ArticlePDF AvailableLiterature Review

Copper‐based nanomaterials for cancer theranostics

Authors:

Abstract

Copper‐based nanomaterials (Cu‐based NMs) with favorable biocompatibility and unique properties have attracted the attention of many biomedical researchers. Cu‐based NMs are one of the most widely studied materials in cancer treatment. In recent years, great progress has been made in the field of biomedicine, especially in the treatment and diagnosis of tumors. This review begins with the classification of Cu‐based NMs and the recent synthetic strategies of Cu‐based NMs. Then, according to the abundant and special properties of Cu‐based NMs, their application in biomedicine is summarized in detail. For biomedical imaging, such as photoacoustic imaging, positron emission tomography imaging, and multimodal imaging based on Cu‐based NMs are summarized, as well as strategies to improve the diagnostic effectiveness. Moreover, a series of unique structures and functions as well as the underlying property activity relationship of Cu‐based NMs were shown to highlight their promising therapeutic performance. Cu‐based NMs have been widely used in monotherapies, such as photothermal therapy (PTT) and chemodynamic therapy (CDT). Moreover, the sophisticated design in composition, structure, and surface fabrication of Cu‐based NMs can endow these NMs with more modalities in cancer diagnosis and therapy. To further improve the efficiency of cancer treatment, combined therapy based on Cu‐based NMs was introduced in detail. Finally, the challenges, critical factors, and future prospects for the clinical translation of Cu‐based NMs as multifunctional theranostic agents were also considered and discussed. The aim of this review is to provide a better understanding and key consideration for the rational design of this increasingly important new paradigm of Cu‐based NMs as theranostic agents. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Diagnostic Tools > In Vivo Nanodiagnostics and Imaging
ADVANCED REVIEW
Copper-based nanomaterials for cancer theranostics
Xiaoyan Zhong
1
| Xingliang Dai
2
| Yan Wang
3
| Hua Wang
4
|
Haisheng Qian
5
| Xianwen Wang
5
1
School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of
Soochow University, Suzhou, China
2
Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
3
Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
4
Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
5
School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China
Correspondence
Xianwen Wang, School of Biomedical
Engineering, Research and Engineering
Center of Biomedical Materials, Anhui
Medical University, Hefei 230032, China.
Email: xianwenwang@ahmu.edu.cn
Funding information
The Basic and Clinical Cooperative
Research and Promotion Program of
Anhui Medical University, Grant/Award
Number: 2021xkjT028; Jiangsu Natural
Science Fund for Young Scholars, Grant/
Award Number: BK20210730; Grants for
Scientific Research of BSKY from Anhui
Medical University, Grant/Award
Number: 1406012201; The Open Fund of
Key Laboratory of Antiinflammatory and
Immune Medicine, Grant/Award Number:
KFJJ-2021-11; The Postdoctoral Science
Foundation of China, Grant/Award
Number: 2021M702383
Edited by: Gareth Williams, Associate
Editor and Gregory M Lanza, Co-Editor-
in-Chief
Abstract
Copper-based nanomaterials (Cu-based NMs) with favorable biocompatibility
and unique properties have attracted the attention of many biomedical
researchers. Cu-based NMs are one of the most widely studied materials in
cancer treatment. In recent years, great progress has been made in the field of
biomedicine, especially in the treatment and diagnosis of tumors. This review
begins with the classification of Cu-based NMs and the recent synthetic strate-
gies of Cu-based NMs. Then, according to the abundant and special properties
of Cu-based NMs, their application in biomedicine is summarized in detail.
For biomedical imaging, such as photoacoustic imaging, positron emission
tomography imaging, and multimodal imaging based on Cu-based NMs are
summarized, as well as strategies to improve the diagnostic effectiveness.
Moreover, a series of unique structures and functions as well as the underlying
property activity relationship of Cu-based NMs were shown to highlight their
promising therapeutic performance. Cu-based NMs have been widely used in
monotherapies, such as photothermal therapy (PTT) and chemodynamic ther-
apy (CDT). Moreover, the sophisticated design in composition, structure, and
surface fabrication of Cu-based NMs can endow these NMs with more modali-
ties in cancer diagnosis and therapy. To further improve the efficiency of can-
cer treatment, combined therapy based on Cu-based NMs was introduced in
detail. Finally, the challenges, critical factors, and future prospects for the clini-
cal translation of Cu-based NMs as multifunctional theranostic agents were
also considered and discussed. The aim of this review is to provide a better
understanding and key consideration for the rational design of this increas-
ingly important new paradigm of Cu-based NMs as theranostic agents.
Xiaoyan Zhong and Xingliang Dai contributed equally to this study.
Received: 31 January 2022 Revised: 14 March 2022 Accepted: 15 March 2022
DOI: 10.1002/wnan.1797
WIREs Nanomed Nanobiotechnol. 2022;e1797. wires.wiley.com/nanomed © 2022 Wiley Periodicals LLC. 1of43
https://doi.org/10.1002/wnan.1797
This article is categorized under:
Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic
Disease
Diagnostic Tools > In Vivo Nanodiagnostics and Imaging
KEYWORDS
cancer imaging, cancer therapy, combination therapy, Cu-based NMs, tumor
microenvironment
1|INTRODUCTION
In recent decades, the emergence of modern therapeutic nanomedicine has aroused intensive research interest in the
scientific community to explore and develop multifunctional biomaterial nanosystems to meet the stringent requirements
of clinical medicine (Wang, Xu, et al., 2021; Wang, Zhong, et al., 2021; Zhuang et al., 2022). This interdisciplinary field
has also facilitated the development of novel therapeutic modalities against a variety of diseases, in which the rapid devel-
opment of nanomaterials is the fundamental/critical basis and prerequisite for determining the ultimate therapeutic effect
(Dong, Feng, Xu, et al., 2020; Yang et al., 2020). Organic nanomaterials have been extensively explored in nanomedicine,
with clinically relevant family members entering the clinical stage. In contrast, inorganic nanomaterials have been
primarily created in the past decade, with remarkable progress in recent years. These inorganic nanoplatforms possess
inherent electronic, photonic, acoustic, and magnetic properties not possessed by conventional organic nanosystems (Ren
et al., 2021; Ruan & Qian, 2021). Among them, copper-based nanomaterials (Cu-based NMs) are the most representative
example in the field of nanomedicine and have attracted extensive attention, mainly due to their unique physicochemical
properties and excellent biocompatibility. In addition, compared with other nanomaterials, Cu-based nanomaterials are
easier to control and tune the structure, composition, morphology, and size during the preparation process, which pro-
vides more opportunities for their different biomedical applications (Liu, Liu, et al., 2020).
There are many different types of Cu-based NMs, mainly including copper oxides (e.g., Cu
2
O, CuO), copper sulfides
(e.g., Cu
2
S, CuS, Cu
9
S
5
), copper selenides (e.g., Cu
2
Se, CuSe, Cu
2x
Se), copper tellurides (e.g., Cu
2
Te, CuTe, Cu
2x
Te),
copper coordination compounds (e.g., Cu-tannic acid), and copper-based nanocomposites (e.g., Fe
3
O
4
@Cu
2x
S, Au-
Cu
9
S
5
). The synthesis strategies of Cu-based NMs are diversified and mainly include solvothermal/hydrothermal
methods, high-temperature thermal decomposition methods, colloidal synthesis methods, microwave-assisted synthetic
methods, cationic exchange methods, and template-oriented synthesis methods (Wu et al., 2022; Yun et al., 2020). Cu-
based NMs have many types and categories, simple preparation, diversified morphology and functions, which make
them play an increasingly powerful role in tumor imaging and therapy (Wang, Ma, et al., 2017; Wang, Shi, et al., 2021).
Using biocompatible molecules and polymers to simply modify the surface of Cu-based NMs can significantly improve
their pharmacokinetic parameters, thereby improving their colloidal stability, reducing toxicity, and prolonging the
blood circulation time (Zhang, Zeng, & Li, 2020). The unique physicochemical properties of Cu-based NMs enable them
to have various applications in tumor imaging and treatment, which are mainly divided into the following aspects:
(1) Cu-based NMs have good near-infrared (NIR) absorption and excellent photothermal performance, which have been
widely used in photothermal therapy (PTT) and photoacoustic (PA) imaging of tumors; (2) Cu-based NMs have rela-
tively large specific surface areas and can be used to load different chemotherapeutic drugs for tumor chemotherapy;
(3) Cu-based NMs can be used as excellent Fenton-like reagents to generate a large number of hydroxyl radicals (OH)
in the presence of H
2
O
2
, which can be used for chemodynamic therapy (CDT) of tumors; (4) Cu-based NMs are also
often used as photosensitizers to produce a large amount of reactive oxygen species (ROS) under light irradiation,
which can be used for photodynamic therapy (PDT) of tumors. In addition, to endow Cu-based NMs with more func-
tions, Cu-based nanocomposites with excellent properties were successfully constructed to realize the combined treat-
ment of tumors (Zhou et al., 2016).
Cu-based NMs have become one of the research hotspots in the field of cancer theranostics due to their unique
physical and chemical properties (Gilam et al., 2016; Goel et al., 2018). Over the past 5 years, more than 15,000 peer-
reviewed copper-based nanomaterials have been reported for cancer treatment and imaging. However, there are only a
few reviews on the application of copper-based NMs in cancer therapy, which are limited to copper sulfide, and there is
2of43 ZHONG ET AL.
a lack of a systematic summary of Cu-based NMs (Dong, Feng, Xu, et al., 2020; Goel et al., 2014; Liu, Liu, et al., 2020;
Yun et al., 2020; Zhang, Zeng, & Li, 2020; Zhou et al., 2016). In recent years, many new cancer therapies based on Cu-
based NMs, such as CDT, radiodynamic therapy (RDT), immunotherapy, and sonodynamic therapy (SDT), have begun
to emerge. In addition, Cu-based NMs have also been reported to regulate the tumor microenvironment (TME) to
enhance cancer treatment (Hao, Zhang, et al., 2021). Therefore, it is urgent and necessary to systematically summarize
the recent progress of Cu-based NMs in cancer treatment. Herein, this review introduces the application progress of
Cu-based NMs in cancer theranostics in detail (Scheme 1). For cancer imaging, PA imaging, positron emission tomogra-
phy (PET) imaging, and multimodal imaging based on Cu-based NMs were summarized, as well as strategies to
improve the diagnostic effectiveness. Next, the application of Cu-based NMs in tumor treatment was introduced, which
was mainly divided into monotherapy and combination therapy. Cu-based NMs have been extensively used in
monotherapy, such as PTT and CDT. To further improve the efficiency of cancer treatment, combined therapy based
on Cu-based NMs was introduced in detail. Finally, the challenges, critical factors, and future prospects for the clinical
translation of Cu-based NMs as multifunctional theranostic agents were also considered and discussed. The aim of this
review is to provide a better understanding and key consideration for the rational design of this increasingly important
new paradigm of Cu-based NMs as theranostic agents.
2|Cu-BASED NMs USED FOR CANCER IMAGING
Cancer imaging provides important evidence for the diagnosis of cancer via different modalities. However, limited to
the lack of sensitivity toward contrast agents (CAs), the lack of specificity to tumors, and the insufficient accumulation
of CAs in the target sites, nanomaterial-based CAs allow us to precisely obtain anatomical information to determine
the stage of cancer (Siddique & Chow, 2020). Cu-based NMs have been applied as multifunctional CAs in many kinds
of imaging modalities. Below, we introduce Cu-based CAs in unimodal and multimodal imaging of cancer (Table 1).
SCHEME 1 Cu-based NMs for cancer theranostics. Reprinted with permission from Cui et al. (2018). American Chemical Society, 2018.
Reprinted with permission from Wang, Zhang, et al. (2021). Copyright 2021. American Association for the Advancement of Science.
Reprinted with permission from Wang, Zhang, et al. (2021). Copyright 2021. Wiley-VCH GmbH. Reprinted with permission from Shrestha
et al. (2019). Copyright 2019. The National Academy of Sciences of the United States of America. Reprinted with permission from Ma
et al. (2019). Copyright 2018. American Chemical Society. Reprinted with permission from Pan et al. (2021). Copyright 2020. Elsevier BV
ZHONG ET AL.3of43
TABLE 1 Cu-based NMs in cancer imaging
Classification Imaging modality Cu-based NMs Parameters References
Single mode
imaging
FL imaging PGC-DOX DOX/IR800 labeling Fu et al. (2021)
Ce6-DNA-zyme/[Cu
(tz)]
Cy5 labeling Liu, Xu, Yang,
et al. (2021)
UCCG UCNP Wang, Chang,
et al. (2021)
IR thermal imaging Cu
2x
Se@FCS 1064-nm laser (1.5 W cm
2
,
10 min)
Zhang, Li,
et al. (2021)
5-FU/Cu-LDH@nAb-
PTX
808-nm laser (0.75 W cm
2
,
3 min)
Liu, Liu, Zhang,
et al. (2021)
AuNRs-CTN@THA 808-nm laser (1 W cm
2
,
2 min)
Gao et al. (2020)
Cu-DhaTph 808-nm laser (1.5 W cm
2
,
10 min)
Feng et al. (2021)
Cu
2
O@CaCO
3
1064-nm laser (0.5 W cm
2
) Chang et al. (2020)
PA imaging CuS@GNPs 980-nm laser (0.8 W cm
2
,
10 min)
Zhang, Xie,
et al. (2021)
Fe-Cu@PANI 808-nm laser
(1.5 W cm
2
, 5 min)
Wang, Zhang,
et al. (2021)
Cu(DTC)
2
[Cu] =5mgkg
1
Pan et al. (2021)
SPECT imaging [
67
Cu]Cu-NOTA-
Pertuzumab
3.7 or 7.4 MBq Hao, Mastren,
et al. (2021)
PET imaging [
64
Cu]CuInS/ZnS 150 μCi in vitro
300 μCi in vivo
Guo et al. (2015)
T
1
-weighted MR imaging M-CSTD.NHAc/Cu(II) [Cu] =8 mM Song et al. (2021)
CuS 16 mg Cu kg
1
Dong et al. (2019)
Cu-TG Yuan et al. (2020)
Multimodal
imaging
FLPA imaging PDC/P@HCuS 680- to 980-nm
Cy7.5-labeling
Sun et al. (2019)
AIBA@FeCuS-FeCO Cy5.5-labeling Sun, An, et al. (2021)
PACT imaging T80-AuPt@CuS 808-nm laser
175 μA, 65 kV
Cai et al. (2021)
IR thermalFL imaging DSF@PVP/Cu-HMPB FITC-labeling Wu et al. (2020)
PT-V@TPDOX DOX delivering Sun, An, et al. (2021)
TPP-ZC-IR-PNPs IR780 delivering
808-nm laser
(1 W cm
2
, 5 min)
Ruttala et al. (2021)
FA-CD@PP-CpG 808-nm laser (0.987 W cm
2
)
PpIX delivering
Chen, Zhou,
et al. (2019)
UCCZ-FA 980-nm laser (1.0 W cm
2
)
Cy5 labeling
Zhang, Sun,
et al. (2020)
IR thermalBL imaging CF 1064-nm laser (1.5 W cm
2
)
luciferase-T24 cells
Zhang, Li,
et al. (2021)
IR thermalPA imaging BP-CuS-FA 808-nm laser (1.0 W cm
2
) Jana et al. (2020)
CuFe
2
O
4
808-nm laser (1 W cm
2
,
5 min)
Liu, Liu, Wan,
et al. (2021)
Cu
9
S
8
808-nm laser (0.5 W cm
2
) Chang et al. (2020)
4of43 ZHONG ET AL.
2.1 |Cu-based NMs used for cancer unimodal imaging
Cu-based NMs as the CAs have been successfully prepared for use in fluorescence (FL) imaging, PA imaging, magnetic
resonance (MR) imaging, positron emission tomography (PET) imaging, and computed tomography (CT) imaging in
cancer unimodal imaging.
Aberrant expression of miRNAs is highly related to the development, progression, and metastasis of cancer. To
directly image low-abundance intracellular miRNAs in vivo for monitoring the pathological condition of cancers, a self-
powered DNA@Cu-MOF nanosystem was prepared for TME hypoxia-responsive in vivo temporalspatial fluorescence
TABLE 1 (Continued)
Classification Imaging modality Cu-based NMs Parameters References
Cu
2
Se 1064-nm laser (0.75 or
1.0 W cm
2
)
Wang, Zhong,
et al. (2019)
PCPT 808-nm laser (1.0 W cm
2
,
7 min)
Liang et al. (2019)
RGD-CuS DENPs/
pDNA
1064-nm laser (0.6 W cm
2
,
5 min)
[Cu] =12 mM
Ouyang et al. (2021)
IR thermalCT imaging Cu
2x
S:Pt(0.3)/PVP 200 μl, 8.5 mg Cu kg
1
Dong et al. (2018)
IR thermalT
1
-MR imaging Cu@Gd
2
O
3
808-nm laser (0.8 W cm
2
) Liu, Xu, Yang,
et al. (2021)
IR thermalPET imaging BP@Cu
0.4
@PEG-RGD 808-nm laser (1.0 W cm
2
,
2 min)
64
Cu labeling (74 MBq μg
1
)
Lyu et al. (2020)
IR thermalX-ray imaging Cu
3
BiS
3
800-nm laser (1.726 W cm
2
) Veeranarayanan
et al. (2018)
IR thermalT
1
-weighted MR
FL imaging
MCIH 808-nm laser (0.76 W cm
2
,
7 min)
ICG delivering
Sun et al. (2020)
IR thermalT
2
-weighted MR
FL imaging
BSA-CuFeS
2
808-nm laser (1.5 W cm
2
)
DiR-labeling
Chen, Luo,
et al. (2019)
IR thermalFLPA imaging Ag
2x
Cu
x
S 635-nm laser (0.40 W cm
2
,
15 min)
Zhao et al. (2020)
PhA@NanoICG 808-nm laser (1.0 W cm
2
)
ICG delivering
Chen, Zuo,
et al. (2021)
IR thermalPA-γimaging CuS/
131
I-PEGDA/
AIPH
915-nm laser (1.0 W cm
2
)
131
I-labeling (50 μCi)
Meng et al. (2018)
IR thermalFLBL imaging AM@DLMSN@CuS/
R848
980-nm laser (1.0 W cm
2
)
IR780 labeling (100 μgkg
1
)
Luciferase-4 T1 cells
Cheng et al. (2020)
IR thermalCTPA imaging CaO
2
-Cu/ICG@PCM 808-nm laser (1.02 W cm
2
,
9 min)
Sun, Bin, et al. (2021)
FLPETPA imaging NCP Cy5.5-labeling
89
Zr-labeling (200 μCi)
IR780 labeling (30 μg)
Deng et al. (2022)
FLCTPA imaging PtCu
3
Cy5.5 labeling Zhong et al. (2020)
FLCTPET imaging Cu(II)NS TCPP delivering
64
Cu (1000 μCi)
Wang et al. (2022)
IR thermalPAFLSPECT/CT
imaging
99m
Tc-M-CuS-PEG 1064-nm laser (0.5 W cm
2
)
Cy5.5 labeling
99m
Tc (800 μCi)
Yi et al. (2021)
ZHONG ET AL.5of43
FIGURE 1 Cu-based NMs for cancer single mode imaging. (a) DNA@Cu-MOF nanosystem for FL imaging of human breast cancer.
Reprinted with permission from Meng et al. (2020). Copyright 2020. American Chemical Society. (b) LDH-CuS NCs for PA imaging of breast
cancer. Reprinted with permission from Liu, Tang, et al. (2020). Copyright 2020. American Chemical Society. (c) CuS NCs for in situ
T
1
-weighted MR imaging of breast cancer. Reprinted with permission from Dong et al. (2019). Copyright 2019. The Royal Society of
Chemistry. (d) [64Cu]CuS-PEG-c(RGDfK) NPs for PET imaging of glioma. Reprinted with permission from Cui et al. (2018). American
Chemical Society, 2018. (e) Cu
2x
S:Pt(0.3)/PVP NPs for CT imaging of breast cancer. Reprinted with permission from Dong et al. (2018). The
Royal Society of Chemistry, 2018
6of43 ZHONG ET AL.
imaging of aberrant miRNAs and hypoxic tumors (Figure 1a; Meng et al., 2020). In this design, upon entering hypoxic
tumors, the DNA@Cu-MOF nanosystem could spontaneously degrade and release Cy3-labeled DNAzyme precursor,
Cy5.5/FAM-labeled Cu-Sub, and Cu
2+
ions. Originally, Cy3 fluorescence was blocked in the DNAzyme precursor; how-
ever, in the presence of miRNA-21, the signal strand was replaced through toehold-mediated strand displacement
hybridization, which resulted in the fluorescence recovery of Cy3, achieving the specific detection of anomalously
expressed miRNAs in vivo. In addition, this displacement procedure also induced the exposure of the recognition site of
Cu-Sub for Cu
2+
-catalyzed Cu-Sub cleavage processes, leading to the recovery of aberrant miRNA expression-
dependent Cy5.5 fluorescence for hypoxic tumor diagnosis.
PA imaging, as another noninvasive modality, has been rapidly developed with superb contrast, spatial resolution,
high penetrability, and sensitivity to tissue (Fu et al., 2019). In recent years, a series of PA-imaging CAs have been
developed to improve imaging performance in biomedical applications. For example, biodegradable layered double
hydroxide (LDH)-copper sulfide (CuS) nanocomposites were successfully prepared for PA imaging of breast cancer
(Figure 1b; Liu, Tang, et al., 2020). Due to the high photothermal conversion efficiency, LDH-CuS NCs not only worked
as CAs for PA imaging but also as photothermal agents for PTT under 808-nm laser irradiation. In addition, the hetero-
junction was helpful for the separation of electron (e
) and hole (h
+
), making LDH-CuS NCs good photodynamic
agents for superoxide anion (
1
O
2
) generation. Moreover, the Cu
+
in the CuS dots could react with H
2
O
2
to generate
OH for CDT. Overall, LDH-CuS NCs mediated PA imaging-guided PTT-PDT-CDT of cancer via NIR-triggered lyso-
some pathway death.
MR imaging, unlike functional imaging modalities such as FL and PA imaging, is a kind of structural imaging with
high image resolution for soft tissues, and it has been widely used clinically. The principle of MR imaging is the process
of aligning and relaxing hydrogen protons under an external magnetic field. The ability of magnetic nanoparticles
(MNPs) enhances the proton relaxation of specific tissues (Sun et al., 2008). In addition to Fe-, Mn-, and Gd-based
MNPs, Cu
2+
ions were also reported to be good CAs for MR imaging. For example, highly active (102) surface-abundant
CuS NCs could be used to conduct pH and NIR light-responsive T
1
-weighted MR imaging of breast cancer (Figure 1c;
Dong et al., 2019). In detail, the high Cu vacancies endowed CuS NCs with a small bandgap for high photothermal con-
version efficiency (46%). Accompanied by the negative formation energy of S vacancies, the (102) surface could be
quickly degraded by pH and 808-nm laser irradiation. The intrinsic high absorbance of CuS NCs could mediate the PTT
of cancer. Then, a large amount of Cu
+
ions were released from CuS NCs to produce high concentrations of superoxide
anion (O
2
) and OH for PDT under 808-nm laser irradiation. Meanwhile, the oxidation of Cu
+
ions into Cu
2+
ions
could be applied for in situ T
1
-weighted MR imaging (MRI), realizing MRI-guided PTT and PDT of cancer.
64
Cu, a radioactive form of Cu, can emit β-ray for PET imaging of cancer. In addition,
64
Cu could be used to build
CuS NPs with no need for a radiometal chelator. Herein,
64
Cu-labeled CuS-PEG nanoparticles with target-directed
c(RGDFK) were prepared via click-chemistry-assisted synthesis for PET imaging of U87 xenograft tumors (Figure 1d;
Cui et al., 2018).
