non-invasive treatment for Choroidal neovascularization (CNV) [102].
Intravenously, it is triggered by shining light on the eyes, avoiding the 3.6. Applications of nanomedicines in pulmonary diseases and heart
destruction of retinal tissue caused by repeated intravitreal injections failure
and conventional laser treatment.
For the treatment of pulmonary diseases, the major obstacle is to
3.3. Applications of nanomedicines in infectious diseases make drugs through pulmonary surfactant without damaging [115]. In
the study of Tian et al., the nanomedicine showed a strong ability to
Nanomedicines are a feasible substitute for antibiotics. Although the cross the pulmonary surfactant film without damaging lung structure
mechanism is not clear, studies have shown that it has a better effect on [116]. Additionally, a new nanomedicine offers the opportunity to

5
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

Fig. 2. Treatment of obesity with PTT.


(A) Gold nanoparticles are injected into adipose
tissue.
(B) Irradiation of epidermis with 800 nm laser
increases the temperature of gold nanoparticles
and liquefies small volumes of fat.
(C) Adipose tissue and gold nanoparticles were
removed by a liposuction program.

improve the effectiveness of influenza virus vaccines. It transports 4. The shortage and research progress of nanomedicines
stimulator of interferon genes (STING) activator through the pulmonary
surfactant to produce a large number of immune mediators, which 4.1. Safety problem
significantly enhance the recruitment and differentiation of CD11b +
dendritic cells (DCs) and CD8 + T cells [117]. The safety of nanodrugs has always been a difficult problem to solve.
The use of drugs targeting the heart can improve efficacy. However, Firstly, the deposition of nanoparticles and activation of macrophages
it is easy to produce adverse reactions by intravenous or oral methods may contribute to adverse immune reactions. Secondly, nanodrugs may
[118–120]. Using the proximity of heart and lung, Miragoli et al. accumulate in liver, spleen and other organs, and may cause cytotoxicity
developed a drug for heart disease with a new mode of transportation. It [7,128]. The cause of the immune response is due to the size of nano­
enters the body by inhalation and is more efficient at transporting than medicine and the heterologous type of nanomedicines. Reducing the size
injections, and can be used to improve heart failure [121]. of nanomedicines via the bottom-up assembly strategy, developing
endogenous nanodrugs [60,70,85], or using cells to provide nanorobots
3.7. Applications of nanodrugs in gene therapy driving force [50], can improve their safety.
The prodrug is the drug delivery system (DDS) that can reduce the
In the process of gene modification with CRISPR, viruses are usually cytotoxicity of drugs [129]. For example, the preparations of nano­
used as drug carriers. In this way, the manufacturing process is complex, prodrug can solve the problems of poor stability and toxicity of borte­
which may lead to off-target effects and cause the immune response zomib (BTZ) in vivo [130]. The efficiency of the precursor
[122]. Light is ideal for regulating CRISPR/Cas9 gene editing, whereas nano-assembly can be refined by adding sulfur/ Selenium/Carbon
most existing light sensors are sensitive only to ultraviolet or visible light linkages to the drug. The circulation time of the drug is prolonged,
and are suitable for shallow tissues. Recently, the first CRISPR/Cas9 which is beneficial to cell uptake [131].
nanoscale vector that can be edited using near-infrared regulatory genes Improving targeting capabilities can reduce drug toxicity. Metallic
has been developed, which can penetrate deeper tissues [123]. platinum compounds are commonly used in clinical chemotherapy for
Replacing it with nanodrugs can solve the problems of cell damage, tumors. However, due to the lack of tumor site selectivity, small mole­
high cost, long time consuming, and even overcome the off-target effect cule platinum-based drugs lead to greater toxicity [7,128].
[124]. In 2019, Shahbazi et al. used a gold nanomedicine as a CRISPR Receptor-mediated targeting can improve drug targeting. The DNA
vector for gene editing of hematopoietic stem cells and proved that there nanocarriers take advantage of the good selectivity of cells expressing
was no toxicity. It is expected to be used for gene editing of the human high epidermal growth factor receptor (EGFR) to enhance the selectivity
immunodeficiency virus (HIV) and hereditary blood diseases [37]. of platinum drugs and reduce the systemic toxicity of platinum drugs for
Moreover, some scientists have proposed another vector to replace the chemotherapy [7,128].
virus. In 2019, Chen et al. designed a nanocapsule with thin glutathione Currently, AI (including machine learning) provides computing tools
(GSH) coating. The experiment showed that the drug had a strong that facilitate the simulation and modeling process in nanotherapeutics
gene-editing ability on cells, and had safety and targeting ability [125]. [132]. For example, an AI toolbox can be combined to understand the
Nanorobots transfected with non-viruses have also been developed to biophysical and chemical properties of the nanoscale interface [133].
gene edit stem cells, further improving the autonomous transport of In-silico methods could decipher the quantitative nanostructure
drugs. For example, magnetosome-like ferromagnetic iron oxide nano­ activity-relationship (Nano-QSAR) [134]. These tools can be used for
chains (MFIONs) can be used to make genetic changes to stem cells that analyzing the toxicology of nanomedicines, and the accuracy is higher
can normally deliver recovery treatments for ischemic stroke [89,126, than experimental animals [135]. This means significant savings in
127]. money and time to predict the biosafety and efficacy [132].

