You are on page 1of 16

pubs.acs.

org/materialsau Review

Combinational Gene Therapy toward Cancer with Nanoplatform:


Strategies and Principles
Jinhui Lin, Xinlian Wang, Dongqi Ni, Yandong Chen, Chunying Chen, and Ying Liu*

Cite This: https://doi.org/10.1021/acsmaterialsau.3c00035 Read Online

ACCESS Metrics & More Article Recommendations


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Cancer remains a significant threat to human health. While


numerous therapies have been developed to combat the disease,
traditional treatments such as chemotherapy and radiotherapy are
suboptimal and associated with significant side effects. Gene therapy is
Downloaded via 2.147.12.71 on October 26, 2023 at 09:16:40 (UTC).

an emerging therapeutic approach that offers improved targeting and


reduced side effects compared with traditional treatments. Using siRNA
and other nucleic acid-based drugs in cancer treatment has generated
significant interest among researchers. Nanocarriers, such as liposomes,
can effectively deliver these agents to tumor sites. However, gene therapy
alone is often insufficient to eradicate tumors, and there is a risk of
recurrence. Therefore, combining gene therapy with other therapies using
nanocarriers, such as phototherapy and magnetic hyperthermia therapy,
can lead to synergistic therapeutic effects through different mechanisms.
In this review, we summarize various ways in which gene therapy can be combined with other therapies and highlight the role of
nanoplatforms in mediating these combined therapies, which would inspire novel design ideas toward combination therapies.
Additionally, bottlenecks and barriers to gene therapy should be addressed in the near future to achieve better clinical efficacy.
KEYWORDS: combinational therapy, gene therapy, tumor treatment, nanoplatform, chemotherapy, phototherapy,
magnetic hyperthermia therapy, immune therapy

1. INTRODUCTION The nanodrug delivery system provides an excellent


Cancer, a major public health problem worldwide, has tended platform for the delivery of gene drugs with its protective
to be the leading cause of death in the 21st century. According properties and targeted effects. Through rational design,
to the American Cancer Society estimates, nearly 600,000 nanomaterials can encapsulate nucleic acids effectively and
people in the United States will die because of cancer in 2022.1 protect gene drugs from degradation. Moreover, some types of
The number of Americans with cancer history is estimated to nanomaterials could help gene drugs escape from lysosome
be more than 22.1 million in 2030 based on the growth and and codelivery other drugs with nucleic acid. Many nano-
aging of the population alone.2 Chemotherapy, radiotherapy, vectors are developed to deliver gene drugs, including
and surgery are the main treatments for tumors, but their liposomes, cationic polymers, etc. Nowadays, novel therapies
effects are not ideal for high mortality and side effects clinically. are urgently needed for cancer treatments. Combining gene
Meanwhile, tumor recurrence and metastasis are still a great therapy with the currently available therapies such as
challenge clinically, while there are no suitable therapeutics. chemotherapy or radiation therapy provides surprising hope
After almost 30 years of development, gene therapy is for better therapeutic effects in patients with advanced-stage
becoming one of the most widely used therapeutic strategies cancers. Nanovectors, which can load more drugs onto
except traditional therapies against cancers and can import targeted organs and deliver them into the cytoplasm, perform
healthy exogenous or therapeutic genes into host cells to kill excellently in combined gene therapy. Based on the therapeutic
cancer cells or protect normal cells.3 Several types of nucleic effect of gene delivery, we summarize the purpose and effects
acid drugs were explored to heal cancer, such as Bcl-2 siRNA
and suicide gene. Nucleic acid drugs can be divided into DNA Received: April 29, 2023
and RNA, interfering with or regulating targeted gene Revised: July 19, 2023
expression in specific cells. Most types of DNA drugs target Accepted: July 20, 2023
the cell nucleus, while RNA drugs target the cytoplasm.
However, gene drugs can be easily degraded in vessels and into
other organs; therefore, gene drugs are limited clinically.
© XXXX The Authors. Published by
American Chemical Society https://doi.org/10.1021/acsmaterialsau.3c00035
A ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Figure 1. Synergistic therapeutic effect of chemotherapy and gene therapy. (A) Scheme of chemotherapy and gene therapy. (B) Nanocomplex
could inhibit the expression of PIKM2, induce mitochondrial exhaustion and collapse, and enhance the cytoplasmic levels of cytochrome C.
Reprinted with permission from ref 22. Copyright 2021, Elsevier B.V. (C) PAP could eliminate the tumor and suppress the expression of HIF-α.
Reprinted with permission from ref 26. Copyright 2018, Elsevier B.V. (D) MDSCs were stained with CD11b and Gr-1 antibody and determined by
flow cytometry. Reprinted with permission from ref 32. Copyright 2019, Elsevier B.V. (E) CCP/PCBP2 siRNA nanocomplex resulted in significant
tumor inhibition via collagen expression. Reprinted with permission under a Creative Commons Attribution 4.0 International License from ref 36.
Copyright 2021, Ivyspring International Publisher.

of gene therapy in combined therapy with a nanoplatform and to reduce cancer-related protein expression.4 Compared to
the mechanism of combined therapy. conventional treatments, gene therapy offers a variety of
advantages for cancer treatment, including high potency and
2. GENE THERAPY FOR TUMORS specificity, low off-target toxicity, and delivery of multiple
2.1. Advantages of Gene Therapy genes that concurrently target cancer tumorigenesis, recur-
The main methods of gene therapy against cancer include rence, and drug resistance.5,6 As a powerful tool, genome
using plasmid DNA containing therapeutic nucleotides to editing technologies have been used widely in gene therapy for
modulate aberrant gene expression and RNA interference like their properties of gene addition, gene ablation, and gene
siRNA, shRNA, miRNA, and antisense oligonucleotide (ASO) “correction” in vivo or in vitro, different from viral vectors that
B https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

can only mediate gene addition.7 For example, adoptive cell their efficiency and safety, as well as exploring the development
therapy uses gene delivery to ex vivo modify the patient’s T of novel nanocarriers.
cells to generate specific antitumor reactivity. It can improve
the antitumor immune response in the body through artificial 3. COMBINED GENE THERAPY WITH OTHER CANCER
methods, achieving a sustained and strong antitumor effect. THERAPIES FOR BETTER THERAPEUTIC EFFECT
Until now, chimeric antigen receptor T cell (CAR-T) therapy,
Single therapies can hardly realize the desired therapeutic
one of the adoptive cell therapies, has been developed to treat
outcomes in clinical trials because of the unfavorable drug
hematologic malignancies and multiple solid tumor types.8
resistance and heterogeneity of tumors. Since then, synergistic
Suicide gene therapy is based on introducing a suicide gene
combination therapies combining two or more therapeutic
into specific tissue, such as a tumor, and triggering cell death
modalities have been receiving more and more attention. In
by expressing specific enzymes, toxins, or pro-apoptotic
cancer therapy, gene therapy is multifunctional, including the
proteins in tumor cells.9,10 It can enhance the sensitivity of
induction of cancer cell apoptosis, inhibition of tumorigenesis
tumor cells to chemotherapy and be controlled by a tumor-
or metastasis, downregulation of heat shock proteins, and
specific promoter to select expression in tumor cells.11
induction of immune response by expressing cytokines.
2.2. Current Challenges of Gene Therapy Nowadays, it is becoming a strong complement to other
There are many barriers inside and outside the cell for nucleic cancer therapy. With the rapid development of nanometer
acid delivery. Naked nucleic acid can be easily degraded in material science, various nanomaterials with the ability to kill
blood for the presence of nucleases and has a very short half- tumor cells and deliver gene drugs have been widely
time in physiological media. Once nucleic acid gets into a cell, developed. These nanomaterials can use magnetic, luminous,
it must escape from the lysosome or will be degraded in the or sound energy to kill cancer cells and deliver therapeutic
lysosome. The tumor microenvironment (TME) can exhibit nucleic acids like siRNA, ASOs, and so on to gain a much
abnormal conditions, including hypoxia, low pH, and high better therapeutic effect.
reactive oxygen species (ROS) levels. Following single therapy, 3.1. Combined Gene Therapy with Chemotherapy
these conditions can promote cancer resistance, tumor Chemotherapy is the most common method of cancer
aggressiveness, metastasis, and recurrence. treatment. The cell’s metabolism can be disturbed or interfered
Moreover, some types of cells in tumor-like cancer stem cells with by delivering chemical drugs into the cell, thus killing
(CSC), hypoxic cells, and S-phase cells have much more robust cells. However, chemotherapy is greatly limited by multidrug
resistance to chemotherapy or radiation therapy, which tend to resistance, high drug dose concentration, low drug perme-
induce cancer recurrence or metastasis.12 For solid tumors, it is ability, and adverse effects.15,16 To improve the antitumor-
difficult for nucleic acid drugs to get into tumors and transfect igenicity, gene therapy is considered to supplement synergistic
target cells because of the complex compounds in solid tumors. chemotherapy. Gene therapy works in conjunction with
The unfavorable physicochemical properties of nucleotides, chemotherapy to increase the death of tumor cells or overcome
such as negative charges, large molecular weight, and size, the above-mentioned mechanisms and break the barriers to
seriously hinder their entry into host cells. Meanwhile, clinical chemotherapy in the below ways (Figure 1A).
gene therapeutics are limited by the low stability of nucleotides 3.1.1. Synergistic Therapeutic Effect of Chemo-
induced by endonuclease in serum.12,13 Therapeutic nucleo- therapy and Gene Therapy. By design of an intelligent
tides must overcome various biological barriers, from their drug delivery platform, chemotherapy and gene therapy can
entry into the bloodstream to their final destination in cancer work together to heal cancer. Several gene medicines have
cells to reach the cancerous site of action. For example, while been found and used in trials with chemotherapy treatment.
injecting plasmid complex by intravenous injection, the gene The tumor necrosis factor-related apoptosis-inducing ligand
complex should overcome the following biobarriers: (1) (TRAIL), a member of the tumor necrosis factor family, can
degradation by serum enzymes, (2) being removed by selectively induce cancer cell apoptosis by binding to the death
phagocytes and organs like kidney and liver, (3) crossing the receptor 4 (DR4), and DR5 is overexpressed in cancer cells.17
vascular endothelial cells, (4) tumor microenvironment, (5) Besides, the TRAIL-based gene therapy produces the by-
crossing the cellular membrane, (6) escape from endosome, stander effect to inhibit tumor growth in vivo.18 However, the
and (7) crossing the karyotheca.13,14 Nanocarriers as following TRAIL-resistance of cancer cells undermines the
promising tools could help nucleic acid drugs overcome the TRAIL-based gene therapy.19 Human antigen R (HuR) is a
biobarriers mentioned above. Several types of nanocarriers potential cancer therapy target associated with tumorigenesis
could be stable in normal tissues and blood and degraded in and metastasis. Amreddy et al. developed a folate receptor-
tumors by the ligands or properties of nanomaterials. Other α(FRA)-targeted polyamidoamine dendrimer-based nanopar-
nanocarriers may help drugs cross vascular cells and escape ticle system (Den-PEI-CDDP-siRNA-FA) for chemo/gene
from cell endosomes. There are some types of nanocarriers combined therapy.20 They used Den-PEI-CDDP-siRNA-FA to
currently used for gene therapy, including liposomes, codeliver anti-HuR siRNA and cis-diamine platinum to treat
polymeric nanoparticles, dendrimers, and viral vectors. Lip- FRA-overexpressing lung cancer cells. They gained a
osomes are spherical vesicles made of phospholipids that can significantly more significant therapeutic effect than individual
encapsulate and deliver DNA or RNA. Polymeric nanoparticles therapeutics. The enhanced mechanism of combined gene
are made of polymers that can bind to nucleic acids. therapy with chemotherapy can be divided into the following
Dendrimers are highly branched, tree-like polymers that can content.
encapsulate and deliver DNA or RNA. Viral vectors are 3.1.2. Fighting Multidrug Resistance in Treating
genetically engineered viruses that can deliver therapeutic Tumor. The use of chemotherapy, one of the most widely
genes to cells. Each type of nanocarrier has its advantages and used and effective treatments for cancers, is seriously limited
limitations, and researchers are continually working to improve by multidrug resistance (MDR), which is developed by the
C https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