In addition to MR imaging, CT imaging also belongs to the structural imaging modality and is widely used for diag-
nosis. It has high spatial and temporal resolution and can provide precise three-dimensional (3D) anatomical informa-
tion of specific tissues. However, it also lacks sensitivity toward CAs, especially for light metal-based NMs. Fortunately,
heavy elements have unique X-ray attenuation properties for enhanced CT images. By incorporating Cu and Pt ele-
ments, Cu
2x
S:Pt(0.3)/PVP NPs were simply constructed for CT imaging of breast cancer (Figure 1e; Dong et al., 2018).
In this work, Pt ions not only functioned as CAs for CT imaging but could also realize chemotherapy by Pt
2+
ions from
GSH-reduced Pt
4+
ions. Combined with the high absorbance within the NIR region, Cu
2x
S:Pt(0.3)/PVP NPs could also
work for PTT under 808-nm laser irradiation.
2.2 |Cu-based NMs used for cancer multimodal imaging
As mentioned above, each imaging modality has its merits and demerits in providing structural and functional informa-
tion. To learn from each other, a multimodal imaging modality is more reasonable and acceptable to offer comprehen-
sive information of the diseases. For instance, to precisely diagnose lymph node metastasis, CuS NPs were labeled with
fluorescent Cy5.5 for α
V
β
3
targeted and noninvasive dual modal FL-CT imaging of metastatic gastric cancer cells in
lymph nodes (Figure 2a; Shi et al., 2018). RGD-CuS-Cy5.5 NPs could readily drain into sentinel lymph nodes (SLNs) to
target MNK45 gastric tumor cells via the interaction between RGD and α
V
β
3
, offering strong fluorescence at 695 nm for
FL imaging and remarkable CT contrast indicative of SLN metastasis. This work combined functional and structural
ZHONG ET AL.7of43
imaging modalities to provide more information on metastatic gastric cancer. In another study, simultaneous
tetramodal imaging, including PET/FL/Cerenkov luminescence (CL)/Cerenkov radiation energy transfer (CRET) imag-
ing, was simply realized by [
89
Zr]CSNC for rapid and accurate delineation of breast tumors (Figure 2b; Goel
et al., 2018). [
89
Zr]CSNC nanoconstructs were prepared by
89
Zr-labeled hollow mesoporous silica nanoshells (HMSN)
FIGURE 2 Cu-based NMs for cancer multimodal imaging. (a) CuS NPs for dual modal FL-CT imaging of human gastric cancer.
Reprinted with permission from Shi et al. (2018). Elsevier Ltd., 2018. (b) CSNCs for four modal FL-CL-PET-CRET imaging of breast cancer.
Reprinted with permission from Goel et al. (2018). WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim, 2017
8of43 ZHONG ET AL.
filled with CuS NPs and meso-tetrakis(4-carboxyphenyl)porphyrin (TCPP) via an electrostatic interaction-driven self-
assembly method. In these nanoconstructs, zirconium-89 (
89
Zr) was used for PET and Cerenkov luminescence
(CL) imaging. TCPP worked not only for FL imaging but also as a photosensitizer for
1
O
2
generation. Interestingly,
CRET could occur between the fluorescent TCPP and radioactively decayed
89
Zr, achieving another diminished
autofluorescence imaging of cancer with improved tumor-to-background contrast.
3|Cu-BASED NMs FOR CANCER MONOTHERAPY
Among the transition-metal-based NMs, the copper age of Cu-involved cancer theranostics has arrived. As an essential
element that participates in biological processes, including angiogenesis and lipid/glucose metabolism, Cu-based NMs
with intriguing theranostic performance have motivated scientists to apply them to the field of cancer (Meng
et al., 2020). In addition to the multimodal imaging properties of Cu-based NMs in visualizing tumors, these NMs have
also shown promise in cancer therapy owing to their unique structures and functions with various physicochemical
properties and bioactivities (Meng et al., 2020). At present, Cu-based cancer monotherapy includes external stimuli
such as light triggered PTT and PDT, ultrasound (US) excited SDT, X-ray initiated RDT, and internal chemical energy-
driven CDT (Table 2; Zhong et al., 2021).
3.1 |Photothermal therapy
The beneficial effect of heat in resisting cancer was first observed in the 19th century owing to fever caused by bacterial
infection. In particular, PTT, which uses NMs capable of generating efficient heat under laser irradiation to achieve
tumor ablation, has become attractive for the last few decades (Jaque et al., 2014). Tremendous efforts have been made
toward improving PTT performance. To date, numerous functional NMs have been reported to be efficient
photothermal transduction agents (PTAs) for PTT, such as metal-based, carbon-based, organic nanoparticle, polymer-
based, and hybrid NMs (Kumari et al., 2021; Zhou et al., 2020; Zhou, Wang, et al., 2021). However, Cu-based PTAs
show advantages in relatively high photothermal conversion efficiency (PCE), thermostability, catalytic performance,
and others. For example, amorphous Ag
2x
Cu
x
S quantum dots (QDs) were characterized with good photothermal/PA
properties, showing a high photothermal conversion efficiency of 44.0% (Figure 3a; Zhao et al., 2020). With the lowest
absorbance within the first biowindow (820 nm), Ag
2x
Cu
x
S QDs could realize infrared (IR) and PA imaging-guided
PTT under a relatively low-power irradiation of a 635-nm laser (0.8 W cm
2
) and demonstrated no long-term toxicity.
Despite the success of diodes as excited light in PTT, the strong extinction coefficients of human tissues within the
visible region limit diode-excited PTT in superficial tumors. Therefore, longer-wavelength NIR light that is adjusted to
the tissue-transparent windows of both the first window (7501000 nm; NIR-I) and second window (10001350 nm;
NIR-II) can penetrate much deeper body tissues (Liu et al., 2019). For this purpose, many types of Cu-involved PTAs
that are responsive to NIR light have been made for PTT. For example, CuS nanocrystals (NCs) were grafted onto the
surface of gelatin nanoparticles (GNPs), forming transformable CuS@GNP structures for the thermal ablation of
human MDA-MB-23 tumors (Figure 3b; Li et al., 2021). Although this nanostructure was approximately 120 nm in
diameter and hardly penetrated into the central zone of the tumor, the overexpressed hydrolases rapidly dissociated the
scaffold to release tiny CuS NCs (8.00 ± 1.46 nm) to efficiently mediate deep PTT of the inner tumor via enzyme-
induced multistage delivery under an NIR laser (980 nm, 0.8 W cm
2
) irradiation. The photothermal conversion effi-
ciency was calculated to be only 10.84%; however, this was fairly efficient for photothermal ablation both in vitro and
in vivo.
In addition to the high concentration of enzymes in the TME, glutathione (GSH) is also highly produced by tumor
cells to maintain redox balance, which is approximately 1000 times higher than that of healthy cells (Yin et al., 2018).
Therefore, GSH can function as another endogenous stimulator to activate tumor-specific therapy. Iron-copper codoped
polyaniline (Fe-Cu@PANI) NPs were prepared to act as GSH-induced absorption spectrum redshift PTAs, from the visi-
ble to NIR region, for PTT of 4T1 breast cancer (Figure 3c; Wang, Zhang, et al., 2021). In this architecture, the Cu
2+
ions were first reduced by GSH, thus depleting GSH, followed by the production of protonated PANI with a maximum
absorption redshifted from 615 to 820 nm. In addition, the higher the concentration of GSH was, the higher the
absorbance at 820 nm, making protonated PANI with ultrahigh photothermal conversion stability even after 6 cycles of
808-nm laser irradiation at a power density of 1.5 W cm
2
. For the assessment of the anticancer effect, Fe-Cu@PANI
ZHONG ET AL.9of43
under laser irradiation could destroy 4T1 cells in vitro and effectively inhibit the growth of tumors in vivo. Moreover,
histopathological analysis further indicated the biosafety of this PTA at the selective dose.
To successfully display the high tissue penetration ability of NIR light, an orthotopic bladder cancer (BCa) model
was used in efficient NIR-II PTT. For the treatment of BCa, the traditional therapeutic option in the clinic is intravesical
instillation. However, this method has limited efficacy because the residence time and transmucosal performance of
therapeutic agents are greatly restricted. To overcome these limitations, fluorinated chitosan (FCS)-modified Cu
2x
Se
(Cu
2x
Se@FCS, denoted CF) NPs were fabricated for BCa treatment after intravesical instillation (Figure 3d; Zhang, Li,
et al., 2021). Why chose fluorinated chitosan (CS), the authors stressed that CS could cause significant necrosis and
deterioration in urothelial cells. In contrast, fluorinated cationic polymers could remarkably deliver drugs to tumors
with high permeability in the bladder wall, which could significantly enhance the therapeutic effect of intravesical
TABLE 2 Cu-based NMs for cancer monotherapy
Classification Cu-based NMs Parameters References
PTT Cu@CPP-t808-nm laser (1.6 W, 10 min) Weng et al. (2020)
Cu
2
O 808-nm laser (1 W cm
2
, 5 min) An et al. (2018)
Au@Cu
2
O 808-nm laser (1 W cm
2
, 5 min) Tao et al. (2019)
CuS 980-nm laser (2.48 W cm
2
, 3 min) Zheng et al. (2021)
CuS 808-nm laser (1 W cm
2
, 5 min) Qi et al. (2020)
CuS-Au 1064-nm laser (1 W cm
2
, 10 min) Wang, Yu, et al. (2020)
Gd/CuS 808-nm laser (0.5 W cm
2
, 10 min) Shi et al. (2019)
Gd/CuS@PEI-FA-PS 1064-nm laser (0.6 W cm
2
, 10 min) Zhang, Sun, et al. (2020)
CuS@MSN-TAT-RGD 980-nm laser (1.5 W cm
2
, 5 min) Li et al. (2018)
Ag
2x
Cu
x
S 635-nm laser (0.40 W cm
2
, 15 min) Zhao et al. (2020)
ZnPP/C
u2x
S 1064-nm laser (0.7 W cm
2
, 10 min) Yi et al. (2021)
CuS@GNPs 980-nm laser (0.8 W cm
2
, 10 min) Zhang, Xie, et al. (2021)
CuSe (2.0 W cm
2
, 10 min) 808-nm laser Wang, Miao, et al. (2017)
Cu
2x
Se (0.75 W cm
2
, 10 min) 1064-nm laser Zhang et al. (2016)
Cu
2x
Se@FCS 1064-nm laser (1.5 W cm
2
, 10 min) Zhang, Li, et al. (2021)
Fe-Cu@PANI 808-nm laser (1.5 W cm
2
, 5 min) Wang, Zhang, et al. (2021)
PDT Cu-Cy 360-nm laser (6 J cm
2
,20mWcm
2
, 5 min) Huang et al. (2019)
Cu-CDs LED (400700 nm) light (40 mW cm
2
) Wang, Xu, et al. (2019)
Lip(ASC/PFH) 670-nm laser (0.48 W cm
2
, 10 min) Liu et al. (2018)
MOF-2 650-nm laser (50 mW cm
2
, 15 min) Zhang et al. (2018)
CuTz-1-O
2
@F127 808-nm laser (0.6 W cm
2
, 20 min) Cai et al. (2019)
PMOF White light (100 mW cm
2
, 5 min) Wang, Zhang, et al. (2021)
CIS 660-nm laser (1 W cm
2
, 10 min) Wang, Ma, et al. (2019)
SDT Cu-Cy 2 W US (2 W, 3 min) Wang et al. (2018)
RDT Cu-Cy X-ray (90 kVp, 30 mA, 5 Gy) Shrestha et al. (2019)
Cu-Cy X-ray (6 MV, 100 MU min
1
, 2 Gy) Chen et al. (2022)
CDT Cu-Cys 5 mg kg
1
Ma et al. (2019)
M-CeO
x
10 mg kg
1
Zhou, Li, et al. (2021)
CP 5 or 10 mg kg
1
Lin et al. (2019)
NCP 3 mg kg
1
Deng et al. (2022)
CPMP 10 or 20 mg kg
1
Yang, Tao, et al. (2021)
M-CSTD.NHAc/Cu(II) 0.8 μmol Song et al. (2021)
Cu-Mn bimetallic complex 4 μM, 8 μM, 16 μM Cao et al. (2019)
10 of 43 ZHONG ET AL.
instillation. Compared with the CS-modified Cu
2x
Se@CS (CC) NPs, the fluorescence signal of CF NPs in the bladder
was 5.3 times higher than that of the nonfluorinated counterparts of CC NPs, revealing the high transmucosal ability of
FCS. After further irradiation with a 1064-nm laser at 1.5 W cm
2
for 10 min, tumor growth was delayed, as visualized
by bioluminescence (BL) imaging. Collectively, these Cu-based NMs preliminarily demonstrated the feasibility of vis
NIR light-excited PTT without significant systematic toxicity to animals.
Exogenous PTAs with high PCE, high absorbance in the NIR I/II regions, high accumulation in the target site, and
high safety determine their potential in cancer PTT. Copper-based NMs have exhibited good performance in the
abovementioned aspects for cancer PTT.
3.2 |Photodynamic therapy
In addition to the PTAs for PTT under light excitation, some photosensitizers (PSs) can also respond to UVvisNIR
light for PDT of cancer. As another light-triggered anticancer technique, PDT, which converts oxygen-containing sub-
stances into ROS to induce cancer cell apoptosis or necrosis, has been shown to have noninvasive features, high selec-
tivity, few side effects, and drug resistance (Veeranarayanan et al., 2018). According to the two different photochemical
reaction processes with or without the electron transfer process, PDT is usually divided into type I and type II PDT.
Type II PDT, which is mainly dependent on O
2,
can generate
1
O
2
, while type I PDT can produce O
2
and OH (Zhao
et al., 2021). Therefore, different light-responsive PSs have been reported to produce ROS to induce cancer cell death
via apoptosis or other pathways. These PSs are usually incorporated with heavy atoms, which made them facing some
drawbacks of dark toxicity, short triplet-state lifetimes, and high cost. Instead, the design of heavy-atom free PSs has
FIGURE 3 Cu-based NMs for PTT. (a) Amorphous Ag
2x
Cu
x
S quantum dots for PTT of liver cancer. Reprinted with permission from
Zhao et al. (2020). Copyright 2020. Elsevier BV. (b) Structure of transformable CuS@GNPs for PTT of breast cancer. Reprinted with
permission from Zhang, Xie, et al. (2021). Copyright 2020. Elsevier BV. (c) Redshifted polymer nanoparticle of Fe-Cu@PANI for PTT of
breast cancer. Reprinted with permission from Wang, Zhang, et al. (2021). Copyright 2021. The American Association for the Advancement
of Science. (d) Cu
2x
Se@FCS for PTT of bladder cancer. Reprinted with permission from Zhang, Li, et al. (2021). Copyright 2021. Wiley-
VCH GmbH
ZHONG ET AL.11 of 43
become popular owing to their superior photophysical and photochemical properties over the past few years (Van-
Nghia et al., 2021). For example, in type II PDT, copper-doped carbon dots (Cu-CDs) were prepared by the pyrolysis
method for PDT of both cervical cancer and neuroblastoma (Figure 4a; Wang, Xu, et al., 2019). Under light-emitting
diode (LED) light (400700 nm) irradiation at 40 mW cm
2
, large amounts of
1
O
2
were generated by Cu-CDs. As rev-
ealed by both ABDA and RB, the
1
O
2
quantum yield of Cu-CDs were calculated to be 0.36, which was comparable to
most conventional PSs. In vitro HeLa cells and SH-SY5Y 3D multicellular spheroids (MCs) mimicking a tissue model
further exhibited an effective tumor suppression effect. As mentioned above, O
2
, as the source of
1
O
2,
plays an essential
role in ROS production; unfortunately, the hypoxic condition in the TME severely reduces the quantum yield of
1
O
2
.To
relieve hypoxia, Au@SiO
2
@Cu
2
O (ASC) was loaded into O
2
self-enriched perfluorohexane (PFH), forming ASC/PFH
nanocomposites for enhanced PDT via a plasmonic resonance energy transfer (PIRET) process (Figure 4b; Liu
et al., 2018). In this work, plasmonic gold (Au) metal nanostructures were incorporated into Cu
2
O semiconductors. By
utilizing the PIRET process from Au to Cu
2
O, ASC showed a high
1
O
2
quantum yield of 0.71, with the help of a
FIGURE 4 Cu-based NMs for PDT. (a) Cu-CD for both human cervical cancer and neuroblastoma. Reprinted with permission from
Wang, Xu, et al. (2019). Copyright 2019. American Chemical Society. (b) ASC/PFH nanocomposites for PDT of breast cancer. Reprinted with
permission from Liu et al. (2018). Copyright 2018. American Chemical Society. (c) MOF-2 of CuL-[AlOH]
2
with Cu
II
as the active center for
GSH depletion enhanced the PDT of liver cancer. Reprinted with permission from Zhang et al. (2018). Copyright 2018. Wiley-VCH Verlag
GmbH & Co. KGaA, Weinheim. (d) CuTz-1-O
2
@F127 for enhanced PDT of both cervical and breast cancer. Reprinted with permission from
Cai et al. (2019). Copyright 2019, WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. (e) In situ synthesis of mitochondria-targeted PMOF
NPs with AIE features for the precise PDT of cervical cancer. Reprinted with permission from Wang, Zhang, et al. (2021). Copyright 2021.
Wiley-VCH GmbH
12 of 43 ZHONG ET AL.
sufficient O
2
supply delivered by PFH under 670-nm laser irradiation at only 0.48 W cm
2
. An in vivo study further
strongly demonstrated hypoxia relief and remarkable PDT therapeutic effects mediated by the ASC/PFH platform.
As the hallmarks of the TME, which contains hypoxia and high concentrations of GSH, reductive GSH within
tumor tissue is not beneficial for ROS accumulation in PDT. As a result, GSH should be considered to be depleted for
enhanced PDT. For this purpose, a nano-MOF of [CuL-(AlOH)
2
]n (MOF-2) was developed for GSH adsorption to
enhance the PDT of liver cancer (Figure 4c; Zhang et al., 2018). Therefore, the active Cu
II
could specifically bind SH to
absorb GSH, thus directly decreasing the intracellular GSH concentration, followed by increasing the ROS level under
650 nm light irradiation due to the contribution of H
6
L of mesotetrakis(4-carboxylphenyl)porphyrin. Interestingly, this
MOF-2-mediated GSH-depleted PDT was comparable to the commercial antitumor drug camptothecin (CPT) in anti-
cancer treatment, making it a new PS candidate or anticancer drug in cancer therapy. However, it has been reported
that the binding ability of Cu
II
sites to sulfhydryl groups is much weaker than that of Cu
I
. Moreover, the authors did
not modulate the hypoxic TME. Not long after this work, smart CuTz-1-O
2
@F127 NPs were designed to synchronously
provide sufficient O
2
and deplete overexpressed GSH for more effective PDT against both cervical and breast cancer
(Figure 4d; Cai et al., 2019). First, in the presence of H
2
O
2
, the CuTz-1 MOF could function as an 808-nm laser-
activated PS to generate OH and O
2
, alleviating hypoxia for type I PDT. Second, they could also adsorb intracellular
GSH at the same time, which was favorable for PDT. Importantly, these NPs were biodegradable, which is of great
importance for the future transformation of nanomedicine in clinics.
Unlike the introduced PSs that were fabricated before biomedical application, the in situ synthesis of PSs with
mitochondria-targeting ability will achieve maximized phototoxic damage to tumor cells but minimized toxicity to nor-
mal cells. To realize this precise PDT, cancer-cell-activated AIE PSs (MOF-199) were reported, a Cu(II)-based MOF as
an inert carrier to deliver PS precursors. Via an in situ click reaction, PSs could be exclusively synthesized with cancer
cells for PDT of cervical cancer (Figure 4e; Wang, Zhang, et al., 2021). In detail, Cu
2+
ions, trimesic acid, and two pho-
tochemically inert precursors of TPA-alkyne-2+and MePy-N
3
were used to form MOF-199 (PMOF) via de novo synthe-
sis. After F-127 coating, the PMOF NPs with a positive charge could target mitochondria. Then, due to the high
concentration of GSH in tumor cells, Cu
2+
ions in PMOF could be reduced to Cu
+
ions, serving as a catalyst source for
the in situ click reaction between TPA-alkyne-2+and MePy-N
3
. In turn, the formed TPATrzPy-3+functioned as a PS
to produce
1
O
2
with bright red fluorescence emission properties. Owing to the existence of quaternary ammonium and
pyridinium units, TPATrzPy-3+was endowed with mitochondria-targeting features to amplify its ROS-mediated cell-
killing efficacy. This strategy provided new insight into the design of PDT systems.
Cu-based inorganic PSs have rarely been reported for PDT, especially those that can respond to NIR light for deep
cancer PDT. Much more effort should be devoted to developing Cu-based PSs for high-efficiency deep PDT of tumors
that are outside the scope of current UVvisNIR light irradiation.
3.3 |Sonodynamic therapy
PDT using light as the excitation source to activate PSs for ROS generation has been extensively studied. Even with NIR
light irradiation, the penetration depth of human tissues is still limited below 1 cm, which makes PDT suitable for the
treatment of superficial tumors (Liang et al., 2020; Yang, Wang, et al., 2021). In contrast to light, US, as a mechanical
wave, is characterized by a high penetration ability relying on different frequencies. To date, US with frequencies rang-
ing from kHz to MHz has been utilized as the trigger of sonosensitizers for SDT in the presence of O
2
or other sub-
strates. It was reported that the penetration depth in soft tissues could reach more than 10 cm, a distance that can meet
the needs of clinical theranostics, such as US imaging and high-intensity focused US (HIFU) therapy. In the process of
SDT, the US-induced cavitation effect and sonoluminescence effects have been widely acknowledged to explain the pos-
sible ROS generation mechanisms, although no definite conclusion has been put forwards (Ouyang et al., 2020).
PSs play a pivotal role in PDT, as do sonosensitizers. The efficacy of SDT is closely related to the properties of
sonosensitizers, which motivated the development of new types of acoustic wave-activated nanomaterials for enhanced
SDT (Lin et al., 2020). From the perspective of construction, sonosensitizers are mainly divided into three categories:
organic, inorganic, and organic/inorganic hybrid sonosensitizers that are mainly based on metallic oxide, noble metal,
carbon, silicon, and metal ions. For Cu-based sonosensitizers, copper-cysteamine (Cu-Cy) was first reported as a new
sensitizer to US for oxidative therapy of breast cancer (Figure 5; Wang et al., 2018). Under US (0.5 W cm
2
) irradiation
for 1 min, the fluorescence intensity of terephthalic acid (TA) gradually increased with increasing Cu-Cy doses, mean-
ing that sonochemical species formed by ultrasonic cavitation. In vitro and in vivo studies further confirmed the
ZHONG ET AL.13 of 43
apoptosis of tumor cells and delayed tumor growth of breast cancer under US irradiation (0.5 W cm
2
, 1.0 MHz). This
study expanded the application of Cu-Cy NPs in the field of SDT. After the discovery that Cu-Cy NPs could respond to
light, X-ray, and microwaves, US-excited Cu-Cy NPs for SDT were first reported in this work. Both ROS and cavitation
effects could be observed in this process. However, whether other types of Cu-based NMs can function as
sonosensitizers should be studied to expand their application in the field of SDT.
3.4 |Radio-dynamic therapy
In addition to US, which has high penetration ability in theranostics, X-ray has also been used for CT imaging and
radiotherapy (RT) in clinical practice and can easily penetrate as deeply as necessary into patients (Zhong et al., 2019).