4.2. Curative effect problem

The factors influencing the efficacy of nanodrugs are circulation,

6
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

accumulation, penetration, internalization, and release. The main ones Therefore, the strategy of active transportation is being explored. It is
are the body’s clearing mechanisms (kidney, liver, and immune system) possible to greatly improve the permeability of nanodrugs by inducing
and the penetration and targeting capabilities of the nanomedicine itself tumor cells to take active antitumor drugs. For example, Zhou et al.
[136]. make use of the role of endocytosis and transport to make the cells in the
Nanomedicines can prolong their circulation time in vivo and adjacent layer, repeat endocytosis and transcytosis of nanomedicine.
enhance the enrichment in the tumor site by modifying PEG. But PEG This mechanism may enable the delivery of nanomedicine across cells
restricts their uptake by tumor cells [137]. The probability of being independent of diffusion [150]. However, nanorobots are likely to
cleared by the immune system can be greatly reduced and the blood achieve greater penetration [46]. Wu et al. are the first to achieve
circulation half-life can be prolonged by coating the nanoparticles with controlled movement of a nanorobot in the eye, solving the problem of
cell membranes. In 2019, Chai et al. encapsulated nanocrystals of brain eye medication that cannot cross multiple biological barriers [44].
tumor drugs with erythrocyte membrane. The nanocrystals were tar­
geted and could be cleared across the blood-brain barrier [11,12]. NPs 5. Conclusion and perspectives
coated with CXCR4-overexpressing nerve stem cell membranes signifi­
cantly improved the efficacy of glibenclamide in stroke treatment, This paper mainly introduces the research progress of several kinds
enhanced nanoparticle penetration in the brain, and led to a new of nanoparticles and their application in treating diseases in recent
approach to improve drug delivery efficiency in ischemic brain. The years. Nanomedicines have made considerable progress in many fields,
drug is expected to be translated into clinical applications to improve the especially in the treatment of cancer. Besides, it is of great significance in
treatment of stroke [138]. some fields. For example, the substitution of antibiotics, and the
improvement of the permeability and targeting ability of nanorobots to
4.2.1. Targeting problem drugs. Their advantages lie in assisting drug targeting, reducing toxicity,
Nanomedicines mainly increase the targeting ability in three di­ controlling drug–releasing, and enhancing drug penetration ability.
rections: cancer cells, tumor immune microenvironment, and peripheral However, the safety and efficacy of nanomedicines cannot be ignored.
immune system [139]. The targeting of nanomedicines has special sig­ The permeability of nanomedicines can be improved by modifying
nificance in the treatment of cancer metastasis but also has some nanodrugs, but it is not enough to transport them by the EPR effect
shortcomings. First, drugs depend on tumor cell markers [140–143]. [154]. In the future, nanomedicines or nanorobots combine with bio­
This restricts the practical application of drugs. Second, drugs that jam logical cells will focus on improving their biocompatibility and efficacy.
the blood supply by targeting the tumor vascular system have the It will make them propelled inside the body without the need for so­
insufficient targeting ability and high effective dose. Third, phage pep­ phisticated instrumentations, space, chemicals, acoustics, and magne­
tide modified nanoparticles usually have the disadvantage of weak tism. It is not only necessary to determine the efficacy of nanomedicines
target binding and cannot specifically target the relevant cell tissue. in vitro, but also to further study the physicochemical properties and
Fourth, the targeting ability of nanodrugs is seriously decreased and the pharmacokinetics of nanomedicine in vivo, so as to transform them into
biochemical and physical properties of NPs are seriously changed after clinical applications.
they are adsorbed by proteins in physiological fluids in vivo to form The development of nanomedicines in the future may not only
protein coronas. require animal experiments, but also the prediction of pharmacokinetic
In view of the above shortcomings, tumor cells with higher enzyme and pharmacotoxicological properties via machine learning and AI
digestion activity than normal liver cells can design drugs targeting toolbox [155,133]. Nanomedicine is likely to develop into an
cancer cells, which can produce tumor-targeting drugs without markers environment-friendly nanomedicine, which is harmless to biology and
[144–146]. The elimination of collagen in the tumor extracellular ma­ has a good effect in vivo. This requires advances in artificial intelligence
trix is beneficial to enhance the targeting of nanomedicine [147]. Using algorithms and medical researchers’ commitment to the clinical trans­
DNA nanorobots to target tumor vascular system can improve the tar­ formation of nanomedicine.
geting ability of nanomedicine, and transport thrombin to block tumor The modification of drug properties by nanomedicines offers
blood vessels [64]. Wu et al. developed a nanoparticle carrier with considerable possibilities, yet few clinical oncologists are aware of the
brain-targeting capability that could cross the blood-brain barrier, importance of antitumor nanodrugs. Therefore, it is necessary to
avoiding the shortcomings of weak effectiveness and specificity of active incorporate nanomedicine into the general medical curriculum, which is
targeting of nanoparticles modified by phage display peptide surface. It conducive to promoting the rapid development of nanomedicine to
can be used for brain-targeted delivery of drugs for central nervous clinical application.
system diseases [148]. Zhao et al. found that the reaction kinetics of In addition to the further improvement of nanomedicines, the future
magnetic nanoparticles with the positive charge was faster, which may in terms of ethics and market of nanomedicines has a way to go. The
be used to solve the problem of the poor targeting of NPs in vivo due to ethical issues raised by the safety and efficacy issues mentioned above
the formation of protein coronas [149]. and the economic benefits of nanomedicines should also be assessed by
assess systems. Otherwise, global equality in access to health care could
4.2.2. The trouble of permeability and diffusion ability be further undermined when nano-diagnosis and nano-treatment are
Although nanodrugs can be aggregated in the tumor site through the promoted. Respect for autonomy, beneficence, non-maleficence, and
EPR effect, most of them remain around the blood vessels and are justice will be major key biomedical ethics that policy makers and reg­
difficult to enter tumor cells through the circulatory system [150]. The ulatory bodies need to carefully implement [156,157].
reason is the obstruction of tumor extracellular matrix (collagen, hyal­
uronic acid, etc.), high pressure, and low permeability in tumors [151, Authors’ contributions
152]. Measures to enhance the EPR effect [153] include degrading
collagen in the tumor matrix [118], reducing the size of nanoparticles, WL wrote the manuscript. JY and XZ discussed and edited the
or modifying functional groups on the surface to increase permeability. manuscript. XZ designed the concept. YQ and XZ supported the funding.
Using the bionic lipoprotein system, tumor stroma can also be destroyed All authors read and approved the final manuscript.
by the photothermal effect [124].
In the last decades, it has been increasingly recognized that there is Funding
large inter- and intra-individual heterogeneity in EPR-mediated tumor
targeting, explaining the heterogeneous outcomes of clinical trials in This work was supported partly by National Natural Science Foun­
which nanomedicine formulations have been evaluated [154]. dation of China (81973908); Guangdong Science and Technology

