Access to this full-text is provided by American Association for the Advancement of Science.
Content available from Biomaterials Research
This content is subject to copyright. Terms and conditions apply.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 1
REVIEW ARTICLE
Copper-Based Nanomedicines for Cuproptosis-
Mediated Effective Cancer Treatment
Dahye Noh1,2, Hokyung Lee1,3, Sangmin Lee3, In-Cheol Sun1,
and Hong Yeol Yoon1,2*
1Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and
Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea. 2Division of
Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Hwarang-
ro14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea. 3Department of Fundamental Pharmaceutical
Sciences, College of Pharmacy, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 02447,
Republic of Korea.
*Address correspondence to: seerou@kist.re.kr
The recent discovery of cuproptosis, a novel copper-ion-induced cell death pathway, has suggested the
novel therapeutic potential for treating heterogeneous and drug-resistant cancers. Currently, copper
ionophore-based therapeutics have been designed to treat cancers, utilizing copper ions as a strategic
tool to impede tumor proliferation and promote cellular demise. However, limitations of copper ionophore-
based therapies include nontargeted delivery of copper ions, low tumor accumulation, and short
half-life. Strategies to enhance specificity involve targeting intracellular cuproptosis mechanisms using
nanotechnology-based drugs. Additionally, the importance of exploring combination therapies cannot
be overstated, as they are a key strategy in improving the efficacy of cancer treatments. Recent studies
have reported the anticancer effects of nanomedicines that can induce cuproptosis of cancer both
invitro and invivo. These cuproptosis-targeted nanomedicines could improve delivery efficiency with the
pharmacokinetic properties of copper ion, resulting in increasing cuproptosis-based anticancer effects.
This review will summarize the intricate nexus between copper ion and carcinogenesis, examining the
pivotal roles of copper homeostasis and its dysregulation in cancer progression and fatality. Furthermore,
we will introduce the latest advances in cuproptosis-targeted nanomedicines for cancer treatment. Finally,
the challenges in cuproptosis-based nanomedicines will be discussed for future development directions.
Introduction
Copper ion is an essential cofactor for numerous enzymes and
plays a crucial role in various metabolic processes [ 1 ]. Addi-
tionally, it acts as a vital component in cytochrome c oxidase,
also referred to as complex IV, within the mitochondrial elec-
tron transport chain (ETC), thereby regulating cell growth and
function through ecient energy conversion [ 2 ]. Inadequate
copper ion levels can impede growth, while excessive exposure
to copper ion can induce oxidative stress, leading to cell death
and tissue damage [ 3 ]. us, it has been shown that disruptions
in copper homeostasis can result in structural abnormalities
or the impairment of essential physiological functions [ 4 ].
Notably, variations in copper ion levels have been observed in
many cancer cells, associated with elevated intratumoral copper
ion concentrations and shis in systemic copper ion distribu-
tion [ 5 ]. ese alterations in copper homeostasis can adversely
aect biological function and contribute to angiogenesis, che-
moresistance, immune evasion, and metastasis, particularly in
cancers [ 6 ].
Recent ndings suggest that copper ion plays a central role
in determining the proliferation (cuproplasia) and cell death
(cuproptosis) [ 7 , 8 ]. While copper ions act as indispensable
cofactor for numerous enzymes and are pivotal in various meta-
bolic processes, disturbances in copper ion homeostasis can
have detrimental eects on biological function [ 1 , 6 ]. A sche-
matic illustration of the biological roles of copper ions is shown
in (Fig. S1). Indeed, the proliferation and metastasis of cancer
cells are widely recognized to be signicantly more dependent
on copper ion than normal tissues [ 5 ]. erefore, a novel form
of copper-ion-induced cell death, cuproptosis, has recently
been utilized for anticancer drug discovery due to the potential
for alternative pathways beyond programmed cell death mecha-
nisms [ 8 , 9 ]. Cuproptosis is triggered by excess copper ions.
High levels of copper ion within cells lead to the direct binding
of lipoylated proteins involved in glycolytic metabolism, dis-
rupting the tricarboxylic acid (TCA) cycle. is disruption
causes aggregation of lipoylated proteins, resulting in the loss
of iron-sulfur (Fe-S) clusters. Subsequently, these processes
promote proteotoxic stress, ultimately leading to cell death [ 8 ].
Given its contribution to cancer proliferation, angiogenesis,
and metastasis, copper-ion-induced cuproptosis has been con-
sidered to hold great potential for cancer treatment. Further-
more, the anticancer drug, disulram (DSF), is a clinically
Citation: NohD, LeeH, LeeS,
SunIC, YoonHY. Copper-Based
Nanomedicines for Cuproptosis-
Mediated Effective Cancer Treatment.
Biomater. Res. 2024;28:Article 0094.
https://doi.org/10.34133/bmr.0094
Submitted 11 June 2024
Revised 9 September 2024
Accepted 24 September 2024
Published 18 October 2024
Copyright © 2024 Dahye Noh etal.
Exclusive licensee Korean Society
for Biomaterials, Republic of Korea.
No claim to original U.S. Government
Works. Distributed under a Creative
Commons Attribution License 4.0
(CC BY 4.0).
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 2
approved copper ionophore, facilitating the redistribution of
copper ion stores across cellular and subcellular compartments
[ 10 – 14 ]. Over the past several years, various copper-related
signal pathways have been explored. As a result, cuproptosis
induced by copper-based therapeutics has been refocused for
cancer therapy due to its unique cell death mechanism, con-
trasting with apoptosis triggered by conventional anticancer
agents [ 15 , 16 ]. Nevertheless, cuproptosis-based cancer treat-
ment has several limitations, including the selective increase
of copper ion concentration in cancer cells, prevention of cop-
per ion damage to normal cells, and extension of cuproptosis
duration [ 17 – 19 ]. Nanomedicine provides an eective treat-
ment approach by leveraging nanoparticles to enhance drug
solubility, extends drug circulation in the bloodstream, and
regulates drug release, enhancing in vivo therapeutic ecacy
and reducing o-target eects [ 20 ]. Copper-based nanomateri-
als oer an innovative option for cupoptosis by facilitating
precise targeting and accumulation at tumor sites through the
enhanced permeability and retention eect [ 21 , 22 ]. Furthermore,
various combination approaches using cuproptosis-targeted
nanomedicines and other therapeutic options, including photo-
dynamic therapy, immunotherapy, gene therapy, and chemo-
therapy, enhance therapeutic outcomes. is review will introduce
copper metabolism with dysregulation in cancer cells and recent
advances in cuproptosis-targeted nanomedicines for eective
cancer treatment. Finally, the future perspectives of cuproptosis-
targeted nanomedicines will be discussed for research and clini-
cal applications.
Copper Homeostasis and Dysregulation in
Cancer Cells
Copper ions are one of the critical trace elements essential for
various physiological functions within the body [ 23 ]. It exists
in 2 distinct ionic forms: Cu
+
(cuprous ion, reduced form) and
Cu
2+
(cupric ion, oxidized form), both actively engaging in the
enzymatic regulation of various cellular physiological processes,
including gene expression, biological metabolic processes,
mitochondrial respiration, and antioxidation [ 24 , 25 ]. e con-
version between Cu+ and Cu2+ gives copper ion unique redox
properties, making it a powerful reactive oxygen species (ROS)
producer and important for ROS scavenging [ 26 , 27 ]. Cancer
cells oen have higher levels of ROS than normal cells due to
several factors, such as increased metabolic activity, gene muta-
tions, and relative hypoxia [ 28 – 30 ]. is imbalance between
oxidants and antioxidants can contribute to the promotion of
tumorigenesis and cancer progression [ 31 , 32 ]. Furthermore,
cancer cells oen exhibit a heightened dependency on copper
ion due to its vital roles in various cellular processes that sup-
port tumor growth and progression [ 4 ]. Elevated copper ion
levels in cancer cells can regulate angiogenesis, metastasis, and
resistance to cell death pathways [ 33 – 35 ]. Copper ion activates
enzymes and signaling pathways that promote these processes,
making it a crucial element for sustaining the malignant phe-
notype of cancer cells [ 36 ]. is section introduces copper
metabolism and homeostasis, examining how copper dysregu-
lation contributes to cancer cells.
Systemic copper metabolism and homeostasis
Copper ion is absorbed from dietary sources primarily by the
small intestine. is process involves the uptake of copper ion
by the Cu transport protein 1 (CTR1) or solute carrier family
31 member 1 (SLC31A1) across the apical membrane of entero-
cytes. Once inside the enterocyte, copper ion is transported
across the cell and exported into the interstitial uid and blood-
stream by a protein called ATPase copper-transporting alpha/
beta (ATP7A/B) located on the basolateral membrane [ 7 , 37 ].
Extracellular copper ions typically exist in the oxidized form,
Cu2+. However, Cu2+ cannot directly enter cells due to its charge
and size. To facilitate cellular uptake, Cu2+ is reduced to Cu+ by
metalloreductases, such as 6-transmembrane epithelial antigen
of the prostate or other reductases on the cell membrane [ 38 ].
e processes and key regulators of copper ion metabolism
are summarized in (Fig. S2). Upon entry into cells, copper ion
interacts with a diverse array of copper chaperone proteins, such
as antioxidant 1 copper chaperone (ATOX1), copper chaperone
for superoxide dismutase, and superoxide dismutase 1 (SOD1),
facilitating its delivery to distinct subcellular compartments,
including mitochondria, the trans-Golgi network, and the
nucleus [ 39 ]. In the mitochondria, copper ion can bind to cyto-
chrome C oxidase and contribute to the respiratory chain and
redox pathways [ 40 ]. In the nucleus, copper ion can regulate
gene expression by engaging with several transcription factors
[ 41 ]. Once copper ion enters the circulatory system, it binds to
various plasma proteins for transport to organs and tissues
throughout the body. Ceruloplasmin is one of the primary cop-
per ion-binding proteins in plasma and plays a crucial role in
copper ion transport and homeostasis. Copper ion can bind to
other plasma proteins such as albumin, trans copper protein,
and metallothionein, which help facilitate its distribution to
various target tissues and organs [ 42 – 44 ]. e absorbed copper
ion is mainly transported to the liver, where hepatocytes are the
major storehouse [ 45 ]. e liver also acts as the primary site for
copper ion removal through hepatobiliary excretion via the cop-
per ion exporter ATPase copper ion transporting beta (ATP7B)
across the bile canalicular membrane of hepatocytes [ 45 – 47 ].