continuous use of chemotherapy drugs. The cancer MDR potential therapeutic target for cancer therapy.31 A multifunc-
requires an increasing dose of chemotherapy drugs, which tional delivery system based on an amphiphilic polymer with
could induce inevitable toxicity to patients. MDR is closely morpholine attached to the pendant side chains (POEG-st-
related to the overexpression of transporters like P- Pmor) for codelivery of IL-36γ expression plasmid and
glycoprotein (P-gp) on the cancer cell membrane. Many doxorubicin (DOX) has been developed to treat cancer. The
drugs, such as doxorubicin (DOX) and paclitaxel (PTX), must nanomicelles are stabilized by charge−charge interactions
enter the cells to play a therapeutic role. At the same time, P- between the cationic polymer and plasmid DNA, which are
gp, one of the ATP-binding cassette (ABC) transporters, can further addressed in vivo. The Dox+IL-36γ/POEG-st-Pmor
actively expel them out of the cells, reducing the efficacy.21 decreases the MDSC in the lung metastasis of breast cancer
One of the strategies against cancer MDR is blocking out the and hence enhances the type I immune response, which raises
energy supply for ATP-binding cassette (ABC) transporters, the efficiency of DOX (Figure 1D).32
achieving a favorable therapeutic effect. A polyelectrolyte One of the main difficulties in treating tumors such as
nanocomplex surface decorated with hyaluronic acid was pancreatic cancer is the nearly impenetrable desmoplastic
developed to deliver DOX and PKM2 siRNA.22 The stroma. It prevents drugs into tumors and supports and
hyaluronic acid of the nanocomplex could target CD44, promotes pancreatic cancer at the same time.33,34 One strategy
which is overexpressed in tumor cells, while the nanocomplex to achieving more drugs into stroma-rich tumors is restoring
could efficiently escape from lysosomal entrapment and release homeostatic stromal function. A gene-/chemo-combined
siRNA into cells. By downregulation of PKM2, the energy strategy was developed.35 The PEGylated PEI-coated gold
supply for ABC transporters in cancer cells has been NPs were developed to codeliver anti-HSP47 siRNA and
suppressed, thus defeating drug resistance. Results showed ATRA to transform activated pancreatic stellate cells into
that combined therapy could enhance and decrease the quiescent pancreatic stellate cells, which could promote
expression of PKM2, induce mitochondrial exhaustion and gemcitabine delivery into a pancreatic tumor. A peptide-
collapse, and release cytochrome C into the cytoplasm (Figure based core-stabilized PCBP2 siRNA nanocomplex was
1B). Another mechanism of MDR is overexpressing growth developed to break type I collagen in pancreatic ductal
factors in cancer cells.23 RNase-resistant RNA nanoparticles adenocarcinoma (PDAC) tumor stroma (Figure 1E).36 After
are modified with an epidermal growth factor receptor (EGFR) siRNA reverses the accumulation of type I collagen, the
to block the growth factor signing pathway. Targeting aptamer antitumor efficacy of gemcitabine was significantly improved.
loading XBP1 siRNA, which could promote sensitization to Cancer-associated fibroblasts could secret a dense desmoplastic
chemotherapy and impede angiogenesis in vivo, was developed matrix or release regulatory molecules, thus enhancing
to kill cancer cells and inhibit tumor growth.24 Hypoxia is a chemotherapy resistance.37 Blocking collagen cross-linking
hallmark feature of the tumor microenvironment, which could and fibronectin fibril assembly via lysyl oxidase (LOX)-
inhibit the efficiency of chemotherapy.25 The aim is to reduce siRNA could enhance doxorubicin sensitization against the
hypoxia’s influence, a novel multifunctional hypoxia-induced triple-negative breast cancer (TNBC) cell, indicating that
size-shrinkable nanoparticle designed to codelivery DOX and combining DOX with LOX siRNA is a promising therapeutic
si-HIF1α.26 The nanoparticle could respond to hypoxia and method.38
degrade the outer PEG to expose positively charged PAMAM, While chemotherapy is a commonly used method for
thus penetrating DOX and si-HIF1α into the tumor core. The treating cancer, it has limitations such as multidrug resistance,
expression of HIF1α induced by hypoxia is downregulated by high drug doses, low drug permeability, and adverse effects.
si-HIF1α, which ultimately enhances the effectiveness of DOX However, gene therapy can be used with chemotherapy to
in treating cancer (Figure 1C). increase the death of tumor cells or overcome these limitations.
3.1.3. Restoring Homeostatic Stromal Function to By the design of intelligent nanomaterials, the synergistic
Promote Therapeutic Efficiency and Drug Delivery. therapeutic effect of chemotherapy and gene therapy can be
Besides the MDR of cancer cells, tumor stroma also plays an achieved in two ways. One way is to combat multidrug
important role in drug resistance.27 To delay the MDR resistance in tumors by disrupting gene expression within the
progression, optimized combined treatments are badly tumor cells. Another way is by restoring the homeostatic
needed.28 The tumor-associated macrophages (TAMs) con- stromal function, which can also promote therapeutic
tribute to drug resistance and relapse via different pathways efficiency and drug delivery. Finally, a combined gene/chemo
and are major components of the tumor microenvironment.29 strategy can effectively treat tumors.
The hyaluronic acid-based nanoparticles were designed to
3.2. Combined Gene Therapy with Phototherapy
encapsulate miR-125b (HA-PEI-miR-125b), which can specif-
ically target TAMs and repolarize macrophages to an immune- Phototherapy can be divided into two main categories:
activating phenotype. Results show that the total number of photothermal therapy (PTT) and photodynamic therapy
CD11b+, F4/80+, and CD206+ macrophages was reduced (PDT). PTT is a widely developed localized cancer therapy
while increasing the number of CD80+ macrophages. However, that utilizes photoinduced heat via near-infrared irradiation to
there is no obvious difference of CD45+ immune cells between cause cancer cell death and avoid damage to other regions
the untreated control and HA-PEI-miR-125b treated group, which are not irradiated.39−41 Compared to traditional
and whether the nanoparticle could increase the number of therapies, the main advantages of PTT are minimal
PD-1+ immune cells by increased infiltrating T cells needs invasiveness, deep tissue penetration, and simple opera-
further evaluation. Later results show that HA-PEI-miR-125b tion.40,42 Another promising cancer therapy based on photo-
could enhance the efficacy of paclitaxel with low toxicity.30 sensitizer (PS) is PDT, which uses visible light to irradiate PS
Myeloid-derived suppressor cells (MDSC) are largely to generate ROS, thereby killing cells.40 PDT is a good choice
immature myeloid cells that inhibit immune effector cell for clinical cancer treatment, particularly for superficial tumors
function in cancer and tumor progression, representing a like esophageal, bladder cancer, and melanoma, because of its
D https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Figure 2. Synergistic therapeutic effect of phototherapy and gene therapy. (A) Scheme of phototherapy and gene therapy. (B) R-nano ICG/S-
siRNA could destroy tumor blood vessels and increase tumor temperature. Mice injected with PBS (1), ICG (2), nanoICG (3), R-nanoICG (4),
and R-nanoICG/S-siRNA (5) and irradiated with an 808 nm laser at a power density of 1.0 W cm−2. Reprinted with permission from ref 49.
Copyright 2018, Wiley-VCH. (C) Representative noninvasive imaging of lung-adenocarcinoma NCI-H889 tumor-bearing nude mice with different
treatments at days 7 and 36 and immunohistochemical analysis of VEGF in lung tissue. Reprinted with permission under a Creative Commons
Attribution 4.0 International License from ref 60. Copyright 2021, Springer Nature. (D, E) DOX-siVEGF-NPs/Ce6-MBs generate more ROS with
laser and inhibit tumor growth. Reprinted with permission from ref 80. Copyright 2020, Elsevier B.V. (F, G) PEG and PEI dual-functionalized BP
nanosheets could target tumor sites and induce cell death. Reprinted with permission from ref 86. Copyright 2018, American Chemical Society.

E https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

eximious features such as low invasiveness, mild killing, terial for PTT.39,55,56 Moreover, BP NS has been efficient PTT
spatiotemporal selectivity, and lower side effects.43,44 One of therapy agents for anticancer treatment because of its ability to
the most significant advantages of phototherapy is its ability to generate singlet oxygen (1O2) with a high quantum yield of
control drug release achieved by visible or infrared light. ∼0.91 under the whole range of visible light.56 In 2018, Wang
However, for those metastatic or disseminated cancers, a single et al. first utilized BP NS for gene-/PTT-combined therapy and
PTT or PDT appears powerless.45 reported it was a promising tool for combined therapy.57 The
Currently, gene therapy is an effective complement to PTT/ negatively charged BP NS was modified with PEI, which
PDT because of its superiority in therapeutic enhancement and enabled it to be loaded with survivin-siRNA. The results of the
decreasing side effects.46−49 Therapeutic nucleic acids, such as Western blot showed that the expression level of survival in
siRNA, have been widely used to combine PTT/PDT to gain a MCF-7 cells (breast cancer cells) was significantly decreased
synergistic therapeutic effect. The synergistic effect of gene by being treated with BP-PEI-siRNA for 24 h, which could
therapy and PTT is mainly achieved in two ways. Either gene inhibit cell growth (44% inhibition rate). With simple design,
therapy and PTT kill tumor cells together, in which gene the BP nanosheets could degrade into phosphate ions in vivo,
therapy can kill the surrounding or PTT-resistant tumor cells, which indicates that the BP-PEI-siRNA complex could be a
or gene therapy can improve the sensitivity of tumor cells to promising tool for clinical applications.
PTT, such as inhibiting the expression of heat shock proteins Furthermore, the inhibition rate of MCF-7 cells increased to
from improving the efficacy of PTT. Several inorganic 64% when the cells were treated with BP-PEI-siRNA for 24 h
materials have been applied in gene/photothermal combined and 1.0 W cm−2 808 nm irradiation for 10 min. In in vivo
therapy, such as gold nanoparticles (GNs), black phosphorus tumor of MCF-7 cells, the combination of BP-PEI-siRNA with
nanosheets (BP NS), carbon-based materials, and 808 nm irradiation had the most potent inhibition of tumor
others.39,50,51 Some inorganic nanomaterials cannot be loaded growth, indicating antisurvivin gene therapy and PTT of BP-
with therapeutic nucleic acids, so modification of these PEI-siRNA had more effective than single therapy. Gold
materials is needed, such as cationic polymer modification nanomaterials, the most widely applicable multifunctional
on the material’s surface to load the nucleic acid by agents in PTT, are usually used in combined therapies based
electrostatic interaction between the cationic polymer and on their versatile surface chemistry.48,51,58 Using gold nano-
the nucleic acid.41 materials, a more effective therapeutic could be achieved. A
3.2.1. Synergistic Therapeutic Effect of Phototherapy gold nanorod was modified by the TAT peptide and thiolated
and Gene Therapy. The most widely used strategy for gene DNA linker to deliver the sgRNA/Cas9 complex, which
therapy combined with PTT/PDT is the synergistic method silenced the tumor-associated gene PLK1.59 A significant
for utilizing antitumor genes with PS together (Figure 2A). inhibiting effect was observed after mild photothermal therapy
Survivin, a member of the inhibitor of apoptosis (IAP) families, combined with gene therapy. A gold nanocage vehicle loading
is overexpressed in most human tumors and is considered an DOX and siRNA was modified with the AS1411 aptamer for
excellent target for cancer gene therapy.52,53 Metal−organic the combination of tumor-responsive genetic therapy, chemo-
nanostructures (MONs) have significant advantages in therapy, and photothermal treatment.60 As shown in Figure
combination therapies for traditional photosensitizers like 2C, a pronounced decrease of VEGF and fewer tumor foci
indocyanine green (ICG) with therapeutic nucleotide.49,54 The were seen in the Au-siRNA-PAA-AS1411 group, which was
Zn(II)-dipicolylamine (Zn-DPA) molecule is a potential treated with a combination of tumor-responsive genetic
siRNA delivery carrier, because of its high affinity for therapy, chemotherapy, and photothermal treatment. By
phosphate-containing molecules. Gold nanorods (GNRs) designing the ROS-responsive materials, NIR spatially
conjugated with Zn-DPA were designed to deliver PLK1- activated siRNA therapy can be achieved to treat tumor sites
siRNA and PTT simultaneously. They found that the with PDT therapy.61 In normal tissue, siRNA molecules
combination of PLK silencing-induced apoptosis and local cannot function due to being trapped and broken down in
PTT had apparent synergy for treating PC-3 tumor.46 A endolysosomes. This is caused by the quaternary ammonium
coordination-driven self-assembly MONs have been developed moiety’s unprotonatable property, keeping the siRNA in an
for gene/photothermal combined therapy.49 The Zn(II)- “off” state. However, when NIR irradiation is applied to tumor
dipicolylamine (Zn-DPA) molecule interacted with ICG and tissue, it triggers the release of siRNA from the endolysosomes
polyvinylpyrrolidone (PVP) polymers to form nanoICG, and into the cytosol. This is achieved through the cleavage of
which was modified with RGD, one of the peptide motifs TK bonds and the photochemical internalization effect,
targeting angiogenesis. Due to the high siRNA transfection, resulting in the “on” state of siRNA activity. By inhibiting
long-lasting tumor accumulation, and good photothermal the glutathione peroxidase 4 gene, siRNA enhances the
properties, the R-nanoICG binding with survivin-siRNA accumulation of ROS, which synergizes with Ce6 photo-
presented apparent synergistic efficacy of PTT and gene dynamic therapy to provide effective antitumor activity in vivo.
therapy. The angiogenesis assay showed that the PTT/gene This is achieved through ROS-sensitive cationic nanocarriers,
therapy group had the lowest level of CD31, which indicated which provide a feasible and controlled strategy for delivering
that combination therapy had the most efficiency for siRNA therapy with synergistic drug effects specific to tumors.
destroying tumor blood vessels (Figure 2B). In the 4T1 The enhanced mechanism of combining gene therapy with
xenograft mouse model, the tumors were entirely eliminated phototherapy can be divided in the following ways.
after combination therapy treatment, while the tumors could 3.2.2. Increasing the Efficacy of Phototherapy. The
not be inhibited by a single therapy. increased thermotolerance induced mainly by the upregulation
BP NS, a kind of attractive two-dimensional (2D) material of heat shock proteins (HSPs) is the main reason for the failure
with photodegradable character, high surface area, and negative of PTT.62 HSPs can act as ATP-mediated molecular
charge, has high photothermal conversion efficiency and chaperones to correct proteins’ misfolding, thereby keeping
significant extinction coefficient, considered a good nanoma- their activities under hyperthermia.62,63 For thermotolerance in
F https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