Applying X-ray as the stimulus of PSs, ROS can also be produced for cancer therapy, forming a new type of X-ray
induced photodynamic therapy (X-PDT), equal to RDT launched in 2006 (Chen & Zhang, 2006). However, most of the
FIGURE 5 Cu-Cy for SDT of breast cancer. Reprinted with permission from Wang et al., (2018). Copyright 2018. WILEY-VCH Verlag
GmbH & Co. KGaA, Weinheim
14 of 43 ZHONG ET AL.
widely used PSs cannot be stimulated by X-ray due to the wide mismatch of the energy gap between them. To convert
high-energy X-ray (keVMeV) into UVvis luminescence (eV), many types of energy transducers have been developed
for RDT, including scintillating NPs (ScNPs), persistent luminescence NPs (PLNPs), aggregation-induced emission
(AIE)-based NPs, nanoscale MOFs (nMOFs), and others (Zhong et al., 2021). However, the majority of the
abovementioned RDT systems, which combine energy transducers and PSs, suffer from the limited loading capacity of
PSs and fluorescence resonance energy transfer (FRET) efficiency, thus leading to nonideal outcomes.
Unlike the FRET-based strategy, we found that the NaCeF
4
:Gd,Tb nanoscintillator itself could function as an X-ray
responsive PS for RDT, with no need to conjugate organic PSs (Zhong et al., 2019). Similarly, among these sensitizers
that can directly respond to X-ray, Cu-Cy nanoparticles not only responded to US mentioned above but also were
responsive to X-ray to generate
1
O
2
for RDT of colorectal cancer (Figure 6a; Shrestha et al., 2019). Based on their previ-
ous pilot study that measured the survival of a large cohort of mice after receiving X-ray activation of Cu-Cy NPs, they
further conjugated a pH-low insertion peptide (pHLIP) onto Cu-Cy NPs to facilitate active targeting of CuCy-pHLIP to
low-pH tumors. Under UV light irradiation at 365 nm, an alternative to X-ray irradiation, Cu-Cy NPs emitted red fluo-
rescence at 607 nm with a shoulder at 633 nm, accompanied by
1
O
2
generation. The mechanism of X-ray-stimulated
FIGURE 6 Cu-based NMs for RDT. (ac) CuCy-pHLIP conjugates for RDT of colorectal cancer. Reprinted with permission from
Shrestha et al. (2019). Copyright 2019. The National Academy of Sciences of the United States of America. (d) Cu-Cy for RDT for liver and
breast cancer. Reprinted with permission from Chen et al. (2022). Copyright 2021. Elsevier BV
ZHONG ET AL.15 of 43
Cu-Cy NPs for
1
O
2
generation was similar to the process of light-activated PDT (Figure 6b). In vivo studies demon-
strated that by pHLIP conjugation, Cu-Cy nanoparticles effectively delayed tumor growth in both male and female mice
(Figure 6c). This might be the reason why pHLIP could enhance the binding of Cu-Cy to tumor cells since
1
O
2
is very
short-lived at 4μs. Therefore, CuCy-pHLIP has been successfully attempted as an X-ray-responsive sensitizer
for RDT.
Excited by the satisfying results of Cu-Cy-based RDT in the laboratory, for the first time, they further used a
linear accelerator and built deep-seated tumor models to mimic the clinical conditions, aiming to judge the
effectiveness and its effects on cell migration and proliferation (Figure 6d; Chen et al., 2022). They found that
Cu-Cy-mediated RDT could inhibit the proliferation and migration of HepG2, SK-HEP-1, Li-7, and 4T1 cells in a
dose-dependent manner. MRI assessment visualized that the growth of deeply located tumors in mice and rabbits
was inhibited by X-ray-irradiated Cu-Cy. In addition, Cu-Cy-mediated RDT inhibited tumor cell proliferation and
migration by upregulating proliferating cell nuclear antigen (PCNA) and E-cadherin. The possible influences of
RDT might also include oxidative stress, killing, and immunity enhancement, which might be beneficial to cancer
treatment. Overall, these systematic studies declared that Cu-Cy NP-mediated RDT has promising prospects in
clinical applications. Unfortunately, there is a lack of research on Cu-based PSs that can respond to X-ray for RDT,
such as Cu-based sonosensitizers.
3.5 |Chemodynamic therapy
According to the position of excitation sources relative to the tumor tissues, ROS can be produced by not only external
stimuli, including light, US, and X-ray but also chemical energy (Zhong et al., 2021). CDT has been recognized as a
tumor-specific treatment with high dependence on the TME. It uses Fenton/Fenton-like agents to convert H
2
O
2
into
cytotoxic OH, inducing tumor cells to undergo apoptosis and necrosis (Tang et al., 2019). Generally, Fenton/Fenton-
like agents are mainly based on transition metal ions of Fe, Co, Ni, Cu, Mn, and heavy metal ions such as Pt and
Ce. For instance, Cu-Cy NPs with multiple faces were used as light-, US-, and X-ray-responsive sensitizers for ROS gen-
eration. In this study, self-assembled Cu-Cys NPs were utilized for GSH-activated CDT of drug-resistant breast cancer
(Ma et al., 2019). In brief, after endocytosis, the Cu
2+
ions in Cu-Cys NPs were first reduced by intracellular GSH to
form Fenton-like agents of Cu
+
ions, followed by interaction with in situ H
2
O
2
to produce OH in the weakly acidic
TME. Compared with DOX administration, drug-resistant breast cancer showed delayed tumor growth, meaning that
they were much more sensitive to Cu-Cys-based CDT.
However, although this work cleverly exploited the high concentration of GSH in the TME to reinforce CDT, the
CDT performance is still limited by certain other issues, including the low content of endogenous H
2
O
2
and the rela-
tively higher pH of solid tumors (6.56.8), which do not meet the strict reaction conditions for most Fenton/Fenton-like
reactions. To supply additional H
2
O
2
into the tumor cells and reduce the dependence of Fenton/Fenton-like agents on
low pH, a series of metal-doped cerium peroxides (M-CeO
x
) with high pH activation and H
2
O
2
self-supply were fabri-
cated for the ultrasensitive CDT of breast cancer (Figure 7a; Zhou, Li, et al., 2021). These doped metal ions included
Fe
2+
,Cu
2+
,Mn
2+
,Co
2+
,Ce
4+
,Cr
3+
,Al
3+
, and Ru
3+
. Compared with undoped CeO
x
, these metal ions endowed it with
high catalytic efficacy at a high pH of 7.0 (40- to 60-fold increase). In addition, as metal peroxides, CeO
x
functioned as
the donor of H
2
O
2
to relieve the insufficient endogenous H
2
O
2
level for enhanced CDT. Therefore, this strategy signifi-
cantly improved tumor cell sensitivity to CDT.
With the same strategy of self-supplying H
2
O
2
, copper peroxide (CP) nanodots were reported much earlier than
CeO
x
by Prof. Chen and coworkers (Lin et al., 2019). After endocytosis, the dissociation of CP nanodots could be
accelerated due to the acidic environment of endo/lysosomes, allowing for the subsequent enhanced CDT. The gen-
erated OH caused tumor cell death via the lysosome-associated pathway via lipid peroxidation (LPO)-mediated
lysosomal membrane permeabilization. As a result, lysosomes have emerged as specific targets for cancer treat-
ment. Despite the appropriate pH (4.55.0) of lysosomes for the Fenton reaction, the catalytic agents will rapidly
escape to the cytoplasm, thus limiting the action time of CDT. Therefore, it is highly meaningful to anchor the cata-
lytic agents inside the acidic lysosome, aiming for a lengthening time to decompose enough catalytic ions for CDT.
More recently, they further prepared a series of nano copper peroxides (NCPs) with tunable acid dissociation con-
stant (pKa) values from 5.2 to 6.2 for H
2
O
2
self-supplying CDT (Figure 7b; Deng et al., 2022). Intracellular traffick-
ing studies revealed that NCP with a lower pKa value (pKa 5.2) retained a longer time in lysosomes, followed by
decomposition of enough Cu
2+
ions and H
2
O
2
for a robust Fenton reaction. Most importantly, the Fenton reaction
16 of 43 ZHONG ET AL.
was strictly processed within the lysosomal compartment, avoiding interference from cytoplasmic GSH. Overall,
this work provides a more precise strategy for enhanced CDT.
Recently, nanozyme-based CDT has become an attractive cancer treatment because of its low side effects and high
efficacy. The construction of a single catalyst with multiple enzyme properties for enhanced CDT is still a major chal-
lenge. For this purpose, seven types of bimetallic nanoparticles were synthesized with MOF (MIL-101) as a stable host
for active metal ion delivery for enhanced CDT of breast cancer (Figure 7c; Yang, Tao, et al., 2021). These seven alloyed
NPs were 6.0 wt%Cu-Pd@MIL-101, 7.5 wt%Cu-Pd@MIL-101, 9.0 wt%Cu-Pd@MIL-101, 9.5 wt%Cu-Pd@MIL-101, 10.1
wt%Cu-Pt@MIL-101, 9.1 wt%Co-Pt@MIL-101, and 8.7 wt%Ni-Ru@MIL-101. Among them, Cu-Pd@MIL-101 with an
alloy loading of 9.5 wt% (9.5% CPMP) was found to have the highest peroxidase (POD) and superoxide dismutase (SOD)
mimic activities and GSH depletion ability. This work was considered another credible strategy for the design of
enhanced CDT.
The bloodbrain barrier (BBB) is a major challenge for the chemotherapy of orthotopic glioma. Similarly,
these chemodynamic agents also hardly cross the BBB to reach tumor tissues. Herein, based on the recognition
between β-cyclodextrin (CD) and adamantane (Ad), supramolecular assembled tectodendrimers with acetyl termini
(M-CSTD. NHAc) were prepared for glioma theranostics (Figure 7d; Song et al., 2021). In this coreshell structure,
thecoreofCSTDswascomposedofCD-modifiedgeneration 5 (G5) poly(amidoamine) (PAMAM) dendrimers and
Ad-functionalized G3 PAMAM dendrimers (G3. NH
2
-Ad) were the shell. Following modification with pyridine,
FIGURE 7 Cu-based NMs for CDT. (a) M-CeO
x
for pH-activated and H
2
O
2
self-supply-mediated CDT of breast cancer. Reprinted with
permission from Zhou, Li, et al. (2021). Copyright 2021. Wiley-VCH GmbH. (b) NCP with the capacity to self-supply H
2
O
2
for specific CDT
for breast cancer. Reprinted with permission from Deng et al. (2022). Copyright 2021. Elsevier Ltd. (c) CPMP for H
2
O
2
supplementation and
GSH depletion enhanced CDT for breast cancer. Reprinted with permission from Yang, Tao, et al. (2021). Copyright 2021. Wiley-VCH
GmbH. (d) M-CSTD. NHAc/Cu(II) complexes self-supply H
2
O
2
for enhanced CDT of orthotopic glioma. Reprinted with permission from
Song et al. (2021). Copyright 2021. Elsevier Ltd
ZHONG ET AL.17 of 43
Cu
2+
ions could be complexed onto the shell, dermorphin could be loaded for BBB crossing, and arginine-
glycine-aspartic acid (RGD) peptide could be further anchored for active targeting to form the final M-CSTD.
NHAc/Cu(II) complexes. After intravenous injection, this intelligent platform could cross the BBB and target
orthotopic glioma for CDT, causing LPO, cell cycle arrest, and cell apoptosis. In vivo CDT efficacy was further
confirmed through the elevated survival rate and relieved histological examinations. Collectively, these Cu-based
chemodynamic agents enhanced the precision and effect of CDT by modulating the hallmarks of the TME,
including pH, H
2
O
2
, and GSH, to relieve harsh conditions or utilize these features to initiate CDT with minimized
side effects on normal cells.
4|Cu-BASED NMs FOR COMBINED CANCER THERAPY
In most cases, the antitumor outcomes of combined therapies are not an additive effect simply by summing the effect of
each therapy alone but rather a synergistic effect (Liu et al., 2019). Cu-based NMs with unique properties have achieved
success in cancer monotherapy, including PTT and dynamic therapies of PDT, SDT, RDT, and CDT. However, the mul-
tiple physical, chemical, and catalytic properties of Cu ions enable Cu-based NMs to wear many hats, which will realize
satisfactory therapeutic effects of cancer combination therapy (Table 3).
4.1 |PTT-based combined cancer therapy
Although some achievements have been made in the field of tumor optical therapy, certain disadvantages or biological
barriers still exist that limit the effectiveness of PTT. For example, the greatest drawback is the limited depth of light
penetration due to the decreased laser intensity, the overexpression of heat shock protein (HSP) that makes cancer cells
resistant to hyperthermia, and even overheating or inhomogeneous heat distribution within healthy tissue or tumors.
However, combining PTT with other treatment modalities would achieve unexpectedly good results because hyperther-
mia can increase regional blood flow, speed up cellular metabolism, and enhance membrane permeability, which is
also beneficial for the cellular uptake of drugs and many types of sensitizers, thus providing favorable conditions for
chemotherapy and many dynamic therapies (Poudel et al., 2019). This is the theoretical basis of combination therapy of
PTT with other therapies. Considerable attention has been devoted to combining PTT with many other treatment
modalities (Xie et al., 2020). Below, we introduce PTT-based combination therapy with chemotherapy, PDT, RT, CDT,
SDT, gene therapy, and immunotherapy.
4.1.1 | Combination of PTT with chemotherapy
For most cancers, chemotherapy is still considered a prominent strategy in the clinic. However, the aimless distribution
of chemodrugs in the body, in other words, the nonspecific target to the tumor site, will cause underdose but induce
systemic toxicity and side effects on healthy cells (Yan et al., 2020). To deliver targeted chemotherapeutic drugs to the
tumor site, drug delivery systems (DDSs) based on organic, inorganic, or hybrid DDSs as trucks would enhance the
drug contents within tumors (Yan et al., 2021). To function as high-efficiency DDSs, the five features principleshould
be satisfied, including long circulation,enhanced tumor accumulation,deep tumor penetration,efficient cellular
internalization, and drug release(Yong et al., 2020).
Cu-based NMs can act not only as PTAs but also as DDSs to combine PTT and chemotherapy to achieve synergetic
anticancer effects. For example, copper-doped layered double hydroxide (Cu-LDH) NPs were chosen as PTA and the
DDS of two FDA-approved chemodrugs, that is, 5-Fluorouracil (5-FU) and albumin-bound paclitaxel (nAb-PTX) for
PTT and chemotherapy of breast cancer (Liu, Liu, Zhang, et al., 2021). The two drugs were functionally complemen-
tary; 5-FU inhibited thymidylate synthase (TS) activity to prevent DNA/RNA synthesis, while PTX inhibited microtu-
bule disassembly to disrupt normal dynamic reorganization of the microtubule network to induce cell apoptosis. 5-FU/
Cu-LDH@nAb-PTX showed pH-sensitive release, and heat facilitated the on-demand release of 5-FU and nAb-PTX.
Under 808-nm laser irradiation at 0.75 W cm
2
for 3 min, a very low dose of 5-FU (0.25 mg kg
1
) and nAb-PTX
(0.50 mg kg
1
), which were 850 times less than those used in other nanodrugs, could effectively eliminate 4T1 tumors
in vivo with no observable side effects.
18 of 43 ZHONG ET AL.
TABLE 3 Cu-based NMs for cancer combination therapy
Classification Cu-based NMs Parameters References
PTTchemotherapy 5-FU/Cu-LDH@nAb-
PTX
808-nm laser (0.75 W cm
2
, 3 min)
5-FU (0.25 mg kg
1
), nAb-PTX
(0.50 mg kg
1
)
Liu, Liu, Zhang,
et al. (2021)
PDC/P@HCuS 1060-nm laser (2.7 W cm
2
, 6 min)
P@HCuS (25 mg kg-
1
), PDC (4 mg kg
1
)
Sun et al. (2019)
BSD 808-nm laser (1.0 W cm
2
, 5 min)
Cu (5 mg kg
1
)
Pan et al. (2021)
DSF@PVP/Cu-HMPB 808-nm laser (1.5 W cm
2
)
DSF@PVP/Cu-HMPB (20 mg kg
1
)
Wu et al. (2020)
P-V@TPDOX 808-nm laser (1 W cm
2
)
200 μgml
1
, 200 μl
Ji et al. (2021)
AuNRs-CTN@THA 808-nm laser (1 W cm
2
, 3 min)
CTN (2 mg kg
1
day
1
6 days)
Gao et al. (2020)
PTTPDT TPP-ZC-IR-PNPs 808-nm laser (1 W cm
2
, 5 min)
ZCNP (5 mg kg
1
), IR 780 (2.5 mg kg
1
)
Ruttala et al. (2021)
B-CuS-FA 808-nm laser (1.0 W cm
2
, 8 min)
200 μgml
1
Jana et al. (2020)
MCNs-Cu 808-nm laser (1 W cm
2
)
10 mg kg
1
You et al. (2020)
Cu-DhaTph 808-nm laser (1.5 W cm
2
, 10 min)
2mgml
1
, 100 μl
Feng et al. (2021)
MCIH 808-nm laser (0.76 W cm
2
, 10 min)
2mgml
1
, 100 μl
Sun et al. (2020)
PhA@NanoICG 808-nm laser (1.0 W cm
2
, 5 min)
630-nm laser (0.5 W cm
2
, 3 min)
ICG (1.0 mg kg
1
), PhA (0.8 mg kg
1
)
Chen, Zuo, et al. (2021)
PTTRT CuS/
131
I-PEGDA 915-nm laser (1.0 W cm
2
,43C for
20 min) 1
915-nm laser (1.0 W cm
2
,43C for
10 min) 2
CuS NPs (9.5 mg kg
1
),
131
I (50 μCi)
Meng et al. (2018)
AuPt@CuS 808-nm laser (1.0 W cm
2
, 10 min)
2.0 mg ml
1
,25μl
X-ray (50 kV, 75 μA, 10 min)
Cai et al. (2021)
PTTCDT BSA-CuFeS
2
808-nm laser (1.5 W cm
2
, 5 min)
15 mg kg
1
Chen, Luo, et al. (2019)
CuFe
2
O
4
808-nm laser (1.0 W cm
2
, 5 min)
10 mg kg
1
Liu, Liu, Wan,
et al. (2021)
C
9
S
8
808-nm laser (0.5 W cm
2
, 5 min)
20 mg kg
1
Wang, An, et al. (2020)
PEG-Cu
2
Se 1064-nm laser (0.75 or 1.0 W cm
2
, 10 min)
5mgkg
1
Wang, Zhong,
et al. (2019)
TRF-mCuGd 808-nm laser (0.8 W cm
2
, 10 min)
2mgkg
1
Zhang, Xie, et al. (2021)
BP@Cu
0.4
@PEG-RGD 808-nm laser (1.0 W cm
2
, 2 min)
100 ppm
Lyu et al. (2020)
PTTSDT PCPT 808-nm laser (1.0 W cm
2
, 7 min)
US (1.0 MHz, 1.0 W cm
2
, 60% duty cycle,
5 min)
20 mg kg
1
Liang et al. (2019)
(Continues)
ZHONG ET AL.19 of 43
In addition to copper-doped LDH, hollow copper sulfide (HCuS) NPs were also proven suitable for drug loading.
However, during circulation before reaching the tumor site, the premature drug is very likely to happen from the naked
HCuS. Therefore, developing a stimuli-responsive gatekeeper on the surface of the pores is a good choice. Moreover,
molecular targeted prodrugs might be more effective in ameliorating chemotherapy. Herein, amphiphilic fluorescence-
labeled copolymer (fPEDC)-coated HCuS NPs were used to load molecular-targeted prodrugs (PDCs), forming
PDC/P@HCuS for chemo-PTT of breast cancer (Sun et al., 2019). In the small-molecule PDC of cRGD-SMCC-DM1,
maytansinoid (DM1) worked as a cytotoxic agent for chemotherapy; cRGD was a homing peptide; and uncleavable
thioether (SMCC) functioned as the linker. Compared with free DM1, peptide-conjugated DM1 (PDCs) could target
integrin α
v
β
3
by cRGD, which was cytotoxic to MDA-MB-231 cells, as they were confirmed to be α
v
β
3
-positive. In the
drug release experiment, DM1 was stable in PDC when dispersed at pH 7.4. At pH 5.2, the cumulative drug released
from PDC/P@HCuS was improved from 34.4% to 61.3% compared with fPEDC noncoated PDC/HCuS, revealing the
pH-responsive feature of fPEDC for controlled drug release. From another point, the weakly alkaline pyridine with
PDC had a relatively low level of protonation in the high-redox TME, which played a more key role in drug release than
pH. Under 1060-nm laser irradiation, this process could be further facilitated for burst release. In vitro studies demon-
strated that this strategy with high cytotoxicity to tumor cells was mainly based on affecting the cell cycle, cytoskeleton,
cellular proteomics, and permeability. In vivo study further indicated that only the combination therapy could actualize
the best efficacy.
Despite the intelligent and responsive properties of the PDC/P@HCuS nanoplatform, the fabrication process was
complex and time consuming. The strategy of in situ drug generation and controllable loading looks more attractive.
TABLE 3 (Continued)
Classification Cu-based NMs Parameters References
PTTgene therapy RGD-CuS DENPs/
pDNA
1064-nm laser (0.6 W cm
2
, 5 min)
[Cu] =(4 mM, 100 μl), pDNA (10 μg)
Ouyang et al. (2021)
PTTimmunotherapy CuS NPs-PEG-Ma 808-nm laser (0.45 W cm
2
, 5 min)
CuS NPs-PEG-Mal (15 mM, 50 μl)
anti-PD-L1 (50 μg)
Wang, He, et al. (2019)
AM@DLMSN@CuS/
R848
980-nm laser (1.0 W cm
2
, 10 min)
CuS (30 mg kg
1
), R848 (3 mg kg
1
)
AUNP-12 (5 mg kg
1
)
Cheng et al. (2020)
CSP@IL-12 1064-nm laser (0.6 W cm
2
, 5 min)
CSP (172.4 μg), IL-12 (10 μg)
Lin et al. (2021)
ChemotherapyCDT Cu-TG 9.2 mg ml
1
,50μl Yuan et al. (2020)
ChemotherapySTCDT PGC-DOX PGC (10 mg kg
1
), DOX (5.7 mg kg
1
) Fu et al. (2021)
Chemo-gas therapyCDT AIBA@FeCuS-FeCO US (1 W cm
2
, 50% duty, 3 min)
US (1 W cm
2
, 100% duty, 5 min)
AIBA@FeCuS-FeCO (500 μgml
1
, 150 μl)
DSF (2 mg kg
1
)
Sun, An, et al. (2021)
PTTPDTchemo-
immunotherapy
FA-CD@PP-CpG 808-nm laser (0.987 W cm
2
, 5 min)
650-nm laser (4.5 mW cm
2
, 5 min)
Chen, Zhou, et al. (2019)
SDTCDT PtCu
3
US (3.0 W cm
2
, 35 kHz, 10 min)
10 mg kg
1
Zhong et al. (2020)
MWDTTchemotherapy RBC-Zr@APC/C MW (0.9 W, 5 min), 50 mg kg
1
Chen, Wu, et al. (2021)
H
2
therapyPTTPDTCDT UCCZ-FA 980-nm laser (1 W cm
2
, 5 min)
1mgml
1
, 200 μl
Wang, Ji, et al. (2020)
PDTCDTICB Cu-TBP-αPD-L1 LED lamp (650 nm, 100 mW cm
2
, 30 min)
TBP (0.2 μmol)
αPD-L1 (75 μg3)
Ni et al. (2019)
PTTPDTCDTCIT
immunotherapy
Cu
2
O@CaCO
3
-anti-
CD47
1064-nm laser (0.5 W cm
2
, 5 min)
Cu
2
O@CaCO
3
(5 mg kg
1
), anti-CD47
(100 μg)
Chang et al. (2020)
20 of 43 ZHONG ET AL.