Alternative pathways for copper ion elimination, such as urine,
sweat, and menses, have minimal impact on copper loss.
e regulation of copper homeostasis relies on the intricate
coordination of its intake, transport, and clearance, which are
pivotal in governing physiological processes [ 48 ]. In the cyto-
plasm, copper ion levels are regulated by various proteins, such
as cuproenzymes, copper chaperones, and membrane trans-
porters [ 5 ]. ese proteins are essential for avoiding the adverse
consequences of copper ion deciency and copper ion over-
load, working together to maintain a precise balance of copper
ion concentrations within cells [ 49 ]. ey also exhibit a high
anity for copper ion binding and facilitate practically irrevers-
ible metal transfer among cognate molecules [ 50 – 52 ].
Reduced copper ion concentrations have been associated
with conditions such as albinism, osteoporosis, and other dis-
eases [ 53 ]. Furthermore, a deciency of copper ion can aect
brain development, highlighting the vital role of copper ion
in the body [ 54 – 56 ]. Maintaining copper ion homeostasis is
essential for cellular functions and is regulated by a network
of proteins that facilitate copper ion uptake, distribution, and
elimination. is is crucial for overall physiological processes
and prevents adverse health outcomes associated with copper
ion deciency or overload.
Copper ion dysregulation in cancer
e association between copper ion and cancer has been estab-
lished over many years, with studies consistently showing
elevated levels of copper ion and ceruloplasmin in cancers or
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 3
the serum of animal models and cancer patients [ 7 , 57 – 60 ].
Copper ion is an essential cofactor for enzymes involved in vital
cellular processes such as mitochondrial respiration, antioxi-
dant defense, hormone synthesis, neurotransmitters, and pig-
ments. However, disturbances in copper ion homeostasis can
lead to oxidative stress and cytotoxicity [ 61 , 62 ]. Recent studies
have revealed elevated levels of copper ion in the serum or
tissues of patients with various malignancies, including colorec-
tal cancer, gallbladder cancer, and thyroid cancers [ 63 – 66 ].
Further more, cancers oen require elevated levels of copper
ions compared to surrounding healthy tissues to support angio-
genesis, growth, metastasis, and resistance to cell death [ 6 , 67 ].
Angiogenesis
Angiogenesis, the process by which new blood vessels sprout
from existing blood vessels, especially capillaries, is essential
for various physiological processes [ 68 ]. In normal physiology,
angiogenesis ensures proper blood supply to tissues and organs,
allowing for their growth, repair, and metabolic needs [ 69 ].
However, intussusceptive angiogenesis has been implicated in
numerous cancer types, including melanoma, colorectal, gli-
oma, and mammary cancers [ 70 – 73 ]. By stimulating the for-
mation of new blood vessels, cancers ensure a steady supply
of oxygen and nutrients, allowing them to grow and metasta-
size [ 74 , 75 ].
Copper ion is closely associated with promoting angiogen-
esis [ 76 ]. Gérard et al. [ 77 ] reported that copper ion promotes
endothelial cell proliferation and enhances angiogenesis in vitro
and in vivo. Specically, copper ion reduced the risk of ischemia
in skin aps and induced the formation of a vascularized cap-
sule around a cross-linked hyaluronic acid (HA)-composed
hydrogel [ 78 – 81 ]. It has been demonstrated that copper ion
plays critical roles in angiogenesis and metastasis by stimulating
the production of several proangiogenic factors, including inter-
leukin-1 (IL-1), IL-6, IL-8, tumor necrosis factor-alpha, angio-
genin, vascular endothelial growth factor (VEGF) [ 82 ], SOD1
[ 83 ], hypoxia-inducible factor-1-alpha (HIF-1α) [ 84 ], and bro-
nectin [ 85 , 86 ].
In other instances, Ungar-Waron et al. [ 87 ] reported on
serum ceruloplasmin levels in rabbits during tumor develop-
ment and regression. Ceruloplasmin levels, the primary copper
ion binder in serum, rose signicantly during tumor progres-
sion and returned to normal during regression but remained
elevated during metastasis. Moreover, cancer angiogenesis is
necessary for tumor growth progression, wherein copper ion
is involved in tumor neovascularization by directly interacting
with angiogenic factors (VEGF and broblast growth factor)
[ 79 , 88 ]. Upon VEGF stimulation, CTR1 undergoes rapid sulfe-
nylation at its cytoplasmic C-terminal Cys189. is event forms
a disulde bond between CTR1 and VEGFR2, leading to their
cointernalization with early endosomes, thereby sustaining
VEGFR2 signaling. Mice with endothelial cell-specic CTR1
deciency or mutations exhibit impaired developmental and
reparative angiogenesis in vivo [ 89 ]. In other words, the bind-
ing of copper ion with angiogenin [ 79 ] suggests that copper
ion-activated angiogenin may interact more eciently with
endothelial cells, potentially enhancing its capacity to promote
the formation of new blood vessels [ 80 ]. Additionally, vascular
copper ion transport systems profoundly inuence the activa-
tion and execution of angiogenesis, serving as multifunctional
regulators of distinct proangiogenic pathways [ 81 ]. e involve-
ment of copper ion may represent a critical mechanism in
regulating the formation of new blood vessel pathways, provid-
ing valuable insights for developing innovative therapies for
cancer treatment.
Drug resistance
Drug resistance represents a signicant challenge in cancer
treatment, impacting a large number of patients with metastatic
cancer [ 90 ]. It can manifest through multiple mechanisms,
including limiting the uptake of drugs, altering drug targets,
inactivating drugs, and actively pumping out drugs [ 91 ]. ese
mechanisms, among others, neutralize chemotherapy, resulting
in treatment failure and disease progression [ 92 , 93 ]. Emerging
evidence suggests that copper ion transport mechanisms could
be implicated in drug resistance [ 94 ]. Majumder et al. [ 95 ]
investigated the correlation between copper ion levels and drug
resistance to identify patients resistant to treatment, aiming to
develop improved therapeutic strategies. ey revealed that
blood serum from tumor-bearing mice displayed increased
copper ion levels compared to healthy normal mice. Moreover,
doxorubicin-resistant Ehrlich ascites carcinoma- or cyclophos-
phamide-resistant Lewis lung carcinoma-bearing mice exhib-
ited higher copper ion levels in the serum compared to those
of drug-sensitive tumor-bearing mice. Furthermore, the analy-
sis of copper ion levels in healthy volunteers and cancer patients
indicated a correlation between copper ion, tumor growth, and
drug resistance. In addition, Jin et al. [ 96 ] investigated how
elevated copper ion levels partially contribute to drug resist-
ance and the repair of damaged DNA in cancer cells. ATOX1-
induced expression of mediator of DNA damage checkpoint 1
(MDC1), a crucial protein involved in double-strand DNA
damage repair. Specically, ATOX1, acting as a copper ion
chaperone, is translocated to the nucleus to target the MDC1
promoter aer exposure to various genotoxic agents, promoting
the transcription of MDC1 in a copper ion-dependent manner.
Consequently, knockout or blockade of ATOX1 rendered can-
cers sensitive to gemcitabine in transplanted cancer mouse
models. ese observations suggest that modulating copper
ion levels may improve the eectiveness of cancer chemother-
apy in drug-resistant patients. By targeting the elevated copper
ion levels associated with drug resistance, it may be possible to
enhance the sensitivity of cancer cells to chemotherapy, thereby
improving treatment outcomes. is approach could lead to
the development of new therapeutic strategies aimed at over-
coming drug resistance in cancer patients.
Immune evasion
Programmed cell death-ligand-1 (PD-L1) expressed in cancers
is believed to suppress tumor-inltrating lymphocytes via pro-
grammed cell death-1, thereby facilitating adaptive immune
resistance [ 97 ]. Interestingly, copper ion contributes to these
immune responses [ 98 ]. Voli et al. [ 15 ] demonstrated a correla-
tion between intratumoral copper ion levels and PD-L1 expres-
sion in cancer cells. Copper ion supplementation boosted
PD-L1 expression at both mRNA and protein levels in cancer
cells. Furthermore, RNA sequencing unveiled that copper ion
regulates critical signaling pathways responsible for PD-L1-
mediated cancer immune evasion. An in-depth analysis of e
Cancer Genome Atlas database and tissue microarrays revealed
a robust association between the CTR1 and PD-L1 expression
across multiple cancers, contrasting with the lack of correlation
in corresponding normal tissues. In addition, Zhou et al. [ 99 ]
reported that DSF-Cu2+ could up-regulate PD-L1 expression
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 4
and inhibit CD8+ T cell inltration and activity by suppressing
poly (ADP-ribose) polymerase 1 activity and inducing glycogen
synthase kinase-3β inactivation via phosphorylation at the Ser9
site. In contrast, the application of copper chelators suppressed
signal transducer and activator of transcription 3 and epidermal
growth factor receptor phosphorylation and enhanced the
ubiquitin-mediated degradation of PD-L1 [ 100 ]. Additionally,
copper chelator tetraethylenepentamine increases mouse survival
by decreasing PD-L1 expression in neuroblastoma xenogras,
consequently boosting the inltration of tumor-inltrating
T cells [ 101 ]. ese ndings imply that copper ion can control
PD-L1 expression and impact cancer immune evasion. erefore,
various approaches that decrease intratumoral copper ion levels
can provide the potential to improve therapeutic ecacy in can-
cer immunotherapy.
Cancer metastasis
Cancer metastasis is a pivotal event in cancer progression, sig-
nicantly contributing to cancer-related mortality. It involves
the spread of cancer cells from the primary tumor to distant
sites, where they establish secondary tumors, complicating
treatment and reducing overall survival rates [ 102 ]. Cancer
cells oen acquire invasive and stem cell-like properties, utiliz-
ing biological processes such as epithelial–mesenchymal transi-
tion, essential for embryogenesis and tissue repair, to promote
metastasis [ 103 – 105 ].
Copper ion promotes cancer metastasis through various
mechanisms, including activating proliferation and metabolic
enzymes [ 106 ]. e dysfunction of copper-containing secretory
enzymes, including superoxide dismutase 3 (SOD3) and lysyl
oxidase (LOX), profoundly impacts cancer metastasis by pro-
moting angiogenesis, epithelial–mesenchymal transition, and
cancer cell invasion [ 107 ]. Loss of SOD3 in tumor tissues
increases oxidative stress, a factor linked to various aspects of
cancer progression [ 108 ]. Conversely, Laukkanen [ 109 ] has
shown that enhancing SOD3 activity or expression through
endogenous administration of recombinant SOD3 or induction
of SOD3 expression can inhibit cancer cell metastasis.
For another example, the copper-dependent amine oxidase
LOX promotes cancer cell invasion and migration [ 110 ].
Extra cellular copper-dependent enzymes belonging to the
LOX family have been implicated in promoting a supportive
extracellular matrix environment that facilitates cancer cell
invasion and metastasis. e increased cross-linking of collagen
bers can enhance the stiness of the extracellular matrix,
providing physical support for cancer cells to migrate through
tissues [ 111 ]. Up-regulation of LOX mRNA and protein expres-
sion has been observed in various cancer types, including head
and neck squamous cell carcinoma (HNSCC) [ 112 ], breast
cancer [ 112 , 113 ], colorectal cancer [ 114 , 115 ], and prostate
cancer [ 116 ].