7
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

Department (2016B030309002 and 2019B090905011); The Fund of [22] K. Hu, L. Xie, Y. Zhang, M. Hanyu, Z. Yang, K. Nagatsu, H. Suzuki, J. Ouyang,
X. Ji, J. Wei, H. Xu, O.C. Farokhzad, S.H. Liang, L. Wang, W. Tao, M.R. Zhang,
Southern Marine Science and Engineering Guangdong Laboratory
Marriage of black phosphorus and Cu(2+) as effective photothermal agents for
(Zhanjiang) (ZJW-2019-007); The Public Service Platform of South PET-guided combination cancer therapy, Nat. Commun. 11 (1) (2020) 2778.
China for R&D Marine Biomedicine Resources (GDMUK201808); [23] S. Wang, J. Shao, Z. Li, Q. Ren, X.F. Yu, S. Liu, Black phosphorus-based
Zhanjiang Science and Technology Plan (2017A06012). multimodal nanoagent: showing targeted combinatory therapeutics against
cancer metastasis, Nano Lett. 19 (8) (2019) 5587–5594.
[24] L. Jin, P. Hu, Y. Wang, L. Wu, K. Qin, H. Cheng, S. Wang, B. Pan, H. Xin,
Consent for publication W. Zhang, X. Wang, Fast-acting black-phosphorus-assisted depression therapy
with low toxicity, Adv. Mater. 32 (2) (2020) e1906050.
[25] J. Hou, H. Wang, Z. Ge, T. Zuo, Q. Chen, X. Liu, S. Mou, C. Fan, Y. Xie, L. Wang,
All authors consent for publication. Treating acute kidney injury with antioxidative black phosphorus nanosheets,
Nano Lett. 20 (2) (2020) 1447–1454.
[26] S. Hocevar, A. Milosevic, L. Rodriguez-Lorenzo, L. Ackermann-Hirschi, I. Mottas,
Declaration of Competing Interest A. Petri-Fink, B. Rothen-Rutishauser, C. Bourquin, M.J.D. Clift, Polymer-coated
gold nanospheres do not impair the innate immune function of human B
lymphocytes in vitro, ACS Nano 13 (6) (2019) 6790–6800.
The authors state that there is no conflict of interest.
[27] A.V. Singh, T. Jahnke, V. Kishore, B.W. Park, M. Batuwangala, J. Bill, M. Sitti,
Cancer cells biomineralize ionic gold into nanoparticles-microplates via secreting
References defense proteins with specific gold-binding peptides, Acta Biomater. 71 (2018)
61–71.
[28] F. Benyettou, A. Ramdas Nair, Y. Dho, T. Prakasam, R. Pasricha, J. Whelan,
[1] C. Battistella, H.A. Klok, Controlling and monitoring intracellular delivery of
H. Traboulsi, J. Mazher, K.C. Sadler, A. Trabolsi, Aqueous synthesis of
anticancer polymer nanomedicines, Macromol. Biosci. 17 (10) (2017), 1700022.
triphenylphosphine-modified gold nanoparticles for synergistic in vitro and in
[2] W. Kreyling, Nanomedicine : an ESF-European Medical Councils (EMRC) forword
vivo photothermal chemotherapy, Chemistry 26 (23) (2020) 5270–5279.
look report, Nanomedicine 2005 (2005) 5–25.
[29] W. Sheng, A.H. Alhasan, G. DiBernardo, K.M. Almutairi, J.P. Rubin, B.
[3] Y. Li, H. Lu, S. Liang, S. Xu, Dual stable nanomedicines prepared by cisplatin-
E. DiBernardo, A. Almutairi, Gold nanoparticle-assisted selective
crosslinked camptothecin prodrug micelles for effective drug delivery, ACS Appl.
photothermolysis of adipose tissue (NanoLipo), plastic and reconstructive
Mater. Interfaces 11 (23) (2019) 20649–20659.
surgery, Glob. Open J. 2 (12) (2014) e283.
[4] X. Wang, C. Ho, Y. Tsatskis, J. Law, Z. Zhang, M. Zhu, C. Dai, F. Wang, M. Tan,
[30] A.V. Singh, Y. Alapan, T. Jahnke, P. Laux, A. Luch, A. Aghakhani, S. Kharratian,
S. Hopyan, H. McNeill, Y. Sun, Intracellular manipulation and measurement with
M.C. Onbasli, J. Bill, M. Sitti, Seed-mediated synthesis of plasmonic gold
multipole magnetic tweezers, Sci. Robot. 4 (28) (2019) eaav6180.
nanoribbons using cancer cells for hyperthermia applications, J. Mater. Chem. B 6
[5] U. Jeong, T. Herricks, E. Shahar, Y. Xia, Amorphous Se: a new platform for
(46) (2018) 7573–7581.
synthesizing superparamagnetic colloids with controllable surfaces, J. Am. Chem.
[31] A.V. Singh, H. Jungnickel, L. Leibrock, J. Tentschert, P. Reichardt, A. Katz,
Soc. 127 (4) (2005) 1098–1099.
P. Laux, A. Luch, ToF-SIMS 3D imaging unveils important insights on the cellular
[6] T. Kaehler, Nanotechnology: basic concepts and definitions, Clin. Chem. 40 (9)
microenvironment during biomineralization of gold nanostructures, Sci. Rep. 10
(1994) 1797–1799.
(1) (2020) 261.
[7] S. Shao, Q. Zhou, J. Si, J. Tang, X. Liu, M. Wang, J. Gao, K. Wang, R. Xu, Y. Shen,
[32] A.V. Singh, M. Batuwangala, R. Mundra, K. Mehta, S. Patke, E. Falletta, R. Patil,
A non-cytotoxic dendrimer with innate and potent anticancer and anti-metastatic
W.N. Gade, Biomineralized anisotropic gold microplate-macrophage interactions
activities, Nat. Biomed. Eng. 1 (9) (2017) 745–757.
reveal frustrated phagocytosis-like phenomenon: a novel paclitaxel drug delivery
[8] A.V. Singh, M.H.D. Ansari, P. Laux, A. Luch, Micro-nanorobots: important
vehicle, ACS Appl. Mater. Interfaces 6 (16) (2014) 14679–14689.
considerations when developing novel drug delivery platforms, Expert Opin.
[33] V. Singh, V. Kumar, S. Kashyap, A.V. Singh, V. Kishore, M. Sitti, P.S. Saxena,
Drug Deliv. 16 (11) (2019) 1259–1275.
A. Srivastava, Graphene oxide synergistically enhances antibiotic efficacy in
[9] Y. Li, H. Lu, S. Liang, S. Xu, Dual stable nanomedicines prepared by cisplatin-
Vancomycin-ResistantStaphylococcus aureus, ACS Appl. Bio. Mater. 2 (3) (2019)
crosslinked camptothecin prodrug micelles for effective drug delivery, ACS Appl.
1148–1157.
Mater. Interfaces 11 (23) (2019) 20649–20659.
[34] M. Saravanan, H. Vahidi, D. Medina Cruz, A. Vernet-Crua, E. Mostafavi,
[10] B. Liu, X. Deng, Z. Xie, Z. Cheng, P. Yang, J. Lin, Thiol-Ene click reaction as a
R. Stelmach, T.J. Webster, M.A. Mahjoub, M. Rashedi, H. Barabadi, Emerging
facile and general approach for surface functionalization of colloidal
antineoplastic biogenic gold nanomaterials for breast cancer therapeutics: a
nanocrystals, Adv. Mater. (Deerfield Beach, Fla.) 29 (36) (2017).
systematic review, Int. J. Nanomed. 15 (2020) 3577–3595.
[11] Z. Chai, D. Ran, L. Lu, C. Zhan, H. Ruan, X. Hu, C. Xie, K. Jiang, J. Li, J. Zhou,
[35] V. Singh, S. Kashyap, U. Yadav, A. Srivastava, A.V. Singh, R.K. Singh, S.K. Singh,
J. Wang, Y. Zhang, R.H. Fang, L. Zhang, W. Lu, Ligand-modified cell membrane
P.S. Saxena, Nitrogen doped carbon quantum dots demonstrate no toxicity under
enables the targeted delivery of drug nanocrystals to glioma, ACS Nano 13 (5)
in vitro conditions in a cervical cell line and in vivo in Swiss albino mice, Toxicol.
(2019) 5591–5601.
Res. (Camb) 8 (3) (2019) 395–406.
[12] Y. Wang, K. Zhang, X. Qin, T. Li, J. Qiu, T. Yin, J. Huang, S. McGinty, G. Pontrelli,
[36] A.V. Singh, T. Jahnke, Y. Xiao, S. Wang, Y. Yu, H. David, G. Richter, P. Laux,
J. Ren, Q. Wang, W. Wu, G. Wang, Biomimetic nanotherapies: red blood cell
A. Luch, A. Srivastava, P.S. Saxena, J. Bill, M. Sitti, Peptide-induced
based core-shell structured nanocomplexes for atherosclerosis management, Adv.
biomineralization of tin oxide (SnO(2)) nanoparticles for antibacterial
Sci. (Weinh) 6 (12) (2019), 1900172.
applications, J. Nanosci. Nanotechnol. 19 (9) (2019) 5674–5686.
[13] J. Wang, P. Li, Y. Yu, Y. Fu, H. Jiang, M. Lu, Z. Sun, S. Jiang, L. Lu, M.X. Wu,
[37] R. Shahbazi, G. Sghia-Hughes, J.L. Reid, S. Kubek, K.G. Haworth, O. Humbert, H.
Pulmonary surfactant–biomimetic nanoparticles potentiate heterosubtypic
P. Kiem, J.E. Adair, Targeted homology-directed repair in blood stem and
influenza immunity, Science 367 (6480) (2020) eaau0810.
progenitor cells with CRISPR nanoformulations, Nat. Mater. 18 (10) (2019)
[14] P.N. Durfee, Y.S. Lin, D.R. Dunphy, A.J. Muniz, K.S. Butler, K.R. Humphrey, A.
1124–1132.
J. Lokke, J.O. Agola, S.S. Chou, I.M. Chen, W. Wharton, J.L. Townson, C.
[38] Y. Xue, B. Dong, X. Liu, F. Wang, J. Yang, D. Liu, Using selenium-conjugated
L. Willman, C.J. Brinker, Mesoporous silica nanoparticle-supported lipid bilayers
polyethylene glycol to enhance the stability of gold nanoparticles in biologically
(Protocells) for active targeting and delivery to individual leukemia cells, ACS
relevant samples, Sci. China-Chem. 62 (2019) 280–286.
Nano 10 (9) (2016) 8325–8345.
[39] S. Zhang, C. Chen, C. Xue, D. Chang, H. Xu, B.J. Salena, Y. Li, Z.S. Wu, Ribbon of
[15] P. Ghasemiyeh, S. Mohammadi-Samani, Solid lipid nanoparticles and
DNA lattice on gold nanoparticles for selective drug delivery to cancer cells,
nanostructured lipid carriers as novel drug delivery systems: applications,
Angew. Chemie 59 (34) (2020) 14584–14592.
advantages and disadvantages, Res. Pharm. Sci. 13 (4) (2018) 288–303.
[40] S.-W. Liu, L. Wang, M. Lin, Y. Liu, L.-N. Zhang, H. Zhang, Tumor photothermal
[16] M. Yu, W. Song, F. Tian, Z. Dai, Q. Zhu, E. Ahmad, S. Guo, C. Zhu, H. Zhong,
therapy employing photothermal inorganic nanoparticles/polymers
Y. Yuan, T. Zhang, X. Yi, X. Shi, Y. Gan, H. Gao, Temperature- and rigidity-
nanocomposites, Chin. J. Polym. Sci. 37 (2) (2019) 115–128.
mediated rapid transport of lipid nanovesicles in hydrogels, Proc. Natl. Acad. Sci.
[41] T. Chen, W. Liu, S. Xiong, D. Li, S. Fang, Z. Wu, Q. Wang, X. Chen, Nanoparticles
U.S.A. 116 (12) (2019) 5362–5369.
mediating the sustained Puerarin release facilitate improved brain delivery to
[17] M.P. Monopoli, C. Aberg, A. Salvati, K.A. Dawson, Biomolecular coronas provide
treat Parkinson’s disease, ACS Appl. Mater. Interfaces 11 (48) (2019)
the biological identity of nanosized materials, Nat. Nanotechnol. 7 (12) (2012)
45276–45289.
779–786.
[42] M. Wang, Y. Zhai, H. Ye, Q. Lv, B. Sun, C. Luo, Q. Jiang, H. Zhang, Y. Xu, Y. Jing,
[18] Z. Zhang, J. Guan, Z. Jiang, Y. Yang, J. Liu, W. Hua, Y. Mao, C. Li, W. Lu, J. Qian,
L. Huang, J. Sun, Z. He, High Co-loading capacity and stimuli-responsive release
C. Zhan, Brain-targeted drug delivery by manipulating protein corona functions,
based on cascade reaction of self-destructive polymer for improved chemo-
Nat. Commun. 10 (1) (2019) 3561.
photodynamic therapy, ACS Nano 13 (6) (2019) 7010–7023.
[19] S.F. Hansen, A. Lennquist, Carbon nanotubes added to the SIN List as a
[43] S. Wannasarit, S. Wang, P. Figueiredo, C. Trujillo, W. Li, A virus-mimicking pH-
nanomaterial of very High Concern, Nat. Nanotechnol. 15 (1) (2020) 3–4.
Responsive acetalated dextran-based membrane-active polymeric nanoparticle
[20] X. Yang, D. Wang, J. Zhu, L. Xue, C. Ou, W. Wang, M. Lu, X. Song, X. Dong,
for intracellular delivery of antitumor therapeutics, Adv. Funct. Mater. 29 (51)
Functional black phosphorus nanosheets for mitochondria-targeting
(2019), 1905352.
photothermal/photodynamic synergistic cancer therapy, Chem. Sci. 10 (13)
[44] Z. Wu, J. Troll, H.H. Jeong, Q. Wei, M. Stang, F. Ziemssen, Z. Wang, M. Dong,
(2019) 3779–3785.
S. Schnichels, T. Qiu, P. Fischer, A swarm of slippery micropropellers penetrates
[21] H. Wang, X. Yang, W. Shao, S. Chen, J. Xie, X. Zhang, J. Wang, Y. Xie, Ultrathin
the vitreous body of the eye, Sci. Adv. 4 (11) (2018) eaat4388.
black phosphorus nanosheets for efficient singlet oxygen generation, J. Am.
Chem. Soc. 137 (35) (2015) 11376–11382.