For example, CuS and copper diethyldithiocarbamate (Cu(DTC)
2
) were coloaded with bovine serum albumin (BSA) via
a one-pot biomineralization-mimicking synthesis, forming BSD NPs, which were prepared for chemo-photothermal
therapy of melanoma (Figure 8a; Pan et al., 2021). By providing Cu
2+
,S
2
, and DTC in water, the PTA of CuS and
chemodrug Cu(DTC)
2
could be concurrently synthesized in situ with BSA as the biomacromolecular template. Of these,
Cu(DTC)
2
was originally the main metabolite of the alcohol-abuse drug disulfiram (DSF), but it was highly cytotoxic to
cancer cells and has been identified as an anticancer drug. Overall, NIR light-irradiated BSD NPs efficiently arrived at
tumor sites and ablated B16 tumors. Similarly, DSF was also used as the prodrug in another study, but the difference
was that DSF and Cu
2+
were separated before reaching the tumor. Only under the stimulus of endogenous mild acidity
within tumors could make cytotoxic bis(N,N-diethyl dithiocarbamato)copper(II) complexes (CuL
2
) be formed via a
DSF-Cu
2+
chelating reaction. This was due to the pH-responsive biodegradation of hollow mesoporous Prussian blue
(HMPB) nanocages (Figure 8b; Wu et al., 2020).
For cancer chemotherapy, multidrug resistance (MDR) usually occurs with poor treatment outcomes. Overcoming
MDR is still a challenge in developing effective DDSs. Herein, a Cu
2x
Se-based photothermal vector (PT-V) was synthe-
sized to effectively deliver triphenylphosphine (TP)-modified DOX (TPDOX), forming PT-V@TPDOX to mitochondria
to overcome MDR cancer (Figure 8c; Ji et al., 2021). This formulation with high toxicity toward MCF-7/ADR cells was
produced via four pathways: (1) folic acid (FA)-receptor mediated endocytosis, (2) photothermally controlled drug
release, (3) TP-directed mitochondrial targeting of TPDOX, and (4) inhibition of P-glycoprotein (P-gp)-mediated drug
FIGURE 8 Cu-based NMs for combined PTT with chemotherapy. (a) In situ drug generation of cu(DTC)
2
for chemo-photothermal
therapy of melanoma. Reprinted with permission from Pan et al., (2021). Copyright 2020. Elsevier BV. (b) DSF@PVP/Cu-HMPB amplified
chemo-photothermal therapy of breast cancer. Reprinted with permission from Wu et al. (2020). Copyright 2020. WILEY-VCH Verlag
GmbH & Co. KGaA, Weinheim. (c) PT-V@TPDOX for combined PTT-chemotherapy of multidrug-resistant human breast cancer. Reprinted
with permission from Sun, An, et al. (2021). Copyright 2020. Elsevier Ltd. (d) Hyaluronic acid-coated hybrid nanocomposite of AuNRs-
CTN@THA for chemo-photothermal therapy of melanoma. Reprinted with permission from Gao et al. (2020). Copyright 2020. The Royal
Society of Chemistry
ZHONG ET AL.21 of 43
efflux for enhanced drug retention. Finally, PT-V@TPDOX showed a powerful inhibition rate of 97.3% against MDR
tumors.
On-demand and stimuli-responsive drug release plays important roles in elevating the efficacy of chemotherapy and
reducing side effects. However, according to the five features principleof deep tumor penetration, DDSs with the abil-
ity to carry chemo-drugs into hypoxic areas that lack or are far from blood vessels are an important parameter to assess
their efficacy in chemotherapy. To validate this possibility, a hybrid nanocomposite (AuNRs-CTN@THA) was designed
for synergetic chemo-PTT of deep tumors (Figure 8d; Gao et al., 2020). Thiolated hyaluronic acid (THA) is a ligand rec-
ognizer that targets CD44 receptors that are highly expressed on B16F10 cells. CTN, a chemotherapeutic agent of [Cu
(ttpy)(NO
3
)
2
] (ttpy =40-p-tolyl-2,20:6,200-terpyridine) (CTN), has high DNA affinity to possibly induce DNA cleavage
activity. Gold nanorods (AuNRs), of course, are famous effective PTAs. All of these factors contributed to photo-
thermally enhanced drug penetration and retention, which was validated by using a 3D tumor spheroid model and mel-
anoma tumor-bearing mice, respectively. This study was a striking example to stress the significance of deep tumor
penetration in chemotherapy. With improved delivery, even distribution, reduced systemic toxicity, and precisely con-
trolled drug release of chemotherapeutics by light, tumor cells appeared more vulnerable to chemotherapy, suggesting
the greater benefits of combined PTT-chemotherapy.
4.1.2 | Combination of PTT with PDT
PTT and PDT are the two types of burgeoning therapeutic modalities triggered by light in cancer therapy. Because of
their similar responsiveness to light, PSs and PTAs, joint PTT with PDT has been considered a feasible way to improve
synergistic anticancer effects (Chu et al., 2022; Gai et al., 2018). One reason is that PTT can increase O
2
perfusion and
nanoagent penetration by hyperthermia effects; another is that some nanoagents possess both PTT and PDT effects
under irradiation with one or two light sources, which simplifies the operation process.
Specific chemical reaction-based bioorthogonal chemistry has emerged as a powerful tool in cancer therapy
(Liu, Hu, Wu, et al., 2021). Due to the highly catalytic Cu(I) and the highly specific bioorthogonality of azides and
alkynes, Cu(I)-catalyzed azide-alkyne 1,3-cycloaddition (CuAAC) reactions have drawn worldwide attention in various
fields, including cancer therapy. Recently, heterogeneous Cu nanoparticles (CuNPs) formulated in situ on mesoporous
carbon nanospheres (MCNs) have been utilized for bioorthogonally assisted PTT and PDT combination therapy of
cervical cancer via an NIR light-promoted CuAAC reaction (Figure 9a; You et al., 2020). In the MCNs-Cu system,
MCNs could act as PSs for PDT, followed by oxidizing Cu(0) to Cu(I) by ROS to enhance the CuAAC reaction. Simulta-
neously, MCNs could also function as PTAs to transform NIR light into heat, which could further accelerate the
catalytic process by increasing the local temperature. As expected, the MCNs-Cu nanocatalyst significantly inhibited
tumor growth and protected healthy tissues. Therefore, this work provides a greater possibility to realize bioorthogonal
chemistry-mediated cancer therapy. In addition to MCNs, another phototherapeutic agent with both sufficient PCE and
highly efficient ROS production was also reported in another study, since this was still a huge challenge. In this study,
carrier-free PhA@NanoICG was synthetized via copper coordination-driven self-assembly for PTT-PDT of breast cancer
(Figure 9b; Chen, Zuo, et al., 2021). Indocyanine green (ICG), as the PTA and PS of pheophorbide A (PhA), was tightly
connected with Cu
2+
and acted as a bridge. Notably, via J-aggregation, the absorption peak of NanoICG was redshifted,
which significantly increased the PCE of ICG for PTT under 808-nm laser irradiation. Once accumulated in tumors,
PhA@NanoICG could be disassembled due to the weak acidity and highly reductive GSH. Since then, the tumors that
lost antioxidant-defense ability would improve the sensitivity to ROS that were generated from PhA under 630-nm laser
irradiation. In summary, a carrier-free nanoagent with dual acidity/GSH responsiveness was successfully developed for
tumor phototherapy.
To achieve synergistic PTT-PDT, three challenges remain: complex conjugation chemistry, absorption wavelength
mismatch, and inadequate biodegradability of the nanoagents. The above two examples solved the first issue with click
chemistry or coordination chemistry, while the remaining two issues should be addressed. Herein, CuS nanodot-
anchored FA-modified black phosphorus nanosheets (BP-CuS-FA) were designed (Figure 9c; Jana et al., 2020). CuS
nanodots with high absorbance in the NIR region contributed to the good performance in PTT. In addition, BP func-
tioned as a PS to generate
1
O
2
for PDT, as revealed by both the DPBF and ABDA probes. Under the guidance of FA,
folate receptor-overexpressing 4T1 tumors were significantly damaged after synergistic PTTPDT treatment. More
importantly, both XPS and NMR spectrum analyses proved that BP nanosheets were dissociated into PO
43
anions
under laser irradiation, enabling the degraded species to be excreted efficiently from the kidney.
22 of 43 ZHONG ET AL.
FIGURE 9 Cu-based NMs for combined PTT with PDT. (a) Heterogeneous MCN-Cu nanocatalyst for PTT-PDT of cervical cancer via
CuAAC reaction. Reprinted with permission from You et al., (2020). Copyright 2020. American Chemical Society. (b) Carrier-free metal-
coordinated PhA@NanoICG for PTT-PDT of breast cancer. Reprinted with permission from Chen, Zuo, et al. (2021). Copyright 2021.
Elsevier Inc. (c) Clearable BP-CuS-FA nanoconjugate for synergistic PTT-PDT of breast cancer. Reprinted with permission from Jana
et al. (2020). Copyright 2020. American Chemical Society. (d) H
2
S-activable PTT-PDT of human colon cancer by Cu(II)-porphyrin-derived
nanoscale COF of Cu-DhaTph. Reprinted with permission from Feng et al. (2021). Copyright 2021. The Royal Society of Chemistry
ZHONG ET AL.23 of 43
In fact, the BP-CuS-FA-based PTT-PDT strategy relied on two separate PTA and PS, and constructing a simple struc-
ture with both faces of PTA and PS through a step-economical approach method is highly imperative and challenging.
To meet this challenge, Cu(II)-porphyrin-derived nanoscale covalent organic frameworks (COFs) of Cu-DhaTph were
fabricated to realize PTT/PDT via an in situ sulfidation reaction by utilizing the high content of H
2
S in colon tumor
sites (Figure 9d; Feng et al., 2021). The concentration of H
2
S in colon tumors ranges from 0.3 to 3.4 mM; specifically,
Cu
2+
with high affinity to S
2
could grab H
2
S to form photothermal CuS for PTT. After CuS formation, the released
DhaTph responded to the same light to generate
1
O
2
for PDT consequently. In this way, endogenous H
2
S-triggered in
situ PDT/PTT was realized for local colon cancer. Under one or two light irradiation cycles, two different physical and
chemical processes could be synchronously initiated, generating heat and oxidase stress to kill cancer cells in a
collaborative way.
4.1.3 | Combining PTT with radiotherapy
RT, which consists of external beam radiotherapy (EBRT) and internal radioisotope therapy (RIT), or brachytherapy,
has been widely used as the mainstream treatment for more than 50% of patients with cancer for curative or palliative
purposes (De Ruysscher et al., 2019). Regardless of the subtype of RT applied, O
2
is essential to fix radioactive ray-
generated ROS to further improve the RT effect. Unfortunately, tumor hypoxia hinders the desired anticancer effect of
RT. It has been reported that PTT with mild heat can increase blood supply by elevating the blood flow rate and vasodi-
lation (Lyu et al., 2020). In turn, NIR light-excited PTT still suffers from limited tissue penetration depth, especially in
inner tumors. Therefore, rational integration of PTT with RT is highly promising for improving antitumor efficacy.
Nanotechnology provides opportunities to bridge RT and photothermal ablation of tumors. For brachytherapy, clini-
cally radioactive sources such as
125
I beads were found to offer inhomogeneous dose distribution or regression by local
implantation. This would result in a steep dose gradient and poor treatment. To improve the homogeneity and retention
of the radiotherapeutic agent
131
I, an NIR-triggered in situ hybrid hydrogel was designed for PTT-enhanced RT of breast
cancer (Figure 10a; Meng et al., 2018). In this system,
131
I-labeled CuS NPs (CuS/
131
I) were the main part that func-
tioned as
131
I-based brachytherapy and CuS-based PTT; poly(ethylene glycol) double acrylates (PEGDA) as a polymeric
matrix served as the host; and the thermal initiator 2,20-azobis[2-(2-imidazolin-2-yl) propane] dihydrochloride (AIPH)
was used to initiate the polymerization of PEGDA upon 980-nm laser irradiation. After in situ gelation, CuS/
131
I NPs
were locally fixed inside the tumor for a long time without obvious leakage into normal organs. Further assisted by
PTT-induced hypoxia relief, a synergistic PTTRT effect was realized by CuS/
131
I-PEGDA/AIPH, which was far better
than that of CuS/
131
I NPs.
In addition to hydrogel-mediated brachytherapy, high-atomic number elemental nanostructure-based NPs have
increasingly positive effects on the sensitivity of cancer cells to EBRT. Combining the photothermal agent of CuS NPs
with noble metal NPs, a heterostructure can be established to enhance the optical absorption cross sections and then
improve the photothermal performance of CuS NPs. Meanwhile, intraparticle charge transfer can be boosted to speed
up chemical cascade reactions induced by radiation for more energy deposition in cancer cells. Based on this principle,
a strong plasmonplasmon coupling T80-AuPt@CuS heterostructure was constructed for radio-photothermal therapy
of breast cancer (Figure 10b; Cai et al., 2021). In this heterostructure, the surface plasmon resonance (SPR) of AuPt NPs
led to a localized electromagnetic field at their interface upon NIR light irradiation, which increased the mass extinc-
tion coefficient of AuPt@CuS to be 4.50 L g
1
cm
1
, much higher than 2.70 L g
1
cm
1
of the CuS NPs. In the study of
radiation-induced chemical cascade reactions for ROS amplification, three different cytotoxic ROS, incluidng OH,
O
2
, and
1
O
2,
were produced under X-ray irradiation. From the perspective of band-edge positions, this was because
both e
cb
and e
aq
were able to directly transfer to O
2
and H
2
O
2
, generating O
2
and OH, respectively. In addition,
h
vb
+
depleted highly reductive GSH for ROS accumulation. By taking terephthalic acid (THA), 3-bis(2-methoxy-4-nitro-
5-sulfopheh-yl)-2H-tetrazolium-5-carboxanilide (XTT), and singlet oxygen sensor green (SOSG) as indicators, OH,
O
2
, and
1
O
2
were indeed produced with high efficiency, respectively. Due to its high performance in photothermal
and photocatalytic processing, synergistic photothermal ablation, and radiotherapy were achieved without incurring
significant side effects.
The above two works successfully utilized CuS NPs plus with β-ray-emitting nuclides or radiosensitizers of heavy
metallic AuPt for synergistic radio-photothermal therapy for breast cancer therapy. PTT has been reported to perform
multiple functions, including preventing DNA damage repair, arresting the cell cycle, dilating blood vessels, and
24 of 43 ZHONG ET AL.
relieving hypoxia. All these changes contribute to the high susceptibility of tumor cells to RT, consequently causing less
damage to the surrounding normal tissues (Xu & Pu, 2021).
4.1.4 | Combination of PTT with CDT
CDT has been proven to have high specificity and selectivity between cancer cells and normal cells (Yang, Gong,
et al., 2021). However, monotherapy alone is still not strong enough to fight against cancer. Accompanied by other
approaches, such as speeding up the reaction rate, supplying additional H
2
O
2
, and modulating GSH and pH would
improve the performance of CDT (Wang, Zhong, et al., 2020). To date, most of the designed Fenton/Fenton-like agents
can generate OH only in a narrow pH range of 34. The pH value of the TME is weakly acidic, which is not beneficial
FIGURE 10 Cu-based NMs for combined PTT with RT. (a) CuS/
131
I-PEGDA/AIPH hydrogel for enhanced photothermal brachytherapy
of breast cancer. Reprinted with permission from Meng et al., (2018). Copyright 2018. American Chemical Society. (b) Plasmonic
T80-AuPt@CuS heterostructure for synergistic radio-photothermal therapy of breast cancer. Reprinted with permission from Cai
et al., (2021). Copyright 2021. American Chemical Society
ZHONG ET AL.25 of 43
for OH generation. Therefore, developing more energetic and weakly acidic adaptable Fenton/Fenton-like agents with
high ROS productivity is a good way to enhance CDT.
By means of hyperthermia-induced elevated Fenton chemical reaction rates, ultrasmall BSA-CuFeS
2
NPs were pre-
pared via an ecofriendly biomineralization strategy for PTT and pH-independent CDT of breast cancer (Chen
et al., 2020; Chen, Luo, et al., 2019; Ruan et al., 2021; Tan et al., 2022). Interestingly, BSA-CuFeS
2
NPs could produce
comparable amounts of OH over wide pH ranges of 7.4, 6.5, 5.4, 4, and 3, indicating that the BSA-CuFeS
2
NPs dis-
played a pH-independent Fenton-like reaction. To explore the Fenton-like reaction mechanism, the dialysate was ana-
lyzed by inductively coupled plasma atomic emission spectroscopy (ICP-AES) at predetermined time intervals. In all
the different pH buffers, no obvious Cu ions could be detected, indicating that BSA-CuFeS
2
NPs integrally served as a
heterogeneous Fenton-like catalyst but not the released Cu ions to boost the production of OH. In addition, under
808-nm laser irradiation, the heat by BSA-CuFeS
2
NPs was able to further improve the OH-producing efficiency, which
synergistically favored the effective suppression of tumor growth. Moreover, BSA-CuFeS
2
NPs of 4.9 nm in size were
quickly cleared out of the body, effectively avoiding long-term and systemic toxicity. In addition to the simple synthesis
of CuFeS
2
NPs via a bioinspired albumin-mediated strategy, they could be formulated in situ from CuFe
2
O
4
NPs in
colon tumors full of H
2
S. As a proof of concept, CuFe
2
O
4
NPs were constructed as the precursor of CuFeS
2
, and upon
meeting endogenous H
2
S, CuFeS
2
NPs could be formulated in situ for PTT-CDT of colon cancer (Figure 11a; Liu, Liu,
Wan, et al., 2021). The tricky things about this work included three strategies: remodeling the reductive nature of TME
FIGURE 11 Cu-based NMs for combined PTT with CDT. (a) H
2
S-activated PTT-CDT of Cu
9
Fe
9
S
16
NPs by in situ sulfuration of
CuFe
2
O
4
NPs for colon cancer. Reprinted with permission from Liu, Liu, Wan, et al. (2021). Copyright 2021. Elsevier BV. (b) Hollow Cu
2
Se
nanozymes for photothermalcatalytic therapy of breast cancer. Reprinted with permission from Wang, Zhong, et al. (2019). Copyright 2019.
American Chemical Society. (c) TRF-mCuGd hierarchical structure for Cu(I) self-supplying CDT with PTT for human breast cancer.
Reprinted with permission from Liu, Xu, Yang, et al. (2021). Copyright 2021. The Royal Society of Chemistry. (d) BP@Cu
0.4
@PEG-RGD
nanostructures for CDT-enhanced PTT of melanoma, lung cancer, and human breast cancer. Reprinted with permission from Lyu
et al. (2020). American Chemical Society
26 of 43 ZHONG ET AL.
by depleting H
2
S, as well as increasing the amount of OH by hyperthermia and H
2
S accelerated valence conversion
between Fe
2+
and Fe
3+
ions. Thus, H
2
S-depleted CuFe
2
O
4
agents activated specific PTT and CDT for synergistic anti-
cancer effects, providing new insight for developing novel treatments for colon cancer.
In addition to modulating pH or redox properties, increasing the specific surface area has been reported to be
a highly efficient catalytic reaction in ROS generation (Zhuang et al., 2012). Inspired by this, hollow Cu
9
S
8
NPs
with abundant active sites were constructed for the photothermal-enhanced CDT of breast cancer (Wang, An,
et al., 2020). Compared with the solid Cu
9
S
8
NPs, the specific surface area of hollow Cu
9
S
8
NPs was substantially
improved from 19.027 to 33.100 m
2
g
1
,asrevealedbyBrunauerEmmettTeller (BET) analysis. To confirm the
better catalytic efficiency of hollow Cu
9
S
8
NPs,theelectronspinresonance(ESR)spectrumwasusedwith5,5-
dimethyl-1-pyrroline-N-oxide (DMPO) as the trapping agent of OH. The ESR spectrum suggested that hollow
Cu
9
S
8
exhibited a better chemodynamic performance at a higher temperature. Both in vitro and in vivo studies
further verified the synergistic therapeutic effect. Similarly, Cu
2
Se nanocubes with hollow structures were also
successfully prepared for PTT-CDT of breast cancer (Figure 11b; Wang, Zhong, et al., 2019). Compared with
NIR-I light, a 1064-nm laser located within NIR-II (10001350 nm) is capable of a higher penetration depth. Via
an anion exchange method with Cu
2
O nanocubes as the template, Cu
2
Se nanocubes were endowed with an opti-
mized PCE of 50.89% in a high NIR II window, which allowed excellent photothermal effects on 4T1 cells under
1064-nm laser irradiation for PTT. In addition, Cu
2
Se nanocubes showed a strong catalytic rate at pH 7.0, acting
as outstanding chemodynamic agents for CDT. Together, Cu
2
Se nanocube-mediated PTT-CDT achieved greatly
enhanced efficacy compared with PTT or CDT alone. Compared with other Cu-based NMs, Cu
2
Se nanomaterials
are particularly interesting due to the physiological effect played by trace elements of Cu and Se in the
human body.
For Cu-based CDT, the formation of Cu
+
, a highly efficient chemodynamic agent, is usually based on the redox
reaction between Cu
2+
and GSH to initiate CDT. However, with the different interactions, the coordination process
between Cu
2+
and GSH occurred much earlier than their redox process, thus leading to the insufficient generation of
Cu
+
ions. To provide enough Cu
+
ions, a highly efficient nanoplatform of a CuO dot-decorated Cu@Gd
2
O
3
hierarchical
core@shell structure was designed to self-supply Cu
+
ions for PTT-enhanced CDT of breast cancer (Figure 11c; Zhang,
Xie, et al., 2021). Under weakly acidic conditions, the copper oxide (CuO) dots (COD) in the shell were first dissolved to
release Cu
2+
ions, followed by reaction with Cu
0
in the core to produce Cu
+
ions. Finally, with the coexistence of Cu
+
and H
2
O
2
, the Fenton-like reaction could be initiated for CDT. In addition, Cu@Gd
2
O
3
had broad absorption between
700 and 900 nm. Under 808-nm laser irradiation (0.8 W cm
2
), an excellent photothermal effect was achieved. Interest-
ingly, by western blot analysis, superoxide dismutase-1 (SOD-1) was activated by Cu@Gd
2
O
3
, leading to more H
2
O
2
generation for the Cu
+
-catalyzed Fenton reaction. In contrast, Cu@Gd
2
O
3
could also inactivate glutathione peroxidase
4 (GPX4), an important protein that works to eliminate intracellular ROS to maintain redox equilibrium. Therefore, the
inactivation of GPX4 further improved the accumulation of OH, further amplifying oxidative stress in MDA-MB-231
and MCF-7/DDP tumor cells. These results indicated that Cu@Gd
2
O
3,
as a nanozyme that mimics the agonist of SOD-1
and the antagonist of GPX4, accelerated cell death.
In the strategy of photothermally enhanced CDT, the photothermal stability of PTAs should also be considered in
addition to the methods for improving Fenton chemistry. However, PTAs with high photothermal stability usually suf-
fer from difficulty in degradation. Conversely, PTAs with rapid degradation ability may not be good at photothermal
stability. Therefore, balancing the two conflicts of high photothermal stability and rapid degradation ability is a difficult
issue for the desired PTT-CDT of cancer. Considering that, BP was an inherent captor for Cu ions, after capture, the
interaction between BP and Cu ions killed two birds with one stone. Surface Cu
2+
ions enhance the photothermal sta-
bility of BP-based PTAs but accelerate the degradation of BP via redox reactions. Based on this theory, Cu
2+
ions were
tightly captured by BP nanosheets (BPNS) via coordination and electrostatic attraction. Forming BP@Cu
nanostructures for complementary PTT and CDT of melanoma, lung cancer, and human breast cancer under 808-nm
laser irradiation (Figure 11d; Lyu et al., 2020). Similar to the abovementioned Cu
2
Se nanocubes, both P as the
macroelement and Cu as the trace element are necessary for human health, and this unique biosafety shows the clinical
potential of BP@Cu-based PTAs.