Furthermore, copper-dependent redox enzymes, such as
the ErbB2-driven cell motility (MEMO1) mediator, play a
crucial role in breast cancer by facilitating cell migration and
invasion. MEMO1 has been demonstrated to promote cell
migration by modulating cytoskeletal dynamics and facilitat-
ing the formation of adhesion sites [ 117 , 118 ]. us, copper
ion is involved in crucial processes, including cellular pro-
liferation, dierentiation, angiogenesis, and cancer invasion/
metastasis [ 119 , 120 ].
To summarize, copper ion plays a dual role in cellular pro-
cesses. It necessarily serves as a catalytic cofactor for multiple
physiologic processes [ 25 ]. On the other hand, excessive copper
ion accumulation can lead to metabolic dysfunction and cell
death [ 39 ]. is underscores the importance of maintaining a
delicate balance in copper ion regulation for optimal physio-
logical function and mitigating adverse eects. erefore, tar-
geting copper ion dysregulation holds promise as a strategy for
cancer treatment.
Cuproptosis is a Novel Therapeutic Target for
Copper-Based Cancer Therapy
e discovery of cuproptosis represents a signicant advance-
ment in cancer treatment. While elesclomol (ES) has been
reported as a drug that trigger apoptosis, its ability to induce
cell death, specically through copper ion loading, represents
a novel pathway that diers from traditional types of cell death,
such as apoptosis, ferroptosis, or necroptosis [ 121 ]. is process
is accompanied by the activation of caspase-3 in the caspase
cascade [ 122 ]. However, ES induces the accumulation of excess
copper ions binding to dihydrolipoamide S-acetyltransferase
(DLAT) in cancer cells results in the down-regulation of Fe-S
cluster proteins and abnormal aggregation of thioctylated
proteins in the TCA cycle, ultimately leading to cuproptosis
of cancer cells [ 8 ]. is cascade of events provides valuable
insights into the cytotoxic mechanism of copper ionophores
and opens up new avenues for treating various diseases, includ-
ing cancers. By targeting the specic vulnerabilities of cancer
cells related to copper metabolism, novel therapeutic strategies
can be developed to exploit this mechanism for more eective
and targeted cancer treatments. Excess amounts of intracellular
copper substances or copper ions can generate hydroxyl radi-
cals and increase intracellular ROS levels, leading to severe
oxidative damage-based apoptosis and ferroptosis [ 123 , 124 ].
Furthermore, copper-induced ROS and copper (II) ions can cause
necroptosis through DNA damage in the cancer cells [ 124 , 125 ].
Copper can increase the NOD-like receptor family pyrin domain-
containing protein 3, cleaved caspase-1, apoptosis-associated
speck-like protein, and IL-1β protein levels, leading to cellular
pyroptosis and inammatory responses [ 126 – 128 ]. erefore,
copper-mediated regulated cell death (RCD) may not func-
tion as an independent mechanism of RCD and can be closely
interrelated with other cell death pathways, implying that vari-
ous therapeutic strategies can be developed by combining
other RCD. is section mainly describes the regulatory mech-
anism of cuproptosis, incorporating insights from gene signal-
ing pathways involved in the interplay between copper ions
and cellular components.
First, researchers are exploring strategies to manipulate
copper metabolism in cancer cells to induce cuproptosis
selectively. is includes the development of copper iono-
phores or other biomaterials that enhance the accumulation
of copper ions within cancer cells. Second, combining cupro-
ptosis-targeting agents with other cancer therapies, such as
chemotherapy or targeted therapies, may enhance therapeutic
ecacy. ese combination applications can provide syner-
gistic eects and overcome resistance mechanisms in cancer
cells. Finally, understanding the molecular mechanisms underly-
ing cuproptosis in specic cancer types can enable the devel-
opment of personalized treatment strategies. More eective
and tailored therapies will be developed by targeting the
unique susceptibilities of individual cancers related to copper
ion metabolism.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 5
Mechanism of cuproptosis
e phenomenon of cell death is caused by excess levels of cop-
per ion in the body called cuproptosis and is associated with
changes in mitochondrial enzymes [ 129 ]. Excessive copper ion
levels can cause oxidative damage to mitochondrial membranes
and interfere with enzymes involved in the TCA cycle, also
known as the citric acid cycle or Krebs cycle. As excess Cu2+
inltrates the cell, it moves to the mitochondria and is converted
to Cu
+
. ese elevated Cu
+
level disrupt both the TCA cycle and
the ETC, leading to cell death due to oligomerization of lipoylated
proteins and depletion of Fe-S cluster proteins [ 130 ]. A study
demonstrated that cells subjected to Cu ionophores exhibited
a time-dependent dysregulation of various TCA cycle-related
metabolites, and the inhibition of ETC complexes I and II sig-
nicantly attenuated Cu ion-induced cell death [ 131 ]. To explore
mechanism of cuproptosis, researchers conducted genome-wide
CRISPR-Cas9 screening. Seven genes have been discovered as
regulators of cuproptosis, namely ferredoxin-1 (FDX1), lipoic
acid synthetase (LIAS), lipoyltransferase 1, dihydrolipoamide
dehydrogenase, DLAT, pyruvate dehydrogenase E1 subunit
alpha 1, and pyruvate dehydrogenase E1 subunit beta [ 8 ] FDX1
is a pivotal enzyme with strong reducing capabilities, responsible
for lipoylating 4 specic enzymes—DLAT, dihydrolipoamide
succinyltransferase, glycine cleavage system protein H, and dihy-
drolipoamide branched chain transacylase E2 (DBT)—within
the mitochondria. is indicates the critical role of lipoylation
in sustaining cellular metabolic pathways and ensuring proper
mitochondrial function [ 132 ].
Interestingly, Cu+ can directly bind to lipoylated TCA cycle
proteins, specically the disulde bond on the terminal cysteine
residue. is interaction triggers disulde bond-dependent
aggregation of these proteins, potentially disrupting the TCA
cycle and resulting in the degradation of Fe-S cluster proteins.
Also, copper ion might impede the ubiquitinated protein degra-
dation function of valosin-containing protein (p97) by interact-
ing with nuclear protein localization protein 4 (Npl4), which
could result in its aggregation or directly binding and inhibiting
its conformational transition, inducing proteotoxic stress, ulti-
mately leading to cell death [ 10 , 133 , 134 ]. Likewise, FDX1/LIAS
act as an upstream regulator of protein lipoylation, and their
dysregulation facilitates the decrease of Fe-S clusters, whereas
the deletion of FDX1 or LIAS results in the accumulation of
α-ketoglutarate and pyruvate, leading to decrease lipoylation and
cell death [ 135 ]. In addition, Tsvetkov et al. [ 8 ] investigated
the impact of intracellular copper ion levels on cuproptosis by
manipulating genes related to copper ion transport in vitro and
in vivo. Overexpression of the copper ion importer SLC31A1
increased cell sensitivity to copper ion-induced protein aggrega
-
tion and degradation of Fe-S cluster proteins. is eect was
partially rescued by copper chelators and depletion of key cupro
-
ptosis regulators, FDX1 or LIAS. Consistently, these ndings
were validated in a mouse model of Wilson's disease mouse
model with Atp7b depletion (Atp7b−/−). e livers of Atp7b−/−
mice exhibited substantial depletion of lipoylated and Fe-S clus-
ter proteins compared to those of Atp7b+/− and wild-type mice.
is observation conrms that the accumulation of intracellular
copper ion is associated with cuproptosis in vivo. e major pro-
cesses and regulators of cuproptosis were illustrated in (Fig. 1 ).
Cuproptosis-based cancer therapy
Cuproptosis, a unique cell death mechanism, has attracted sig-
nicant interest in cancer research. is is because it has the
potential to inhibit cancer cell proliferation and even reverse
resistance to anticancer drugs, oering new treatment options.
To induce cuproptosis, delivering Cu2+ or Cu+ to cancer cells
is necessary. Various approaches have been developed for the
intracellular delivery of copper ion, including ionophores,
metal-organic frameworks (MOFs), hydrogels, and inorganic
nanoparticles ( Table ).
Among them, ionophores can bind metal ions reversibly
under physiological conditions. is not only promotes the
transmembrane transport of metal ions in the electrically neu-
tral and lipophilic state but can also change the biodistribution
of metal ions by releasing them under specic stimuli, such as
local low metal ion concentrations or biological reduction
[ 130 , 136 , 137 ]. ES, DSF, and NSC319726 are all considered cop-
per ionophores with the potential to kill cancer cells [ 138 , 139 ].
Specically, NP@ESCu, a nanoparticle utilizing the traditional
ionophore ES, has facilitated the targeted delivery of copper
ion to mitochondria [ 140 ]. ES can reversibly bind with Cu2+,
resulting in the formation of an ES–Cu2+ complex. is com-
plex facilitates the direct transportation of copper ion into
mitochondria. Within the mitochondria, FDX1 is responsible
for reducing Cu2+ to Cu+ and subsequently releasing it. e
increased level of Cu+ directly binds to lipoylated DLAT, lead-
ing to the lipoylated proteins aggregation and loss of Fe-S clus-
ter proteins, ultimately resulting in cuproptosis [ 8 , 130 , 137 ].
Additionally, DSF, which has antiproliferative eects, has shown
promise for cancer treatment by inducing cuproptosis beyond its
role as a copper ion carrier [ 8 , 141 ]. DSF has also been studied for
its ability to reverse drug resistance, making cancer cells more sen-
sitive to chemotherapy [ 142 ]. Furthermore, DSF has been found
to have immunomodulatory eects, potentially enhancing the
body’s immune response against cancer cells [ 143 ].
Despite promising preclinical ndings demonstrating ben-
ecial anticancer eects through copper chelation or induction
of copper ion-dependent cell death via copper ionophores,
several limitations remain in the clinical translation, including
their lack of specicity and short half-life. Developing novel
approaches is essential to address these limitations and advance
the clinical translation of treatments targeting copper-dependent
mechanisms for eective cancer therapy.
Cuproptosis-Targeted Nanomedicines for
Combination Cancer Therapy
For eective cancer therapy, cuproptosis-based cancer treat-
ment still needs to overcome several limitations, including the
selective increase of copper ion levels in cancer cells, preventing
copper ion damage to healthy cells, and extending the dura-
tion of cuproptosis [ 18 , 19 ]. Nanomedicine emerges as a cru-
cial solution for tackling this challenge, given its eectiveness
in enhancing drug solubility, prolonging circulation time,
enabling targeted drug delivery, and minimizing unexpected
side eects [ 144 – 146 ]. Consequently, the recent elucidation of
this phenomenon has led to signicant progress in developing
nanomaterials based on cuproptosis and has shown promising
progress in applications for cancer eradication. For example,
copper and copper oxide nanoparticles have garnered signi-
cant attention in cancer biology, presenting a wide array of
advantages such as enhancing drug stability, ensuring appropri-
ate biodistribution, rening therapeutic index, and facilitating
targeted delivery of active agents to specic sites through active
or passive targeting mechanisms [ 147 – 151 ].