8
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

[45] F. Soto, J. Wang, R. Ahmed, U. Demirci, Medical micro/nanorobots in precision stabilized polymeric micelles potentiate docetaxel therapy in advanced-stage
medicine, Adv. Sci. (Weinh) 7 (21) (2020), 2002203. gastrointestinal cancer, Biomaterials 266 (2021), 120432.
[46] Z. Wu, L. Li, Y. Yang, P. Hu, Y. Li, S.Y. Yang, L.V. Wang, W. Gao, A microrobotic [73] B. Sun, C. Luo, X. Zhang, M. Guo, M. Sun, H. Yu, Q. Chen, W. Yang, M. Wang,
system guided by photoacoustic computed tomography for targeted navigation in S. Zuo, P. Chen, Q. Kan, H. Zhang, Y. Wang, Z. He, J. Sun, Probing the impact of
intestines in vivo, Sci. Robot 4 (32) (2019) eaax0613. sulfur/selenium/carbon linkages on prodrug nanoassemblies for cancer therapy,
[47] D. Wang, C. Gao, C. Zhou, Z. Lin, Q. He, Leukocyte membrane-coated liquid metal Nat. Commun. 10 (1) (2019) 3211.
nanoswimmers for actively targeted delivery and synergistic chemophotothermal [74] Z. Tian, L. Xu, Q. Chen, R. Feng, H. Lu, H. Tan, J. Kang, Y. Wang, H. Yan,
therapy, Research 2020 (2020), 3676954. Treatment of surgical brain injury by immune tolerance induced by peripheral
[48] Y. Tu, F. Peng, J.M. Heuvelmans, S. Liu, R.J.M. Nolte, D.A. Wilson, Motion intravenous injection of biotargeting nanoparticles loaded with brain antigens,
control of polymeric nanomotors based on host-guest interactions, Angew. Front. Immunol. 10 (2019) 743.
Chemie 58 (26) (2019) 8687–8691. [75] W. Jiang, L. Yin, H. Chen, A.V. Paschall, L. Zhang, W. Fu, W. Zhang, T. Todd, K.
[49] D.D. Jin, Y.U. Jiangfan, T.Y. Huang, H.L. Duan, L. Zhang, Magnetic micro-/ S. Yu, S. Zhou, Z. Zhen, M. Butler, L. Yao, F. Zhang, Y. Shen, Z. Li, A. Yin, H. Yin,
nanoscale swimmers: current status and potential applications, Chin. ence Bull. X. Wang, F.Y. Avci, X. Yu, J. Xie, NaCl nanoparticles as a cancer therapeutic, Adv.
62 (2–3) (2017) 136–151. Mater. 31 (46) (2019), e1904058.
[50] A.V. Singh, M.H.D. Ansari, M. Mahajan, S. Srivastava, S. Kashyap, P. Dwivedi, [76] Y. Wang, T. Yang, Q. He, Strategies for engineering advanced nanomedicines for
V. Pandit, U. Katha, Sperm cell driven microrobots-emerging opportunities and gas therapy of cancer, Sci. Rev. 7 (9) (2020) 1485–1512.
challenges for biologically inspired robotic design, Micromachines (Basel) 11 (4) [77] J. Galon, D. Bruni, Approaches to treat immune hot, altered and cold tumours
(2020) 448. with combination immunotherapies, Nat. Rev. Drug Discov. 18 (3) (2019)
[51] J.F. Liu, Z. Lan, C. Ferrari, J.M. Stein, E. Higbee-Dempsey, L. Yan, 197–218.
A. Amirshaghaghi, Z. Cheng, D. Issadore, A. Tsourkas, Use of oppositely polarized [78] X. Li, X. Wang, A. Ito, N.M. Tsuji, A nanoscale metal organic frameworks-based
external magnets to improve the accumulation and penetration of magnetic vaccine synergises with PD-1 blockade to potentiate anti-tumour immunity, Nat.
nanocarriers into solid tumors, ACS Nano 14 (1) (2020) 142–152. Commun. 11 (1) (2020) 3858.
[52] M. Chen, J. Wu, P. Ning, J. Wang, Z. Ma, L. Huang, G.R. Plaza, Y. Shen, C. Xu, [79] C.W. Ng, J. Li, K. Pu, Recent progresses in phototherapy-synergized cancer
Y. Han, M.S. Lesniak, Z. Liu, Y. Cheng, Remote Control of Mechanical Forces via immunotherapy, Adv. Funct. Mater. 28 (46) (2018) 1804688.1–1804688.20.
Mitochondrial-Targeted Magnetic Nanospinners for Efficient Cancer Treatment, [80] B. Zhou, J. Song, M. Wang, X. Wang, J. Wang, E.W. Howard, F. Zhou, J. Qu, W.
Small 16 (3) (2020), e1905424. R. Chen, BSA-bioinspired gold nanorods loaded with immunoadjuvant for the
[53] Z. Zhou, Z. Shen, X. Chen, Tale of two magnets: an advanced magnetic targeting treatment of melanoma by combined photothermal therapy and immunotherapy,
system, ACS Nano 14 (1) (2020) 7–11. Nanoscale 10 (46) (2018) 21640–21647.
[54] H.W. Huang, F.E. Uslu, P. Katsamba, E. Lauga, M.S. Sakar, B.J. Nelson, Adaptive [81] H. Huang, C.T. Jiang, S. Shen, A. Liu, Y.J. Gan, Q.S. Tong, S.B. Chen, Z.X. Gao, J.
locomotion of artificial microswimmers, Sci. Adv. 5 (1) (2019) eaau1532. Z. Du, J. Cao, J. Wang, Nanoenabled reversal of IDO1-mediated
[55] X. Zhong, X. Wang, G. Zhan, Ya. Tang, Y. Yao, Z. Dong, L. Hou, H. Zhao, S. Zeng, immunosuppression synergizes with immunogenic chemotherapy for improved
J. Hu, L. Cheng, X. Yang, NaCeF4:Gd,Tb Scintillator as an X-ray responsive cancer therapy, Nano Lett. 19 (8) (2019) 5356–5365.
photosensitizer for multimodal imaging-guided synchronous Radio/ [82] L. Tian, Y. Wang, L. Sun, J. Xu, Y. Chao, K. Yang, S. Wang, Z. Liu, Cerenkov
Radiodynamic therapy, Nano Lett. 19 (11) (2019) 8234–8244. luminescence-induced NO release from 32P-Labeled ZnFe(CN)5NO nanosheets to
[56] V. Ntziachristos, J. Ripoll, L.V. Wang, R. Weissleder, Looking and listening to enhance radioisotope-immunotherapy, Matter 1 (4) (2019) 1061–1076.
light: the evolution of whole-body photonic imaging, Nat. Biotechnol. 23 (3) [83] D. Cui, J. Li, X. Zhao, K. Pu, R. Zhang, Semiconducting polymer nanoreporters for
(2005) 313–320. near-infrared chemiluminescence imaging of immunoactivation, Adv. Mater. 32
[57] X. Yan, Q. Zhou, M. Vincent, Y. Deng, J. Yu, J. Xu, T. Xu, T. Tang, L. Bian, Y.-X. (6) (2020), e1906314.
J. Wang, K. Kostarelos, L. Zhang, Multifunctional biohybrid magnetite [84] X. Li, L. Liu, S. Li, Y. Wan, J.X. Chen, S. Tian, Z. Huang, Y.F. Xiao, X. Cui, C. Xiang,
microrobots for imaging-guided therapy, Sci. Robot. 2 (12) (2017) eaaq1155. Q. Tan, X.H. Zhang, W. Guo, X.J. Liang, C.S. Lee, Biodegradable pi-conjugated
[58] Z. Wu, J. Troll, H.-H. Jeong, Q. Wei, M. Stang, F. Ziemssen, Z. Wang, M. Dong, oligomer nanoparticles with high photothermal conversion efficiency for Cancer
S. Schnichels, T. Qiu, P. Fischer, A swarm of slippery micropropellers penetrates theranostics, ACS Nano 13 (11) (2019) 12901–12911.
the vitreous body of the eye, Sci. Adv. 4 (11) (2018) eaat4388. [85] R. Xing, Q. Zou, C. Yuan, L. Zhao, R. Chang, X. Yan, Self-assembling endogenous