Cu-based NMs showed outstanding catalytic performance in Fenton-like reactions by reducing the reliance on
low pH, accelerating the reaction rate by PTT, and modulating the TME of self-supplying H
2
O
2
and GSH depletion.
Additionally, considering their good biocompatibility and biodegradability, they have potential in future
cancer CDT.
ZHONG ET AL.27 of 43
4.1.5 | Combination of PTT with SDT
SDT, with advantages in overcoming the drawbacks of PDT, shows broad and promising application in combating can-
cer. However, as a kind of ROS-based dynamic therapy, SDT efficacy also highly relies on O
2
assistance since it plays a
vital role in transforming into
1
O
2
and O
2
. To supply sufficient O
2
for enhanced SDT, herein, Janus composed of the
hollow semiconductors CuS and Pt (Pt-CuS) was successfully synthesized to load tetra-(4-aminophenyl) porphyrin
(TAPP) for PTT-augmented SDT of colon cancer (Figure 12; Liang et al., 2019). In this design, hollow CuS with a large
inner cavity functioned not only as the DDS for TAPP delivery but also as PTA for PTT under 808-nm laser irradiation.
By depositing Pt, the photothermal performance of CuS was significantly enhanced due to the effect of the local electric
field enhancement. Moreover, Pt also possessed catalase (CAT)-mimicking activity by decomposing endogenous H
2
O
2
to O
2
for hypoxia relief. Importantly, 808-nm laser-irradiated Pt-CuS could further accelerate the catalytic activity of Pt
to elevate the O
2
level, thus facilitating SDT efficacy. Upon further coating with a thermally sensitive copolymer named
poly(oligo(ethylene oxide) methacrylate-co-2-(2-methoxyethoxy) ethyl methacrylate (p-(OEOMA-co-MEMA)),
temperature-responsive TAPP release could be realized. In vivo synergistic PTT-SDT almost completely eliminated the
tumors without obvious reoccurrence at a highly safe dose.
In principle, both US and NIR light could disturb the integrity and enhance the permeability of cell membranes via
thermal effect-based lipid arrangements. In some cases, photothermal agents could also mediate US-excited ROS pro-
duction, bestowing the convenience of operating combined PTT and SDT. Similar to SDT and RDT, Cu-based NMs have
rarely been reported to act as both PTAs and sonosensitizers, which limits their application in combined PTTSDT.
4.1.6 | Combination of PTT with gene therapy
Despite the success of nanomaterial-based anticancer therapy in one solid tumor mimicking a primary tumor, tumor
metastasis is the main reason for the decline in survival rates. Therefore, complete containment of the primary tumor
FIGURE 12 Cu-based NMs for combined PTT with SDT. Hollow PCPT Janus architecture for synergistic PTT-SDT of colon cancer.
Reprinted with permission from Liang et al. (2019). Copyright 2019. American Chemical Society
28 of 43 ZHONG ET AL.
in its early stages and timely suppression of tumor metastasis would improve the survival of patients. However, the syn-
chronous inhibition of primary and metastatic tumors is still a challenging task.
Based on the theranostic nanoplatform, this task would be solved for improved survival rates. PTT, as a local treat-
ment modality, is powerful in destroying the cell membrane structure and degenerating nuclear DNA, RNA, and pro-
teins of primary tumors. For example, CuS NPs have attracted immense interest for the PTT of tumors (Nikam
et al., 2020). For the inhibition of cancer metastasis, genetherapyhasbeenproventobeaneffectivemethodamong
various newly developed methods (Gilam et al., 2016). Gene therapy is mainly divided into DNA therapeutics and
RNA therapeutics, with the former including plasmid DNA (pDNA), oligodeoxynucleotides, and DNA aptamers and
the latter including miRNA, siRNA, ribozymes, and circular RNAs (Mirza & Karim, 2021). However, DDSs-mediated
gene delivery suffers from limited transfection efficacy and poor gene manipulation duetoinadequatecellular
uptake. Fortunately, hyperthermia could improve the targeting of nucleotides to the nucleus by loosening the cell
membrane. Taking advantage of individual PTT and gene therapy, a synergistic antitumor effect would be achieved
via their interplay. Herein, CuS nanoparticles complexed with plasmid DNA-encoding hypermethylation in cancer
1 (pDNA-HIC1) were entrapped by a dendrimer, forming RGD-CuS DENPs/pDNA polyplexes for simultaneous
photothermal-gene therapy of breast cancer (Figure 13; Ouyang et al., 2021). With RGD navigation, CuS NPs generat-
ing heat significantly delayed tumor growth under 1064-nm laser irradiation. Moreover, the delivered pDNA-HIC1
could inhibit cancer cell invasion and metastasis in a serum-enhancing manner via upregulated expression of HIC1,
a tumor suppressor gene, by methylation that could be promoted in triple-negative breast cancer (TNBC). In vivo
FIGURE 13 Cu-based NMs for combined PTT with gene therapy. RGD-CuS DENPs/pDNA polyplexes directed combined PTT/gene
therapy of human breast cancer. Reprinted with permission from Ouyang et al. (2021). Copyright 2021. American Chemical Society
ZHONG ET AL.29 of 43
studies demonstrated that NIR light-irradiated RGD-CuS DENPs/pDNA polyplexes effectively inhibited primary
tumor and lung metastasis for prolonged survival. Cu-based NMs are easily prepared to increase the transfection effi-
ciency of genes by manipulating physical and chemical properties, showing promise in highly effective PTT-gene
therapy against cancer.
4.1.7 | Combination of PTT with immunotherapy
PTT by NIR light irradiating localized tumors can stimulate the host's immune system by causing the release of tumor
antigens after tumor cell apoptosis and necrosis, which in turn promotes antigen presentation to T cells to induce a dis-
tal effect. However, PTT alone does not efficiently control tumor metastasis and recurrence just by activating the host's
immune system because different tumor cells can escape recognition and response of the immune system via specific
inhibitory signaling pathways.
Cancer immunotherapy has become a research hotspot and has achieved tremendous progress in recent decades. At
present, including four types of immune checkpoint blockade (ICB) therapy, cytokine therapy, cancer vaccines, and chi-
meric antigen receptor (CAR)-T cell therapy, immunotherapy has exhibited encouraging clinical responses in some
lucky patients (Chen, Chen, & Liu, 2019). However, the abovementioned strategies suffer from many shortcomings,
including low clinical objective response rates, risk of causing autoimmune diseases, off-target side effects, cytokine
release syndrome, and complicated manufacturing processes with extremely high costs. By learning from each other's
strengths, there is great hope that combining PTT with immunotherapy would make cancers curable in the years to
come. Immune checkpoints, with the negative regulatory ability of immune activation to make tumors immune-resis-
tant, limit antitumor responses with unprecedented rates of long-lasting tumor responses in patients. Considering that
many immune checkpoints, including cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell
death 1 (PD-1), are initiated by ligandreceptor interactions, they can be blocked by antibodies or modulated by recom-
binant forms of ligands or receptors, forming ICB therapy (Pardoll, 2012). Herein, with the aid of the outstanding
hyperthermic performance of CuS NPs, anti-PD-L1-mediated ICB was combined with CuS NP-PEG-Mal-based PTT for
breast cancer (Figure 14a; Wang, He, et al., 2019). In this study, CuS NPs not only contributed to the thermal ablation
of tumors for PTT but also worked as an antigen-capturing agent to adsorb and transport antigens released during
hyperthermia to DCs for immune activation by surface modification of maleimide PEG (PEG-Mal). Moreover, with
anti-PD-L1 administration, both primary and distant tumors showed significantly slower growth. In another study, CuS
NPs were also used with PD-1 blockade for photothermal ablation and immune remodeling of triple-negative breast
cancer (TNBC) (Figure 14b; Cheng et al., 2020). The differences compared with the above work contain three points.
First, with dendritic large-pore mesoporous silica nanoparticles (DLMSNs) as the DDSs, CuS NPs were deposited in situ
inside the large pores of DLMSNs, and the immune adjuvant resiquimod R848 could be loaded. Second, the 4T1 cell
membrane homogenously covered the surfaces of DLMSNs to improve the homing ability to tumors and conjugation
with peptides. Third, the anti-PD-1 peptide of AUNP-12 was linked by an acid-labile benzoic-imine bond consisting of
PEG. After step-by-step preparation, the final functional AM@DLMSN@CuS/R848 was applied to holistically treat met-
astatic TNBC. Under 980-nm laser irradiation, hyperthermia was generated, followed by tumor antigen generation,
increased R848 release, weakly acidic TME responsive AUNP-12 detachment, all these changes synergistically exerted
tumor vaccination, and T lymphocyte activation to prevent TNBC recurrence and metastasis.
In addition to ICB, cytokine therapy as a chief participant in regulating immune responses also shows promise in
cancer immunotherapy. Cytokines participate in regulating the survival, activation, and differentiation of T cells.
Inflammatory cytokines such as interleukin 1β(IL1β), IL2, IL12, IL18, tumor necrosis factor α(TNF-α), and interferon-
γ(IFN-γ) can promote innate and adaptive immune responses, playing a positive role in fighting against immune resis-
tance (Wang & Mooney, 2018). Among these cytokines, IL-12 can interact with other members of the immune system.
It has been reported that IL-12 promotes not only type 1 helper T (Th1) cell differentiation and the proliferation of natu-
ral killer (NK) cells and cytotoxic T lymphocytes (CTLs) but also the secretion of IL-2, TNF-α, IFN-γ, granulocyte-
macrophage colony-stimulating factor (GM-CSF), perforin, and granzyme B, systematically enhancing antitumor
immune responses. However, fetal adverse effects appeared with systemic administration of IL-12, but intratumoral
injection of the IL-12 gene exhibited many superiorities of sustained local expression and sustained effective concentra-
tion of local IL-12 compared with direct delivery of recombinant IL-12 protein. Considering these facts, mesoporous
SiO
2
was chosen as the DDSs for PTA/gene codelivery to realize CuS-based NIR-II PTT and in situ cytokine therapy
through local generation of IL-12 (Figure 14c; Lin et al., 2021). In the CSP@IL-12 nanocomplex, SiO
2
was
30 of 43 ZHONG ET AL.
functionalized with poly((2-di-methylamino)ethyl methacrylate) (PDMAEMA) to have positive charges. Then, plasmid
DNA encoding the IL-12 gene with negative electrons was attracted to realize in situ expression of the recombinant IL-
12 protein. Combined with 1064-nm laser irradiation, antitumor photoimmunotherapy was systemically improved.
In addition, clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR associated 9 (Cas9) technol-
ogy has been combined with CuS NPs for photothermal immunotherapy (Guo et al., 2014). Cu-based NM-mediated
hyperthermia also controls the expression of several thermosensitive gene elements, such as the HSP promoter, for
enhanced PTT immunotherapy (Sun, Zhang, et al., 2021).
4.2 |Chemotherapy-based combined cancer therapy
Insufficient drug accumulation in tumor cells is the major cause of chemotherapy failure. Although the strategy of
DDSs-assisted chemotherapeutics has shown promise in delivering enhanced drug contents, only 0.7% of the
FIGURE 14 Cu-based NMs for combined PTT with immunotherapy. (a) CuS NPs-PEG-Ma with anti-PD-L1 for combined PTT and ICB
of breast cancer. Reprinted with permission from Wang, He, et al. (2019). Copyright 2019. American Chemical Society.
(b) AM@DLMSN@CuS/R848 biomimetic nanoplatform for PTT and immune remodeling of TNBC. Reprinted with permission from Cheng
et al. (2020). Copyright 2020. American Chemical Society. (c) Combined PTT with in situ cytokine therapy for melanoma. Reprinted with
permission from Lin et al. (2021). Copyright 2021. The Royal Society of Chemistry
ZHONG ET AL.31 of 43
administered NMs could truly arrive at the solid tumor (Wilhelm et al., 2016). Therefore, it remains a great challenge to
improve the targeting ability of chemotherapeutics via localized on-demand release, controlled release, and other
routes, including combination with other treatments, to improve the therapeutic effect.
In this regard, a metal-drug coordination nanoplatform of Cu-TG was developed to achieve efficient treatment of
breast cancer via CDT-chemotherapy (Yuan et al., 2020). Ingeniously, 6-thioguanine (6-TG), as a chemotherapy drug
with abundant N and S, could act as the ligand to bond Cu ions, forming Cu-TG with a high drug loading rate (60.1%)
and 100% utilization rate. Moreover, the sulfhydryl group of 6-TG guaranteed the coexistence of Cu
+
/Cu
2+
, which endo-
wed Cu-TG with multiple nanozyme performances. First, Cu-TG with glutathione peroxidase (GSH-Px)-like activity could
deplete GSH to trigger the release of 6-TG and Cu ions, thereby enhancing the T
1
relaxation rates of Cu ions for T
1
-
weighted MRI and chemotherapy. Second, upon entering tumor cells, the released Cu
+
ions, as Fenton-like agents, could
react with endogenous H
2
O
2
to efficiently produce OH for CDT, showing horseradish peroxidase (HRP)-like activity.
Moreover, O
2
could also be generated under this condition, making Cu-TG highly efficient catalase (CAT)-like activities.
Based on the above properties, Cu-TG exhibited self-reinforcing circular catalysis for synergistic therapeutic outcomes.
Despite the multifunctional nanozyme performance of Cu-TG, the insufficient content of H
2
O
2
inside tumor cells was not
solved in this work. Glucose oxidase (GOx), a kind of natural oxidoreductase, has been proven to be an excellent H
2
O
2
producer due to its high catalytic capacity against β-D-glucose for adequate glucose supply. Herein, for improved
CDT-chemotherapy of breast cancer, an H
2
O
2
self-supplying nanoplatform was prepared through the one-step, biomi-
metic mineralization method. Using PEG-modified GOx as a template, copper-doped calcium phosphate (CuCaP) NPs
could be anchored; further loading with DOX successfully produced biodegradable DOX-loaded CuCaP (PGC-DOX;
Figure 15a; Fu et al., 2021). In this intelligent nanotheranostic, the shortcomings of GOx, including poor stability, short
in vivo half-life, and systemic toxicity, could be significantly reduced by biomineralization. In addition, CaP could sponta-
neously decompose to Ca
2+
and PO
43
, which participate in normal metabolism under acidic TME triggering. After the
collapse of CaP, the cargos in PGC-DOX were consequently released, with GOx exhausting O
2
and supplying H
2
O
2
for
starvation therapy (ST); Cu
2+
ions depleting GSH to activate H
2
O
2
self-reinforcing and GSH depletion amplified CDT;
and DOX for chemotherapy. Taken together, the prepared PGC-DOX realized cooperative cancer therapy by integrating
the three functions.
DDSs that can improve the drug contents in targeted tumors highly rely on the enhanced permeability and retention
(EPR) effect. To amplify the EPR effect for more drug accumulation, physical or pharmacological approaches, including
light/US irradiation and vasodilators, have been proven effective in elevating the endocytosis of NMs (Dhaliwal &
Zheng, 2019). Moreover, these external stimuli can also provide precise spatiotemporal control of drug release at desired
sites of interest. Among various vasodilators, some gases, such as NO and CO, which are endogenous gas transmitters,
are capable of augmenting the EPR effect via vasodilation (Szabo, 2016). Consequently, to achieve US/gas-mediated
CDT-chemotherapy enhancement, hybrid AIBA@FeCuS-FeCO NPs with DSF were fabricated for gas-chemo-
chemodynamic therapy of orthotopic gastric tumors (Figure 15b; Sun, An, et al., 2021). This nanoplatform could
enhance cancer therapy through four different pathways. First, upon US irradiation, the thermal effect could trigger the
decomposition of thermally responsive 2, 20-azobis(2-methylpropionamidine) dihydrochloride (AIBA) into N
2
and alkyl
radicals, with N
2
further driving AIBA@FeCuS-FeCO degradation, thus eliciting alkyl radical-induced cytotoxicity inde-
pendent of O
2
. Second, the generated alkyl radicals induce the decomposition of the CO donor of Fe
3
(CO)
12
, releasing
CO for vasorelaxation to enhance the EPR effect. Third, the alkyl radicals would also break up FeCuS into Fe
2+
and
Cu
2+
ions, and with Fe
2+
interaction with H
2
O
2
,OH-mediated CDT would lead to ferroptosis. Fourth, the released
Cu
2+
chelates with DSF, forming CuL
2
for cancer therapy. By virtue of precise spatiotemporal control, good US-
activated multimodal therapies were realized.
Docetaxel (DTX), a traditional chemotherapeutic drug, was found to decrease the proportion of myeloid-derived
suppressor cells (MDSCs) and induce MDSC transformation from an M2-like phenotype to an M1-like phenotype
(Millrud et al., 2018). M1-type macrophages are desired because they can produce both proinflammatory cytokines and
inducible nitric oxide synthase (iNOS) to kill cancer cells. In contrast, M2-type macrophages promote tumor angiogene-
sis. Therefore, DTX can not only kill cancer cells but also polarize tumor-associated macrophages (TAMs). For breast
cancer, immunotherapy is usually functionally limited owing to cold tumorscharacterized by low PD-L1 expression
and CTL infiltration but high levels of MDSCs that are resistant to the immune system (Galon & Bruni, 2019). From
the perspective of DTX in modulating the tumor immune microenvironment, herein, DTX-loaded nanocomposites of
FA-CuS/DTX@PEI-PpIX-CpG, denoted FA-CD@PP-CpG, were designed for synergistic phototherapy and DTX-based
chemotherapy-enhanced immunotherapy of breast cancer (Figure 15c; Chen, Zhou, et al., 2019). In this nanoplatform,
CuS, a famous PTA, and protoporphyrin IX (PpIX), a widely used PS, could be excited by 808- and 650-nm laser for heat
32 of 43 ZHONG ET AL.
and
1
O
2
generation-based PTT and PDT, respectively. Moreover, on the one hand, the level of CTL infiltration was pro-
moted; on the other hand, MDSCs were effectively polarized toward the M1 phenotype, and these regulatory effects
contributed to the enhanced efficacy against breast cancer. Moreover, further combined with anti-PD-L1 antibody-
based ICB, these nanocomposites offered efficient efficacy in inhibiting breast cancer.
Cu-based NMs have shown excellent anticancer activity by acting as DDSs, multistimuli responsive sensitizers, and
catalysts for chemotherapy, radiotherapy, thermal therapy, and dynamic therapy, showing the potential of these
nanostructures in future cancer treatment.
FIGURE 15 Cu-based NMs for chemotherapy-based combined cancer therapy. (a) Nanocatalytic PGC-DOX with H
2
O
2
self-supply and
GSH-elimination abilities for STCDTchemotherapy of breast cancer. Reprinted with permission from Fu et al. (2021). Copyright 2021.
Wiley-VCH GmbH. (b) AIBA@FeCuS-FeCO with DSF for US-activated gas-chemo-chemodynamic therapy of orthotopic gastric tumors.
Reprinted with permission from Sun, An, et al. (2021). Copyright 2021. American Chemical Society. (c) FA-CD@PP-CpG with aPD-L1 for
PDTPTT chemotherapy and immunotherapy of breast cancer. Reprinted with permission from Chen, Zhou, et al. (2019). Copyright 2019.
WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
ZHONG ET AL.33 of 43
4.3 |Other forms of synergistic cancer therapy
Encouraged by various kinds of Cu-based NMs in different types of treatment modalities, some other synergistic thera-
pies worth mentioning will be introduced in this section. ROS-based dynamic therapy relies heavily on the TME due to
the harsh conditions of hypoxia and high content of reductive GSH, which are not harmful for ROS generation and
accumulation. A nanoagent that can respond to both high-tissue penetrated external stimuli and in situ internal chemi-
cal energy for ROS generation, accompanied by TME modulation ability, would be more attractive in cancer therapy.
By simply synthesizing intermetallic compounds (IMCs) via a one-step solvothermal method, PtCu
3
nanocages were
uniformly synthesized for GSH depletion-amplified sono-chemodynamic therapy of breast cancer (Zhong
et al., 2020). Under US irradiation, PtCu
3
nanocages as inorganic sonosensitizers could produce two types of ROS,
including
1
O
2
and OH, due to the partial formation of Cu
2
O, a kind of PS on their surface. In addition, due to the
existence of Cu
+
,PtCu
3
nanocages could mimic peroxidase (POD), functioning as Fenton-like agents for CDT by
highly generating OH. Interestingly, they as nanozymes could also mimic GSH-Px, depleting 3 mM GSH only by
0.1 mM PtCu
3
nanocages and 0.1 mM H
2
O
2
, showing outstanding performance in the potential of GSH-depleted ROS
accumulation.Bothinvitroandinvivostudiesdemonstrated a significant growth delay, showing promise in
nanozyme-based cancer therapy.
With research interest focusing on microwaves (MWs), Prof. Meng's group developed exogenous stimulation of
MW-mediated microwave dynamic therapy (MDT) and microwave thermal therapy (MWTT) for cancer therapy.
MW-excited ROS generation for MDT shows an outstanding advantage that it can achieve deep tumor treatment
without interference from gas and bone. Similar to PSs and sonosensitizers, microwave sensitizers are also indispens-
ableforMDTs.Forexample,theydevelopedPEG-IL-LM-ZrO
2
SNPs for ROS generation by transferring electrons
from Ga to the surrounding H
2
OandO
2
. However, this sensitizer-mediated MDT still faces some challenges. First,
the quantum yield of ROS generation needs to be improved. Second, ROS-induced cell death and vascular occlusion
prompt surviving tumor cells to secrete vascular endothelial growth factor (VEGF), facilitating the metastasis and
recurrence of tumors. Finally, the sufficient accumulation of microwave sensitizers at the tumor should also be
considered.
Based on the above challenges, the construction of MDT sensitizers that can meet the above requirements is badly
needed. Herein, they designed red blood cell (RBC) membrane-coated ZrO
2
@CuO/Cu-MOF-apatinib-PCM (RBC-
Zr@APC/C) NPs for effective MWTTMDT- chemotherapy of breast cancer (Figure 16a; Chen, Wu, et al., 2021). In this
study, mesoporous hollow ZrO
2
nanospheres were first fabricated via the template-sacrifice method. Then, as a truck,
they codelivered CuO/Cu-MOF, tetradecanol (PCM), and apatinib to the tumors. Finally, by covering with immune-
escape RBCs for stealth, this sensitizer could be highly internalized by tumor cells. Upon MW irradiation, Zr@C/C NPs
could produce O
2
and O
2
,OH and
1
O
2
, especially O
2
, for hypoxia-relieved MDT-MWTT (MWDTT). In addition,
blocked apatinib will be released from the mesoporous shell due to the MWTT-induced switch of PCM in a
temperature-controlled manner. Apatinib, as an antiangiogenic drug, significantly downregulated the expression of
VEGF and platelet endothelial cell adhesion molecule (CD31), thus inhibiting tumor angiogenesis after MWDTT. This
study further expanded the application of Cu-based NMs in combined cancer therapy.
In recent years, gas therapy (GT) using several gaseous molecules (e.g., NO, CO, H
2
S, N
2
, and H
2
) with physiological
modulation functions has emerged as a promising field for cancer therapy (He, 2017). The underlying mechanism of
GT was largely due to interference with tumor cell metabolism. Compared with normal cells, the Warburg effect
appears in cancer cells with energy consumption via a glycolysis route rather than via the aerobic respiration method.