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 6
In another study, a photothermal-activated nanosystem
infused with copper (Au@mesoporous silica nanoparticles
[MSN]-Cu/polyethylene glycol [PEG]/DSF) has been engi-
neered, comprising mesoporous silica-coated gold nanorods
(Au@MSN) functioning as photothermal agents [ 152 ]. is
system exhibits excellent loading capacity for Cu2+ and the anti-
cancer drug DSF, facilitating synergistic treatment involving
cuproptosis, apoptosis, and photothermal therapy (PTT). In the
following section, we provide the recent strategies employing
anticancer nanomaterials based on cuproptosis and categorize
potential synergistic treatment approaches for improved cancer
treatment.
Since the concept of cuproptosis, increasing evidence has
demonstrated the anticancer potential of cuproptosis, and much
eort has been devoted to the design and development of
various copper-based nanomaterials for cancer treatment. e
nanomaterials up-regulate local copper ion concentration by
delivering copper ions to the tumor site. Increasing copper ion
levels represents a novel approach for copper-ion-interference
therapy, disrupting copper homeostasis and addressing diseases
through various cell death pathways. For example, Jia et al.
[ 153 ] developed a brain-targeted nanoplatform (HFn-Cu-
REGO NPs), incorporating human H-ferritin (HFn), rego-
rafenib, and Cu2+, enabling site-specic delivery and modulation
of autophagy and cuproptosis against glioblastoma multiforme
(GBM) (Fig. S3A). Eective management of persistent GBM
oen requires prolonged chemotherapy postsurgery to eliminate
residual cancerous tissues [ 154 ]. While temozolomide is the
primary chemotherapeutic agent for GBM therapy, its treatment
outcomes remain unsatisfactory [ 155 ].
However, regorafenib, an oral multikinase inhibitor, exhibits
signicantly superior eects in suppressing GBM compared to
temozolomide [ 156 ]. HFn, due to its selective accumulation in
GBM facilitated by transferrin receptor 1 (TfR1)-mediated active
targeting and pH-responsive delivery characteristics, serves as
an ideal carrier for targeted drug delivery [ 157 ]. Regorafenib
inhibits autophagosome-lysosome fusion, leading to lethal autoph-
agy arrest in GBM cells [ 156 ]. Cu
2+
facilitates the encapsulation
of regorafenib within HFn through coordination interaction
and disrupts copper homeostasis, inducing cuproptosis (Fig.
S3B) [ 8 , 158 ]. Dynamic light scattering analysis conrmed the
narrow size distribution of HFn with a mean diameter of approxi-
mately 15.5 nm (Fig. S3C).
To explore the potential involvement of cuproptosis in HFn-
Cu-REGO-mediated cell death, immunouorescence analysis
was conducted to assess the endogenous levels of lipoic acid
and FDX1 in GBM cells treated with regorafenib, HFn-Cu, and
HFn-Cu-REGO NPs for 24 h. e immunouorescence results
revealed a notable accumulation of FDX1 in the groups treated
with HFn-Cu and HFn-Cu-REGO NPs (Fig. S3D). Treatments
involving HFn-Cu and HFn-Cu-REGO NPs demonstrated a
signicant rise in intracellular copper ion levels within GBM
cells (Fig. S3E and F). Moreover, these treatments induced uc-
tuations in the expression levels of various Cu2+ transporters
in both U251 and U87 cells (Fig. S3G and H). As shown in (Fig.
S3I), the group treated with HFn-Cu-REGO NPs exhibited
signicant cancer damage, attributed to ecient delivery and
therapeutic synergy between regorafenib and Cu
2+
. Additionally,
treatment with HFn-Cu and HFn-Cu-REGO NPs intensied
staining of lipoic acid and FDX1 in brain tumor tissues (F ig. S3J).
Fig.1. The schematic of cuproptosis mechanism. Cu ionophores like ES and DSF initially bind to extracellular copper ions, aiding in their internalization into intracellular
compartments. Once inside, copper ion interacts with lipoylated mitochondrial enzymes involved in TCA cycle, such as DLAT. This interaction triggers the aggregation of these
proteins. FDX1/LIAS acts as an upstream regulator of protein lipoylation, promoting the aggregation of mitochondrial proteins. This process leads to the loss of Fe-S clusters
and the inactivation of Npl4-p97. These aberrant events induce proteotoxic stress, ultimately resulting in cell death. STEAP, 6-transmembrane epithelial antigen of the prostate.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 7
Table. Application of cuproptosis-targeted nanomedicines used for cancer therapy
Cuproptosis-
targeted nano-
medicines Nanomaterials Cancer Type Therapeutic effect
Combination
therapy Yea r Ref.
GOx@ [Cu(tz)] Coordination
polymers
Bladder
cancer
Glucose and GSH depletion enhance GOx@
[Cu(tz)]-mediated cuproptosis, inducing mito-
chondrial protein aggregation. Elevated H2O2 lev-
els boost type I PDT efficacy. GOx@[Cu(tz)] shows
low systemic toxicity and inhibits tumor growth in
mice with bladder cancers.
PDT 2022 [183]
CuET NPs - Non-small
cell lung
cancer
(NSCLC)
CuET overcomes cisplatin resistance in NSCLC
via cuproptosis, possessing lower reduction
potential and inertness with GSH. It conquers drug
resistance in A549/DDP cells, showing potent
anticancer activity unaffected by GSH levels. CuET
NPs induce cell death via cuproptosis and dem-
onstrate superior biosafety in a cisplatin-resistant
cancer model.
-2022 [142]
DOX@Fe/CuTH Hollow
amorphous
metal organic
framework
(HaMOF)
Breast
cancer
DOX@Fe/CuTH demonstrates catalytic therapeu-
tic properties triggered by the tumor microenvi-
ronment. It amplifies cellular oxidative stress by
increasing H2O2 production and depleting GSH
simultaneously. This induces mitochondrial dys-
function and down-regulates ATP7A expression,
resulting in Cu2+ overload and cellular cuproptosis.
Chemotherapy 2022 [184]
Au@MSN-Cu/
PEG/DSF
Mesoporous
silica-coated
Au nanorods
Breast
cancer
Combined with PTT, Au@MSN-Cu/PEG/DSF
effectively killed cancer cells, inhibited growth
(inhibition rate up to 80.1%), and reduced DLAT,
LIAS, and Npl4.
PTT 2023 [152]
HFn-Cu-REGO
NPs
- Glioblasto-
ma (GBM)
HFn enables targeted delivery to GBM via TfR1.
Regorafenib halts autophagy, inducing cell death.
Cu2+ enhances drug encapsulation and triggers
cuproptosis, augmenting regorafenib's efficacy
against GBM.
Chemotherapy 2023 [153]
Cu2O@
CuBTC-DSF@
HA nanocom-
posites
(CCDHs)
Metal–organic
frameworks
(MOFs)
Breast
cancer
CCDHs degrade rapidly in acidic conditions,
releasing Cu+ and DSF. Cu+ disrupts the TCA cycle,
causing mitochondrial dysfunction and initiating
cuproptosis. Simultaneously, Cu+ undergoes a
Fenton-like reaction, generating ROS, and convert-
ing back to Cu2+. DSF chelates Cu2+, forming
CuET, which reduces Cu2+ to Cu+ and boosts ROS
production. ROS exacerbates mitochondrial dam-
age, contributing to cuproptosis cell death and
enhancing the anticancer effect synergistically.
-2023 [185]
BSO-CAT@
MOF-199 @DDM
(BCMD)
MOFs Glioblasto-
ma (GBM)
BCMD-mediated cuproptosis can induce immuno-
genic cell death (ICD), promoting the infiltration of
cytotoxic T lymphocytes and reversing the immu-
nosuppressive microenvironment of glioblastoma
to enhance tumoricidal immunity.
Immuno-
therapy
2023 [186]
Cu2(PO4)(OH)
NPs
H2S-respon-
sive copper
hydroxyphos-
phate nanopar-
ticles
Colon
cancer
Cu2(PO4)(OH) NP, the synergy of increased
endocytosis and decreased exportation results
in maximum copper ion overload. Coupled with
efficient copper ion release, this disrupts the
mitochondrial tricarboxylic acid cycle and down-
regulates iron-sulfur cluster proteins, ultimately
initiating cuproptosis.
-2023 [167]
(Continued)
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 8
Cuproptosis-
targeted nano-
medicines Nanomaterials Cancer Type Therapeutic effect
Combination
therapy Yea r Ref.
Cu2O@
TBP-2(PTC)
Cuprous oxide Breast
cancer
PTC triggers cuproptosis in cancer cells both
invitro and invivo, markedly inhibits lung metasta-
sis of breast cancer, boosts central memory
T cell count in peripheral blood, and prevents
tumor rechallenge.
PDT 2023 [178]
CS/MTO-Cu@
AMI
MOFs Breast
cancer
Coordinated Cu2+ of CS/MTO-Cu@AMI triggers
cuproptosis, activating AMPK-mediated PD-L1
degradation, disrupting energy supply, and ampli-
fying oxidative stress, sensitizing chemo-
immunotherapy. AMI complements by suppress-
ing macropinocytosis and exosome release,
enhancing Cu2+ mediated therapy.
Chemothera-
py/ immuno-
therapy
2023 [187]
CuMoO4
Nanodots
- Breast
cancer
CuMoO4 nanodots efficiently convert light to heat,
inhibiting cancer cell regulation to oxidative stress
and promoting sustained photothermal synergis-
tic ferroptosis. They trigger immune responses
and induce both immunogenic cell death and
cuproptosis in cancer cells.
PTT 2023 [188]
HD/BER/GOx/
Cu hydrogel
Hyaluronic
acid-dopamine
(HD) polymer
Breast
cancer
The HD/BER/GOx/Cu hydrogel reduces the
frequency of dosing in local cancer therapy and
mitigates invasiveness-related inconveniences.
It could be used to shrink breast cancer size
before surgery and suppress tumor growth in
clinical settings.
CDT/CT/ ST 2023 [189]
NP@ESCu Reactive
oxygen species
(ROS)-sensi-
tive polymer
Bladder
cancer
Intracellular ROS-triggered release of ES
and Cu synergistically eliminate cancer cells
through cuproptosis and stimulate immune
responses. Invitro, NP@ESCu effectively deliv-
ers Cu, inducing cuproptosis. In a mouse model
of subcutaneous bladder cancer, NP@ESCu
induces cuproptosis and remodels the tumor
microenvironment. NP@ESCu enhances the
anticancer activity of αPD-L1.