[59] Z. Guler Gokce, S. Zuhal Birol, T. Eren, S. Ercelen Ceylan, Biophysical biliverdin as a versatile near-infrared photothermal nanoagent for cancer
characterization of quaternary pyridinium functionalized polynorbornenes for theranostics, Adv. Mater. 31 (16) (2019), e1900822.
DNA complexation and their cellular interactions, Biopolymers 107 (4) (2017) [86] C. Liu, S. Zhang, J. Li, J. Wei, K. Mullen, M. Yin, A. Water-Soluble, NIR-absorbing
e23005. quaterrylenediimide chromophore for photoacoustic imaging and efficient
[60] A. Vikram Singh, P. Laux, A. Luch, S. Balkrishnan, S. Prasad Dakua, Bottom-UP photothermal cancer therapy, Angew. Chemie 58 (6) (2019) 1638–1642.
assembly of nanorobots: extending synthetic biology to complex material design, [87] J. Wang, J. Sun, Y. Wang, T. Chou, Q. Zhang, B. Zhang, L. Ren, H. Wang, Gold
Front. Nanosci. Nanotechnol. 5 (1) (2019) 1–2. nanoframeworks with mesopores for Raman–photoacoustic imaging and photo-
[61] J.H. Reif, DNA robots sort as they walk, Science 357 (6356) (2017) 1095–1096. chemo tumor therapy in the second near-infrared biowindow, Adv. Funct. Mater.
[62] S. Ranallo, C. Prévost-Tremblay, A. Idili, A. Vallée-Bélisle, F. Ricci, Antibody- 30 (9) (2020), 1908825.
powered nucleic acid release using a DNA-based nanomachine, Nat. Commun. 8 [88] C. Zhang, W. Sun, Y. Wang, F. Xu, J. Qu, J. Xia, M. Shen, X. Shi, Gd-/CuS-Loaded
(1) (2017) 15150. functional nanogels for MR/PA imaging-guided tumor-targeted photothermal
[63] Z. Di, J. Zhao, H. Chu, W. Xue, Y. Zhao, L. Li, An acidic-microenvironment-Driven therapy, ACS Appl. Mater. Interfaces 12 (8) (2020) 9107–9117.
DNA nanomachine enables specific ATP imaging in the extracellular milieu of [89] H. Zhang, W. Zeng, C. Pan, L. Feng, M. Ou, X. Zeng, X. Liang, M. Wu, X. Ji, L. Mei,
tumor, Adv. Mater. (Deerfield Beach, Fla.) 31 (33) (2019), e1901885. SnTe@MnO2-SP nanosheet–based intelligent nanoplatform for second near-
[64] S. Li, Q. Jiang, S. Liu, Y. Zhang, Y. Tian, C. Song, J. Wang, Y. Zou, G.J. Anderson, infrared light–mediated cancer theranostics, Adv. Funct. Mater. 29 (37) (2019),
J.Y. Han, Y. Chang, Y. Liu, C. Zhang, L. Chen, G. Zhou, G. Nie, H. Yan, B. Ding, 1903791.
Y. Zhao, A DNA nanorobot functions as a cancer therapeutic in response to a [90] C. Liu, S. Zhang, J. Li, J. Wei, K. Müllen, M. Yin, A. Water-Soluble, NIR-absorbing
molecular trigger in vivo, Nat. Biotechnol. 36 (3) (2018) 258–264. quaterrylenediimide chromophore for photoacoustic imaging and efficient
[65] W. Ma, Y. Zhan, Y. Zhang, X. Shao, X. Xie, C. Mao, W. Cui, Q. Li, J. Shi, J. Li, photothermal cancer therapy, Angew. Chemie Int. Ed. 58 (6) (2019) 1638–1642.
C. Fan, Y. Lin, An intelligent DNA nanorobot with in vitro enhanced protein [91] H. Peng, J. Tang, R. Zheng, G. Guo, A. Dong, Y. Wang, W. Yang, Nuclear-targeted
lysosomal degradation of HER2, Nano Lett. 19 (7) (2019) 4505–4517. multifunctional magnetic nanoparticles for photothermal therapy, Adv. Healthc.
[66] A.V. Singh, V. Kishore, G. Santomauro, O. Yasa, J. Bill, M. Sitti, Mechanical Mater. 6 (7) (2017), 1601289.
coupling of puller and pusher active microswimmers influences motility, [92] Z. Xie, T. Fan, J. An, W. Choi, Y. Duo, Y. Ge, B. Zhang, G. Nie, N. Xie, T. Zheng,
Langmuir 36 (19) (2020) 5435–5443. Y. Chen, H. Zhang, J.S. Kim, Emerging combination strategies with phototherapy
[67] H. Wang, M. Pumera, Micro/nanomachines and living biosystems: from simple in cancer nanomedicine, Chem. Soc. Rev. 49 (22) (2020) 8065–8087.
interactions to microcyborgs, Adv. Funct. Mater. 28 (25) (2018), 1705421. [93] D. Cui, J. Huang, X. Zhen, J. Li, Y. Jiang, K. Pu, A semiconducting polymer nano-
[68] S.J. Park, M. Gazzola, K.S. Park, S. Park, V. Di Santo, E.L. Blevins, J.U. Lind, P. prodrug for hypoxia-activated photodynamic cancer therapy, Angew. Chemie 58
H. Campbell, S. Dauth, A.K. Capulli, F.S. Pasqualini, S. Ahn, A. Cho, H. Yuan, B. (18) (2019) 5920–5924.
M. Maoz, R. Vijaykumar, J.W. Choi, K. Deisseroth, G.V. Lauder, L. Mahadevan, K. [94] S. Chakrabortty, B.K. Agrawalla, A. Stumper, N.M. Vegi, S. Fischer, C. Reichardt,
K. Parker, Phototactic guidance of a tissue-engineered soft-robotic ray, Science M. Kogler, B. Dietzek, M. Feuring-Buske, C. Buske, S. Rau, T. Weil, Mitochondria
353 (6295) (2016) 158–162. targeted protein-ruthenium photosensitizer for efficient photodynamic
[69] J. Shao, M. Xuan, H. Zhang, X. Lin, Z. Wu, Q. He, Chemotaxis-guided hybrid applications, J. Am. Chem. Soc. 139 (6) (2017) 2512–2519.
neutrophil micromotors for targeted drug transport, Angew. Chem. Int. Ed. Engl. [95] L.B. Josefsen, R.W. Boyle, Unique diagnostic and therapeutic roles of porphyrins
56 (42) (2017) 12935–12939. and Phthalocyanines in photodynamic therapy, imaging and theranostics,
[70] A.V. Singh, M.H.D. Ansari, P. Laux, A. Luch, Micro-nanorobots: important Theranostics 2 (9) (2012) 916–966.
considerations when developing novel drug delivery platforms, Expert Opin. Drug [96] W. Deng, K.J. McKelvey, A. Guller, A. Fayzullin, J.M. Campbell, S. Clement,
Deliv. 16 (11) (2019) 1259–1275. A. Habibalahi, Z. Wargocka, L. Liang, C. Shen, V.M. Howell, A.F. Engel, E.
[71] A. Gothwal, I. Khan, U. Gupta, Polymeric micelles: recent advancements in the M. Goldys, Application of mitochondrially targeted nanoconstructs to
delivery of anticancer drugs, Pharm. Res. 33 (1) (2016) 18–39. neoadjuvant X-ray-Induced photodynamic therapy for rectal cancer, ACS Cent.
[72] C. Liang, X. Bai, C. Qi, Q. Sun, X. Han, T. Lan, H. Zhang, X. Zheng, R. Liang, Sci. 6 (5) (2020) 715–726.
J. Jiao, Z. Zheng, J. Fang, P. Lei, Y. Wang, D. Mockel, J.M. Metselaar, G. Storm, [97] W. Deng, W. Chen, S. Clement, A. Guller, Z. Zhao, A. Engel, E.M. Goldys,
W.E. Hennink, F. Kiessling, H. Wei, T. Lammers, Y. Shi, B. Wei, Pi electron- Controlled gene and drug release from a liposomal delivery platform triggered by
X-ray radiation, Nat. Commun. 9 (1) (2018) 2713.