These gases maintain bioenergetic homeostasis to block the survival pathway for cancer cells while protecting normal
cells, showing an anti-Warburg effect. Therefore, GT for cancer has been considered far superior to traditional thera-
pies. Among them, small gas molecules of H
2
with inert and life element properties endow hydrogen therapy with
exceptional treatment performance (Wu et al., 2019). However, direct delivery of H
2
by DDSs from outside the body
would not transport enough H
2
into tumors due to its low solubility and high diffusivity. Therefore, developing NM-
based gas generators in stimuli-responsive ways would improve the efficacy of gas targeting ability. Herein, an NIR
light-driven H
2
-releasing donor of NaGdF
4
:Yb,Tm/g-C
3
N
4
/Cu
3
P (UCC) was encapsulated with zeolitic imidazolate
framework-8 (ZIF-8), forming UCNPs/g-C
3
N
4
/Cu
3
P@ZIF-8 NPs (UCCZ NPs) for H
2
-mediated cascade-amplifying GT
PTTPDTCDT of breast cancer (Figure 16b; Wang, Ji, et al., 2020). Due to the acid responsibility of the ZIF-8 shell and
folate receptor-mediated endocytosis, CSNPs could be selectively engulfed by tumor cells to release UCC. Under
980-nm laser irradiation, NaGdF
4
:Yb,Tm (UCNPs) could emit purple and blue light, which in turn excited g-C
3
N
4
to
generate e
in the conduction band (CB) and excited Cu
3
P to generate h
+
in the valence band (VB), respectively.
34 of 43 ZHONG ET AL.
Generally, e
and h
+
will quickly recombine via the Z-scheme pathway, thus maintaining the stronger oxidizability of
h
+
in the VB of g-C
3
N
4
and the higher reducibility of e
in the CB of Cu
3
P. Moreover, the generated strong oxidizing
h
+
in the VB of g-C
3
N
4
could be reduced by GSH in the TME, thus providing isolated e
in the CB of Cu
3
P to reduce
H
2
O for H
2
production via in situ water splitting-mediated gas therapy. In this process,
1
O
2
and O
2
could also be pro-
duced for PDT via energy or e
transfer to O
2
. At the same time, Cu
3
P could not only dominate PTT but also act as a
GSH-activated Fenton-like agent for photothermally enhanced CDT (ECDT). Significantly, the four types of treatment
modalities were not mutually independent. In detail, both PDT- and CDT-mediated oxidative stress would induce the
expression of stress proteins that would make cancer cells tolerate heat effects from PTT. Fortunately, H
2
can inhibit
the expression of these stress-induced proteins, making ROS-based PDT/CDT more effective in combination with PTT.
In addition, GSH depletion by h
+
in the VB of g-C
3
N
4
or Cu
2+
in Cu
3
P would further enhance PDT. Collectively, the
four synergetic GTPTTPDTCDT treatment modes were realized in a H
2
-mediated cascade amplification process.
Advanced tumors are skilled in dysregulating, hijacking, and depleting immune-related cells, cytokines, and mole-
cules to escape immune surveillance and elimination by immune cells. Although ICB therapy benefits a small subset of
patients, this reality of coldtumors makes immunotherapy feel quite helpless. Turning tumors from coldto hot
would improve the responsiveness of tumors to ICB. Immunogenic RT, PDT, CDT, and chemotherapy have been uti-
lized as immunomodulatory adjuvants to augment ICB. As a hallmark of many cancers, high serum levels of estradiol
(E2) directly affect cell proliferation, cell cycle arrest, and ultimately tumorigenesis (Revankar et al., 2005). From
FIGURE 16 Cu-based NMs for other synergistic therapies. (a) RBC-Zr@APC/C NPs for microwave dynamic thermal therapy and
chemotherapy of breast cancer. Reprinted with permission from Chen, Wu, et al. (2021). Copyright 2021. Elsevier Ltd. (b) H
2
generated
UCCZ-FA for cascade-amplifying synergetic hydrogen therapyPTTPDTCDT of breast cancer. Reprinted with permission from Zhang,
Sun, et al. (2020). Copyright 2020. American Chemical Society. (c) Cu-TBP with α-PD-L1 for synergistic radical therapy of ovarian carcinoma
via PDT-CDT-ICB. Reprinted with permission from Ni et al. (2019). Copyright 2019. Elsevier Inc. (d) Cu
2
O@CaCO
3
nanocomposites with
anti-CD47 antibody for synergistic PTTPDTCDTCITimmunotherapy of colorectal cancer. Reprinted with permission from Chang
et al. (2020). Copyright 2020. Wiley-VCH GmbH
ZHONG ET AL.35 of 43
clinical experience, hormone therapy has been widely used in estrogen receptor-abundant tumors. Interestingly, in the
presence of Cu
2+
-based catalysis, estrogens with DNA will form stable adducts while generating ROS. This phenome-
non could be embezzled to conduct ROS-based cancer therapy by supplying additional Cu
2+
to tumors. Herein, Cu
2+
ions were coordinated with 5,10,15,20-tetrabenzoatoporphyrin (H
4
TBP) ligands, forming a nanoscale metalorganic
framework (nMOF) of Cu-TBP. Additionally, with an anti-PD-L1 antibody, synergistic PDT-CDT-ICB of melanoma and
ovarian carcinoma was achieved (Figure 16c; Ni et al., 2019). In this design, Cu
2+
ions catalyzed E2 metabolism to gen-
erate OH, O
2
, and H
2
O
2
for E2-induced CDT and radical therapies via the Cu-E2 catalytic redox cycle, whereas LED
light-triggered H
4
TBP generated
1
O
2
and Cu-TBP generated O
2
-mediated PDT. In vivo studies showed that B16F10
and SKVO-3 tumors with high E2 levels were sensitive to Cu-TBP-mediated CDT/PDT. Further combined with α-PD-
L1, a strong abscopal effect was observed in the bilateral B16F10 tumor model. This clever combination broadened the
therapeutic effects of CBI on hormonally dysregulated tumor phenotypes.
Ion interference therapy (IIT), originating from the discovery of the clinically common phenomenon of calcifica-
tionin certain tumor types after RT or chemotherapy, has been proven to induce cancer cell death by altering the
metal content balance, including ferroptosis and calcicoptosis (Zhang et al., 2019). Calcium overload-based IIT was
named Ca
2+
interference therapy (CIT). As weak acidity is the typical hallmark of tumors, many Ca
2+
ion-releasing
NMs, including CaO
2
and CaCO
3
, have been reported for CIT (Dong, Feng, Hao, et al., 2020; Zhang et al., 2019). For
example, a Cu
2
O@CaCO
3
@HA (CCH) nanostructure was combined with an anti-CD47 antibody for colorectal cancer
(CRC) turn-ontherapy via synergistic PTT-PDT-CDT-CIT immunotherapy (Figure 16d; Chang et al., 2020). Under
the specific physiological feature of H
2
S overexpression in CRC, the pH-triggered decomposition of CaCO
3
would lead
to the release of Ca
2+
ions for CIT and the core of Cu
2
O for the following functions. Hereafter, with H
2
S sulfuration,
Cu
2
O transformed into metabolizable Cu
31
S
16
nanocrystals for strong NIR light (1064 nm)-irradiated PTT, PDT and
enhanced CDT. Furthermore, in combination with CD47 blockade (anti-CD47), protumoral M2 TAMs could be polar-
ized to the tumoricidal M1 phenotype, reprogramming the immunosuppressive TME to initiate T cell-mediated
immune responses. Therefore, CCH as a TME-triggered turn-ontherapeutic tool was successfully constructed for the
precise targeted therapy of specific CRCs.
5|CONCLUSIONS AND PERSPECTIVES
Cu-based NMs have attracted increasing attention not only in biomedical communities due to their easily modulated
nanostructure and versatile composition but also have high therapeutic properties and imaging functions in the bio-
medical field due to their unique physicochemical properties and biological effects. Therefore, in this review, we sys-
tematically summarized the recent application progress of Cu-based NMs in cancer imaging and tumor therapy. For
cancer imaging, PA imaging, PET imaging, and multimodal imaging based on Cu-based NMs were summarized, as well
as strategies to improve the diagnostic effectiveness. Next, the application of Cu-based NMs in cancer treatment was
introduced, which was mainly divided into monotherapy and combination therapy. Although the application of Cu-
based NMs in tumor imaging and therapy has achieved remarkable achievements and progress in the past few years,
there are still some intractable problems and challenges that need to be overcome to achieve clinical translation.
First, the biosafety of Cu-based NMs needs to be systematically evaluated. The traditional view is that Cu, as a tran-
sition metal element, is more toxic than Fe or Mn, so the application of Cu-based NMs in biomedicine is relatively lim-
ited. Fortunately, the as-prepared Cu-based NMs have good biocompatibility and biosafety, which solves the important
toxicity problem of Cu in biomedicine. Notably, the prerelease of Cu ions from Cu-based NMs should be avoided prior
to exerting therapeutic properties to avoid potential toxicity. However, the long-term retention of Cu-based NMs in vivo
can raise toxicity concerns. Therefore, it is very challenging to prepare ultrasmall or biodegradable Cu-based NMs for
cancer theranostics.
Second, it is necessary to develop Cu-based NMs with high photothermal performance and light penetration ability.
Due to the scattering of light in the tissue, its penetration depth is low, so it can only treat diseases on the surface of the
skin and has limited therapeutic effects on deep-seated diseases. Through precise modulation of the nanostructure,
composition, and physicochemical properties, the photothermal conversion efficiency of Cu-based NMs is enhanced,
and the photoresponse wavelength has been extended to the second NIR biological window, thereby effectively enhanc-
ing the penetration of light into the tissue penetration depth. In addition, the combination of PTT and other treatment
modalities can achieve the effect of synergistic treatment, which further overcomes the problem of low treatment effi-
ciency caused by the low tissue penetration ability of phototherapy.
36 of 43 ZHONG ET AL.
Third, the large-scale fabrication and surface engineering of Cu-based NMs need to be addressed to meet the
requirements of clinical translation. At present, the preparation of Cu-based NMs is still in the laboratory stage, and
there are still many problems in large-scale production, which need to be given more attention in the future.
Last, it is of great significance to prepare Cu-based NMs with modulated TME properties. The complex TME impairs
the therapeutic performance of Cu-based NMs, although some Cu-based NMs have also been reported to have the abil-
ity to modulate the TME, mainly focusing on depleting GSH and reacting with H
2
O
2
to generate OH for CDT, but these
materials alone are not sufficient for high-efficiency tumor treatment.
Most importantly, if these bottlenecks are successfully overcome, the therapeutics of Cu-based NMs will lead to sig-
nificant progress in clinical application. We believe that Cu-based NMs will become highly competitive and promising
multifunctional nanomaterials for cancer imaging therapy in the future.
ACKNOWLEDGMENTS
This work was supported by the Basic and Clinical Cooperative Research and Promotion Program of Anhui Medical
University (2021xkjT028), the Open Fund of Key Laboratory of Antiinflammatory and Immune Medicine (KFJJ-
2021-11), Grants for Scientific Research of BSKY from Anhui Medical University (1406012201), a Jiangsu Natural Sci-
ence Fund for Young Scholars (BK20210730), and the Postdoctoral Science Foundation of China (2021M702383).
CONFLICT OF INTEREST
The authors have declared no conflicts of interest for this article.
AUTHOR CONTRIBUTIONS
Xiaoyan Zhong: Writing original draft (lead). Xingliang Dai: Writing original draft (equal). Yan Wang: Resources
(equal). Hua Wang: Supervision (equal). Haisheng Qian: Supervision (equal). Xianwen Wang: Conceptualization
(lead); funding acquisition (lead); supervision (lead); writing review and editing (supporting).
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this review.
ORCID
Xiaoyan Zhong https://orcid.org/0000-0003-4725-6452
Xianwen Wang https://orcid.org/0000-0002-6343-440X
RELATED WIRES ARTICLES
Multifunctional MnO2 nanoparticles for tumor microenvironment modulation and cancer therapy
REFERENCES
An, L., Wang, X., Rui, X., Lin, J., Yang, H., Tian, Q., Tao, C., & Yang, S. (2018). The in situ sulfidation of Cu
2
O by endogenous H
2
S for colon
cancer theranostics. Angewandte Chemie-International Edition,57, 1578215786.
Cai, R., Xiang, H., Yang, D., Lin, K.-T., Wu, Y., Zhou, R., Gu, Z., Yan, L., Zhao, Y., & Tan, W. (2021). Plasmonic AuPt@CuS Heterostructure
with enhanced synergistic efficacy for Radiophotothermal therapy. Journal of the American Chemical Society,143, 1611316127.
Cai, X., Xie, Z., Ding, B., Shao, S., Liang, S., Pang, M., & Lin, J. (2019). Monodispersed copper(I)-based nano metal-organic framework as a
biodegradable drug carrier with enhanced photodynamic therapy efficacy. Advanced Science,6, 1900848.
Cao, S., Fan, J., Sun, W., Li, F., Li, K., Tai, X., & Peng, X. (2019). A novel Mn-Cu bimetallic complex for enhanced chemodynamic therapy
with simultaneous glutathione depletion. Chemical Communications,55, 1295612959.
Chang, M. Y., Hou, Z. Y., Jin, D. Y., Zhou, J. J., Wang, M., Wang, M. F., Shu, M. M., Ding, B. B., Li, C. X., & Lin, J. (2020). Colorectal tumor
microenvironment-activated bio-decomposable and metabolizable Cu2O@CaCO(3)nanocomposites for synergistic oncotherapy.
Advanced Materials,32, 11.
Chen, B., Zhang, C., Wang, W., Chu, Z., Zha, Z., He, X., Zhou, W., Liu, T., Wang, H., & Qian, H. (2020). Ultrastable AgBiS
2
hollow
nanospheres with cancer cell-specific cytotoxicity for multimodal tumor therapy. ACS Nano,14, 1491914928.
Chen, L., Zhou, L., Wang, C., Han, Y., Lu, Y., Liu, J., Hu, X., Yao, T., Lin, Y., Liang, S., Shi, S., & Dong, C. (2019). Tumor-targeted drug and
CpG delivery system for phototherapy and docetaxel-enhanced immunotherapy with polarization toward M1-type macrophages on triple
negative breast cancers. Advanced Materials,31, 1904997.
Chen, L., Zuo, W., Xiao, Z., Jin, Q., Liu, J., Wu, L., Liu, N., & Zhu, X. (2021). A carrier-free metal-coordinated dual-photosensitizers
nanotheranostic with glutathione-depletion for fluorescence/photoacoustic imaging-guided tumor phototherapy. Journal of Colloid and
Interface Science,600, 243255.
ZHONG ET AL.37 of 43
Chen, Q., Chen, M., & Liu, Z. (2019). Local biomaterials-assisted cancer immunotherapy to trigger systemic antitumor responses. Chemical
Society Reviews,48, 55065526.
Chen, Q., Luo, Y., Du, W. X., Liu, Z., Zhang, S. J., Yang, J. H., Yao, H. L., Liu, T. Z., Ma, M., & Chen, H. R. (2019). Clearable theranostic plat-
form with a pH-independent chemodynamic therapy enhancement strategy for synergetic photothermal tumor therapy. ACS Applied
Materials & Interfaces,11, 1813318144.
Chen, W., & Zhang, J. (2006). Using nanoparticles to enable simultaneous radiation and photodynamic therapies for cancer treatment. Jour-
nal of Nanoscience and Nanotechnology,6, 11591166.
Chen, X., Liu, J., Li, Y., Pandey, N. K., Chen, T., Wang, L., Amador, E. H., Chen, W., Liu, F., Xiao, E., & Chen, W. (2022). Study of copper-
cysteamine based X-ray induced photodynamic therapy and its effects on cancer cell proliferation and migration in a clinical mimic set-
ting. Bioactive Materials,7, 504514.
Chen, Z., Wu, Q., Guo, W., Niu, M., Tan, L., Wen, N., Zhao, L., Fu, C., Yu, J., Ren, X., Liang, P., & Meng, X. (2021). Nanoengineered biomi-
metic cu-based nanoparticles for multifunational and efficient tumor treatment. Biomaterials,276, 121016.
Cheng, Y., Chen, Q., Guo, Z., Li, M., Yang, X., Wan, G., Chen, H., Zhang, Q., & Wang, Y. (2020). An intelligent biomimetic nanoplatform for
holistic treatment of metastatic triple-negative breast cancer via photothermal ablation and immune remodeling. ACS Nano,14, 15161
15181.
Chu, Z., Tian, T., Tao, Z., Yang, J., Chen, B., Chen, H., Wang, W., Yin, P., Xia, X., & Wang, H. (2022). Upconversion nanoparticles@AgBiS
2
core-shell nanoparticles with cancer-cell-specific cytotoxicity for combined photothermal and photodynamic therapy of cancers. Bioactive
Materials,17,7180.
Cui, L., Xiong, C., Zhou, M., Shi, S., Chow, D. S. L., & Li, C. (2018). Integrin alpha v beta 3-targeted cu-64 CuS nanoparticles for PET/CT
imaging and photothermal ablation therapy. Bioconjugate Chemistry,29, 40624071.
De Ruysscher, D., Niedermann, G., Burnet, N. G., Siva, S., Lee, A. W. M., & Hegi-Johnson, F. (2019). Radiotherapy toxicity. Nature Reviews
Disease Primers,5, 20.
Deng, H., Yang, Z., Pang, X., Zhao, C., Tian, J., Wang, Z., & Chen, X. (2022). Self-sufficient copper peroxide loaded pKa-tunable
nanoparticles for lysosome-mediated chemodynamic therapy. Nano Today,42, 101337.
Dhaliwal, A., & Zheng, G. (2019). Improving accessibility of EPR-insensitive tumor phenotypes using EPR-adaptive strategies: Designing a
new perspective in nanomedicine delivery. Theranostics,9, 80918108.
Dong, C., Feng, W., Xu, W., Yu, L., Xiang, H., Chen, Y., & Zhou, J. (2020). The coppery age: Copper (Cu)-involved nanotheranostics.
Advanced Science,7, 2001549.
Dong, L., Li, K., Wen, D., Lu, Y., Du, K., Zhang, M., Gao, X., Feng, J., & Zhang, H. (2019). A highly active (102) surface-induced rapid degra-
dation of a CuS nanotheranostic platform for in situ T-1-weighted magnetic resonance imaging-guided synergistic therapy. Nanoscale,
11, 1285312857.
Dong, L., Zhang, P., Xu, X., Lei, P., Du, K., Zhang, M., Wang, D., Feng, J., Li, W., & Zhang, H. (2018). Simple construction of Cu2-xS: Pt
nanoparticles as nanotheranostic agent for imaging-guided chemo-photothermal synergistic therapy of cancer. Nanoscale,10, 10945
10951.
Dong, Z., Feng, L., Hao, Y., Li, Q., Chen, M., Yang, Z., Zhao, H., & Liu, Z. (2020). Synthesis of CaCO
3
-based nanomedicine for enhanced
sonodynamic therapy via amplification of tumor oxidative stress. Chem,6, 13911407.
Feng, J., Ren, W.-X., Kong, F., & Dong, Y.-B. (2021). A covalent organic framework-based nanoagent for H2S-activable phototherapy against
colon cancer. Chemical Communications,57, 72407243.
Fu, L. H., Wan, Y. L., Qi, C., He, J., Li, C. Y., Yang, C., Xu, H., Lin, J., & Huang, P. (2021). Nanocatalytic theranostics with glutathione deple-
tion and enhanced reactive oxygen species generation for efficient cancer therapy. Advanced Materials,33, 11.
Fu, Q., Zhu, R., Song, J., Yang, H., & Chen, X. (2019). Photoacoustic imaging: Contrast agents and their biomedical applications. Advanced
Materials,31, 1805875.
Gai, S. L., Yang, G. X., Yang, P. P., He, F., Lin, J., Jin, D. Y., & Xing, B. G. (2018). Recent advances in functional nanomaterials for light-
triggered cancer therapy. Nano Today,19, 146187.
Galon, J., & Bruni, D. (2019). Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nature
Reviews Drug Discovery,18, 197218.
Gao, L., Zhang, L., Zhu, X., Chen, J., Zhao, M., Li, S., Yu, C., Hu, L., Qiao, H., & Guo, Z. (2020). Hyaluronic acid functionalized gold
nanorods combined with copper-based therapeutic agents for chemo-photothermal cancer therapy. Journal of Materials Chemistry B,8,
48414845.
Gilam, A., Conde, J., Weissglas-Volkov, D., Oliva, N., Friedman, E., Artzi, N., & Shomron, N. (2016). Local microRNA delivery targets Palla-
din and prevents metastatic breast cancer. Nature Communications,7, 12868.
Goel, S., Chen, F., & Cai, W. (2014). Synthesis and biomedical applications of copper sulfide nanoparticles: From sensors to theranostics.
Small,10, 631645.
Goel, S., Ferreira, C. A., Chen, F., Ellison, P. A., Siamof, C. M., Barnhart, T. E., & Cai, W. (2018). Activatable hybrid nanotheranostics for
tetramodal imaging and synergistic photothermal/photodynamic therapy. Advanced Materials,30, 1704367.
Guo, L., Yan, D. D., Yang, D., Li, Y., Wang, X., Zalewski, O., Yan, B., & Lu, W. (2014). Combinatorial photothermal and immuno cancer
therapy using chitosan-coated hollow copper sulfide nanoparticles. ACS Nano,8, 56705681.
38 of 43 ZHONG ET AL.
Guo, W., Sun, X., Jacobson, O., Yan, X., Min, K., Srivatsan, A., Niu, G., Kiesewetter, D. O., Chang, J., & Chen, X. (2015). Intrinsically radioac-
tive Cu-64 CuInS/ZnS quantum dots for PET and optical imaging: Improved radiochemical stability and controllable Cerenkov lumines-
cence. ACS Nano,9, 488495.
Hao, G. Y., Mastren, T., Silvers, W., Hassan, G., Oz, O. K., & Sun, X. K. (2021). Copper-67 radioimmunotheranostics for simultaneous immu-
notherapy and immuno-SPECT. Scientific Reports,11, 11.
Hao, Y.-N., Zhang, W.-X., Gao, Y.-R., Wei, Y.-N., Shu, Y., & Wang, J.-H. (2021). State-of-the-art advances of copper-based nanostructures in
the enhancement of chemodynamic therapy. Journal of Materials Chemistry B,9, 250266.
He, Q. (2017). Precision gas therapy using intelligent nanomedicine. Biomaterials Science,5, 22262230.
Huang, X., Wan, F., Ma, L., Phan, J. B., Lim, R. X., Li, C., Chen, J., Deng, J., Li, Y., Chen, W., & He, M. (2019). Investigation of copper-
cysteamine nanoparticles as a new photosensitizer for anti-hepatocellular carcinoma. Cancer Biology & Therapy,20, 812825.
Jana, D., Jia, S., Bindra, A. K., Xing, P., Ding, D., & Zhao, Y. (2020). Clearable black phosphorus Nanoconjugate for targeted cancer Photo-
theranostics. ACS Applied Materials & Interfaces,12, 1834218351.
Jaque, D., Maestro, L. M., del Rosal, B., Haro-Gonzalez, P., Benayas, A., Plaza, J. L., Rodriguez, E. M., & Sole, J. G. (2014). Nanoparticles for
photothermal therapies. Nanoscale,6, 94949530.
Ji, C. D., Cheng, W. Y., Hu, Y. S., Liu, Y. F., Liu, F. Y., & Yin, M. Z. (2021). A nano vector with photothermally enhanced drug release and
retention to overcome cancer multidrug resistance. Nano Today,36,7.
Kumari, S., Sharma, N., & Sahi, S. V. (2021). Advances in cancer therapeutics: Conventional thermal therapy to nanotechnology-based
photothermal therapy. Pharmaceutics,13, 1174.