Immuno-
therapy
2023 [140]
TP-M-Cu-MOF/
siATP7a NP
Copper-based
metal organic
framework
(Cu-MOF)
Small cell
lung cancer
(SCLC)
TP-M-Cu-MOF/siATP7a demonstrated potent
gene silencing, specifically hindering copper
ion trafficking, enhancing copper ion intake,
inducing cuproptosis, and enhancing therapeu-
tic efficacy in mice with SCLC brain metastasis
tumors.
Gene therapy 2023 [190]
Au25(NAMB)18
NCs-Cu2 + @
SA/NHGs
Nanohybrid
gels
Hepato-
cellular
carcinoma
(HCC)
The release of Cu2+ from the nanohybrid gels
induced cuproptosis and catalyzed H2O2 to gen-
erate O2, enhancing PDT. Cu2+ depleted GSH,
forming Cu+, which produced hydroxyl radicals
to kill cancer cells, synergizing with enhanced
PDT and CDT.
PTT/PDT/CDT 2023 [191]
CCNAs Copper-coordi-
nated nanoas-
semblies
Prostate
cancer
The inclusion of Cu2+ in the CCNAs not only
amplified the photodynamic process by
catalyzing oxygen generation but also facili-
tated the aggregation of toxic mitochondrial
proteins, ultimately triggering cuproptosis in
cancer cells.
PDT/ immuno-
therapy
2024 [192]
Table. (Continued)
(Continued)
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 9
Consequently, mice treated with HFn-Cu-REGO NPs exhibited
delayed tumor growth and the lowest bioluminescence levels
compared to other treatment groups (Fig. S3K). is synergy
with regorafenib-mediated lethal autophagy arrest enhances
the therapeutic impact against GBM [ 159 ].
In other cases, Zhao et al. prepared hydrogen sulde (H
2
S)-
responsive copper hydroxyphosphate nanoparticles (Cu2(PO4)
(OH) NPs) to enhance cellular uptake and reduce the eux of
copper ions [ 160 , 161 ]. is approach can achieve copper ion
overload because excess intracellular copper ions tend to
be pumped out of the cells by the copper exporter ATP7A
[ 162 , 163 ]. Upon exposure to an H2S-rich microenvironment
in colon cancer, Cu2(PO4)(OH) NPs transform smaller copper
sulde NPs, enhancing cellular uptake [ 164 – 166 ]. is process
improves Fenton activity and copper ion dissociation [ 167 ].
ROS generated through the Fenton reaction activate inam-
masomes and Caspase-1 proteins, leading to gasdermin D
cleavage and induction of pyroptosis [ 168 – 171 ]. ese ROS
also aect mitochondrial function and down-regulate the cop-
per exporter ATP7A, further reducing copper excretion. e
synergy of enhanced endocytosis and reduced exportation
results in maximal copper ion overload. is, coupled with the
ecient release of copper ions, disrupts the mitochondrial TCA
cycle and down-regulates Fe-S cluster proteins, ultimately
inducing cuproptosis. Since both pyroptosis and cuproptosis
are eective mechanisms for cell death induction, this research
oers a novel approach for achieving eective cancer-targeted
therapy through H2S-activated copper ion overload using
simple Cu2(PO4)(OH) NPs [ 167 ]. e ndings demonstrated
the potential of inducing cuproptosis in cancer cells using
copper-based nanomaterials as a promising strategy for cancer
treatment.
Because cancer cells are commonly heterogeneous, relying
solely on chemotherapy may prove less eective and less toler-
able for certain cancers. us, implementing combination thera-
pies through various methods is essential to enhance treatment
Cuproptosis-
targeted nano-
medicines Nanomaterials Cancer Type Therapeutic effect
Combination
therapy Yea r Ref.
CuSiO3@Au-Pd
NMs
Multifunctional
copper phyllo-
silicate-based
nanomotors
Breast
cancer
The generation of ·OH by CuSiO3@Au-Pd NMs
through Fenton-like reactions and the thermal en-
ergy produced by the photothermal effect coopera-
tively induced apoptosis in cancer cells. Moreover,
these thermal gradients facilitated the movement
of CuSiO3@Au-Pd NMs via thermophoresis, promot-
ing deeper penetration into tumor tissue.
PTT/CDT 2024 [193]
CuO2/DDP@
SiO2
Silica-coated
copper perox-
ide (CuO2)
Hepato-
cellular
carcinoma
(HCC)
Depleting GSH sensitizes cancer cells to CuO2/
DDP@SiO2-induced cuproptosis, causing
lipoylated mitochondrial protein aggregation.
Invitro, reduced GSH binding boosts intracellular
cisplatin levels. CuO2 down-regulates MRP2 via
O2-dependent HIF-1 inactivation, blocking cisplatin
efflux and enhancing its anticancer effect both
invitro and invivo.
Chemothera-
py/CDT
2024 [194]
HA-CuH-PVP
(HCP).
HA combined
with PVP
Breast
cancer
HCP effectively damages tumor cells by generat-
ing Cu+ and hydrogen (H2), inducing apoptosis
and cuproptosis, and shows promising invivo
antitumor and metastasis-inhibiting potential.
Apoptosis 2024 [179]
Cu(I) NP Cu(I) self-as-
semble with a
ROS-sensitive
polymer
Pancreatic
cancer
ROS-sensitive copper complex nanoparticles (Cu(I)
NP) that deliver copper complexes to cancer cells,
inducing cuproptosis. In a pancreatic cancer mouse
model, Cu(I) NP accumulated in tumors, inhibited
growth, and enhanced immune responses.
Immuno-
therapy
2024 [180]
CuP/Er Nanoscale
coordination
polymers
(NCPs)
Breast can-
cer/ colon
cancer
CuP/Er nanoparticles codeliver copper and erastin
to synergize cuproptosis and ferroptosis, enhanc-
ing T cell response and effectively regressing
cancers with immune checkpoint blockade.
Ferroptosis/
immuno-
therapy
2024 [181]
MetaCell Thermosensi-
tive liposomal
bimetallic Fe-
Cu MOFs (Lip@
Fe-Cu-MOFs)
Breast
cancer
MetaCell system integrates cuproptosis and fer-
roptosis using Fe-Cu-MOFs to catalyze Fenton-like
reactions, producing radicals and depleting GSH
in tumor cells, effectively eliminating solid tumors,
preventing recurrence, and extending survival.
PTT/ferrop-
tosis
2024 [182]
Table. (Continued)
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 10
outcomes [ 172 , 173 ]. Specically, the discovery of cuproptosis
contributing to tumor immunogenicity suggests its potential
to augment the eectiveness of immune checkpoint blockade
(ICB) therapy [ 174 , 175 ]. ICB has emerged as a pivotal approach
in cancer immunotherapy, enhancing and activating the anti-
cancer immune responses in the body [ 176 ]. Recent studies
indicated that reduced intracellular copper ion levels promoted
ubiquitin-mediated degradation of PD-L1 in cancer cells,
increasing antitumor immunity [ 15 ]. Consequently, there is
the possibility of a synergistic eect between cuproptosis and
ICB therapy.
Guo et al. [ 140 ] designed a ROS-sensitive polymer known
as PHPM has been developed for the coencapsulation of ES and
copper ion, resulting in the formation of nanoparticles termed
NP@ESCu. e design involves an amphiphilic biodegradable
polymer termed PHPM, as shown in (Fig. 2 A). PHPM is notable
for containing ROS-sensitive thioketal bonds in its main chain,
along with pendant pairs of carboxylic acids. ES and Cu+ are
promptly released upon entering cancer cells, triggered by the
abundance of intracellular ROS. Increasing the presence of cop-
per ionophores like ES in tumor tissues can expedite the delivery
of copper ion into cancer cells, enhancing the antitumor eect
through cuproptosis [ 177 ].
Additionally, transporting copper ion into cancer cells with
ionophores can elevate PD-L1 expression (Fig. 2 B) [ 15 ]. e
anticancer activity of NP@ESCu was evaluated using a live–
dead staining assay on 3-dimensional (3D) tumor spheroids,
demonstrating that the highest number of dead cells (indicated
by red staining) was observed in the 3D tumor spheroids
treated with NP@ESCu (Fig. 2 C). Furthermore, the apoptotic
Fig.2.Cuproptosis induction via ROS-responsive nanoparticles incorporating ES and copper ion in conjunction with αPD-L1 for augmented cancer immunotherapy.
(A) Schematic illustration of NP@ESCu to induce cuproptosis. (B) Design of converting immune “cold tumors” to “hot tumors” through NP@ESCu induces cuproptosis,
eliciting strong antitumor immune responses invivo, further maximized by combining with αPD-L1. (C) Representative CLSM images of 3D cell spheroids from BIU-
87 cells subjected to various treatments, stained with calcein-AM/PI. Scale bars: 100 μm. (D) Representative flow cytometry profiles illustrating the apoptotic rate
of BIU-87 cells under various treatment treatments. (E) Tumor growth curves and (F) Exvivo tumor weight of mice under various treatments. (G) Biodistribution
following intravenous injection of NP@ESCu@Cy7.5 at different time points. (H) Exvivo imaging and (I) Mean fluorescence intensity of NP@ESCu@Cy7.5 in major
organs and tumor (I, intestine; S, spleen; H, heart; Lu, lung; K, kidney; Li, liver; T, tumor). Reproduced from [140] with permission from Wiley-VCH, Copyright 2023.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 11
rate of BIU-87 cells treated with various drugs was investigated
using an Annexin V-FITC and propidium iodide (PI) double-
staining assay. e ndings revealed that the apoptotic rate of
BIU-87 cells treated with NP@ESCu was 44.9%, surpassing that
of cells treated with ES (29.8%) by 1.5 times and exceeding that
of cells treated with NP@ES (35.5%) by approximately 1.3 times
in a culture medium containing CuCl2 (0.1 μm) (Fig. 2 D). e
anticancer eect of ES was minimal on day 12, similar to NP@
ES in vivo. In contrast, NP@ESCu exhibited the most signi-
cant anticancer eect (Fig. 2 E and F). Remarkably, even 36 h
postinjection of NP@ESCu@Cy7.5, a robust uorescence signal
persisted at the tumor site, suggesting rapid accumulation and
prolonged retention of NP@ESCu@Cy7.5 in the tumor micro-
environment (Fig. 2 G). Aer 36 h, mice were sacriced, and
NP@ESCu@Cy7.5 biodistribution was assessed ex vivo. e
strongest uorescence signal was observed in tumors, indicat-
ing eective tumor targeting (Fig. 2 H and I). Taken together,
NP@ESCu demonstrated rapid and ecient targeting and accu-
mulation at tumor sites, highlighting its potential for clinical
applications. Hence, combining copper ionophores with immu-
notherapeutic agents like anti-PD-L1 antibody (αPD-L1) may
oer a more eective cancer therapy strategy.