9
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

[98] W. Zhang, X. Hu, Q. Shen, D. Xing, Mitochondria-specific drug release and [120] S. Li, Z. Zhang, W.-F. Lai, L. Cui, X. Zhu, How to overcome the side effects of
reactive oxygen species burst induced by polyprodrug nanoreactors can enhance tumor immunotherapy, Biomed. Pharmacother. 130 (2020), 110639.
chemotherapy, Nat. Commun. 10 (1) (2019) 1704. [121] M. Miragoli, P. Ceriotti, M. Iafisco, M. Vacchiano, N. Salvarani, A. Alogna,
[99] D. Zhong, W. Li, Y. Qi, J. He, M. Zhou, Photosynthetic biohybrid nanoswimmers P. Carullo, G.B. Ramirez-Rodriguez, T. Patricio, L.D. Esposti, F. Rossi,
system to alleviate tumor hypoxia for FL/PA/MR Imaging-guided enhanced F. Ravanetti, S. Pinelli, R. Alinovi, M. Erreni, S. Rossi, G. Condorelli, H. Post,
radio-photodynamic synergetic therapy, Adv. Funct. Mater. 30 (17) (2020), A. Tampieri, D. Catalucci, Inhalation of peptide-loaded nanoparticles improves
1910395. heart failure, Sci. Transl. Med. 10 (424) (2018) eaan6205.
[100] D.A. Srinivasarao, G. Lohiya, D.S. Katti, Fundamentals, challenges, and [122] W.L. Chew, M. Tabebordbar, J.K. Cheng, P. Mali, E.Y. Wu, A.H. Ng, K. Zhu, A.
nanomedicine-based solutions for ocular diseases, Wiley interdisciplinary J. Wagers, G.M. Church, A multifunctional AAV-CRISPR-Cas9 and its host
reviews, Nanomed. Nanobiotechnol. 11 (4) (2019) e1548. response, Nat. Methods 13 (10) (2016) 868–874.
[101] Y. Ye, J. He, Y. Qiao, Y. Qi, H. Zhang, H.A. Santos, D. Zhong, W. Li, S. Hua, [123] Y. Lyu, S. He, J. Li, Y. Jiang, H. Sun, Y. Miao, K. Pu, A photolabile semiconducting
W. Wang, A. Grzybowski, K. Yao, M. Zhou, Mild temperature photothermal polymer nanotransducer for near-infrared regulation of CRISPR/Cas9 gene
assisted anti-bacterial and anti-inflammatory nanosystem for synergistic editing, Angew. Chemie 58 (50) (2019) 18197–18201.
treatment of post-cataract surgery endophthalmitis, Theranostics 10 (19) (2020) [124] H. Yin, C.Q. Song, S. Suresh, Q. Wu, S. Walsh, L.H. Rhym, E. Mintzer, M.
8541–8557. F. Bolukbasi, L.J. Zhu, K. Kauffman, H. Mou, A. Oberholzer, J. Ding, S.Y. Kwan, R.
[102] Y. Wang, C.H. Liu, T. Ji, M. Mehta, W. Wang, E. Marino, J. Chen, D.S. Kohane, L. Bogorad, T. Zatsepin, V. Koteliansky, S.A. Wolfe, W. Xue, R. Langer, D.
Intravenous treatment of choroidal neovascularization by photo-targeted G. Anderson, Structure-guided chemical modification of guide RNA enables
nanoparticles, Nat. Commun. 10 (1) (2019) 804. potent non-viral in vivo genome editing, Nat. Biotechnol. 35 (12) (2017)
[103] P. Jelinkova, A. Mazumdar, V.P. Sur, S. Kociova, K. Dolezelikova, A.M.J. Jimenez, 1179–1187.
Z. Koudelkova, P.K. Mishra, K. Smerkova, Z. Heger, M. Vaculovicova, A. Moulick, [125] G. Chen, A.A. Abdeen, Y. Wang, P.K. Shahi, S. Robertson, R. Xie, M. Suzuki, B.
V. Adam, Nanoparticle-drug conjugates treating bacterial infections, J. Control. R. Pattnaik, K. Saha, S. Gong, A biodegradable nanocapsule delivers a Cas9
Release 307 (2019) 166–185. ribonucleoprotein complex for in vivo genome editing, Nat. Nanotechnol. 14 (10)
[104] Y. Qiao, Y. Ping, H. Zhang, B. Zhou, F. Liu, Y. Yu, T. Xie, W. Li, D. Zhong, (2019) 974–980.
Y. Zhang, K. Yao, H.A. Santos, M. Zhou, Laser-activatable CuS nanodots to treat [126] A.V. Singh, M.H. Dad Ansari, C.B. Dayan, J. Giltinan, S. Wang, Y. Yu, V. Kishore,
multidrug-resistant Bacteria and release copper ion to accelerate healing of P. Laux, A. Luch, M. Sitti, Multifunctional magnetic hairbot for untethered
infected chronic nonhealing wounds, ACS Appl. Mater. Interfaces 11 (4) (2019) osteogenesis, ultrasound contrast imaging and drug delivery, Biomaterials 219
3809–3822. (2019), 119394.
[105] S. Yu, G. Li, P. Zhao, Q. Cheng, Q. He, D. Ma, W. Xue, NIR-laser-controlled [127] A.V. Singh, Z. Hosseinidoust, B.W. Park, O. Yasa, M. Sitti, Microemulsion-based
hydrogen-releasing PdH nanohydride for synergistic hydrogen-photothermal Soft Bacteria-Driven microswimmers for active cargo delivery, ACS Nano 11 (10)
antibacterial and wound-healing therapies, Adv. Funct. Mater. 29 (50) (2019), (2017) 9759–9769.
1905697. [128] T. Wu, J. Liu, M. Liu, S. Liu, S. Zhao, R. Tian, D. Wei, Y. Liu, Y. Zhao, H. Xiao,
[106] M. Liang, M. Zhang, S. Yu, Q. Wu, K. Ma, Y. Chen, X. Liu, C. Li, F. Wang, Silver- B. Ding, A nanobody-conjugated DNA nanoplatform for targeted platinum-drug
laden black phosphorus nanosheets for an efficient in vivo antimicrobial delivery, Angew. Chemie 58 (40) (2019) 14224–14228.
application, Small 16 (13) (2020), e1905938. [129] X. Han, K. Cheng, Y. Xu, Y. Wang, H. Min, Y. Zhang, X. Zhao, R. Zhao, G.
[107] S. Ma, S.M. Motevalli, J. Chen, M.Q. Xu, Y. Wang, J. Feng, Y. Qiu, D. Han, M. Fan, J. Anderson, L. Ren, G. Nie, Y. Li, Modularly designed peptide nanoprodrug
M. Ding, L. Fan, W. Guo, X.J. Liang, F. Cao, Precise theranostic nanomedicines for augments antitumor immunity of PD-L1 checkpoint blockade by targeting
inhibiting vulnerable atherosclerotic plaque progression through regulation of indoleamine 2,3-dioxygenase, J. Am. Chem. Soc. 142 (5) (2020) 2490–2496.
vascular smooth muscle cell phenotype switching, Theranostics 8 (13) (2018) [130] X. Hu, Z. Chai, L. Lu, H. Ruan, R. Wang, C. Zhan, C. Xie, J. Pan, M. Liu, H. Wang,
3693–3706. W. Lu, Bortezomib dendrimer prodrug-based nanoparticle system, Adv. Funct.
[108] Z. Wu, C. Chen, B. Zhang, L. Tang, W. Shi, D. Liao, G. Di, J.R. Davis, H. Wang, Mater. 29 (14) (2019), 1807941.
EGFP-EGF1-conjugated poly(lactic-co-glycolic acid) nanoparticles, a new [131] B. Sun, C. Luo, X. Zhang, M. Guo, J. Sun, Probing the impact of sulfur/selenium/
diagnostic tool and drug carrier for atherosclerosis, Int. J. Nanomed. 14 (2019) carbon linkages on prodrug nanoassemblies for cancer therapy, Nat. Commun. 10
2609–2618. (1) (2019) 3211.