many cancer cells, HSP70 and HSP90 are crucial proteins.63,64 Furthermore, the Hela tumor growth was almost completely
Applying siRNA against the HSP family can improve cells’ suppressed under the treatment of PP-NG-siHSP70 (laser),
sensitivity to PTT and enhance the efficiency of PTT.58,63,65 and more surprisingly, two-thirds of the tumors completely
Bcl-2-associated athanogene domain 3 (BAG3), acting as a disappeared. Moreover, PP-NG-siHSP70 plus laser could
molecular cochaperone of the HSP family, including HSP70, is induce the most effective inhibition of HSP70 expression
also an antiapoptotic protein.66−68 The evidence showed that and increase the level of caspase-3 expression. This triple-shield
the BAG3 overexpression was closely related to the tolerance complex PP-NG-siHSP70 showed the capability of strongly
of PTT, radiation therapy, and chemotherapy.69 The ablating solid tumors under mild conditions. Further, this gene
combination of BAG3-siRNA and gold nanorods (GNRs) in delivery system could be expanded to other combined
gene and photothermal therapy was studied. Treating GNRs- therapies such as gene/photothermal/immune combined
mediated PTT can significantly increase the expression of therapy.63
BAG3 and HSP families (HSP27, HSP60, HSP70, and PDT could be strengthened by downregulating the selected
HSP90) in Cal-27 cells, which indicates the PTT-induced gene. Recently, chemophotodynamic combination therapy has
heat shock response. However, applying BAG3-siRNA could become an ideal strategy. However, tumor cells treated by
significantly inhibit the heat shock response, which could PDT will induce an abnormal increase in angiogenesis for
strengthen cells’ sensitivity to PTT. The GNRs-siBAG3 generating reactive oxygen species (ROS). A complex
complex showed a stronger BAG3 silencing capability than composed of two compartments was developed to suppress
lipofectamine 2000-siBAG3 and a higher efficiency of PTT tumor angiogenesis.80 The first compartment is nanoparticle
than GNRs themselves, which completely suppressed the loading DOX and siRNA silencing the VEGF gene, while the
growth of Cal-27 tumors in vivo. other part is a Ce6-encapsulating microbubble. Three therapies
The generation of HSPs after PTT relies on an abundant showed significantly lower tumor growth, indicating that
ATP-meditated energy metabolism.70 Therefore, weakening overexpression of VEGF was successfully prevented, and
the energy metabolism seems to be a potential solution to further angiogenesis was restrained (Figure 2D and E).
downregulate HSPs expression during PTT.65 Pyruvate kinase Survivin, an antiapoptosis protein, was overexpressed in
M2 (PKM2) is involved in ATP production in cancer cells.71 tumor cells after treatment, thus leading to therapeutic
In recent work, a combined strategy of gene therapy and PTT resistance in tumors.81,82 To downregulate the survival level
was reported.64 Simply put, the spherical dendrimer-polypep- in cancer cells, Jin et al. designed an upconversion nanoplat-
tide (DPP) was synthesized through the PAMAM-mediated form for enhanced PDT.83 They used upconversion nano-
ring-opening polymerization of N-carboxy anhydride. Then, particles adsorbing a long single-stranded DNA (ssDNA) with
DPP was used to load ICG and PKM2-siRNA and finally was multiple copies of the aptamer (AS1411) and DNAzyme to
coated with human serum albumin. Compared to commercial specifically target nucleolin overexpressed on cancer cells
transfection reagents (25 kDa PEI and Lipofectamine 2000), specifically. The AS1411 aptamer could encapsulate the
DPP had a dramatically more substantial cellular uptake photosensitizer 5,10,15,20-tetrakis (1-methylpyridinium-4-yl)
capacity and a higher PKM2 silencing efficiency (∼87%) in porphyrin (TMPyP4) as well as recognize the nucleolin
MCF-7 cells. Both qPCR and Western blot in vivo and in vitro overexpressed in tumor. The upconversion nanoplatform could
showed the same results: the inhibition of PKM2 expression trigger PDT not only under NIR light but also with high
could remarkably inhibit the production of HSP70 and HSP90 recognition and loading capacity. The combined therapy
due to the lack of ATP. The triple synergy of siPKM2-induced dramatically silences surviving gene expression, hence enhanc-
cell starvation, downregulated HSP expression, and PTT ing the efficiency of PDT.
showed surprisingly anticancer efficacy for in vivo MCF-7 3.2.3. Inhibiting Growth and Metastasis of Tumor
xenograft model. The tumors of the combined therapy group Cells to Assist PDT/PTT in Treatment of Tumors. Cancer
were completely suppressed within the observation period, and metastasis is the leading cause of tumor recurrence. Though
the survival rate of the combined therapy group was some single therapies like PTT, 84 PDT, MHT, and
satisfactorily 100% within the observation period of 50 days, sonodynamic therapy (SDT) have been considered promising
which is significantly higher than those of other groups. therapies because of their stable spatiotemporal control, they
The local PTT inevitably induces the heat damage of are designed as localized anticancer treatments that are
surrounded normal cells, which is likely to cause cancer cell powerless against the cancers that transfer or survive after
metastasis and local inflammation.72−74 Low-temperature PTT treatment.40,43
seems an excellent choice to avoid the side effects because it Human telomerase reverses transcriptase (hTERT) is
relies on low-temperature heat-induced cellular apopto- closely related to cancer metastasis and growth.85 A dual-
sis.72,75,76 The efficacy of low-temperature PTT to solid functionalized BP NS modified with poly(ethylene glycol)
tumors is still undesirable due to increased thermotolerance (PEG) and PEI (PPBP) was designed to deliver hTERT
induced by upregulated expression of intracellular HSPs.77,78 A siRNA for gene/photodynamic/photothermal combination
general strategy was investigated for combining low-temper- therapy.86 Through the detection of the degradation of
ature PTT and gene therapy against HSP70.63 First, a siRNA- PPBP under different conditions, it is found that PPBP
bearing self-assembled nucleic acid nanogel based on nucleic degrades more significantly under a combination treatment of
acid hybridization between DNA-grafted polycaprolactone H2O2 and PBS of pH 5.0, compared with a single treatment of
(DNA-g-PCL) and siRNA linker was developed.79 The H2O2 or PBS of pH 5.0, indicating PPBP can be specifically
siRNA-bearing nucleic acid nanogel was coated with polydop- degraded in the microenvironment of acidic lysosome and high
amine and surface PEGylated, named PP-NG. The in vivo photoinduced ROS. si-hTERT synergizes with PDT and PTT
efficacy of PP-NG-siHSP70 combined with low-temperature on Hela cells. Moreover, results showed that the introduction
PTT was then evaluated on HeLa tumor xenografts. of si-hTERT can effectively cooperate with PTT/PDT therapy
G https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Figure 3. Synergistic therapeutic effect of MHT and gene therapy. (A) Scheme of magnetic hyperthermia therapy and gene therapy. (B) The shape
of differently shaped M-MSNs, plots of inverse transverse relaxation time (1/T2) versus Fe concentration of the M-MSNs, and photographs of
HepG2 tumor-bearing mice in each group and MR images of mice from different groups at day 7 and 14 after treatment (tumors marked by the
white arrow). Reprinted with permission from ref 104. Copyright 2018, Elsevier B.V. (C) The effects of let-7a gene and FACS analysis of
combination-treated cells compared to controls. Reprinted with permission from ref 109. Copyright 2014, Wiley-VCH. (D) RT-PCR
demonstrating the successful synthesis of the sTRAIL-EGFP plasmid transfected into A2780 ovarian cancer cells, and tumor volume was followed
over 2 weeks. The size of the tumors decreased significantly when treated with the engineered AD-MSCs. Reprinted with permission from ref 110.
Copyright 2016, Elsevier B.V.

to inhibit the metastasis and growth of A549 cells in vivo As described above, the TRAIL gene has been used wildly in
through downregulating hTERT (Figure 2F and 2G).86 gene therapy. Single gene therapy will lead to uncontrollable
H https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