Ning et al. [ 178 ] developed a cuprous oxide nanoparticle
(Cu2O)/TBP-2 cuproptosis sensitization system coated with
platelet vesicles, abbreviated as PTC. PTC was formed through
the physical extrusion of the AIE photosensitizer (TBP-2),
Cu2O, and platelet vesicle. PTC exhibits enhanced long-term
blood circulation and tumor-targeting capabilities (Fig. 3 A).
As shown in (Fig. 3 B), the PTC surface features a detailed gray
cell membrane structure, indicating successful encapsulation
Fig.3.Enhancing cuproptosis in breast cancer metastasis inhibition and rechallenge resistance through combination therapy with type-I AIE photosensitizer loaded biomimetic
system. (A) Schematic illustration of a type-I AIE photosensitizer integrated into a biomimetic system to trigger cancer cuproptosis. (B) Transmission electron microscopy images of
Cu2O and PTC. Scale bars: 50 nm. (C) Alterations in the concentration of copper ions within cells following incubation with various formulations. (D) DLAT fluorescence images of
cancer cells posttreatment as specified. DLAT aggregation is highlighted by white arrows. PC: PM-coated Cu2O without TBP-2. Scale bars: 15 μm. (E) DLAT fluorescence images
of cancer cells post PTC+L treatment with varying concentrations of Cu2O. DLAT aggregation is highlighted by white arrows. Scale bars: 15 μm. (F) Analysis of cellular apoptosis
in 4T1 cells following treatment with various formulations. (G) Changes of tumor volume in mice after undergoing diverse treatments for tumor metastasis. (H and I) The ratio of
central memory T cells (TCM, CD62L+, CD44+) to CD3+, CD8+ T cells in the bloodstream 14 d post PTC treatment for tumor rechallenge assessed via flow cytometry. Reproduced
from [178] with permission from American Chemical Society, Copyright 2023.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 12
with a platelet membrane. Also, the copper ion content within
tumor cells notably surged in the PTC group within 2 h, reaching
around 6 pg/cell. In contrast, the intracellular copper ion content
in tumor cells of the erythrocyte membrane-coated RTC group
remained approximately at 2 pg/cell. is observation further
validated the tumor-targeting ecacy of PTC (Fig. 3 C). en,
the uorescence of the dihydrolipoyllysine-residue acetyltrans-
ferase component of the pyruvate dehydrogenase complex, mito-
chondrial DLAT, and the expression of cuproptosis-related
proteins were examined in tumor cells treated with dierent
materials. During cuproptosis, copper ions directly bind to
lipoylated components of the TCA cycle, causing aggregation
of the fatty acylated proteins and loss of Fe-S proteins. Both the
PC+L and PTC groups exhibited varying degrees of DLAT
aggregation, with the PTC+L group showing higher DLAT
aggregation. Additionally, an increase in PTC concentration cor-
related with increased aggregation (Fig. 3 D and E). PTC+L treat-
ment is also capable of inducing apoptosis in tumor cells (Fig.
3 F). Subsequently, they assessed the antitumor ecacy of PTC
in vivo, observing a partial tumor suppression rate in both the
PC+L and PTC groups. Notably, the PTC+L group demon-
strated pronounced tumor growth when exposed to light, indi-
cating that PTC exhibited a high tumor growth under light
conditions (Fig. 3 G). In addition, 14 d aer the initial treatment,
peripheral blood was collected from mice, and the ratio of cen-
tral memory T cells (TCM, CD3+, CD8+, CD62L+, and CD44+)
to CD3+, CD8+ T cells was analyzed using ow cytometry. e
ndings revealed that the TCM content was most elevated fol-
lowing PTC+L treatment, suggesting a robust immune memory
response triggered by PTC+L therapy (Fig. 3 H and I). us,
PTC treatment eectively targets and induces cuproptosis in
cancer cells both in vitro and in vivo, signicantly suppressing
lung metastasis of breast cancer and increasing central memory
T cell counts in peripheral blood, resulting in preventing recur-
rence aer tumor rechallenge. is strategic design of combina-
tion therapy induces various forms of cell death linked to ROS
and oers signicant benets for cancer treatment.
Although cuproptosis has received enormous attention for
cancer treatment, specic activation of cuproptosis with cop-
per utilization in the tumor microenvironment remains chal-
lenging. To address this limitation, He et al. [ 179 ] developed
HA-coated acid-degradable copper hydride (HCP) nanopar-
ticles using microuidic synthesis. Surface HA on HCP enabled
targeting CD44 receptors on 4T1 breast tumor cells, resulting
in internalized via receptor-mediated endocytosis and induc-
ing signicant tumor cell damage by simultaneously releasing
Cu+ ions and hydrogen (H2). e Cu+ ions triggered apoptosis
by Fenton-like reactions and induced cuproptosis by aggregat-
ing mitochondrial proteins. Additionally, released H2 increased
both cell death by promoting mitochondrial dysfunction
and intracellular redox imbalance. Finally, HCP signicantly
inhibited tumor growth and prevented lung metastases, show-
ing potent therapeutic ecacy by copper-based antitumor
materials.
Hu et al. [ 180 ] developed stimuli-responsive copper com-
plex nanoparticles (Cu(I) NP) designed to deliver copper (I)
ions into cancer cells for cuproptosis-based cancer immu-
notherapy (Fig. 4 A). Dinitrogen-diphosphine chelated Cu(I)
complex ([Cu(2,9-dimethyl-1,10-phenanthroline)(N,N-bis-
((diphenylphosphaneyl)methyl)pyridin-3-amine)](BF4)) was
synthesized and then nanoparticulated with a ROS-sensitive
polymer through electrostatic and hydrophilic interactions,
resulting in forming Cu(I) NP. ioketal moieties in ROS-
sensitive polymer enabled rapid dissociation of Cu(I) NP aer
incubation with H
2
O
2
, resulting in 91.4% of Cu(I) being released
within 36 h. Cu(I) NP showed time-dependent cellular uptaken
in human pancreatic cancer cells, MIAPACA-2, and eective
penetration in human pancreatic cancer cells (PANC-1)-based
3D multicellular tumor spheroids. Cu(I) NP exhibited a higher
antitumor eect in dierent pancreatic cancer cell lines, wherein
the half-maximal inhibitory concentration (IC50) value of Cu(I)
NP was 0.16 μM only, as showed 1/33 of oxaliplatin (Oxa).
Furthermore, live–dead cell staining on MIAPACA-2 cells
showed that Cu(I) NP had a superior cell-killing eect compared
to CuCl, Oxa, or Cu(I) (Fig. 4 B). Notably, Cu(I) NP-treated
MIAPACA-2 cells showed signicantly reduced FDX1, LIAS, and
DLAT protein expression level, supporting an induction of
cuproptosis (Fig. 4 C). Addi tionally, confocal laser scanning
microscopy (CLSM) revealed pronounced DLAT aggregation in
Cu(I) NP-treated cells compared to phosphate-buered saline
(PBS) controls (Fig. 4 D). Next, they demonstrated that Cu(I) NP
could induce immunogenic cell death (ICD). Cu(I) NP-treated
cells showed 2.4- and 2.8-fold higher CRT translocation than
those treated with CuCl and PBS, respectively (Fig. 4 E and F).
Adenosine triphosphate (ATP) release analysis showed that Cu(I)
NP-treated cells released 41.4 nM of ATP, signicantly higher
than that of CuCl (15.3 nM) and PBS (5.9 nM) (Fig. 4 G). In a
subcutaneous PANC-02 model mice model, Cy7.5-labeled Cu(I)
NP (Cy7.5@Cu(I) NP) was mainly accumulated in the liver and
tumor tissue aer intravenous injection (Fig. 4 H). Furthermore,
Cu(I) NP exhibited eectively reduced tumor growth compared
to other treatments (Fig. 4 I). Cu(I) NP-treated mice showed an
increased proportion of mature dendritic cells (CD80+ CD86+)
in tumor and spleen tissues, indicating enhanced antigen pre-
sentation and dendritic cell maturation (Fig. 4 J). Furthermore,
CD8
+
T cell inltration was 1.5 times higher in Cu(I) NP-treated
tumors than those treated with Oxa, showing enhanced antitu-
mor immunity (Fig. 4 K). Finally, Cu(I) NP elicited higher trans-
formation of the M2 phenotype of macrophages into the M1
phenotype in tumor tissue, indicating reprogramming of the
immunosuppressive tumor microenvironment (Fig. 4 L). ese
studies showed that specic activation of copper-based nano-
medicines in tumor microenvironment could enhance anticancer
eects by cuproptosis.
Li et al. [ 181 ] developed a bifunctional CuP/Er nanoparticle
that combines copper ions and peroxide in the core with erastin
(Er) on the shell, designed to synergize the eects of cupropto-
sis and ferroptosis for enhanced cancer treatment. e CuP/Er
nanoparticles sensitized 4T1 breast tumor cells to cuproptosis
by disrupting their reliance on aerobic glycolysis and inhibiting
the TCA cycle, leading to the oligomerization of lipoylated TCA
proteins in mitochondria. Simultaneously, CuP/Er promoted
ferroptosis by increasing ROS production and creating an intra-
cellular redox imbalance. is dual action depleted glutathione
(GSH), enhanced lipid peroxidation, and caused severe mito-
chondrial damage, resulting in signicant tumor growth inhibi-
tion in mouse models of 4T1 breast and MC38 colon cancer.
Interestingly, CuP/Er induced immunogenic cell death, improv-
ing antigen presentation and up-regulating PD-L1 expression
in tumor cells. e combination of CuP/Er with a αPD-L1
showed potent tumor regression and prevented metastasis by
synergizing T cell proliferation and reinvigoration. As another
combination approach of cuproptosis and ferroptosis, Chen
et al. developed a thermosensitive liposomal bimetallic Fe-Cu
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 13
MOFs (Lip@Fe-Cu-MOFs, named MetaCell) to simultaneously
activate cuproptosis and ferroptosis by laser irradiation [ 182 ].