Li, N., Sun, Q., Yu, Z., Gao, X., Pan, W., Wan, X., & Tang, B. (2018). Nuclear-targeted photothermal therapy prevents cancer recurrence with
near-infrared triggered copper sulfide nanoparticles. ACS Nano,12, 51975206.
Li, X., Pan, Z. a., Xiang, C., Yuan, Y., Chen, J., Qing, G., Ma, J., Liang, X.-J., Wu, Y., and Guo, W. (2021). Structure transformable
nanoparticles for photoacoustic imaging-guided photothermal ablation of tumors via enzyme-induced multistage delivery. Chemical
Engineering Journal 421, 127747.
Liang, S., Deng, X., Ma, P. a., Cheng, Z., and Lin, J. (2020). Recent advances in nanomaterial-assisted combinational sonodynamic cancer
therapy. Advanced Materials 32, 2003214.
Liang, S., Deng, X. R., Chang, Y., Sun, C. Q., Shao, S., Xie, Z. X., Xiao, X., Ma, P. A., Zhang, H. Y., Cheng, Z. Y., & Lin, J. (2019). Intelligent
hollow Pt-CuS Janus architecture for synergistic catalysis-enhanced sonodynamic and photothermal cancer therapy. Nano Letters,19,
41344145.
Lin, L.-S., Huang, T., Song, J., Ou, X.-Y., Wang, Z., Deng, H., Tian, R., Liu, Y., Wang, J.-F., Liu, Y., Yu, G., Zhou, Z., Wang, S., Niu, G.,
Yang, H.-H., & Chen, X. (2019). Synthesis of copper peroxide nanodots for H
2
O
2
self-supplying chemodynamic therapy. Journal of the
American Chemical Society,141, 99379945.
Lin, X., Song, J., Chen, X., & Yang, H. (2020). Ultrasound-activated sensitizers and applications. Angewandte Chemie-International Edition,
59, 1421214233.
Lin, X., Wang, X., Li, J., Cai, L., Liao, F., Wu, M., Zheng, D., Zeng, Y., Zhang, Z., Liu, X., Wang, J., & Yao, C. (2021). Localized NIR-II photo-
immunotherapy through the combination of photothermal ablation and in situ generated interleukin-12 cytokine for efficiently eliminat-
ing primary and abscopal tumors. Nanoscale,13, 17451758.
Liu, C., Dong, H., Wu, N., Cao, Y., & Zhang, X. (2018). Plasmonic resonance energy transfer enhanced photodynamic therapy with Au@S-
iO
2
@Cu
2
O/perfluorohexane nanocomposites. ACS Applied Materials & Interfaces,10, 69917002.
Liu, C.-G., Tang, H.-X., Zheng, X., Yang, Y., Zhang, Y., Zhang, J.-T., Kankala, R. K., Wang, S.-B., Liu, G., & Chen, A.-Z. (2020). Near-infra-
red-activated lysosome pathway death induced by ROS generated from layered double hydroxide-copper sulfide nanocomposites. ACS
Applied Materials & Interfaces,12, 4067340683.
Liu, D., Liu, M., Wan, Y., Zhou, X., Yang, S., An, L., Huang, G., & Tian, Q. (2021). Remodeling endogenous H2S microenvironment in colon
cancer to enhance chemodynamic therapy. Chemical Engineering Journal,422, 130098.
Liu, J., Hu, F., Wu, M., Tian, L., Gong, F., Zhong, X., Chen, M., Liu, Z., & Liu, B. (2021). Bioorthogonal coordination polymer nanoparticles
with aggregation-induced emission for deep tumor-penetrating radio- and radiodynamic therapy. Advanced Materials,33, 2007888.
Liu, J., Liu, K., Zhang, L., Zhong, M., Hong, T., Zhang, R., Gao, Y., Li, R., Xu, T., & Xu, Z. P. (2021). Heat/pH-boosted release of
5-fluorouracil and albumin-bound paclitaxel from cu-doped layered double hydroxide nanomedicine for synergistical chemo-photo-
therapy of breast cancer. Journal of Controlled Release,335,4958.
Liu, K., Liu, K., Liu, J., Ren, Q., Zhao, Z., Wu, X., Li, D., Yuan, F., Ye, K., & Li, B. (2020). Copper chalcogenide materials as photothermal
agents for cancer treatment. Nanoscale,12, 29022913.
Liu, S.-Y., Xu, Y., Yang, H., Liu, L., Zhao, M., Yin, W., Xu, Y.-T., Huang, Y., Tan, C., Dai, Z., Zhang, H., Zhang, J.-P., & Chen, X.-M. (2021).
Ultrathin 2D copper(I) 1,2,4-triazolate coordination polymer nanosheets for efficient and selective gene silencing and photodynamic
therapy. Advanced Materials,33, 2100849.
Liu, Y., Bhattarai, P., Dai, Z., & Chen, X. (2019). Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer.
Chemical Society Reviews,48, 20532108.
Lyu, M., Zhu, D. M., Duo, Y. H., Li, Y., & Quan, H. (2020). Bimetallic nanodots for tri-modal CT/MRI/PA imaging and hypoxia-resistant
thermoradiotherapy in the NIR-II biological windows. Biomaterials,233, 11.
ZHONG ET AL.39 of 43
Ma, B., Wang, S., Liu, F., Zhang, S., Duan, J., Li, Z., Kong, Y., Sang, Y., Liu, H., Bu, W., & Li, L. (2019). Self-assembled copper amino acid
nanoparticles for in situ glutathione ANDH
2
O
2
sequentially triggered chemodynamic therapy. Journal of the American Chemical Soci-
ety,141, 849857.
Meng, X., Zhang, K., Yang, F., Dai, W., Lu, H., Dong, H., & Zhang, X. (2020). Biodegradable metal-organic frameworks power DNAzyme for
in vivo temporal-spatial control fluorescence imaging of aberrant microRNA and hypoxic tumor. Analytical Chemistry,92, 83338339.
Meng, Z. Q., Chao, Y., Zhou, X. F., Liang, C., Liu, J. J., Zhang, R., Cheng, L., Yang, K., Pan, W., Zhu, M. F., & Liu, Z. (2018). Near-infrared-
triggered in situ gelation system for repeatedly enhanced photothermal brachytherapy with a single dose. ACS Nano,12, 94129422.
Millrud, C. R., Mehmeti, M., & Leandersson, K. (2018). Docetaxel promotes the generation of anti-tumorigenic human macrophages. Experi-
mental Cell Research,362, 525531.
Mirza, Z., & Karim, S. (2021). Nanoparticles-based drug delivery and gene therapy for breast cancer: Recent advancements and future chal-
lenges. Seminars in Cancer Biology,69, 226237.
Ni, K., Aung, T., Li, S., Fatuzzo, N., Liang, X., & Lin, W. (2019). Nanoscale metal-organic framework mediates radical therapy to enhance
cancer immunotherapy. Chem,5, 18921913.
Nikam, A. N., Pandey, A., Fernandes, G., Kulkarni, S., Mutalik, S. P., Padya, B. S., George, S. D., & Mutalik, S. (2020). Copper sulphide based
heterogeneous nanoplatforms for multimodal therapy and imaging of cancer: Recent advances and toxicological perspectives. Coordina-
tion Chemistry Reviews,419, 213356.
Ouyang, J., Tang, Z., Farokhzad, N., Kong, N., Kim, N. Y., Feng, C., Blake, S., Xiao, Y., Liu, C., Xie, T., & Tao, W. (2020). Ultrasound medi-
ated therapy: Recent progress and challenges in nanoscience. Nano Today,35, 100949.
Ouyang, Z. J., Li, D., Xiong, Z. J., Song, C., Gao, Y., Liu, R. N., Shen, M. W., & Shi, X. Y. (2021). Antifouling dendrimer-entrapped copper sul-
fide nanoparticles enable photoacoustic imaging-guided targeted combination therapy of tumors and tumor metastasis. ACS Applied
Materials & Interfaces,13, 60696080.
Pan, Q., Peng, X., Cun, J.-E., Li, J., Pu, Y., & He, B. (2021). In-situ drug generation and controllable loading: Rational design of copper-based
nanosystems for chemo-photothermal cancer therapy. Chemical Engineering Journal,409, 128222.
Pardoll, D. M. (2012). The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer,12, 252264.
Poudel, K., Gautam, M., Jin, S. G., Choi, H.-G., Yong, C. S., & Kim, J. O. (2019). Copper sulfide: An emerging adaptable nanoplatform in can-
cer theranostics. International Journal of Pharmaceutics,562, 135150.
Qi, C., Jiang, C., Fu, L.-H., Sun, T., Wang, T., Lin, J., Nie, Z., & Huang, P. (2020). Melanin-instructed biomimetic synthesis of copper sulfide
for cancer phototheranostics. Chemical Engineering Journal,388, 124232.
Ren, X., Huang, X., Wu, Q., Tan, L., Fu, C., Chen, Y., & Meng, X. (2021). Nanoscale metal organic frameworks inhibition of pyruvate kinase
of M2. Chinese Chemical Letters,32, 30873089.
Revankar, C. M., Cimino, D. F., Sklar, L. A., Arterburn, J. B., & Prossnitz, E. R. (2005). A transmembrane intracellular estrogen receptor
mediates rapid cell signaling. Science,307, 16251630.
Ruan, J., Liu, H., Chen, B., Wang, F., Wang, W., Zha, Z., Qian, H., Miao, Z., Sun, J., & Tian, T. (2021). Interfacially engineered
Zn
x
Mn
1x
S@polydopamine hollow nanospheres for glutathione depleting photothermally enhanced chemodynamic therapy. ACS Nano,
15, 1142811440.
Ruan, J., & Qian, H. (2021). Recent development on controlled synthesis of Mn-based nanostructures for bioimaging and cancer therapy.
Advanced Therapeutics,4, 2100018.
Ruttala, H. B., Ramasamy, T., Ruttala, R. R. T., Tuan Hiep, T., Jeong, J.-H., Choi, H.-G., Ku, S. K., Yong, C. S., & Kim, J. O. (2021). Mitochon-
dria-targeting multi-metallic ZnCuO nanoparticles and IR780 for efficient photodynamic and photothermal cancer treatments. Journal
of Materials Science & Technology,86, 139150.
Shi, H., Sun, Y., Yan, R., Liu, S., Zhu, L., Liu, S., Feng, Y., Wang, P., He, J., Zhou, Z., & Ye, D. (2019). Magnetic semiconductor Gd-doping
CuS nanoparticles as activatable nanoprobes for bimodal imaging and targeted photothermal therapy of gastric tumors. Nano Letters,19,
937947.
Shi, H., Yan, R., Wu, L., Sun, Y., Liu, S., Zhou, Z., He, J., & Ye, D. (2018). Tumor-targeting CuS nanoparticles for multimodal imaging and
guided photothermal therapy of lymph node metastasis. Acta Biomaterialia,72, 256265.
Shrestha, S., Wu, J., Sah, B., Vanasse, A., Cooper, L. N., Ma, L., Li, G., Zheng, H. B., Chen, W., & Antosh, M. P. (2019). X-ray induced photo-
dynamic therapy with copper-cysteamine nanoparticles in mice tumors. Proceedings of the National Academy of Sciences of the
United States of America,116, 1682316828.
Siddique, S., & Chow, J. C. L. (2020). Application of nanomaterials in biomedical imaging and cancer therapy. Nanomaterials,10, 1700.
Song, C., Ouyang, Z., Gao, Y., Guo, H., Wang, S., Wang, D., Xia, J., Shen, M., & Shi, X. (2021). Modular design of multifunctional core-
shell tecto dendrimers complexed with copper(II) for MR imaging-guided chemodynamic therapy of orthotopic glioma. Nano Today,
41, 101325.
Sun, C., Lee, J. S. H., & Zhang, M. (2008). Magnetic nanoparticles in MR imaging and drug delivery. Advanced Drug Delivery Reviews,60,
12521265.
Sun, H., Zhang, Q., Li, J., Peng, S., Wang, X., & Cai, R. (2021). Near-infrared photoactivated nanomedicines for photothermal synergistic can-
cer therapy. Nano Today,37, 101073.
Sun, Q., Wang, Z., Liu, B., Jia, T., Wang, C., Yang, D., He, F., Gai, S., & Yang, P. (2020). Self-generation of oxygen and simultaneously
enhancing photodynamic therapy and MRI effect: An intelligent nanoplatform to conquer tumor hypoxia for enhanced phototherapy.
Chemical Engineering Journal,390, 124624.
40 of 43 ZHONG ET AL.
Sun, Q. Q., Bin, L., Wang, Z., Feng, L. L., Zhao, R. X., Dong, S. M., Dong, Y. S., Zhong, L., Gai, S. L., & Yang, P. (2021). H2O2/O-2 self-
supplementing and GSH-depleting Ca2+nanogenerator with hyperthermia-triggered, TME-responsive capacities for combination cancer
therapy. Chemical Engineering Journal,425, 16.
Sun, Y., An, C., Wu, L., Zeng, W., Wang, J., Wang, Y., He, J., Gao, G., & Ye, D. (2021). Degradable FeCuS-lipid nanoparticles confer
ultrasound-activated CO release and O-2-independent radical production for synergistic therapy. ACS Nano,15, 1629816313.
Sun, Y., Liang, Y., Dai, W., He, B., Zhang, H., Wang, X., Wang, J., Huang, S., & Zhang, Q. (2019). Peptide-drug conjugate-based
nanocombination actualizes breast cancer treatment by maytansinoid and photothermia with the assistance of fluorescent and photo-
acoustic images. Nano Letters,19, 32293237.
Szabo, C. (2016). Gasotransmitters in cancer: From pathophysiology to experimental therapy. Nature Reviews Drug Discovery,15, 185203.
Tan, C., Zhou, Z., Wang, Y., Peng, F., Meng, F., Zha, J., Ma, L., Du, Y., Peng, N., & Ma, L. (2022). Intercalation-activated layered MoO
3
nanobelts as biodegradable nanozymes for tumor-specific photo-enhanced catalytic therapy. Angewandte Chemie International Edition,
61, 202115939.
Tang, Z., Liu, Y., He, M., & Bu, W. (2019). Chemodynamic therapy: Tumour microenvironment-mediated Fenton and Fenton-like reactions.
Angewandte Chemie-International Edition,58, 946956.
Tao, C., An, L., Lin, J., Tian, Q., & Yang, S. (2019). Surface plasmon resonance-enhanced photoacoustic imaging and photothermal therapy
of endogenous H
2
S-triggered Au@Cu
2
O. Small,15, 1903473.
Van-Nghia, N., Yan, Y., Zhao, J., & Yoon, J. (2021). Heavy-atom-free photosensitizers: From molecular design to applications in the photody-
namic therapy of cancer. Accounts of Chemical Research,54, 207220.
Veeranarayanan, S., Mohamed, M. S., Poulose, A. C., Rinya, M., Sakamoto, Y., Maekawa, T., & Kumar, D. S. (2018). Photodynamic therapy
at ultra-low NIR laser power and X-ray imaging using Cu3BiS3 nanocrystals. Theranostics,8, 52315245.
Wang, H., Guo, J., Lin, W., Fu, Z., Ji, X., Yu, B., Lu, M., Cui, W., Deng, L., Engle, J. W., Wu, Z., Cai, W., & Ni, D. (2022). Open-shell nano-
sensitizers for glutathione responsive cancer sonodynamic therapy. Advanced Materials, e2110283.
Wang, H., & Mooney, D. J. (2018). Biomaterial-assisted targeted modulation of immune cells in cancer treatment. Nature Materials,17,
761772.
Wang, H., Xu, Z., Li, Q., & Wu, J. (2021). Application of metal-based biomaterials in wound repair. Engineered Regeneration,2, 137153.
Wang, J., Xu, M., Wang, D., Li, Z., Primo, F. L., Tedesco, A. C., & Bi, H. (2019). Copper-doped carbon dots for optical bioimaging and photo-
dynamic therapy. Inorganic Chemistry,58, 1339413402.
Wang, M., Chang, M., Li, C., Chen, Q., Hou, Z., Xing, B., & Lin, J. (2021). Tumor-microenvironment-activated reactive oxygen species ampli-
fier for enzymatic cascade cancer starvation/chemodynamic/immunotherapy. Advanced Materials,34, 2106010.
Wang, P., Wang, X., Ma, L., Sahi, S., Li, L., Wang, X., Wang, Q., Chen, Y., Chen, W., & Liu, Q. (2018). Nanosonosensitization by using
copper-cysteamine nanoparticles augmented sonodynamic cancer treatment. Particle & Particle Systems Characterization,35, 1700378.
Wang, Q., Ji, Y., Shi, J., & Wang, L. (2020). NIR-driven water splitting H-2 production nanoplatform for H-2-mediated cascade-amplifying
synergetic cancer therapy. ACS Applied Materials & Interfaces,12, 2367723688.
Wang, R., He, Z., Cai, P., Zhao, Y., Gao, L., Yang, W., Zhao, Y., Gao, X., & Gao, F. (2019). Surface-functionalized modified copper sulfide
nanoparticles enhance checkpoint blockade tumor immunotherapy by photothermal therapy and antigen capturing. ACS Applied Mate-
rials & Interfaces,11, 1396413972.
Wang, S., Zhang, L., Zhao, J., He, M., Huang, Y., & Zhao, S. (2021). A tumor microenvironment-induced absorption red-shifted polymer
nanoparticle for simultaneously activated photoacoustic imaging and photothermal therapy. Science Advances,7, eabe3588.
Wang, X., Ma, Y., Chen, H., Wu, X., Qian, H., Yang, X., & Zha, Z. (2017). Novel doxorubicin loaded PEGylated cuprous telluride nanocrystals
for combined photothermal-chemo cancer treatment. Colloids and Surfaces B: Biointerfaces,152, 449458.
Wang, X., Miao, Z., Ma, Y., Chen, H., Qian, H., & Zha, Z. (2017). One-pot solution synthesis of shape-controlled copper selenide
nanostructures and their potential applications in photocatalysis and photothermal therapy. Nanoscale,9, 1451214519.
Wang, X., Shi, Q., Zha, Z., Zhu, D., Zheng, L., Shi, L., Wei, X., Lian, L., Wu, K., & Cheng, L. (2021). Copper single-atom catalysts with
photothermal performance and enhanced nanozyme activity for bacteria-infected wound therapy. Bioactive Materials,6, 43894401.
Wang, X., Zhong, X., Li, J., Liu, Z., & Cheng, L. (2021). Inorganic nanomaterials with rapid clearance for biomedical applications. Chemical
Society Reviews,50, 86698742.
Wang, X., Zhong, X., Liu, Z., & Cheng, L. (2020). Recent progress of chemodynamic therapy-induced combination cancer therapy. Nano
Today,35, 100946.
Wang, X. W., Zhong, X. Y., Lei, H. L., Geng, Y. H., Zhao, Q., Gong, F., Yang, Z. J., Dong, Z. L., Liu, Z., & Cheng, L. (2019). Hollow Cu
2
Se
nanozymes for tumor photothermal-catalytic therapy. Chemistry of Materials,31, 61746186.
Wang, Y., An, L., Lin, J., Tian, Q., & Yang, S. (2020). A hollow Cu9S8 theranostic nanoplatform based on a combination of increased active
sites and photothermal performance in enhanced chemodynamic therapy. Chemical Engineering Journal,385, 123925.
Wang, Z., Ma, Y., Wang, H., Wang, F., Xu, T., & Gu, Y. (2019). CXCR4-enriched nano-trap targeting CXCL12 in lung for early prevention
and enhanced photodynamic therapy of breast cancer metastasis. Advanced Functional Materials,29, 1905480.
Wang, Z., Yu, N., Li, X., Yu, W., Han, S., Ren, X., Yin, S., Li, M., & Chen, Z. (2020). Galvanic exchange-induced growth of au nanocrystals on
CuS nanoplates for imaging guided photothermal ablation of tumors. Chemical Engineering Journal,381, 122613.
Weng, Y., Guan, S., Wang, L., Lu, H., Meng, X., Waterhouse, G. I. N., & Zhou, S. (2020). Defective porous carbon polyhedra decorated with
copper nanoparticles for enhanced NIR-driven photothermal cancer therapy. Small,16, 1905184.
ZHONG ET AL.41 of 43
Wilhelm, S., Tavares, A. J., Dai, Q., Ohta, S., Audet, J., Dvorak, H. F., & Chan, W. C. W. (2016). Analysis of nanoparticle delivery to tumours.
Nature Reviews Materials,1, 16014.
Wu, K., Zhu, D., Dai, X., Wang, W., Zhong, X., Fang, Z., Peng, C., Wei, X., Qian, H., & Chen, X. (2022). Bimetallic oxide Cu
1.5
Mn
1.5
O
4
cage-
like frame nanospheres with triple enzyme-like activities for bacterial-infected wound therapy. Nano Today,43, 101380.
Wu, W., Yu, L., Pu, Y., Yao, H., Chen, Y., & Shi, J. (2020). Copper-enriched prussian blue nanomedicine for in situ disulfiram toxification
and photothermal antitumor amplification. Advanced Materials,32, 2000542.
Wu, Y., Yuan, M., Song, J., Chen, X., & Yang, H. (2019). Hydrogen gas from inflammation treatment to cancer therapy. ACS Nano,13,85058511.
Xie, Z., Fan, T., An, J., Choi, W., Duo, Y., Ge, Y., Zhang, B., Nie, G., Xie, N., Zheng, T., Chen, Y., Zhang, H., & Kim, J. S. (2020). Emerging
combination strategies with phototherapy in cancer nanomedicine. Chemical Society Reviews,49, 80658087.
Xu, C., & Pu, K. (2021). Second near-infrared photothermal materials for combinational nanotheranostics. Chemical Society Reviews,50,
11111137.
Yan, J., Chen, J., Zhang, N., Yang, Y., Zhu, W., Li, L., & He, B. (2020). Mitochondria-targeted tetrahedral DNA nanostructures for doxorubi-
cin delivery and enhancement of apoptosis. Journal of Materials Chemistry B,8, 492503.
Yan, J., Zhan, X., Zhang, Z., Chen, K., Wang, M., Sun, Y., He, B., & Liang, Y. (2021). Tetrahedral DNA nanostructures for effective treatment
of cancer: Advances and prospects. Journal of Nanobiotechnology,19,116.
Yang, N., Gong, F., Zhou, Y., Hao, Y., Dong, Z., Lei, H., Zhong, L., Yang, X., Wang, X., & Zhao, Y. (2021). A general in-situ reduction method
to prepare core-shell liquid-metal/metal nanoparticles for photothermally enhanced catalytic cancer therapy. Biomaterials,277, 121125.
Yang, N., Li, W., Gong, F., Cheng, L., Dong, Z., Bai, S., Xiao, Z., Ni, C., & Liu, Z. (2020). Injectable nonmagnetic liquid metal for Eddy-
thermal ablation of tumors under alternating magnetic field. Small Methods,4, 2000147.
Yang, P., Tao, J., Chen, F., Chen, Y., He, J., Shen, K., Zhao, P., & Li, Y. (2021). Multienzyme-mimic ultrafine alloyed nanoparticles in metal
organic frameworks for enhanced chemodynamic therapy. Small,17, 2005865.
Yang, Y., Wang, X., Qian, H., & Cheng, L. (2021). Titanium-based sonosensitizers for sonodynamic cancer therapy. Applied Materials Today,
25, 101215.
Yi, X., Shen, M., Liu, X., Gu, J., Jiang, Z., Xu, L., & Yang, K. (2021). Diagnostic radionuclides labeled on biomimetic nanoparticles for
enhanced follow-up photothermal therapy of cancer. Advanced Healthcare Materials,10, 2100860.
Yin, C., Tang, Y., Li, X., Yang, Z., Li, J., Li, X., Huang, W., & Fan, Q. (2018). A single composition architecture-based nanoprobe for rati-
ometric photoacoustic imaging of glutathione (GSH) in living mice. Small,14, 1703400.