expression of TRAIL in vivo, which phototherapy could solve NCT02033447). In order to be clinically applicable, the
for its property of controlled drug release. Meanwhile, the temperature of MHT (43−46 °C) and the accumulation of
recurrence of tumor cells after PTT could be killed by TRAIL MNPs in tissue must be strictly controlled.93 However, since
treatment. For example, Qiao et al. developed semiconducting the frequency f and field amplitude H of AMF (f × H < 5 ×
nanoparticles decorated with caged TRAIL-expressing plasmid 109 Am−1s−1) is strictly limited by technology and biomedical
under HSP70 protomer named SPN@HSP70-TRAIL-GFP- reasons and the thermal conversion efficiency of the existing
(SPNHT).87 SPNHT has great potential to induce cell MNPs is unable to meet the satisfaction, the therapeutic
apoptosis in vitro and in vivo with a laser, indicating that efficacy of single MHT is still limited and unsatisfactory.94−96
combined therapy could effectively restrain tumor recurrence Hence, many combined strategies are developed to enhance
after PTT treatment. the efficacy of MHT, such as PTT/MHT, PDT/MHT,
3.2.4. Protecting Normal Cells and Reducing the chemotherapy/MHT, gene therapy/MHT, and so on.94 In
Toxicity of PTT/PDT. The traditional treatment and some order to achieve codelivery of heat magnetic nanoparticles and
other new therapies like PTT and PDT have high antitumor gene drugs, there are three main strategies as following:
efficiency, but at the same time, they are likely to be harmful to delivering MNPs and therapeutic oligonucleotide separately;
normal healthy cells.88 As two significant photo sources are modifying the surface of MNPs with gene vectors like cationic
used in phototherapy, short-wavelength UV/visible light polymers which are able to load negative oligonucleotide;
cannot penetrate deeply into tissue. At the same time, high- using MNPs themselves to load pDNA/polymer complex.97−99
power-density near-infrared (NIR) light can cause undesired It was reported that a mild multiple MHT-mediated TRAIL
heat damage. To solve this problem, a ROS-degradable release system was a good solution for TRAIL-resistant cancer
polycation was designed to codeliver a photosensitizer (PS) cells. A biodegradable TRAIL/SPION nanocomplex hydrogel
and gene drug, which uses far-red light (661 nm) at low optical (T/S-NH) designed for combined therapy with MHT and
power density.89 The polycation could release the P53 gene simultaneously MHT-induced TRAIL release.100
under exposure to far-red light and work together with ROS to 3.3.1. Synergistic Therapeutic Effect of MHT and
induce the death of cells. Gene Therapy. The unique magnetism gives MNPs
In most cases, HSP can be an unfavorable factor during enhanced cellular internalization, controllable tumor accumu-
phototherapy. However, utilizing this property can also be an lation under external magnetic fields, and control of target gene
excellent way to take control of gene expression. A multifunc- expression under AMF (Figure 3A). Through multiple surface
tional nanocomposite named PBDTQ, which loads a plasmid modifications of viral or nonviral vectors, MNPs can load
inserted CD-TK double suicide gene with HSP70 promoter, therapeutic nucleotides to achieve the combined gene/
could achieve heat-controlled gene expression.90 Furthermore, magnetic hyperthermia therapy.101 In the earliest examples of
the suicide gene expression of cytosine deaminase (CD) and the combined therapy, they injected 20 μg of pGadTNF into
herpes simplex virus type-I thymidine kinase (TK) could turn tumors by a lipofection method for TNF-α gene therapy and,
prodrugs, 5-fluorocytosine (5-FC) and ganciclovir (GCV), after 1 day, injected magnetite cationic liposomes into tumors
loaded in PBDTQ into their cytotoxic forms of 5-fluorouracil for MHT under AMF (118 kHz and 30.6 kA/m) for 30 min.
(5-Fu) and ganciclovir-triphosphate (GCV-TP) to minimize The hyperthermia (46 °C) and overexpression of TNF-α (3-
collateral damage and nontargeted side effect. fold vs control group) resulted in potent inhibition of U251-SP
In summary, gene therapy is an effective complement to tumor growth (0.5 ± 0.4 cm3 on day 32), which was
phototherapy as it enhances therapeutic efficacy and reduces significantly smaller than that of single MHT (2.0 ± 1.1 cm3
side effects. For metastatic cancers, a single PTT or PDT may on day 32) or TNF-α gene therapy (2.9 ± 1.1 cm3 on day
not be effective. Gene therapy, specifically the use of a 32).98
therapeutic nucleic acid such as siRNA, has been widely used Among the numerous gene therapy clinical trials, the herpes
to combine with PTT/PDT to gain a synergistic therapeutic simplex virus thymidine kinase (HSV-TK) gene is the best-
effect. The synergistic effect of gene therapy and PTT is
characterized suicide gene for tumor cell killing. The HSV-TK
achieved in two ways: either they kill tumor cells together, or
can make nontoxic ganciclovir monophosphate into toxic
gene therapy can improve the sensitivity of tumor cells to PTT,
ganciclovir triphosphate, leading to a bystander effect.102 Here
such as by inhibiting the expression of heat shock proteins.
25 kDa PEI-Fe3O4 MNs were developed to deliver the HSV-
The inhibition of tumor growth and metastasis is also possible
TK suicide gene induced by an α-fetoprotein promoter and
with the combination of gene therapy and phototherapy. To
hypoxia enhancement for the combined gene/magnetic
reduce the toxicity of phototherapy, far-red light at low optical
hyperthermia therapy of hepatocellular carcinoma (HCC).
power density can be used, and gene expression can be
They found a synergistic therapeutic effect of the combined
controlled by utilizing the properties of heat shock proteins.
therapy.103 Sphere-like and rod-like shape-controlled magnetic
3.3. Combined Gene Therapy with Magnetic mesoporous silica nanoparticles (M-MSNs) were developed to
Hyperthermia Therapy/Magnetothermal Therapy integrate gene therapy, MHT, and magnetic resonance imaging
Magnetic hyperthermia therapy (MHT), another kind of together.104 Modified with PEG-g-PLL, the positively charged
promising hyperthermia therapy for localized treatment of M-MSNs were used to load ganciclovir and the HSV-TK
solid cancer, utilizes an external alternating magnetic field plasmid. Both shapes of M-MSNs with ganciclovir and pTK
(AMF) to selectively heat magnetic nanoparticles (MNPs or demonstrated a highly effective combined suicide gene/
MNs), inducing tumor cell death/apoptosis.91 Magnetic magnetic hyperthermia therapy of HCC (Figure 3B).
particle imaging (MPI) can help track the MNPs’ delivery Compared to sphere-like M-MSNs, rod-like M-MSNs showed
and carefully control the release of drugs.92 Due to its safety, higher magnetic targeting and combined therapeutic perform-
high efficacy, and excellent tissue penetration, MHT has ance (90% inhibition in vivo). Gene therapy could enhance the
entered the clinical trial stage (ClinicalTrials.gov Identifier: effect of MHT in several ways, as described below.
I https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

3.3.2. Improving the Sensitivity of Tumor Cells to deliver and activate heat-inducible TRAIL plasmid with HSP70
MHT. Gene therapy, as an exciting tumor treatment, can B’ promoter in mesenchymal stem cells.110 MCNPs can deliver
improve the sensitivity of tumor cells to hyperthermia induced a heat-inducible plasmid encoding TRAIL and activate TRAIL
by MHT by regulating heat resistance-related genes like HSPs. secretion in engineered AD-MSCs with mild magnetic
The inhibition of HSPs expression makes it possible for a hyperthermia. Quantifying luminescence intensity shows that
practical therapeutic effect of magnetic nanoparticle complex the engineered AD-MSCs are significantly better than
under mild heating or low concentration, which can decrease treatment with a single dose of recombinant TRAIL (Figure
the risk of inflammation and damage to surrounding normal 3D).
cells.105 In Court et al.’s study, they detected the gene In short, gene therapy can improve the sensitivity of tumor
expression of ovarian cancer cell line HeyA8 cells after cells to hyperthermia induced by MHT by regulating heat
treatment of MHT (43 °C, 30 min) through microarray resistance-related genes like HSPs. The inhibition of HSPs
analysis and found that the top 20 upregulated genes contained expression makes it possible for a practical therapeutic effect of
HSP70 (HSPA6, HSPA7, HSPA4L) and BAG3.106 Then, they magnetic nanoparticle complexes under mild heating or low
investigated the combination of HSPA6-siRNA and MHT to concentration, which can decrease the risk of inflammation and
treat ovarian cancer cells in vitro and in vivo. Four kinds of damage to surrounding normal cells. Various studies have
anti-HSPA6 siRNA were selected to inhibit HSPA6 expression reported that combined therapy has shown significant and
in ovarian cancer cells (A27880 cP20 and HeyA8), and all four broader effects compared to single therapy. Meanwhile, the
HSPA6-siRNA-showed a noticeable improvement to CMDx- development of combined therapy should consider the rational
IO based MHT in vitro. The similar positive impact of combination of MNs, surface decorations, nucleic acid types,
HSPA6-siRNA on MHT was verified in an in vivo and genes.
subcutaneous ovarian tumor model. 3.4. Combined Gene Therapy with Immune therapy
Besides siRNA therapy, micro-RNAs are reported to
combine with MHT against HSP. Compared to the gene The immune system is the key to detecting and eradicating
therapy based on siRNA, which only modulates single HSP- cancer cells in vivo, involving a series of complex mechanisms.
related targets, the gene therapy based on miRNA can On the one hand, it can effectively prevent the occurrence and
modulate multiple targets on the same or similar pathways, development of malignant tumors. However, on the other
indicating a more significant and broader effect.107 Lethal-7a hand, it will also promote the selection of immune-tolerant
miRNA (let-7a), a tumor suppressor, is downregulated in tumor cells. After three stages of tumor development
several human cancers, including brain cancers. Let-7a is (elimination, equilibrium, and escape), tumors in the body
associated with DNA repair and cell survival mechanisms by have evolved into tumor cells that can evade the immune
targeting factors such as RAS, MYC, and cyclin D, also related response, resulting in uncontrollable cancer in the body.111
to downstream regulation of the HSP family.108 The combined The immune system has been considered to be a key point for
therapy of miRNA gene therapy with ZnFe2O4-based MHT cancer healing and detection. Immune cells can be engaged in
was developed.109 The negatively charged ZnFe2O4 MNPs a battle with cancer cells. Other cases can promote the
were modified with 10 kDa branched PEI to load let-7a metastasis and recurrence of tumors after three phases of
miRNA through a two-step layer-by-layer process. To evaluate eliminating, balancing, and escaping tumor cells, which involve
the therapeutic enhancement of let-7a to MHT, U87- complex mechanisms. The mechanisms for the escape of
EGFRvIII cells were treated with let-7a, magnetic hyper- cancer cells from the immune system have been widely studied,
thermia, or the combination. The combined treatment group including the change of immune checkpoint proteins like PD-
exhibited lower cell viability (34%) than the single treatment 1, and CTLA-4, manipulation of cytokine expression, and
groups (let-7a treatment 68.9%, magnetic hyperthermia alteration of antigens.111,112 Newer therapies combined with
63.14%). A mechanistic study of combined therapy showed gene therapy and immunotherapy have flourished in recent
the upregulation of caspase-3 and downregulation of PI3K, years and made encouraging progress, including OV therapy,
HSPs (HSP27, HSP70, HSP72, and HSP90), and let-7a targets adoptive immunotherapy, and tumor vaccine. Besides, certain
(RAS, MYC, and IGF1R), which suggested that let-7a could materials exhibit immunomodulatory effects that can poten-
enhance the MHT to glioblastoma multiforme through tially synergize with gene therapy drugs to enhance their
inducing cell apoptosis and increasing the sensitivity of cells efficacy.113
to magnetic hyperthermia (Figure 3C). The MNPs can be 3.4.1. Synergistic Therapeutic Effect of Immunother-
further modified with targeting ligands or PEG to improve the apy and Gene therapy. Immunotherapy is a promising
targeting and biocompatibility. therapeutic that has been investigated widely in recent years.
Nevertheless, due to the limitations of magnetic materials The main types of cellular immunotherapy include CAR T,
and gene vectors, the combination of gene therapy and MHT TCR-transduced T cells (TCR-T), tumor-infiltrating lympho-
still receives less attention. The development of the combined cytes (TIL), and natural killer (NK) cells. CAR T therapy is a
therapy should consider the rational combination of MNs, hot field in clinical practice. Five cell therapies approved by the
surface decorations, nucleic acid types, and genes. Recently, U.S. Food and Drug Administration (FDA) come from this
magnetic hyperthermia-mediated gene therapy has been field. Checkpoint inhibitors (ICI) have revolutionized cancer
receiving increased attention. In this gene therapy, MNs treatment and have become the standard of care for many
enhance tumor accumulation, therapeutic gene release, and cancer patients. Although immunotherapy has been applied in
magnetic hyperthermia-activated gene expression.101 For terminal cancer, it is greatly limited by an immunosuppressive
example, a novel mild magnetic hyperthermia-activated stem tumor microenvironment for low pH, hypoxia, etc. Gene
cell-based gene therapy was developed to heal ovarian cancer. therapy could steadily activate immune cells in the tumor
Here, 25 kDa PEI-modified magnetic core−shell nanoparticles microenvironment or enhance the vaccine effect, thus gaining
composd a ZnFe2O4 core and mesoporous silica shell to the strengthening therapeutic effect (Figure 4A).
J https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Figure 4. Synergistic therapeutic effect of immunotherapy and gene therapy. (A) Scheme of immunotherapy and gene therapy. (B) Western
blotting of OVA proteins in RAW264.7 and DC2.4, showing the expression of OVA in GLP-O and GLP-RONP groups and H&E images of lung
tissues collected on day 40, showing that the GLP-RO Gel could efficiently prevent the formation of metastasis. Reprinted with permission from ref
118. Copyright 2021, American Chemical Society. (C) The polarization states of macrophages stimulated with supernatants derived from
transfected B16F10 cells were assessed by FCM. The PD-L1 expression levels in tumors subjected to different treatments were assessed by FCM.
Codelivery of CCL19 pDNA and BMS-1 in RGD-DMA nanoparticles reshaped the TME and significantly inhibited the abdominal dissemination
of cancer. Reprinted with permission from ref 121. Copyright 2021, Wiley-VCH. (D) Immunohistochemical images showing the expression of PD-
L1 (brown) in CT26 tumor tissue. Red arrows indicate PD-L1 protein. Western blot analysis of PD-L1 expression in CT26 tumor tissue collected
from mice in the indicated treatment groups and Statistical analysis of the number of TUNEL+ cells per field for B16 and CT26 tumor. Scale bar =
50 μm. Reprinted with permission from ref 123. Copyright 2020, American Chemical Society.