Fe-Cu-MOFs were rst synthesized using iron and copper
metal ions via coordination bonding with organic ligands.
en, Fe-Cu-MOFs were encapsulated into thermosensitive
liposomes, followed by delivering them into live neutrophils,
resulting in MetaCell (Fig. 5 A). MetaCell could inltrate the
tumors as well as respond to inammation, releasing Fe-Cu-
MOFs and inducing photo-triggered cuproptosis with ferrop-
tosis (Fig. 5 B). e C6-labeled Lip@Fe-Cu-MOFs showed
intracellular localization in MetaCell that they were rapidly
uptaken by neutrophils at 2 h posttreatment and distributed
throughout the cytoplasm (Fig. 5 C). MetaCell combined with
phorbol-12-myristate-13-acetate, which a compound for induc-
ing oxidative stress and inammation, demonstrated a strong
anticancer eect under near-infrared (NIR) irradiation (Fig.
5 D). Moreover, MetaCell led disruption of intracellular metal
homeostasis aer laser irradiation, decreasing in FDX1 and
LIAS levels and inducing cell death by cuproptosis (Fig. 5 E).
Aer intravenous injection into 4T1 orthotopic breast tumor
model, DiR-labeled MetaCell showed superior tumor-targeting
ability compared to DiR-labeled Lip@Fe-Cu-MOFs for 7 d,
resulting in exhibiting 1.55-fold higher uorescence intensity
than DiR-labeled Lip@Fe-Cu-MOFs (Fig. 5 F and G). Conse-
quently, MetaCell (2 × 107 cells per mouse) showed eective
tumor ablation eects, aer exposure to NIR laser irradiation
(808 nm, 1.5 W/cm2) for 5 min, 24 h postinjection. Notably,
MetaCell showed higher antitumor eects compared to Lip@
Fe-Cu-MOFs, indicating enhanced antitumor eects by neu-
trophil. Furthermore, DLAT levels increased while GPX4 levels
Fig.4.Stimulus-responsive copper complex nanoparticles as inducers of cuproptosis for enhanced cancer immunotherapy. (A) Schematic illustration of Cu(I) NP preparation
and mechanism for cuproptosis-induced immunotherapy. (B) CLSM images of live/dead staining in MIAPACA-2 cells following different treatments. Scale bars: 50 μm.
(C) Western blot analysis of LIAS, FDX1, and DLAT expression levels in MIAPACA-2 cells. (D) CLSM images showing DLAT aggregation in MIAPACA-2 cells following Cu(I) NP
treatment. Scale bars: 20 μm. (E) Flow cytometry analysis of CRT exposure under different treatments and (F) corresponding quantification. (G) Quantitative analysis of ATP
levels in the supernatant of MIAPACA-2 cells under various treatments. (H) Invivo biodistribution of Cy7.5@Cu(I) NP by IVIS and exvivo imaging of tumors and major organs
48 h postadministration. (I) Tumor growth inhibition curve in mice following various treatments. (J) Flow cytometry results of CD80+ CD86+ dendritic cells gated on CD11c+
in tumors. (K) Flow cytometry results of CD8+ and CD4+ T cells gated on CD3+ cells in tumors. (L) Flow cytometry profiles of M1 and M2 macrophage phenotypes at tumor
sites under various treatments. Reproduced from [180] with permission from Wiley-VCH, Copyright 2024.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 14
decreased, indicating that the treatment exerted its antitumor
eect through a synergistic cuproptosis-ferroptosis mechanism
by Lip@Fe-Cu-MOFs (Fig. 5 H).
Although various combination strategies of cuproptosis-
targeted nanomedicines showed enhanced therapeutic ecacy
in various cancer treatments, these studies collectively suggest
further researches are needed in relation to the combination of
dierent regimes and cell death mechanisms with copper strat-
egies for further optimizing therapeutic ecacy.
Conclusions and Future Perspectives
Cuproptosis is one of the programmed cell deaths triggered by
the accumulation of copper ions in the cells. is concept has
gained attention in cancer therapy as a potential strategy for
enhancing treatment ecacy. Since cuproptosis was introduced
in 2022, numerous research studies have reported its cellular
regulatory mechanisms and signaling pathways. Furthermore,
numerous studies have demonstrated the close relationship
Fig.5.Lip@Fe-Cu-MOFs-internalized neutrophil (MetaCell)-mediated synergistic cuproptosis and ferroptosis-based treatment of malignancies. (A) Design and preparation of
MetaCell. (B) Schematic illustration of synergistic anticancer effects of cuproptosis and ferroptosis induced by MetaCell. (C) CLSM images of MetaCell after incubation with
Lip@Fe-Cu-MOFs. Scale bars: 10 μm. (D) Invitro anticancer effects of MetaCell on 4T1 cells under various treatments. (E) Western blot analysis of DLAT, LIAS, FDX1, and GPX4
expression levels in 4T1 cells, wherein (1) was treated with neutrophils or (2) MetaCell with phorbol-12-myristate-13-acetate (PMA) and NIR irradiation. (F) Invivo tumor targeting
ability of MetaCell targeting in 4T1 tumor-bearing. (G) Biodistribution of DiR-labeled Lip@Fe-Cu-MOFs and MetaCell in tumors and major organs on 7 d postadministration.
(H) Representative tumor tissue section images stained with hematoxylin and eosin, ROS, GPX4, and DLAT after treatment. Scale bars: 100 μm. Reproduced from [182] with
permission from the American Association for the Advancement of Science, Copyright 2024.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 15
between cancer and copper, as disrupted copper homeostasis
is frequently observed in various malignant cancers, wherein
dysregulated copper homeostasis can drive cancer metastasis,
immune evasion, angiogenesis, and drug resistance. us, har
-
nessing cuproptosis may provide novel strategies for cancer
treatment to overcome resistance by apoptosis and avoid the
risk of necrosis-related inammation. is review comprehen-
sively introduces dysregulated copper homeostasis in cancer
and cuproptosis-mediated therapeutic strategies in nanomedi-
cines to provide insights into novel designs and therapeutic
applications.
Despite the therapeutic potential of cuproptosis-based
nanomedicines for cancer treatment, several limitations remain
to be addressed through in-depth studies. Firstly, the mecha-
nism of cuproptosis is still being explored, requiring the exact
mechanism with associated molecular pathways. Moreover,
limited specic biomarkers in pathological conditions that can
evaluate therapeutic ecacy in cancer treatment may hinder
applications of cuproptosis-based nanomedicines in the clinic.
e complicated and compensated cuproptosis mechanism can
provide novel designs of nanomedicines and therapeutic strate-
gies in combination with other regimes. Secondly, establishing
the eective range of intracellular copper ion concentration for
cuproptosis may closely relate to eliciting therapeutic ecacy
and reducing adverse side eects. Because cuproptosis based
on intracellular copper metabolism, it can induce cytotoxicity
in both cancer cells and normal cells, resulting in acting as a
double-edged sword. us, control of intracellular copper ion
concentration should be carefully optimized. irdly, nano-
medicines designed for copper ion delivery to cancer cells are
carefully considered for inducing the physicochemical proper-
ties of copper. For example, copper/copper oxide nanoparticles
could provide copper ions in the cells, but copper ions induced
a Fenton reaction using hydrogen peroxide, generating a cyto-
toxic hydroxyl radical. Furthermore, the intracellular turnover
mechanism of Cu2+ to Cu+ in target cancer cells should be
considered for an eective cuproptosis based on DLAT aggre-
gation and Fe-S cluster loss caused by excessive Cu+ in cancer
cells. us, a rational design and related dose optimization are
needed for nanomedicines to induce an eective cuproptosis.
Lastly, various combination approaches using other therapeutic
regimes based on dierent cell death mechanisms can be con-
sidered to improve the anticancer ecacy of cuproptosis. In
conclusion, cuproptosis is a promising cell death mechanism,
and the development of nanomedicines that can precisely con-
trol this process may oer an eective anticancer treatment
strategy. Combined with an in-depth understanding of the
biological pathways of cuproptosis and rational designs of
copper-based nanomedicines will provide the potential for
eective cancer treatment.
Acknowledgments
Funding: is work was supported by the National Research
Foundation of Korea (NRF) grant (RS-2024-00355467) funded
by the Korea government (MSIT) and by grants from the
Intramural Research Program of the Korea Institute of Science
and Technology (KIST).
Author contributions : D.N. prepared the gure conguration
and draed the manuscript, H.L.prepared the table congura-
tion. S.L. and I.-C.S. revised the manuscript. H.Y.Y. proposed
the project and revised the manuscript. All the authors carried
out reference searching and checking. All authors read and
approved the nal manuscript.
Competing interests: e authors declare that they have no
competing interests.
Data Availability
Data sharing is not applicable to this article as no datasets were
generated or analyzed during the current study.
Supplementary Materials
Figs. S1 to S3
References
1. Ruiz LM, Libedinsky A, Elorza AA. Role of copper on
mitochondrial function and metabolism. Front Mol Biosci.
2021;8:Article 711227.
2. Steensholt G. On the eect of copper on cytochrome oxidase.
Acta Physiol Scand. 1947;14(4):335–339.
3. Xue Q, Kang R, Klionsky DJ, Tang D, Liu J, Chen X. Copper
metabolism in cell death and autophagy. Autophagy.
2023;19(8):2175–2195.
4. Lelièvre P, Sancey L, Coll J-L, Deniaud A, Busser B. e
multifaceted roles of copper in cancer: A trace metal element
with dysregulated metabolism, but also a target or a bullet for
therapy. Cancers. 2020;12(12):3594.
5. Chen L, Min J, Wang F. Copper homeostasis and cuproptosis
in health and disease. Signal Transduct Target er.
2022;7(1):378.
6. Wang X, Zhou M, Liu Y, Si Z, Si Z. Cope with copper: From
copper linked mechanisms to copper-based clinical cancer
therapies. Cancer Lett. 2023;561:216157.
7. Ge EJ, Bush AI, Casini A, Cobine PA, Cross JR, DeNicola GM,
Dou QP, Franz KJ, Gohil VM, Gupta S, et al. Connecting
copper and cancer: From transition metal signalling to
metalloplasia. Nat Rev Cancer. 2022;22(2):102–113.
8. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A,
Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R,
Spangler RD, et al. Copper induces cell death by targeting
lipoylated tca cycle proteins. Science. 2022;375(6586):
1254–1261.
9. Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X,
Shi S. Targeting cell death pathways for cancer therapy:
Recent developments in necroptosis, pyroptosis, ferroptosis,
and cuproptosis research. J Hematol Oncol. 2022;15(1):174.
10. Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D,
Gursky J, Ozdian T, Bartkova J, Turi Z, Moudry P, et al.
Alcohol-abuse drug disulram targets cancer via p97
segregase adaptor npl4. Nature. 2017;552(7684):194–199.
11. Chen D, Cui QC, Yang H, Dou QP. Disulram, a clinically
used anti-alcoholism drug and copper-binding agent, induces
apoptotic cell death in breast cancer cultures and xenogras
via inhibition of the proteasome activity. Cancer Res.
2006;66(21):10425–10433.