Yong, T. Y., Wang, D. D., Li, X., Yan, Y. C., Hu, J., Gan, L., & Yang, X. L. (2020). Extracellular vesicles for tumor targeting delivery based on
five features principle. Journal of Controlled Release,322, 555565.
You, Y., Cao, F., Zhao, Y., Deng, Q., Sang, Y., Li, Y., Dong, K., Ren, J., & Qu, X. (2020). Near-infrared light dual-promoted heterogeneous
copper nanocatalyst for highly efficient bioorthogonal chemistry in vivo. ACS Nano,14, 41784187.
Yuan, H., Zhao, Y., Yang, C., Zhang, C., Yang, Y., Meng, H., Huan, S., Song, G., & Zhang, X. (2020). Copper-thioguanine metallodrug with
self-reinforcing circular catalysis for activatable MRI imaging and amplifying specificity of cancer therapy. Science China-Chemistry,63,
924935.
Yun, B., Zhu, H., Yuan, J., Sun, Q., & Li, Z. (2020). Synthesis, modification and bioapplications of nanoscale copper chalcogenides. Journal of
Materials Chemistry B,8, 47784812.
Zhang, C., Sun, W., Wang, Y., Xu, F., Qu, J., Xia, J., Shen, M., & Shi, X. (2020). Gd-/CuS-loaded functional nanogels for MR/PA imaging-
guided tumor-targeted photothermal therapy. ACS Applied Materials & Interfaces,12, 91079117.
Zhang, G., Xie, W., Xu, Z., Si, Y., Li, Q., Qi, X., Gan, Y., Wu, Z., & Tian, G. (2021). CuO dot-decorated cu@Gd2O3 core-shell hierarchical
structure for cu(i) self-supplying chemodynamic therapy in combination with MRI-guided photothermal synergistic therapy. Materials
Horizons,8, 10171028.
Zhang, H., Zeng, X., & Li, Z. (2020). Copper-chalcogenide-based multimodal nanotheranostics. ACS Applied Bio Materials,3, 65296537.
Zhang, M., Song, R. X., Liu, Y. Y., Yi, Z. G., Meng, X. F., Zhang, J. W., Tang, Z. M., Yao, Z. W., Liu, Y., Liu, X. G., & Bu, W. B. (2019). Cal-
cium-overload-mediated tumor therapy by calcium peroxide nanoparticles. Chem,5, 21712182.
Zhang, S., Li, G., Deng, D., Dai, Y., Liu, Z., & Wu, S. (2021). Fluorinated chitosan mediated synthesis of copper selenide nanoparticles with
enhanced penetration for second near-infrared photothermal therapy of bladder cancer. Advanced Therapeutics,4, 2100043.
Zhang, S., Sun, C., Zeng, J., Sun, Q., Wang, G., Wang, Y., Wu, Y., Dou, S., Gao, M., & Li, Z. (2016). Ambient aqueous synthesis of ultrasmall
PEGylated Cu
2x
Se nanoparticles as a multifunctional theranostic agent for multimodal imaging guided photothermal therapy of cancer.
Advanced Materials,28, 89278936.
Zhang, W., Lu, J., Gao, X., Li, P., Zhang, W., Ma, Y., Wang, H., & Tang, B. (2018). Enhanced photodynamic therapy by reduced levels of
intracellular glutathione obtained by employing a nano-MOF with Cu-II as the active center. Angewandte Chemie-International Edition,
57, 48914896.
Zhao, X., Liu, J., Fan, J., Chao, H., & Peng, X. (2021). Recent progress in photosensitizers for overcoming the challenges of photodynamic
therapy: From molecular design to application. Chemical Society Reviews,50, 41854219.
Zhao, Y., Song, M., Yang, X., Yang, J., Du, C., Wang, G., Yi, J., Shan, G., Li, D., Liu, L., Yan, D., Li, Y., & Liu, X. (2020). Amorphous
Ag2-xCuxS quantum dots: all-in-onetheranostic nanomedicines for near-infrared fluorescence/photoacoustics dual-modal-imaging-
guided photothermal therapy. Chemical Engineering Journal,399, 125777.
Zheng, Z., Yu, P., Cao, H., Cheng, M., Zhou, T., Lee, L. E., Ulstrup, J., Zhang, J., Engelbrekt, C., & Ma, L. (2021). Starch capped atomically
thin CuS nanocrystals for efficient photothermal therapy. Small,17, 2170249.
42 of 43 ZHONG ET AL.
Zhong, X., Wang, X., Cheng, L., Tang, Y. a., Zhan, G., Gong, F., Zhang, R., Hu, J., Liu, Z., and Yang, X. (2020). GSH-depleted PtCu3 nan-
ocages for chemodynamic-enhanced sonodynamic cancer therapy. Advanced Functional Materials,30, 1907954.
Zhong, X., Wang, X., Li, J., Hu, J., Cheng, L., & Yang, X. (2021). ROS-based dynamic therapy synergy with modulating tumor cell-
microenvironment mediated by inorganic nanomedicine. Coordination Chemistry Reviews,437, 213828.
Zhong, X., Wang, X., Zhan, G., Tang, Y., Yao, Y., Dong, Z., Hou, L., Zhao, H., Zeng, S., Hu, J., Cheng, L., & Yang, X. (2019). NaCeF4:Gd,Tb
scintillator as an X-ray responsive photosensitizer for multimodal imaging-guided synchronous radio/radiodynamic therapy. Nano Let-
ters,19, 82348244.
Zhou, H., Li, X., Niu, D., Li, Y., Liu, X., Li, C., Si, W., Cao, J., Song, Y., Wen, G., Niu, Z., & Zhang, L. (2021). Ultrasensitive chemodynamic
therapy: Bimetallic peroxide triggers high pH-activated, synergistic effect/H
2
O
2
self-supply-mediated cascade Fenton chemistry.
Advanced Healthcare Materials,10, 2002126.
Zhou, M., Tian, M., & Li, C. (2016). Copper-based nanomaterials for cancer imaging and therapy. Bioconjugate Chemistry,27, 11881199.
Zhou, Z., Li, B., Shen, C., Wu, D., Fan, H., Zhao, J., Li, H., Zeng, Z., Luo, Z., & Ma, L. (2020). Metallic 1T phase enabling MoS
2
nanodots as
an efficient agent for photoacoustic imaging guided photothermal therapy in the near-infrared-ii window. Small,16, 2004173.
Zhou, Z., Wang, X., Zhang, H., Huang, H., Sun, L., Ma, L., Du, Y., Pei, C., Zhang, Q., & Li, H. (2021). Activating layered metal oxide
nanomaterials via structural engineering as biodegradable nanoagents for photothermal cancer therapy. Small,17, 2007486.
Zhuang, J., Tian, Q., Zhou, H., Liu, Q., Liu, P., & Zhong, H. (2012). Hierarchical porous TiO
2
@C hollow microspheres: One-pot synthesis
and enhanced visible-light photocatalysis. Journal of Materials Chemistry,22, 70367042.
Zhuang, Y., Han, S., Fang, Y., Huang, H., & Wu, J. (2022). Multidimensional transitional metal-actuated nanoplatforms for cancer che-
modynamic modulation. Coordination Chemistry Reviews,455, 214360.
AUTHOR BIOGRAPHIES
Xiaoyan Zhong, PhD, received her PhD degree from Huazhong University of Science and Technology
(HUST) in 2020, and then joined the Department of Toxicology in School of Public Health, Suzhou
Medical College of Soochow University as a Postdoctor. Her research interest is the development and
biosafety assessments of multifunctional inorganic nanomaterials for applications in reactive oxygen
species-based cancer theranostics.
Xianwen Wang, PhD, obtained his PhD degree under the guidance of Prof. Liang Cheng and
Prof. Zhuang Liu in the Institute of Functional Nano & Soft Materials (FUNSOM) at Soochow
University in 2021. Then he joined Anhui Medical University as a specially-appointed professor
and doctoral supervisor. His current research interest is the development of multifunctional
nanomaterials for biomedical application.
How to cite this article: Zhong, X., Dai, X., Wang, Y., Wang, H., Qian, H., & Wang, X. (2022). Copper-based
nanomaterials for cancer theranostics. WIREs Nanomedicine and Nanobiotechnology, e1797. https://doi.org/10.
1002/wnan.1797
ZHONG ET AL.43 of 43
... Yet another example of copper-based nanomaterials for theragnostic include the polyvinylpyrrolidone-modified copper sulfide nanocrystals (CuS NCs) developed by Dong et al. (89) and the poly(vinylpyrrolidone)-protected Cu 1.2 O nanoparticles produced by Gao and coworkers(41). For more examples of copper-based nanomaterials applied in theragnostic and detailed approaches and effects, the reader may check the review written by Zhong and others (84). Figure 5 summarizes the main therapies and diagnostic approaches involving copper-based nanomaterials. ...
Chapter
Copper-based nanomaterials have emerged as a new platform in various scientific domains due to their unique physicochemical, biological, and compatibility properties. This review explores the multifaceted applications of copper nanomaterials in the areas of biosensing, drug delivery, cancer therapy, and environmental remediation. The innovative role of copper nanomaterials in biosensors and their exceptional sensitivity and specificity in detecting biomolecules are discussed. Moreover, the potential applications of copper-based nanomaterials in drug delivery systems, highlighting the controlled and targeted release of therapeutic agents’ approaches are explored. In the domain of cancer theragnostic, copper nanomaterials exhibit promising results thus offering practical tools for early diagnosis and advanced treatment modalities. Furthermore, the antimicrobial properties of copper nanoparticles are explored, emphasizing their importance in combating microbial threats and promoting a new era in antimicrobial research. Copper-based nanomaterials could have a significant role in environmental and bioremediation applications. The strong catalytic activity of copper nanoparticles makes them ideal candidates for environmental remediation. In summary, this comprehensive review consolidates the latest advances in copper nanomaterials research, providing valuable information for scientists and researchers involved in diverse fields to promote the development of new applications and technologies.
... Upconversion luminescent materials convert lowenergy photons into high-energy (short wavelength) emission and have the advantages of excellent stability, high sensitivity, low toxicity, strong penetrability, and excellent biocompatibility [31,38]. As efficient photothermal transduction agents and response sensitizers, copper-based nanoparticles have displayed good therapeutic performance in phototherapy, sonodynamic therapy, radiodynamic therapy, and chemodynamic therapy, in addition to their common biosensing and imaging functions [103]. Quantum dots exhibit electroluminescence and optical properties as they release energy or light upon returning to the ground state after absorbing an energy photon and undergoing an electron jump [26,63]. ...
Article
Full-text available
Cancer remains one of the primary factors leading to premature death in humans. Conventional therapeutic approaches result in significant harm to normal tissue cells and impede the effective treatment of cancer. In recent years, the benefits of inorganic nanomaterials in cancer research have become increasingly evident, and they are expected to provide new ideas for diagnosing and treating cancer. This study aims to reveal the technological trajectory and important applications of inorganic nanomaterials in cancer diagnosis and treatment better to understand their technological development trends and technical priorities. A patent citation network was constructed to find the milestone patents of inorganic nanomaterials in the field of cancer by main path–derived path analysis, and a citation analysis method based on the PageRank (PR) algorithm was used to identify the high PR value patents of inorganic nanotechnology in cancer. The study found that the overall technological development trajectory of inorganic nanomaterials in cancer is linear, with a focus on enhancing technical aspects such as biological detection and imaging, drug delivery, phototherapy, magnetic hyperthermia, and radiotherapy, progressively enhancing the diagnostic and therapeutic effects, and realizing the integration of diagnosis and treatment. Gold nanomaterials are essential in cancer diagnosis and treatment. Carbon-based nanomaterials, magnetic nanomaterials, upconversion luminescent materials, copper-based nanoparticles, quantum dots, and other inorganic nanomaterials will become significant R&D directions.
Article
Full-text available
Hematological malignancies (HMs) encompass a diverse group of blood neoplasms with significant morbidity and mortality. Immunotherapy has emerged as a validated and crucial treatment modality for patients with HMs. Despite notable advancements having been made in understanding and implementing immunotherapy for HMs over the past decade, several challenges persist. These challenges include immune-related adverse effects, the precise biodistribution and elimination of therapeutic antigens in vivo, immune tolerance of tumors, and immune evasion by tumor cells within the tumor microenvironment (TME). Nanotechnology, with its capacity to manipulate material properties at the nanometer scale, has the potential to tackle these obstacles and revolutionize treatment outcomes by improving various aspects such as drug targeting and stability. The convergence of nanotechnology and immunotherapy has given rise to nano-immunotherapy, a specialized branch of anti-tumor therapy. Nanotechnology has found applications in chimeric antigen receptor T cell (CAR-T) therapy, cancer vaccines, immune checkpoint inhibitors, and other immunotherapeutic strategies for HMs. In this review, we delineate recent developments and discuss current challenges in the field of nano-immunotherapy for HMs, offering novel insights into the potential of nanotechnology-based therapeutic approaches for these diseases.
Article
Full-text available
In the realm of cancer treatment, traditional modalities like radiotherapy and chemotherapy have achieved certain advancements but continue to grapple with challenges including harm to healthy tissues, resistance to treatment, and adverse drug reactions. The swift progress in nanotechnology recently has opened avenues for investigating innovative approaches to cancer therapy. Especially, chemodynamic therapy (CDT) utilizing metal nanomaterials stands out as an effective cancer treatment choice owing to its minimal side effects and independence from external energy sources. Transition metals like manganese are capable of exerting anti-tumor effects through a Fenton-like mechanism, with their distinctive magnetic properties playing a crucial role as contrast agents in tumor diagnosis and treatment. Against this backdrop, this review emphasizes the recent five-year advancements in the application of manganese (Mn) metal ions within nanomaterials, particularly highlighting their unique capabilities in catalyzing CDT and enhancing MRI imaging. Initially, we delineate the biomedical properties of manganese, followed by an integrated discussion on the utilization of manganese-based nanomaterials in CDT alongside multimodal therapies, and delve into the application and future outlook of manganese-based nanomaterial-mediated MRI imaging techniques in cancer therapy. By this means, the objective is to furnish novel viewpoints and possibilities for the research and development in future cancer therapies.
Article
Full-text available
Deleterious effects to normal tissues and short biological half‐life of sonosensitizers limit the applications of sonodynamic therapy (SDT). Herein we synthesize a new sonosensitizer (Cu(II)NS) that consists of porphyrins, the chelated Cu2+, and PEG to overcome the challenges of SDT. As Cu2+ contains 27 electrons, Cu(II)NS have an unpaired electron (open shell), resulting in a doublet ground state and little sonosensitivity. Overexpressed glutathione (GSH) in the tumor can reduce Cu2+ to generate Cu(I)NS, leading to a singlet ground state and recuperative sonosensitivity. Additionally, PEG endows Cu(II)NS with increased blood biological half‐life and enhanced tumor accumulation, further increasing the effect of SDT. Through regulating the valence state of Cu, we achieve cancer SDT with enhanced therapeutic index. This article is protected by copyright. All rights reserved
Article
Full-text available
The existence of natural van der Waals gaps in layered materials allows them to be easily intercalated with varying guest species, offering an appealing strategy to optimize their physicochemical properties and application performance. Herein, we report the activation of layered MoO3 nanobelts via aqueous intercalation as an efficient biodegradable nanozyme for tumor‐specific photo‐enhanced catalytic therapy. The long MoO3 nanobelts are grinded and then intercalated with Na⁺ and H2O to obtain the short Na⁺/H2O co‐intercalated MoO3−x (NH−MoO3−x) nanobelts. In contrast to the inert MoO3 nanobelts, the NH−MoO3−x nanobelts exhibit excellent enzyme‐mimicking catalytic activity for generation of reactive oxygen species, which can be further enhanced by the photothermal effect under a 1064 nm laser irradiation. Thus, after bovine serum albumin modification, the NH−MoO3−x nanobelts can efficiently kill cancer cells in vitro and eliminate tumors in vivo facilitating with 1064 nm laser irradiation.
Article
Full-text available
[email protected]2 core-shell nanoparticles that AgBiS2 coated on the surface of upconversion nanoparticles (UCNPs) was successfully prepared through an ion exchange reaction. The photothermal conversion efficiency of AgBiS2 can be improved from 14.7% to 45% due to the cross relaxation between Nd ions and AgBiS2. The doping concentration of Nd ions played a critical role in the production of reactive oxygen species (ROS) and enhanced the photothermal conversion efficiency. The NaYF4:Yb/Er/[email protected]4:Nd nanoparticles endows strong upconversion emissions when the doped concentration of Nd ions is 1% in the inner core, which excites the AgBiS2 shell to produce ROS for photodynamic therapy (PDT) of cancer cells. As a result, the as-prepared NaYF4:Yb/Er/[email protected]4:[email protected]2 core-shell nanoparticles showed combined photothermal/photodynamic therapy (PTT/PDT) against malignant tumors. This work provides an alternative near-infrared light-active multimodal nanostructures for applications such as fighting against cancers.
Article
Full-text available
Recently, DNA nanostructures with vast application potential in the field of biomedicine, especially in drug delivery. Among these, tetrahedral DNA nanostructures (TDN) have attracted interest worldwide due to their high stability, excellent biocompatibility, and simplicity of modification. TDN could be synthesized easily and reproducibly to serve as carriers for, chemotherapeutic drugs, nucleic acid drugs and imaging probes. Therefore, their applications include, but are not restricted to, drug delivery, molecular diagnostics, and biological imaging. In this review, we summarize the methods of functional modification and application of TDN in cancer treatment. Also, we discuss the pressing questions that should be targeted to increase the applicability of TDN in the future. Graphical Abstract
Article
Full-text available
Starch Capped CuS Nanocrystals In article number 2103461, Christian Engelbrekt, Lixin Ma, and co-workers report a one-pot synthesis of starch-capped ultrathin CuS nanocrystals with efficient cancer photothermal therapy capacity. The starch layer endows CuS nanocrystals with enhanced photothermal efficacy by increasing the free carrier density and effectively assigning the localized surface plasmon resonance at specific optical wavelength.
Article
Specific cytotoxicity for catalytic nanomedicine triggered by the tumor microenvironment (TME) has attracted increasing interest. In this work, we prepared AgBiS 2 hollow nanospheres with narrow bandgaps via rapid precipitation in a weakly polar solvent, which lowered the intrinsic energy gap for the active production of highly reactive hydroxyl radicals (• OH), especially in the TME. The as-prepared AgBiS 2 hollow nanospheres exhibited enhanced optical absorption and high photothermal conversion efficiency (44.2%). In addition, the hollow structured AgBiS 2 nanospheres were found to have a peroxidase-mimicking feature to induce cancer cell-specific cytotox-icity while exhibiting negligible cytotoxicity toward normal cells, which might be attributed to the efficient production of highly reactive • OH originating from the overexpression H 2 O 2 in the TME caused by surface catalysis. In particular, the cancer cell-specific cytotoxicity of the nanospheres was greatly enhanced both in vitro and in vivo upon irradiation with a near-infrared (NIR) laser (808 nm). The above-mentioned features of the hollow structured AgBiS 2 will make it a promising candidate for tumor therapy.
Article
The existence of natural van der Waals gaps in layered materials allows them to be easily intercalated with varying guest species, offering an appealing strategy to optimize their physicochemical properties and application performance. Herein, we report the activation of layered MoO3 nanobelts via aqueous intercalation as an efficient biodegradable nanozyme for tumor‐specific photo‐enhanced catalytic therapy. The long MoO3 nanobelts are grinded and then intercalated with Na+ and H2O to obtain the short Na+/H2O co‐intercalated MoO3 nanobelts (NH‐MoO3). In contrast to the inert MoO3 nanobelts, the NH‐MoO3 nanobelts exhibit excellent enzyme‐mimicking catalytic activity for generation of reactive oxygen species, which can be further enhanced by the photothermal effect under a 1064 nm laser irradiation. Thus, after bovine serum albumin modification, the NH‐MoO3 nanobelts can efficiently kill cancer cells in vitro and eliminate tumors in vivo facilitating with 1064 nm laser irradiation.
Article
Copper-based nanomaterials with intrinsic enzyme-like activity have shown increasing potential as new broad-spectrum antibiotics. However, due to its low catalytic activity, poor glutathione (GSH) depletion capacity, and complex material design, their feasibility is still far from satisfactory. Herein, bimetallic oxide Cu1.5Mn1.5O4 cage-like frame nanospheres (CFNSs) were successfully prepared by a two-step approach for the first time, including gas-assisted soft template solvothermal preparation of Cu-Mn hydroxide hollow sphere (Cu-Mn-OH HSs) precursors, and then calcination to synthesize Cu1.5Mn1.5O4 CFNSs. This ingenious, simple, and rapid material synthesis strategy could obtain well-dispersed and extremely uniform Cu1.5Mn1.5O4 CFNSs with a special mesoporous cavity structure, making its performance close to the requirements of practical applications. Interestingly, the as-prepared Cu1.5Mn1.5O4 CFNSs showed enhanced triple enzyme-like activities (oxidase-, peroxidase-, and glutathione peroxidase-like), which could significantly promote the generation of reactive oxygen species (ROS) due to the increased exposure of active edge sites. Cu1.5Mn1.5O4 CFNSs could effectively kill bacteria by combining OXD-like, POD-like, and GSH-Px-like nanozyme activities. More importantly, in vivo wound healing showed that Cu1.5Mn1.5O4 CFNSs could be conveniently used for wound disinfection. In addition, Cu1.5Mn1.5O4 CFNSs exhibited excellent biosafety without observable toxicity or side effects in mice. This work emphasizes the potential application of bimetallic oxides with triple enzyme-like activities in antibacterial therapy.
Article
Cancer is one of the deadliest diseases threatening human health worldwide. As the understanding of the tumor microenvironment (TME) becomes more comprehensive, many promising TME-enabled nanotherapies have been developed, such as dynamic therapy and immunotherapy. Transitional metal elements also play an indispensable regulatory role in TME by exerting crucial physiological functions in the human body, thereby affecting the formation and progression of tumors, and exhibiting significant potential for anticancer strategies. They can spontaneously activate in situ chemical regulation at the tumor site based on the Fenton/Fenton-like reaction to interfere with the TME redox potential. Based on this, the concept of chemodynamic therapy (CDT) as one of the most popular therapeutic methods came into being. This article introduces the transitional metal-based chemodynamic adjustment mechanisms and systematically reviews the latest CDT-relevant antineoplastic nanosystems, with their applications based on different dimensions. Finally, the opportunities and challenges of transitional metal-based chemodynamic modulation strategies in future development and clinical translation are examined.
Article
Chemodynamic therapy (CDT) has recently gained much attention for Fenton chemistry-mediated cancer treatment, but the anti-tumor efficacy of CDT suffers from insufficient amount of endogenous H2O2 and inefficient decomposition of metal oxides to catalytic ions. Although tremendous progress has been made to increase the amount of H2O2 in the tumor region, the antitumor activity of CDT remains limited due to the suboptimal ionization to release enough amounts of catalytic ions for converting endogenous H2O2 to reactive oxygen species (e.g. highly toxic hydroxyl radical ·OH). Here, a series of nanoparticles with tunable acid dissociation constant (pKa) values from 5.2 to 6.2 were prepared to load H2O2 self-supplying copper peroxide, which can be used to trap copper peroxide in acidic lysosome to produce ample catalytic ions that convert self-supplied H2O2 into ·OH by a robust Fenton reaction. The highly reactive ·OH effectively permeate the lysosomal membrane through lipid peroxidation and thus kill tumor cells in a lysosome-mediated manner. Most importantly, the Fenton reaction is processed inside the lysosomal compartment, which avoids the cytoplasmic antioxidants such as glutathione (GSH) to scavenge ·OH. Overall, this work provides a new strategy to enhance CDT efficacy.