3.4.2. Using an Oncolytic Virus to Combat Tumors. to 2% in the control group.116 Stephen Russell at Mayo Clinic
Immunity therapy is usually combined with oncolytic virus developed a recombinant oncolytic vesicular stomatitis virus
therapy because oncolytic virus (OVs) could induce (VSV) expressing the antiviral cytokine interferon-β to protect
immunogenic cell death, release tumor-associated antigens noncancerous cells from being killed.116,117 However, the
(TAAs), disrupt immunosuppression within tumors, and immune response induced by OVs or the cytokines expressed
promote antitumor immune responses.114 The Phase I study by OVs is likely to be systemic, increasing the risk of OVs’
of Delta-24-RGD oncolytic adenovirus showed a significant toxicity. A modified oncolytic adenovirus Delta-24-RGDOX
antitumor efficiency because of a direct oncolytic effect and a was developed to express an immune costimulatory OX40L
following antitumor immune response.115 The oncolytic virus/ (OX40 ligand) which can bind a unique costimulatory OX40
immune combined therapies of Delta-24-RGD and anti-PD-1 on the T cell membrane and help T cells to recognize TAAs.
antibodies are undergoing Phase II clinical trials (Clinical- The treatment induced complete regression in the long term
Trials.gov identifier: NCT02798406). Through modifying surviving mice treated with the combination. Combining OX,
antitumor cytokine or antiviral cytokine genes, oncolytic immune therapy, and the unique gene OX40L gave Delta-24-
virus therapy combined with gene therapy can improve the RGDOX a specific immune response to cancer cells,
relative dismal efficacy of a single oncolytic virus agent or significantly reducing the potential immunotoxicity of OVs.114
reduce the virus-induced toxicity. Amgen’s T-Vec (talimogene 3.4.3. Nucleic Acid-Based Vaccine. Nanovaccine is a
laherparepvec), the first approved oncolytic virus by FDA, was new therapeutic that can activate the immune system by
modified with the immunostimulatory cytokine granulocyte- presenting an antigen to lymphocytes, thus killing tumor cells.
macrophage colony-stimulating factor (GM-CSF) gene and The former vaccine usually contains an antigen alone or with
gained a highly therapeutic effect in melanoma patients with a an adjuvant, which cannot be recognized by lymphocytes
durable response rate of 16% in advanced melanoma patients effectively. With nanocarriers, vaccines could effectively target
K https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

specific organs and transform them into cells. The nucleic acid accumulation of the OMV-PD1 at the tumor and the vesicles’
vaccine, especially the mRNA vaccine, has become one of the efficient binding to PD-L1 on tumor cells deplete tumor cell
most potent tools against diseases such as covid-19 and cancer PD-L1 and ultimately block the PD1/PD-L1 inhibitory axis.
because of its long-term effect and safety. Once mRNA enters The combination of immune activation and blockage of PD1/
the cell, it can express specific proteins continuously, thus PD-L1 leads to a significant reduction in tumor growth in
making immune cells produce antibodies. A nanovaccine mouse melanoma and colorectal cancer models. The
formed with ovalbumin mRNA and adjuvants (R848) was engineered bacterial extracellular vesicle-based system provides
loaded in injectable hydrogel to gain the enhanced effect.118 an exciting opportunity to further develop tumor immuno-
The nanovaccine could generate an OVA 30 days after one therapy strategies (Figure 4D).
injection and target lymph nodes. Results show that generating In summary, combining immunotherapy with gene therapy
tumor antigen antibodies in the serum could effectively prevent can enhance the therapeutic effect of cancer treatment.
metastasis formation (Figure 4B). To encapsulate individual Oncolytic virus therapy combined with gene therapy has also
tumor mRNA into cationic lipid nanoparticles, the RNA-NPs shown promise in disrupting immunosuppression within
could activate systemic and intratumoral myeloid cells.119 tumors and promoting antitumor immune responses. Nucleic
Positive charge lipid nanoparticles encapsulating untargeted acid-based vaccines, such as mRNA vaccines, have become
tumor RNA can activate the response to immunotherapy. powerful tools against cancer due to their long-term effect and
Codelivery of anti-PD-L1 mAbs and RNA-NPs could increase safety. Changing the tumor microenvironment by gene therapy
peripheral/intratumoral PD-1+ CD8+ cells and elicits anti- can also enhance the effectiveness of immunotherapy.
tumor activity in “cold” tumor models resistant to ICI
monotherapy. Nanovaccine could also work together with
implantable materials to gain an enhanced efficacy. A blood 4. CONCLUSION AND FUTURE PERSPECTIVE
clot scaffold loading liposome vaccine and siRNAs was Combined gene therapy has many advantages that a single
developed to recruit and induce the maturation of DCs and therapeutic cannot achieve, such as preventing tumor meta-
reduce immunosuppressive signals in mature DCs120. By stasis and recurrence, enhanced therapeutic efficiency, etc. By
loading PD-L1/TIM-3 siRNA, the scaffold could reduce the encapsulating drugs into nanocarriers, the delivery system
expression of immunosuppressive molecules in DCs thus could achieve targeting ability, lysosome escape, and other
stimulating a strong immune response against tumors. It can functions. Understanding the mechanisms of synergistic
also treat different tumors by changing tumor antigens. The therapeutic effects is crucial for designing therapeutic schemes
system can be optimized by encapsulating components into the and the forms of nanocarriers. However, there are still several
same nanoparticle in the future.120 barriers to combining gene therapy with nanoplatform. First,
3.4.4. Changing Tumor Microenvironment from Cold the metabolism and toxicity of different nanocarriers must be
to Hot. Besides using nucleic acids to stimulate the immune researched systematically. Although many nanocarriers have
system against the tumor, utilizing ICI, cytokines, and other been researched, a smaller number of carriers have been
immunostimulatory molecules could also heal cancer by the approved by the FDA. Before clinical use, it is crucial to
immune system. However, the immunosuppressive tumor thoroughly investigate the interaction between various nano-
microenvironment significantly limits these traditional immu-
materials and organisms as well as determine whether these
notherapies for their low pH, hypoxia, etc. By transferring a
materials can be degraded in vivo. These factors must be
specific gene or interfering with gene expression, the tumor
clearly understood to ensure safe and effective use. Second, the
microenvironment could be changed, enhancing the effect of
immunotherapy. The nanoparticles named RGD-DMA efficiency of gene transformations still has much room for
encapsulating CCL19-encoding plasmid DNA (CCL19 improvement. How to synthesize a nanocarrier that could
pDNA) and immune checkpoint ligand PD-L1 inhibitor deliver specific nucleic acids into cells with no apparent toxicity
(BMS-1) were developed to overcome the immunosuppressive is a big challenge. Besides, the transform of gene drugs is
TME and gain enhanced efficacy.121 CCL-19 could stimulate T critical for evaluation of dosage of administration and the
cell proliferation, thus increasing IFN-γ secretion in the TME, proportion of drugs contained in the delivery system. Third,
while BMS-1 could inhibit the expression of PD-L1 more the long-term effects of combined therapy should be
efficiently. Nanoparticles could reshape the TME and inhibit investigated, which should observe if gene drugs have other
cancer’s abdominal dissemination (Figure 4C). Tumor- effects on normal organs and if it is suitable for long-term use.
targeted lipid-dendrimer-calcium-phosphate (TT-LDCP) Gene therapy is a highly sought-after treatment option due to
nanoparticles with thymine functionalization were designed its ability to provide a cure with just one dose or treatment,
to gain the enhanced gene delivery capacity and adjuvant unlike other therapies. However, to ensure optimal therapeutic
immune properties by activating the stimulator of interferon results, it is crucial to assess the appropriate interval of time for
genes (STING) pathway.122 TT-LDCP NPs could deliver combined therapy prior to clinical use.
siRNA against PD-L1 and plasmid DNA, which could encode
IL-2 in tumors, activate CD8+ T cells, augment the efficacy of
cancer immunotherapy, and suppress tumor progression. A
■ AUTHOR INFORMATION
Corresponding Author
bacterial outer membrane vesicle-based immunotherapeutic
agent has been studied for tumor immunotherapy.123 It Ying Liu − CAS Key Laboratory for Biomedical Effects of
activates the immune system and disrupts the PD-L1-mediated Nanomaterials and Nanosafety and CAS Center for
immunosuppressive tumor microenvironment. PD1-bearing Excellence in Nanoscience, National Center for Nanoscience
OMVs significantly enhance the antitumor immune response. and Technology of China, Beijing 100190, P.R. China;
OMV-PD1 stimulates the tumor infiltration of immune cells orcid.org/0000-0002-6540-0473; Phone: +8610
and a systemic antitumor immune response. The strong 82545526; Email: liuy@nanoctr.cn
L https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Authors (4) Liu, Y.; Bhattarai, P.; Dai, Z.; Chen, X. Photothermal therapy
and photoacoustic imaging via nanotheranostics in fighting cancer.
Jinhui Lin − CAS Key Laboratory for Biomedical Effects of Chem. Soc. Rev. 2019, 48 (7), 2053−2108.
Nanomaterials and Nanosafety and CAS Center for (5) Ni, D.; Lin, J.; Zhang, N.; Li, S.; Xue, Y.; Wang, Z.; Liu, Q.; Liu,
Excellence in Nanoscience, National Center for Nanoscience K.; Zhang, H.; Zhao, Y.; Chen, C.; Liu, Y. Combinational application
and Technology of China, Beijing 100190, P.R. China; of metal-organic frameworks-based nanozyme and nucleic acid
University of Chinese Academy of Sciences, Beijing 100049, delivery in cancer therapy. WIREs Nanomedicine and Nanobiotechnol-
P.R. China ogy 2022, 14 (3), No. e1773.
Xinlian Wang − CAS Key Laboratory for Biomedical Effects of (6) Huang, J.; Xiao, Z.; Chen, G.; Li, T.; Peng, Y.; Shuai, X. A pH-
Nanomaterials and Nanosafety and CAS Center for sensitive nanomedicine incorporating catalase gene and photo-
Excellence in Nanoscience, National Center for Nanoscience sensitizer augments photodynamic therapy and activates antitumor
and Technology of China, Beijing 100190, P.R. China; immunity. Nano Today 2022, 43, 101390.
(7) Dunbar, C. E.; High, K. A.; Joung, J. K.; Kohn, D. B.; Ozawa, K.;
University of Chinese Academy of Sciences, Beijing 100049,
Sadelain, M. Gene therapy comes of age. Science 2018, 359 (6372),
P.R. China No. eaan4672.
Dongqi Ni − CAS Key Laboratory for Biomedical Effects of (8) D’Aloia, M. M.; Zizzari, I. G.; Sacchetti, B.; Pierelli, L.; Alimandi,
Nanomaterials and Nanosafety and CAS Center for M. CAR-T cells: the long and winding road to solid tumors. Cell
Excellence in Nanoscience, National Center for Nanoscience Death & Disease 2018, 9 (3), 282.
and Technology of China, Beijing 100190, P.R. China; (9) Zhao, N.; Yan, L.; Xue, J.; Zhang, K.; Xu, F.-J. Degradable one-
University of Chinese Academy of Sciences, Beijing 100049, dimensional dextran-iron oxide nanohybrids for MRI-guided syner-
P.R. China gistic gene/photothermal/magnetolytic therapy. Nano Today 2021,
Yandong Chen − CAS Key Laboratory for Biomedical Effects 38, 101118.
of Nanomaterials and Nanosafety and CAS Center for (10) Li, Y.; Liu, L.; Ji, W.; Peng, H.; Zhao, R.; Zhang, X. Strategies
Excellence in Nanoscience, National Center for Nanoscience and materials of ″SMART″ non-viral vectors: Overcoming the
barriers for brain gene therapy. Nano Today 2020, 35, 101006.
and Technology of China, Beijing 100190, P.R. China
(11) Qiu, N.; Wang, G.; Wang, J.; Zhou, Q.; Guo, M.; Wang, Y.; Hu,
Chunying Chen − CAS Key Laboratory for Biomedical Effects X.; Zhou, H.; Bai, R.; You, M.; Zhang, Z.; Chen, C.; Liu, Y.; Shen, Y.
of Nanomaterials and Nanosafety and CAS Center for Tumor-Associated Macrophage and Tumor-Cell Dually Transfecting
Excellence in Nanoscience, National Center for Nanoscience Polyplexes for Efficient Interleukin-12 Cancer Gene Therapy. Adv.
and Technology of China, Beijing 100190, P.R. China; Mater. 2021, 33 (2), 2006189.
University of Chinese Academy of Sciences, Beijing 100049, (12) Song, G.; Cheng, L.; Chao, Y.; Yang, K.; Liu, Z. Emerging
P.R. China; orcid.org/0000-0002-6027-0315 Nanotechnology and Advanced Materials for Cancer Radiation
Therapy. Adv. Mater. 2017, 29 (32), 1700996.
Complete contact information is available at: (13) Shen, J.; Zhang, W.; Qi, R.; Mao, Z.-W.; Shen, H. Engineering
https://pubs.acs.org/10.1021/acsmaterialsau.3c00035 functional inorganic-organic hybrid systems: advances in siRNA
therapeutics. Chem. Soc. Rev. 2018, 47 (6), 1969−1995.
Author Contributions (14) Teo, P. Y.; Cheng, W.; Hedrick, J. L.; Yang, Y. Y. Co-delivery of
CRediT: Jinhui Lin formal analysis (equal), validation (equal), drugs and plasmid DNA for cancer therapy. Adv. Drug Delivery Rev.
2016, 98, 41−63.
writing-original draft (lead); Xinlian Wang resources (support- (15) Jiang, D. M.; Gupta, S.; Kitchlu, A.; Meraz-Munoz, A.; North, S.
ing); Dongqi Ni writing-original draft (supporting); Yandong A.; Alimohamed, N. S.; Blais, N.; Sridhar, S. S. Defining cisplatin
Chen resources (supporting), funding acquisition (support- eligibility in patients with muscle-invasive bladder cancer. Nature
ing); Chunying Chen funding acquisition (lead), writing- Reviews Urology 2021, 18 (2), 104−114.
review & editing (equal); Ying Liu conceptualization (lead), (16) Liu, S.; Khan, A. R.; Yang, X.; Dong, B.; Ji, J.; Zhai, G. The
funding acquisition (equal), writing-review & editing (lead). reversal of chemotherapy-induced multidrug resistance by nano-
medicine for cancer therapy. J. Controlled Release 2021, 335, 1−20.
Notes (17) Belkahla, H.; Herlem, G.; Picaud, F.; Gharbi, T.; Hémadi, M.;
The authors declare no competing financial interest. Ammar, S.; Micheau, O. TRAIL-NP hybrids for cancer therapy: a
review. Nanoscale 2017, 9 (18), 5755−5768.