[109] Y. Xue, X. Xu, X.Q. Zhang, O.C. Farokhzad, R. Langer, Preventing diet-induced [132] A.V. Singh, M.H.D. Ansari, D. Rosenkranz, R.S. Maharjan, F.L. Kriegel, K. Gandhi,
obesity in mice by adipose tissue transformation and angiogenesis using targeted A. Kanase, R. Singh, P. Laux, A. Luch, Artificial intelligence and machine learning
nanoparticles, Proc. Natl. Acad. Sci. U.S.A. 113 (20) (2016) 5552–5557. in computational nanotoxicology: unlocking and empowering nanomedicine,
[110] W. Sheng, W.J. Seare, B. DiBernardo, A.H. Alhasan, E. Cory, P. Chasan, R.L. Sah, Adv. Healthc. Mater. 9 (17) (2020), e1901862.
K.M. Almutairi, A. Almutairi, A single-blind study evaluating the efficacy of gold [133] A.V. Singh, D. Rosenkranz, M.H.D. Ansari, R. Singh, A. Kanase, S.P. Singh,
nanoparticle photothermal-assisted liposuction in an ex vivo human tissue model, B. Johnston, J. Tentschert, P. Laux, A. Luch, Artificial intelligence and machine
Aesthet. Surg. J. 38 (11) (2018) 1213–1224. learning empower advanced biomedical material design to toxicity prediction,
[111] T. Binderup, R. Duivenvoorden, F. Fay, M.M.T. van Leent, J. Malkus, S. Baxter, Adv. Intell. Syst. (2020), 2000084.
S. Ishino, Y. Zhao, B. Sanchez-Gaytan, A.J.P. Teunissen, Y.C.A. Frederico, J. Tang, [134] A.V. Singh, T. Jahnke, S. Wang, Y. Xiao, Y. Alapan, S. Kharratian, M.C. Onbasli,
G. Carlucci, S. Lyashchenko, C. Calcagno, N. Karakatsanis, G. Soultanidis, M. K. Kozielski, H. David, G. Richter, J. Bill, P. Laux, A. Luch, M. Sitti, Anisotropic
L. Senders, P.M. Robson, V. Mani, S. Ramachandran, M.E. Lobatto, B.A. Hutten, J. gold nanostructures: optimization via in silico modeling for hyperthermia, Acs
F. Granada, T. Reiner, F.K. Swirski, M. Nahrendorf, A. Kjaer, E.A. Fisher, Z. Appl. Nano Mater. 1 (11) (2018) 6205–6216.
A. Fayad, C. Perez-Medina, W.J.M. Mulder, Imaging-assisted [135] T. Hartung, Perspectives on in vitro to in vivo extrapolations, Appl. In Vitro
nanoimmunotherapy for atherosclerosis in multiple species, Sci. Transl. Med. 11 Toxicol. 4 (4) (2018) 305–316.
(506) (2019) eaaw7736. [136] K.M. Tsoi, S.A. MacParland, X.-Z. Ma, V.N. Spetzler, J. Echeverri, B. Ouyang, S.
[112] J. Ma, S. Zhang, J. Liu, F. Liu, F. Du, M. Li, A.T. Chen, Y. Bao, H.W. Suh, J. Avery, M. Fadel, E.A. Sykes, N. Goldaracena, J.M. Kaths, J.B. Conneely, B.A. Alman,
G. Deng, Y. Zhou, P. Wu, K. Sheth, H. Wang, J. Zhou, Targeted drug delivery to M. Selzner, M.A. Ostrowski, O.A. Adeyi, A. Zilman, I.D. McGilvray, W.C.W. Chan,
stroke via chemotactic recruitment of nanoparticles coated with membrane of Mechanism of hard-nanomaterial clearance by the liver, Nat. Mater. 15 (11)
engineered neural stem cells, Small 15 (35) (2019), e1902011. (2016) 1212–1221.
[113] T. Zhang, F. Li, Q. Xu, Q. Wang, X. Jiang, Z. Liang, H. Liao, X. Kong, J. Liu, H. Wu, [137] Y. Xue, B. Dong, X. Liu, F. Wang, J. Yang, D. Liu, Using selenium-conjugated
D. Zhang, C. An, L. Dong, Y. Lu, H. Cao, D. Kim, J. Sun, T. Hyeon, J. Gao, D. Ling, polyethylene glycol to enhance the stability of gold nanoparticles in biologically
Ferrimagnetic nanochains-based mesenchymal stem cell engineering for highly relevant samples, Sci. China Chem. 62 (2) (2018) 280–286.
efficient post-stroke recovery, Adv. Funct. Mater. 29 (24) (2019), 1900603. [138] J. Ma, S. Zhang, J. Liu, F. Liu, F. Du, M. Li, A.T. Chen, Y. Bao, H.W. Suh, J. Avery,
[114] A. Galstyan, J.L. Markman, E.S. Shatalova, A. Chiechi, A.J. Korman, R. Patil, G. Deng, Y. Zhou, P. Wu, K. Sheth, H. Wang, J. Zhou, Targeted drug delivery to
D. Klymyshyn, W.G. Tourtellotte, L.L. Israel, O. Braubach, V.A. Ljubimov, L. stroke via chemotactic recruitment of nanoparticles coated with membrane of
A. Mashouf, A. Ramesh, Z.B. Grodzinski, M.L. Penichet, K.L. Black, E. Holler, engineered neural stem cells, Small 15 (35) (2019), e1902011.
T. Sun, H. Ding, A.V. Ljubimov, J.Y. Ljubimova, Blood–brain barrier permeable [139] Y. Shi, T. Lammers, Combining nanomedicine and immunotherapy, Acc. Chem.
nano immunoconjugates induce local immune responses for glioma therapy, Nat. Res. 52 (6) (2019) 1543–1554.
Commun. 10 (1) (2019) 3850. [140] G. Lu, H. Luo, X. Zhu, Targeting the GRP78 pathway for cancer therapy, Front.
[115] X. Liang, D. Li, S. Leng, X. Zhu, RNA-based pharmacotherapy for tumors: from Med. 7 (2020) 351.
bench to clinic and back, Biomed. Pharmacother. 125 (2020), 109997. [141] K. Li, H. Luo, H. Luo, X. Zhu, Clinical and prognostic pan-cancer analysis of m6A
[116] F. Tian, X. Lin, R.P. Valle, Y.Y. Zuo, N. Gu, Poly(amidoamine) Dendrimer as a RNA methylation regulators in four types of endocrine system tumors, Aging
respiratory nanocarrier: insights from experiments and molecular dynamics (Albany NY) 12 (2020) 1–13.
simulations, Langmuir 35 (15) (2019) 5364–5371. [142] B. Guo, D. Li, L. Du, X. Zhu, piRNAs: biogenesis and their potential roles in cancer,
[117] J. Wang, P. Li, Y. Yu, Y. Fu, H. Jiang, M. Lu, Z. Sun, S. Jiang, L. Lu, M.X. Wu, Cancer Metastasis Rev. 39 (2) (2020) 567–575.
Pulmonary surfactant-biomimetic nanoparticles potentiate heterosubtypic [143] J. Liu, D. Li, H. Luo, X. Zhu, Circular RNAs: the star molecules in cancer, Mol.
influenza immunity, Science 367 (6480) (2020) eaau0810. Aspects Med. 70 (2019) 141–152.
[118] Q. Yin, Z. Pei, H. Wang, Y. Zhao, Cyclosporine A-nanoparticles enhance the [144] D. Huo, J. Zhu, G. Chen, Q. Chen, C. Zhang, X. Luo, W. Jiang, X. Jiang, Z. Gu,
therapeutic benefit of adipose tissue-derived stem cell transplantation in a swine Y. Hu, Eradication of unresectable liver metastasis through induction of tumour
myocardial infarction model, Int. J. Nanomed. 9 (2014) 17–26. specific energy depletion, Nat. Commun. 10 (1) (2019) 3051.
[119] S. Tan, D. Li, X. Zhu, Cancer immunotherapy: pros, cons and beyond, Biomed. [145] K. Li, H. Luo, L. Huang, H. Luo, X. Zhu, Microsatellite instability: a review of what
Pharmacother. 124 (2020), 109821. the oncologist should know, Cancer Cell Int. 20 (2020) 16.