■ ACKNOWLEDGMENTS
This work was financially supported by the National Natural
(18) Zhong, H.-h.; Wang, H.-y.; Li, J.; Huang, Y.-z. TRAIL-based
gene delivery and therapeuticstrategies. Acta Pharmacologica Sinica
2019, 40 (11), 1373−1385.
(19) Xu, F.; Zhong, H.; Chang, Y.; Li, D.; Jin, H.; Zhang, M.; Wang,
Science Foundation of China (31971318, 22027810, H.; Jiang, C.; Shen, Y.; Huang, Y. Targeting death receptors for drug-
32101091) and China Postdoctoral Science Foundation resistant cancer therapy: Codelivery of pTRAIL and monensin using
(2021M690043). dual-targeting and stimuli-responsive self-assembling nanocomposites.
Biomaterials 2018, 158, 56−73.
■ REFERENCES
(1) Siegel, R. L.; Miller, K. D.; Fuchs, H. E.; Jemal, A. Cancer
(20) Amreddy, N.; Babu, A.; Panneerselvam, J.; Srivastava, A.;
Muralidharan, R.; Chen, A.; Zhao, Y. D.; Munshi, A.; Ramesh, R.
Chemo-biologic combinatorial drug delivery using folate receptor-
statistics, 2022. CA: A Cancer Journal for Clinicians 2022, 72 (1), 7− targeted dendrimer nanoparticles for lung cancer treatment. Nano-
33. medicine: Nanotechnology, Biology and Medicine 2018, 14 (2), 373−
(2) Miller, K. D.; Nogueira, L.; Mariotto, A. B.; Rowland, J. H.; 384.
Yabroff, K. R.; Alfano, C. M.; Jemal, A.; Kramer, J. L.; Siegel, R. L. (21) Qiu, L.; Chen, T.; Ocsoy, I.; Yasun, E.; Wu, C.; Zhu, G.; You,
Cancer treatment and survivorship statistics, 2019. CA: A Cancer M.; Han, D.; Jiang, J.; Yu, R.; Tan, W. A cell-targeted, size-
Journal for Clinicians 2019, 69 (5), 363−385. photocontrollable, nuclear-uptake nanodrug delivery system for drug-
(3) Pan, L.; Liu, J.; Shi, J. Cancer cell nucleus-targeting resistant cancer therapy. Nano Lett. 2015, 15 (1), 457−63.
nanocomposites for advanced tumor therapeutics. Chem. Soc. Rev. (22) Shu, J.; Li, X.; Dang, J.; Liu, Y.; Duan, S.; Zhu, R.; Yin, L.;
2018, 47 (18), 6930−6946. Chen, Y. Drug Resistance Reversal by Interventing Cancer

M https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Bioenergetics with Spherical Helical Polypeptide-Potented Gene (39) Qin, L.; Jiang, S.; He, H.; Ling, G.; Zhang, P. Functional black
Silencing. Chemical Engineering Journal 2021, 414 (10), 128545. phosphorus nanosheets for cancer therapy. J. Controlled Release 2020,
(23) Bukowski, K.; Kciuk, M.; Kontek, R. Mechanisms of Multidrug 318, 50−66.
Resistance in Cancer Chemotherapy. International Journal of (40) Fusco, L.; Gazzi, A.; Peng, G.; Shin, Y.; Vranic, S.; Bedognetti,
Molecular Sciences 2020, 21 (9), 3233. D.; Vitale, F.; Yilmazer, A.; Feng, X.; Fadeel, B.; Casiraghi, C.; Delogu,
(24) Zhang, L.; Mu, C.; Zhang, T.; Yang, D.; Wang, C.; Chen, Q.; L. G. Graphene and other 2D materials: a multidisciplinary analysis to
Tang, L.; Fan, L.; Liu, C.; Shen, J.; Li, H. Development of targeted uncover the hidden potential as cancer theranostics. Theranostics
therapy therapeutics to sensitize triple-negative breast cancer 2020, 10 (12), 5435−5488.
chemosensitivity utilizing bacteriophage phi29 derived packaging (41) Kim, J.; Kim, J.; Jeong, C.; Kim, W. J. Synergistic nanomedicine
RNA. J. Nanobiotechnol. 2021, 19 (1), 13. by combined gene and photothermal therapy. Adv. Drug Delivery Rev.
(25) Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. 2016, 98, 99−112.
Role of hypoxia in cancer therapy by regulating the tumor (42) Kim, N. Y.; Blake, S.; De, D.; Ouyang, J.; Shi, J.; Kong, N. Two-
microenvironment. Molecular Cancer 2019, 18 (1), 157. Dimensional Nanosheet-Based Photonic Nanomedicine for Com-
(26) Xie, Z.; Guo, W.; Guo, N.; Huangfu, M.; Liu, H.; Lin, M.; Xu, bined Gene and Photothermal Therapy. Frontiers in Pharmacology
W.; Chen, J.; Wang, T.; Wei, Q.; Han, M.; Gao, J. Targeting tumor 2020, 10, na DOI: 10.3389/fphar.2019.01573.
hypoxia with stimulus-responsive nanocarriers in overcoming drug (43) Cai, Y.; Ni, D.; Cheng, W.; Ji, C.; Wang, Y.; Müllen, K.; Su, Z.;
resistance and monitoring anticancer efficacy. Acta biomaterialia 2018, Liu, Y.; Chen, C.; Yin, M. Enzyme-Triggered Disassembly of Perylene
71, 351. Monoimide-based Nanoclusters for Activatable and Deep Photo-
(27) Valkenburg, K. C.; de Groot, A. E.; Pienta, K. J. Targeting the dynamic Therapy. Angew. Chem., Int. Ed. 2020, 59 (33), 14014−
tumour stroma to improve cancer therapy. Nat. Rev. Clin Oncol 2018, 14018.
15 (6), 366−381. (44) Chen, W. H.; Luo, G. F.; Zhang, X. Z. Recent Advances in
(28) Ni, Y.; Zhou, X.; Yang, J.; Shi, H.; Li, H.; Zhao, X.; Ma, X. The Subcellular Targeted Cancer Therapy Based on Functional Materials.
Role of Tumor-Stroma Interactions in Drug Resistance Within Adv. Mater. 2019, 31 (3), No. 1802725.
Tumor Microenvironment. Frontiers in Cell and Developmental Biology (45) Nam, J.; Son, S.; Ochyl, L. J.; Kuai, R.; Schwendeman, A.;
2021, 9 (1206), na DOI: 10.3389/fcell.2021.637675. Moon, J. J. Chemo-photothermal therapy combination elicits anti-
(29) Larionova, I.; Cherdyntseva, N.; Liu, T.; Patysheva, M.; Rakina, tumor immunity against advanced metastatic cancer. Nat. Commun.
M.; Kzhyshkowska, J. Interaction of tumor-associated macrophages 2018, 9 (1), 1074.
and cancer chemotherapy. OncoImmunology 2019, 8 (7), (46) Min, K. H.; Kim, Y. H.; Wang, Z.; Kim, J.; Kim, J. S.; Kim, S.
No. e1596004. H.; Kim, K.; Kwon, I. C.; Kiesewetter, D. O.; Chen, X. Engineered
(30) Parayath, N. N.; Gandham, S. K.; Leslie, F.; Amiji, M. M. Zn(II)-Dipicolylamine-Gold Nanorod Provides Effective Prostate
Improved anti-tumor efficacy of paclitaxel in combination with Cancer Treatment by Combining siRNA Delivery and Photothermal
MicroRNA-125b-based tumor-associated macrophage repolarization Therapy. Theranostics 2017, 7 (17), 4240−4254.
in epithelial ovarian cancer - ScienceDirect. Cancer letters 2019, 461, (47) Zhang, Y.; Ren, K.; Zhang, X.; Chao, Z.; Yang, Y.; Ye, D.; Dai,
1−9. Z.; Liu, Y.; Ju, H. Photo-tearable tape close-wrapped upconversion
(31) Tcyganov, E.; Mastio, J.; Chen, E.; Gabrilovich, D. I. Plasticity nanocapsules for near-infrared modulated efficient siRNA delivery
of myeloid-derived suppressor cells in cancer. Current Opinion in and therapy. Biomaterials 2018, 163, 55−66.
Immunology 2018, 51, 76−82. (48) Wang, S. J.; Tian, Y.; Tian, W.; Sun, J.; Zhao, S.; Liu, Y.; Wang,
(32) Chen, Y.; Sun, J.; Huang, Y.; Liu, Y.; Liang, L.; Yang, D.; Lu, B.; C. Y.; Tang, Y. X.; Ma, X. Q.; Teng, Z. G.; Lu, G. M. Selectively
Li, S. Targeted codelivery of doxorubicin and IL-36γ expression Sensitizing Malignant Cells to Photothermal Therapy Using a CD44-
plasmid for an optimal chemo-gene combination therapy against Targeting Heat Shock Protein 72 Depletion Nanosystem. ACS Nano
cancer lung metastasis. Nanomedicine: Nanotechnology, Biology and 2016, 10 (9), 8578−8590.
Medicine 2019, 15, 129−141. (49) Chu, C.; Ren, E.; Zhang, Y.; Yu, J.; Lin, H.; Pang, X.; Zhang, Y.;
(33) Hwang, R. F.; Moore, T.; Arumugam, T.; Ramachandran, V.; Liu, H.; Qin, Z.; Cheng, Y.; Wang, X.; Li, W.; Kong, X.; Chen, X.; Liu,
Amos, K. D.; Rivera, A.; Ji, B.; Evans, D. B.; Logsdon, C. D. Cancer- G. Zinc(II)-Dipicolylamine Coordination Nanotheranostics: Toward
associated stromal fibroblasts promote pancreatic tumor progression. Synergistic Nanomedicine by Combined Photo/Gene Therapy.
Cancer Res. 2008, 68 (3), 918−26. Angew. Chem., Int. Ed. Engl. 2019, 58 (1), 269−272.
(34) Chen, X.; Jia, F.; Li, Y.; Deng, Y.; Huang, Y.; Liu, W.; Jin, Q.; Ji, (50) Taghavi, S.; Abnous, K.; Taghdisi, S. M.; Ramezani, M.;
J. Nitric oxide-induced stromal depletion for improved nanoparticle Alibolandi, M. Hybrid carbon-based materials for gene delivery in
penetration in pancreatic cancer treatment. Biomaterials 2020, 246, cancer therapy. J. Controlled Release 2020, 318, 158−175.
119999. (51) Zhang, Y.; Yang, L.; Yang, C.; Liu, J. Recent advances of smart
(35) Han, X.; Li, Y.; Xu, Y.; Zhao, X.; Zhang, Y.; Yang, X.; Wang, Y.; acid-responsive gold nanoparticles in tumor therapy. Wiley Interdiscip
Zhao, R.; Anderson, G. J.; Zhao, Y.; Nie, G. Reversal of pancreatic Rev. Nanomed Nanobiotechnol 2020, 12, No. e1619.
desmoplasia by re-educating stellate cells with a tumour micro- (52) Wang, Z. Q.; Wang, L. C.; Prabhakar, N.; Xing, Y. X.;
environment-activated nanosystem. Nat. Commun. 2018, 9 (1), 3390. Rosenholm, J. M.; Zhang, J. X.; Cai, K. Y. CaP coated mesoporous
(36) Li, Y.; Zhao, Z.; Lin, C.-Y.; Liu, Y.; Staveley-OCarroll, K. F.; Li, polydopamine nanoparticles with responsive membrane permeation
G.; Cheng, K. Silencing PCBP2 normalizes desmoplastic stroma and ability for combined photothermal and siRNA therapy. Acta
improves the antitumor activity of chemotherapy in pancreatic cancer. Biomaterialia 2019, 86, 416−428.
Theranostics 2021, 11 (5), 2182−2200. (53) Altieri, D. C. Validating survivin as a cancer therapeutic target.
(37) Wu, F.; Yang, J.; Liu, J.; Wang, Y.; Mu, J.; Zeng, Q.; Deng, S.; Nat. Rev. Cancer 2003, 3 (1), 46−54.
Zhou, H. Signaling pathways in cancer-associated fibroblasts and (54) Chu, C.; Su, M.; Zhu, J.; Li, D.; Cheng, H.; Chen, X.; Liu, G.
targeted therapy for cancer. Signal Transduction and Targeted Therapy Metal-Organic Framework Nanoparticle-Based Biomineralization: A
2021, 6 (1), 218. New Strategy toward Cancer Treatment. Theranostics 2019, 9 (11),
(38) Saatci, O.; Kaymak, A.; Raza, U.; Ersan, P. G.; Akbulut, O.; 3134−3149.
Banister, C. E.; Sikirzhytski, V.; Tokat, U. M.; Aykut, G.; Ansari, S. A.; (55) Shao, J.; Xie, H.; Huang, H.; Li, Z.; Sun, Z.; Xu, Y.; Xiao, Q.;
Dogan, H. T.; Dogan, M.; Jandaghi, P.; Isik, A.; Gundogdu, F.; Yu, X. F.; Zhao, Y.; Zhang, H.; Wang, H.; Chu, P. K. Biodegradable
Kosemehmetoglu, K.; Dizdar, O.; Aksoy, S.; Akyol, A.; Uner, A.; black phosphorus-based nanospheres for in vivo photothermal cancer
Buckhaults, P. J.; Riazalhosseini, Y.; Sahin, O. Targeting lysyl oxidase therapy. Nat. Commun. 2016, 7, 12967.
(LOX) overcomes chemotherapy resistance in triple negative breast (56) Wang, H.; Yang, X. Z.; Shao, W.; Chen, S. C.; Xie, J. F.; Zhang,
cancer. Nat. Commun. 2020, 11 (1), 2416. X. D.; Wang, J.; Xie, Y. Ultrathin Black Phosphorus Nanosheets for

N https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

Efficient Singlet Oxygen Generation. J. Am. Chem. Soc. 2015, 137 (73) Martin, S. J.; Henry, C. M.; Cullen, S. P. A Perspective on
(35), 11376−11382. Mammalian Caspases as Positive and Negative Regulators of
(57) Wang, H.; Zhong, L.; Liu, Y.; Xu, X.; Xing, C.; Wang, M.; Bai, Inflammation. Mol. Cell 2012, 46 (4), 387−397.
S. M.; Lu, C. H.; Yang, H. H. A black phosphorus nanosheet-based (74) Bonfil, R. D.; Bustuoabad, O. D.; Ruggiero, R. A.; Meiss, R. P.;
siRNA delivery system for synergistic photothermal and gene therapy. Pasqualini, C. D. Tumor Necrosis Can Facilitate the Appearance of
Chem. Commun. (Camb) 2018, 54 (25), 3142−3145. Metastases. Clin. Exp. Metastasis 1988, 6 (2), 121−129.
(58) Wang, Z.; Li, S.; Zhang, M.; Ma, Y.; Liu, Y.; Gao, W.; Zhang, J.; (75) Li, W. T.; Peng, J. R.; Tan, L. W.; Wu, J.; Shi, K.; Qu, Y.; Wei,
Gu, Y. Laser-Triggered Small Interfering RNA Releasing Gold X. W.; Qian, Z. Y. Mild photothermal therapy/photodynamic
Nanoshells against Heat Shock Protein for Sensitized Photothermal therapy/chemotherapy of breast cancer by Lyp-1 modified Docetax-
Therapy. Adv. Sci. (Weinh) 2017, 4 (2), 1600327. el/IR820 Co-loaded micelles. Biomaterials 2016, 106, 119−133.
(59) Tang, W.; Han, L.; Lu, X.; Wang, Z.; Liu, F.; Li, Y.; Liu, S.; Liu, (76) Ding, Y.; Du, C.; Qian, J. W.; Dong, C. M. NIR-Responsive
S.; Tian, R.; Liu, J.; Ding, B. A Nucleic Acid/Gold Nanorod-Based Polypeptide Nanocomposite Generates NO Gas, Mild Photothermia,
Nanoplatform for Targeted Gene Editing and Combined Tumor and Chemotherapy to Reverse Multidrug-Resistant Cancer. Nano
Therapy. ACS Appl. Mater. Interfaces 2021, 13 (18), 20974−20981. Lett. 2019, 19 (7), 4362−4370.
(60) Yang, Y.; Han, Y.; Sun, Q.; Cheng, J.; Yue, C.; Liu, Y.; Song, J.; (77) Yang, Y.; Zhu, W.; Dong, Z.; Chao, Y.; Xu, L.; Chen, M.; Liu,
Jin, W.; Ding, X.; de la Fuente, J. M.; Ni, J.; Wang, X.; Cui, D. Au- Z. 1D Coordination Polymer Nanofibers for Low-Temperature
siRNA@ aptamer nanocages as a high-efficiency drug and gene Photothermal Therapy. Adv. Mater. 2017, 29 (40), 1703588.
delivery system for targeted lung cancer therapy. J. Nanobiotechnol. (78) Zhou, J.; Li, M. H.; Hou, Y. H.; Luo, Z.; Chen, Q. F.; Cao, H.
2021, 19 (1), 54. X.; Huo, R. L.; Xue, C. C.; Sutrisno, L.; Hao, L.; Cao, Y.; Ran, H. T.;
(61) Deng, S. H.; Wang, S. Y.; Xiao, Z. C.; Cheng, D. Lu, L.; Li, K.; Cai, K. Y. Engineering of a Nanosized Biocatalyst for
Unprotonatable and ROS-Sensitive Nanocarrier for NIR Spatially Combined Tumor Starvation and Low-Temperature Photothermal
Activated siRNA Therapy with Synergistic Drug Effect. SMALL 2022, Therapy. ACS Nano 2018, 12 (3), 2858−2872.
18 (41), 2203823. (79) Ding, F.; Mou, Q. B.; Ma, Y.; Pan, G. F.; Guo, Y. Y.; Tong, G.
(62) Li, G. C.; Mivechi, N. F.; Weitzel, G. Heat-Shock Proteins, S.; Choi, C. H. J.; Zhu, X. Y.; Zhang, C. A Crosslinked Nucleic Acid
Thermotolerance, And Their Relevance to Clinical Hyperthermia. Int. Nanogel for Effective siRNA Delivery and Antitumor Therapy. Angew.
J. Hyperthermia 1995, 11 (4), 459−488. Chem.-Int. Ed. 2018, 57 (12), 3064−3068.
(63) Ding, F.; Gao, X. H.; Huang, X. G.; Ge, H.; Xie, M.; Qian, J. (80) Jang, Y.; Kim, D.; Lee, H.; Jang, H.; Park, S.; Kim, G. E.; Lee,
W.; Song, J.; Li, Y. H.; Zhu, X. Y.; Zhang, C. Polydopamine-coated H. J.; Kim, H. J.; Kim, H. Development of an ultrasound triggered
nucleic acid nanogel for siRNA-mediated low-temperature photo- nanomedicine-microbubble complex for chemo-photodynamic-gene
therapy. Nanomedicine: Nanotechnology, Biology and Medicine 2020,
thermal therapy. Biomaterials 2020, 245, 119976.
(64) Dang, J.; Ye, H.; Li, Y.; Liang, Q.; Li, X.; Yin, L. Multivalency- 27, 102194.
(81) Pu, Y.; Wu, W.; Xiang, H.; Chen, Y.; Xu, H. CRISPR/Cas9-
assisted membrane-penetrating siRNA delivery sensitizes photo-
based genome editing for multimodal synergistic cancer nanotherapy.
thermal ablation via inhibition of tumor glycolysis metabolism.
Nano Today 2023, 48, 101734.
Biomaterials 2019, 223, 119463.
(82) Haque, F.; Shu, D.; Shu, Y.; Shlyakhtenko, L. S.; Rychahou, P.
(65) Chen, W. H.; Luo, G. F.; Lei, Q.; Hong, S.; Qiu, W. X.; Liu, L.
G.; Mark Evers, B.; Guo, P. Ultrastable synergistic tetravalent RNA
H.; Cheng, S. X.; Zhang, X. Z. Overcoming the Heat Endurance of
nanoparticles for targeting to cancers. Nano Today 2012, 7 (4), 245−
Tumor Cells by Interfering with the Anaerobic Glycolysis Metabolism
257.
for Improved Photothermal Therapy. ACS Nano 2017, 11 (2), 1419− (83) Jin, Y.; Wang, H.; Li, X.; Zhu, H.; Sun, D.; Sun, X.; Liu, H.;
1431. Zhang, Z.; Cao, L.; Gao, C.; Wang, H.; Liang, X.-J.; Zhang, J.; Yang,
(66) Kogel, D.; Linder, B.; Brunschweiger, A.; Chines, S.; Behl, C. At X. Multifunctional DNA Polymer-Assisted Upconversion Therapeutic
the Crossroads of Apoptosis and Autophagy: Multiple Roles of the Nanoplatform for Enhanced Photodynamic Therapy. ACS Appl.
Co-Chaperone BAG3 in Stress and Therapy Resistance of Cancer. Mater. Interfaces 2020, 12 (24), 26832−26841.
Cells 2020, 9 (3), 574. (84) Lal, S.; Clare, S. E.; Halas, N. J. Nanoshell-Enabled
(67) Rauch, J. N.; Tse, E.; Freilich, R.; Mok, S. A.; Makley, L. N.; Photothermal Cancer Therapy: Impending Clinical Impact. Acc.
Southworth, D. R.; Gestwicki, J. E. BAG3 Is a Modular, Scaffolding Chem. Res. 2008, 41 (12), 1842−1851.
Protein that physically Links Heat Shock Protein 70 (Hsp70) to the (85) Liu, T.; Li, W.; Lu, W.; Chen, M.; Luo, M.; Zhang, C.; Li, Y.;
Small Heat Shock Proteins. J. Mol. Biol. 2017, 429 (1), 128−141. Qin, G.; Shi, D.; Xiao, B.; Qiu, H.; Yu, W.; Kang, L.; Kang, T.; Huang,
(68) Fang, X.; Bogomolovas, J.; Wu, T. B.; Zhang, W.; Liu, C. Z.; W.; Yu, X.; Wu, X.; Deng, W. RBFOX3 Promotes Tumor Growth and
Veevers, J.; Stroud, M. J.; Zhang, Z. Y.; Ma, X. L.; Mu, Y. X.; Lao, D. Progression via hTERT Signaling and Predicts a Poor Prognosis in
H.; Dalton, N. D.; Gu, Y. S.; Wang, C. L.; Wang, M.; Liang, Y.; Lange, Hepatocellular Carcinoma. Theranostics 2017, 7 (12), 3138−3154.
S.; Ouyang, K. F.; Peterson, K. L.; Evans, S. M.; Chen, J. Loss-of- (86) Chen, L.; Chen, C.; Chen, W.; Li, K.; Chen, X.; Tang, X.; Xie,
function mutations in co-chaperone BAG3 destabilize small HSPs and G.; Luo, X.; Wang, X.; Liang, H.; Yu, S. Biodegradable Black
cause cardiomyopathy. J. Clin. Invest. 2017, 127 (8), 3189−3200. Phosphorus Nanosheets Mediate Specific Delivery of hTERT siRNA
(69) Wang, B. K.; Yu, X. F.; Wang, J. H.; Li, Z. B.; Li, P. H.; Wang, for Synergistic Cancer Therapy. ACS Appl. Mater. Interfaces 2018, 10
H. Y.; Song, L.; Chu, P. K.; Li, C. Z. Gold-nanorods-siRNA nanoplex (25), 21137−21148.
for improved photothermal therapy by gene silencing. Biomaterials (87) Qiao, S.; Xin, F.; Wu, M.; Zheng, Y.; Zhao, B.; Zhang, C.; Liu,
2016, 78, 27−39. X.; Wei, Z.; Liu, J. A remotely controlled NIR-II photothermal-
(70) He, X.; Hao, Y.; Chu, B.; Yang, Y.; Sun, A.; Shi, K.; Yang, C.; sensitive transgene system for hepatocellular carcinoma synergistic
Zhou, K.; Qu, Y.; Li, H.; Qian, Z. Redox–activatable photothermal therapy. J. Mater. Chem. B 2021, 9 (25), 5083−5091.
therapy and enzyme-mediated tumor starvation for synergistic cancer (88) Zhou, R.; Liu, X.; Wu, Y.; Xiang, H.; Cao, J.; Li, Y.; Yin, W.; Zu,
therapy. Nano Today 2021, 39, 101174. Y.; Li, J.; Liu, R.; Zhao, F.; Liu, Z.; Chen, C.; Gu, Z.; Yan, L.; Zhao, Y.
(71) Israelsen, W. J.; Vander Heiden, M. G. Pyruvate kinase: Suppressing the Radiation-Induced Corrosion of Bismuth Nano-
Function, regulation and role in cancer. Semin. Cell Dev. Biol. 2015, particles for Enhanced Synergistic Cancer Radiophototherapy. ACS
43, 43−51. Nano 2020, 14 (10), 13016−13029.
(72) Melamed, J. R.; Edelstein, R. S.; Day, E. S. Elucidating the (89) Wang, J.; He, H.; Xu, X.; Wang, X.; Chen, Y.; Yin, L. Far-red
Fundamental Mechanisms of Cell Death Triggered by Photothermal light-mediated programmable anti-cancer gene delivery in coopera-
Therapy. ACS Nano 2015, 9 (1), 6−11. tion with photodynamic therapy. Biomaterials 2018, 171, 72−82.