10
W. Lu et al. Biomedicine & Pharmacotherapy 134 (2021) 111103

[146] X. Zhu, H. Luo, Y. Xu, Transcriptome analysis reveals an important candidate [151] Z. Zou, T. Tao, H. Li, X. Zhu, mTOR signaling pathway and mTOR inhibitors in
gene involved in both nodal metastasis and prognosis in lung adenocarcinoma, cancer: progress and challenges, Cell Biosci. 10 (2020) 31.
Cell Biosci. 9 (2019) 92. [152] Z. Tang, D. Li, S. Hou, X. Zhu, The cancer exosomes: clinical implications,
[147] X. Dong, H.J. Liu, H.Y. Feng, S.C. Yang, X.L. Liu, X. Lai, Q. Lu, J.F. Lovell, H. applications and challenges, Int. J. Cancer 146 (11) (2020) 2946–2959.
Z. Chen, C. Fang, Enhanced drug delivery by nanoscale integration of a nitric [153] L. Duan, L. Yang, J. Jin, F. Yang, D. Liu, K. Hu, Q. Wang, Y. Yue, N. Gu, Micro/
oxide donor to induce tumor collagen depletion, Nano Lett. 19 (2) (2019) nano-bubble-assisted ultrasound to enhance the EPR effect and potential
997–1008. theranostic applications, Theranostics 10 (2) (2020) 462–483.
[148] L.P. Wu, D. Ahmadvand, J. Su, A. Hall, X. Tan, Z.S. Farhangrazi, S.M. Moghimi, [154] T. Lammers, Smart drug delivery systems: back to the future vs. clinical reality,
Crossing the blood-brain-barrier with nanoligand drug carriers self-assembled Int. J. Pharm. 454 (1) (2013) 527–529.
from a phage display peptide, Nat. Commun. 10 (1) (2019) 4635. [155] A.V. Singh, M.H.D. Ansari, D. Rosenkranz, R.S. Maharjan, F.L. Kriegel, K. Gandhi,
[149] J. Zhao, S. Wu, J. Qin, D. Shi, Y. Wang, Electrical-charge-mediated cancer cell A. Kanase, R. Singh, P. Laux, A. Luch, Artificial intelligence and machine learning
targeting via protein corona-decorated superparamagnetic nanoparticles in a in computational nanotoxicology: unlocking and empowering nanomedicine,
simulated physiological environment, ACS Appl. Mater. Interfaces 10 (49) (2018) Adv. Healthc. Mater. 9 (17) (2020), e1901862.
41986–41998. [156] D.J. Adams, The Valley of Death in anticancer drug development: a reassessment,
[150] Q. Zhou, S. Shao, J. Wang, C. Xu, J. Xiang, Y. Piao, Z. Zhou, Q. Yu, J. Tang, X. Liu, Trends Pharmacol. Sci. 33 (4) (2012) 173–180.
Z. Gan, R. Mo, Z. Gu, Y. Shen, Enzyme-activatable polymer-drug conjugate [157] R. Bosetti, S.L. Jones, Cost-effectiveness of nanomedicine: estimating the real size
augments tumour penetration and treatment efficacy, Nat. Nanotechnol. 14 (8) of nano-costs, Nanomedicine (Lond) 14 (11) (2019) 1367–1370.
(2019) 799–809.

11

You might also like