O https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX
ACS Materials Au pubs.acs.org/materialsau Review

(90) Zhang, X.; Yang, Y.; Kang, T.; Wang, J.; Yang, G.; Yang, Y.; Lin, (108) Roush, S.; Slack, F. J. The let-7 family of microRNAs. Trends
X.; Wang, L.; Li, K.; Liu, J.; Ni, J.-S. NIR-II Absorbing Semi- Cell Biol. 2008, 18 (10), 505−16.
conducting Polymer-Triggered Gene-Directed Enzyme Prodrug (109) Yin, P. T.; Shah, B. P.; Lee, K. B. Combined magnetic
Therapy for Cancer Treatment. Small 2021, 17 (23), 2100501. nanoparticle-based microRNA and hyperthermia therapy to enhance
(91) Revia, R. A.; Zhang, M. Magnetite nanoparticles for cancer apoptosis in brain cancer cells. Small 2014, 10 (20), 4106−4112.
diagnosis, treatment, and treatment monitoring: recent advances. (110) Yin, P. T.; Shah, S.; Pasquale, N. J.; Garbuzenko, O. B.;
Mater. Today (Kidlington) 2016, 19 (3), 157−168. Minko, T.; Lee, K.-B. Stem cell-based gene therapy activated using
(92) Tay, Z. W.; Chandrasekharan, P.; Chiu-Lam, A.; Hensley, D. magnetic hyperthermia to enhance the treatment of cancer.
W.; Dhavalikar, R.; Zhou, X. Y.; Yu, E. Y.; Goodwill, P. W.; Zheng, B.; Biomaterials 2016, 81, 46−57.
Rinaldi, C.; Conolly, S. M. Magnetic Particle Imaging-Guided Heating (111) Kennedy, L. B.; Salama, A. K. S. A review of cancer
in Vivo Using Gradient Fields for Arbitrary Localization of Magnetic immunotherapy toxicity. CA Cancer J. Clin 2020, 70 (2), 86−104.
Hyperthermia Therapy. ACS Nano 2018, 12 (4), 3699−3713. (112) Bhatia, A.; Kumar, Y. Cellular and molecular mechanisms in
(93) Kumar, C. S.; Mohammad, F. Magnetic nanomaterials for cancer immune escape: a comprehensive review. Expert Rev. Clin
hyperthermia-based therapy and controlled drug delivery. Adv. Drug Immunol 2014, 10 (1), 41−62.
Deliv Rev. 2011, 63 (9), 789−808. (113) Dai, H.; Fan, Q.; Wang, C. Recent applications of
(94) Liu, X.; Zhang, Y.; Wang, Y.; Zhu, W.; Li, G.; Ma, X.; Zhang, immunomodulatory biomaterials for disease immunotherapy. Explo-
Y.; Chen, S.; Tiwari, S.; Shi, K.; Zhang, S.; Fan, H. M.; Zhao, Y. X.; ration 2022, 2 (6), 20210157−20210157.
Liang, X. J. Comprehensive understanding of magnetic hyperthermia (114) Jiang, H.; Rivera-Molina, Y.; Gomez-Manzano, C.; Clise-
for improving antitumor therapeutic efficacy. Theranostics 2020, 10 Dwyer, K.; Bover, L.; Vence, L. M.; Yuan, Y.; Lang, F. F.; Toniatti, C.;
(8), 3793−3815. Hossain, M. B.; Fueyo, J. Oncolytic Adenovirus and Tumor-Targeting
(95) Jordan, A.; Scholz, R.; Wust, P.; Fahling, H.; Felix, R. Magnetic Immune Modulatory Therapy Improve Autologous Cancer Vacci-
fluid hyperthermia (MFH): Cancer treatment with AC magnetic field nation. Cancer Res. 2017, 77 (14), 3894−3907.
induced excitation of biocompatible superparamagnetic nanoparticles. (115) Lang, F. F.; Conrad, C.; Gomez-Manzano, C.; Yung, W. K. A.;
J. Magn. Magn. Mater. 1999, 201, 413−419. Sawaya, R.; Weinberg, J. S.; Prabhu, S. S.; Rao, G.; Fuller, G. N.;
(96) Hergt, R.; Dutz, S. Magnetic particle hyperthermia� Aldape, K. D.; Gumin, J.; Vence, L. M.; Wistuba, I.; Rodriguez-
biophysical limitations of a visionary tumour therapy. J. Magn. Canales, J.; Villalobos, P. A.; Dirven, C. M. F.; Tejada, S.; Valle, R. D.;
Magn. Mater. 2007, 311 (1), 187−192. Alonso, M. M.; Ewald, B.; Peterkin, J. J.; Tufaro, F.; Fueyo, J. Phase I
(97) Cheng, L.; Ke, Y.; Yu, S.; Jing, J. Co-delivery of doxorubicin and Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus:
recombinant plasmid pHSP70-Plk1-shRNA by bacterial magneto- Replication and Immunotherapeutic Effects in Recurrent Malignant
somes for osteosarcoma therapy. Int. J. Nanomedicine 2016, 11, 5277− Glioma. J. Clin Oncol 2018, 36 (14), 1419−1427.
5286. (116) Sheridan, C. First oncolytic virus edges towards approval in
(98) Ito, A.; Shinkai, M.; Honda, H.; Kobayashi, T. Heat-inducible surprise vote. Nat. Biotechnol. 2015, 33 (6), 569−570.
TNF-alpha gene therapy combined with hyperthermia using magnetic (117) Heiber, J.; Hyun, J.; Obuchi, M.; Barber, G. N. Development
nanoparticles as a novel tumor-targeted therapy. Cancer Gene Ther. of recombinant vesicular stomatitis virus for use as an oncolytic vector
2001, 8 (9), 649−654. in cancer therapy. Cytokine 2009, 48 (1−2), 47−47.
(99) Walther, W.; Stein, U. Heat-responsive gene expression for (118) Yin, Y.; Li, X.; Ma, H.; Zhang, J.; Yu, D.; Zhao, R.; Yu, S.; Nie,
gene therapy. Adv. Drug Deliv Rev. 2009, 61 (7−8), 641−9. G.; Wang, H. In Situ Transforming RNA Nanovaccines from
(100) Zhang, Z. Q.; Song, S. C. Multiple hyperthermia-mediated Polyethylenimine Functionalized Graphene Oxide Hydrogel for
release of TRAIL/SPION nanocomplex from thermosensitive Durable Cancer Immunotherapy. Nano Lett. 2021, 21 (5), 2224−
polymeric hydrogels for combination cancer therapy. Biomaterials 2231.
2017, 132, 16−27. (119) Sayour, E. J.; Grippin, A.; De Leon, G.; Stover, B.; Rahman,
(101) Moros, M.; Idiago-Lopez, J.; Asin, L.; Moreno-Antolin, E.; M.; Karachi, A.; Wummer, B.; Moore, G.; Castillo-Caro, P.;
Beola, L.; Grazu, V.; Fratila, R. M.; Gutierrez, L.; de la Fuente, J. M. Fredenburg, K.; Sarkisian, M. R.; Huang, J.; Deleyrolle, L. P.;
Triggering antitumoural drug release and gene expression by magnetic Sahay, B.; Carrera-Justiz, S.; Mendez-Gomez, H. R.; Mitchell, D. A.
hyperthermia. Adv. Drug Deliv Rev. 2019, 138, 326−343. Personalized Tumor RNA Loaded Lipid-Nanoparticles Prime the
(102) Yu, S.; Yi, M.; Qin, S.; Wu, K. Next generation chimeric Systemic and Intratumoral Milieu for Response to Cancer
antigen receptor T cells: safety strategies to overcome toxicity. Mol. Immunotherapy. Nano Lett. 2018, 18 (10), 6195−6206.
Cancer 2019, 18 (1), 125. (120) Chen, Y.; Zhang, Y.; Wang, B.; Fan, Q.; Yang, Q.; Xu, J.; Dai,
(103) Yuan, C.; An, Y.; Zhang, J.; Li, H.; Zhang, H.; Wang, L.; H.; Xu, F.; Wang, C. Blood Clot Scaffold Loaded with Liposome
Zhang, D. Magnetic nanoparticles for targeted therapeutic gene Vaccine and siRNAs Targeting PD-L1 and TIM-3 for Effective DC
delivery and magnetic-inducing heating on hepatoma. Nanotechnology Activation and Cancer Immunotherapy. ACS Nano 2023, 17 (1),
2014, 25 (34), 345101. 760−774.
(104) Wang, Z.; Chang, Z. M.; Lu, M. M.; Shao, D.; Yue, J.; Yang, D. (121) Yu, T.; Nie, W.; Hong, Z.; He, Y.; Chen, J.; Mi, X.; Yang, S.;
Li, X.; Wang, B.; Lin, Y.; Gao, X. Synergy of Immunostimulatory
A.; Zheng, X.; Li, M. Q.; He, K.; Zhang, M.; Chen, L.; Dong, W. F.
Genetherapy with Immune Checkpoint Blockade Motivates Immune
Shape-controlled magnetic mesoporous silica nanoparticles for
Response to Eliminate Cancer. Adv. Funct. Mater. 2021, 31 (22),
magnetically-mediated suicide gene therapy of hepatocellular
2100715.
carcinoma. Biomaterials 2018, 154, 147−157.
(122) Huang, K.-W.; Hsu, F.-F.; Qiu, J. T.; Chern, G.-J.; Lee, Y.-A.;
(105) Noh, S.-h.; Moon, S. H.; Shin, T.-H.; Lim, Y.; Cheon, J.
Chang, C.-C.; Huang, Y.-T.; Sung, Y.-C.; Chiang, C.-C.; Huang, R.-L.;
Recent advances of magneto-thermal capabilities of nanoparticles:
Lin, C.-C.; Dinh, T. K.; Huang, H.-C.; Shih, Y.-C.; Alson, D.; Lin, C.-
From design principles to biomedical applications. Nano Today 2017,
Y.; Lin, Y.-C.; Chang, P.-C.; Lin, S.-Y.; Chen, Y. Highly efficient and
13, 61−76.
tumor-selective nanoparticles for dual-targeted immunogene therapy
(106) Court, K. A.; Hatakeyama, H.; Wu, S. Y.; Lingegowda, M. S.;
against cancer. Science Advances 2020, 6 (3), No. eaax5032.
Rodriguez-Aguayo, C.; Lopez-Berestein, G.; Ju-Seog, L.; Rinaldi, C.;
(123) Li, Y.; Zhao, R.; Cheng, K.; Zhang, K.; Wang, Y.; Zhang, Y.;
Juan, E. J.; Sood, A. K.; Torres-Lugo, M. HSP70 Inhibition Li, Y.; Liu, G.; Xu, J.; Xu, J.; Anderson, G. J.; Shi, J.; Ren, L.; Zhao, X.;
Synergistically Enhances the Effects of Magnetic Fluid Hyperthermia Nie, G. Bacterial Outer Membrane Vesicles Presenting Programmed
in Ovarian Cancer. Mol. Cancer Ther 2017, 16 (5), 966−976. Death 1 for Improved Cancer Immunotherapy via Immune Activation
(107) Bader, A. G.; Brown, D.; Winkler, M. The Promise of and Checkpoint Inhibition. ACS Nano 2020, 14 (12), 16698−16711.
MicroRNA Replacement Therapy. Cancer Res. 2010, 70 (18), 7027−
7030.

P https://doi.org/10.1021/acsmaterialsau.3c00035
ACS Mater. Au XXXX, XXX, XXX−XXX

You might also like