12. Wu W, Yu L, Jiang Q, Huo M, Lin H, Wang L, Chen Y, Shi J.
Enhanced tumor-specic disulram chemotherapy by insitu
cu2+ chelation-initiated nontoxicity-to-toxicity transition.
J Am Chem Soc. 2019;141(29):11531–11539.
13. Liu W, Xiang H, Tan M, Chen Q, Jiang Q, Yang L, Cao Y,
Wang Z, Ran H, Chen Y. Nanomedicine enables drug-
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 16
potency activation with tumor sensitivity and hyperthermia
synergy in the second near-infrared biowindow. ACS Nano.
2021;15(4):6457–6470.
14. Wu W, Yu L, Pu Y, Yao H, Chen Y, Shi J. Copper-enriched
prussian blue nanomedicine for insitu disulram toxication
and photothermal antitumor amplication. Adv Mater.
2020;32(17):2000542.
15. Voli F, Valli E, Lerra L, Kimpton K, Saletta F, Giorgi FM,
Mercatelli D, Rouaen JR, Shen S, Murray JE, et al.
Intratumoral copper modulates pd-l1 expression
and inuences tumor immune evasion. Cancer Res.
2020;80(19):4129–4144.
16. Kahlson MA, Dixon SJ. Copper-induced cell death. Science.
2022;375(6586):1231–1232.
17. Zhong X, Dai X, Wang Y, Wang H, Qian H, Wang X. Copper-
based nanomaterials for cancer theranostics. Wiley Interdiscip
Rev Nanomed Nanobiotechnol. 2022;14(4):Article e1797.
18. Khalaj M, Kamali M, Khodaparast Z, Jahanshahi A. Copper-
based nanomaterials for environmental decontamination–an
overview on technical and toxicological aspects. Ecotoxicol
Environ Saf. 2018;148:813–824.
19. Zhou M, Tian M, Li C. Copper-based nanomaterials
for cancer imaging and therapy. Bioconjug Chem.
2016;27(5):1188–1199.
20. Farokhzad OC, Langer R. Impact of nanotechnology on drug
delivery. ACS Nano. 2009;3(1):16–20.
21. Vincent J, Lau KS, Evyan YC-Y, Chin SX, Sillanpää M,
Chia CH. Biogenic synthesis of copper-based nanomaterials
using plant extracts and their applications: Current and future
directions. Nanomater. 2022;12(19):3312.
22. Liu T, Xiao B, Xiang F, Tan J, Chen Z, Zhang X, Wu C,
Mao Z, Luo G, Chen X, et al. Ultrasmall copper-based
nanoparticles for reactive oxygen species scavenging and
alleviation of inammation related diseases. Nat Commun.
2020;11(1):2788.
23. MC Linder. Biochemistry of copper. Berlin (Germany):
Springer Science & Business Media; 2013.
24. Grubman A, White AR. Copper as a key regulator of cell
signalling pathways. Expert Rev Mol Med. 2014;16:Article e11.
25. Chen J, Jiang Y, Shi H, Peng Y, Fan X, Li C. e molecular
mechanisms of copper metabolism and its roles in human
diseases. Pugers Arch. 2020;472(10):1415–1429.
26. Nývltová E, Dietz J, Seravalli J, Khalimonchuk O, Barrientos A.
Coordination of metal center biogenesis in human cytochrome
c oxidase. Nat Commun. 2022;13(1):3615.
27. Arredondo M, Núñez MT. Iron and copper metabolism. Mol
Aspects Med. 2005;26(4−5):313–327.
28. Storz P. Reactive oxygen species in tumor progression. Front
Biosci. 2005;10(1–3):1881–1896.
29. Szatrowski TP, Nathan CF. Production of large amounts
of hydrogen peroxide by human tumor cells. Cancer Res.
1991;51(3):794–798.
30. Babior BM. NADPH oxidase: An update. Blood.
1999;93(5):1464–1476.
31. Nakamura H, Takada K. Reactive oxygen species in
cancer: Current ndings and future directions. Cancer Sci.
2021;112(10):3945–3952.
32. Perillo B, Di Donato M, Pezone A, Di Zazzo E, Giovannelli P,
Galasso G, Castoria G, Migliaccio A. ROS in cancer therapy:
e bright side of the moon. Exp Mol Med. 2020;52(2):192–203.
33. Yoshii J, Yoshiji H, Kuriyama S, Ikenaka Y, Noguchi R,
Okuda H, Tsujinoue H, Nakatani T, Kishida H, Nakae D, et al.
e copper-chelating agent, trientine, suppresses tumor
development and angiogenesis in the murine hepatocellular
carcinoma cells. Int J Cancer. 2001;94(6):768–773.
34. Finney L, Mandava S, Ursos L, Zhang W, Rodi D, Vogt S,
Legnini D, Maser J, Ikpatt F, Olopade OI, et al. X-ray
uorescence microscopy reveals large-scale relocalization
and extracellular translocation of cellular copper during
angiogenesis. Proc Natl Acad Sci USA. 2007;104(7):2247–2252.
35. Yu Z, Cao W, Ren Y, Zhang Q, Liu J. ATPase copper
transporter A, negatively regulated by miR-148a-3p,
contributes to cisplatin resistance in breast cancer cells. Clin
Transl Med. 2020;10(1):57–73.
36. Quist DA, Diaz DE, Liu JJ, Karlin KD. Activation of
dioxygen by copper metalloproteins and insights from model
complexes. J Biol Inorg Chem. 2017;22(2−3):253–288.
37. Kaler SG. ATP7A-related copper transport diseases—
Emerging concepts and future trends. Nat Rev Neurol.
2011;7(1):15–29.
38. Galler T, Lebrun V, Raibaut L, Faller P, Wezynfeld NE.
How trimerization of CTR1 N-terminal model peptides
tunes Cu-binding and redox-chemistry. Chem Commun.
2020;56(81):12194–12197.
39. Cobine PA, Moore SA, Leary SC. Getting out what you
put in: Copper in mitochondria and its impacts on
human disease. Biochim Biophys Acta Mol Cell Res.
2021;1868(1):118867.
40. Cui L, Gouw AM, LaGory EL, Guo S, Attarwala N, Tang Y,
Qi J, Chen Y-S, Gao Z, Casey KM, et al. Mitochondrial
copper depletion suppresses triple-negative breast cancer in
mice. Nat Biotechnol. 2021;39(3):357–367.
41. Itoh S, Kim HW, Nakagawa O, Ozumi K, Lessner SM,
Aoki H, Akram K, McKinney RD, Ushio-Fukai M, Fukai T.
Novel role of antioxidant-1 (Atox1) as a copper-dependent
transcription factor involved in cell proliferation. J Biol Chem.
2008;283(14):9157–9167.
42. Harris ED. Cellular copper transport and metabolism. Annu
Rev Nutr. 2000;20:291–310.
43. Ramos D, Mar D, Ishida M, Vargas R, Gaite M, Montgomery A,
Linder MC. Mechanism of copper uptake from blood
plasma ceruloplasmin by mammalian cells. PLOS ONE.
2016;11(3):Article e0149516.
44. Weiss KC, Linder MC. Copper transport in rats involving a
new plasma protein. Am J Physiol. 1985;249(1):E77–E88.
45. Roberts EA, Sarkar B. Liver as a key organ in the supply,
storage, and excretion of copper. Am J Clin Nutr.
2008;88(3):851S–854S.
46. Wang X, Guo L, Zhang S, Chen Y, Chen Y-T, Zheng B, Sun J,
Qian Y, Chen Y, Yan B, et al. Copper sulde facilitates
hepatobiliary clearance of gold nanoparticles through
the copper-transporting ATPase ATP7B. ACS Nano.
2019;13(5):5720–5730.
47. Wijmenga C, Klomp LW. Molecular regulation of copper
excretion in the liver. Proc Nutr Soc. 2004;63(1):31–39.
48. Yang L, Yang P, Lip GYH, Ren J. Copper homeostasis and
cuproptosis in cardiovascular disease therapeutics. Trends
Pharmacol Sci. 2023;44(9):573–585.
49. Tsang T, Davis CI, Brady DC. Copper biology. Curr Biol.
2021;31(9):R421–R427.
50. González-Guerrero M, Argüello JM. Mechanism of cu+-
transporting ATPases: Soluble Cu+ chaperones directly
transfer Cu+ to transmembrane transport sites. Proc Natl
Acad Sci USA. 2008;105(16):5992–5997.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 17
51. O'Halloran TV, Culotta VC. Metallochaperones, an
intracellular shuttle service for metal ions. J Biol Chem.
2000;275(33):25057–25060.
52. Banci L, Bertini I, Cantini F, Cio-Baoni S. Cellular copper
distribution: A mechanistic systems biology approach. Cell
Mol Life Sci. 2010;67(15):2563–2589.
53. Borobia M, Villanueva-Saz S, Ruiz M, de Arcaute A,
Fernández MTV, González JM, Navarro T, Benito AA,
Arnal JL, De Las Heras M, et al. Copper poisoning, a deadly
hazard for sheep. Animals. 2022;12(18):2388.
54. Stelmashook EV, Alexandrova OP, Genrikhs EE, Novikova SV,
Salmina AB, Isaev NK. Eect of zinc and copper ions on
cadmium-induced toxicity in rat cultured cortical neurons.
J Trace Elem Med Biol. 2022;73:Article 127012.
55. Chen X, Cai Q, Liang R, Zhang D, Liu X, Zhang M, Xiong Y,
Xu M, Liu Q, Li P, et al. Copper homeostasis and copper-
induced cell death in the pathogenesis of cardiovascular
disease and therapeutic strategies. Cell Death Dis.
2023;14(2):105.
56. Tang X, Yan Z, Miao Y, Ha W, Li Z, Yang L, Mi D. Copper in
cancer: From limiting nutrient to therapeutic target. Front
Oncol. 2023;13:1209156.
57. Baszuk P, Marciniak W, Derkacz R, Jakubowska A,
Cybulski C, Gronwald J, Dębniak T, Huzarski T, Białkowska K,
Pietrzak S, et al. Blood copper levels and the occurrence of
colorectal cancer in Poland. Biomedicines. 2021;9(11):1628.
58. Liao Y, Zhao J, Bulek K, Tang F, Chen X, Cai G, Jia S, Fox PL,
Huang E, Pizarro TT, et al. Inammation mobilizes copper
metabolism to promote colon tumorigenesis via an IL-17-
STEAP4-XIAP axis. Nat Commun. 2020;11:900.
59. Nayak SB, Bhat VR, Upadhyay D, Udupa SL. Copper and
ceruloplasmin status in serum of prostate and colon cancer
patients. Indian J Physiol Pharmacol. 2003;47(1):108–110.
60. Blockhuys S, Celauro E, Hildesjö C, Feizi A, Stål O,
Fierro-González J, Wittung-Stafshede P. Dening the human
copper proteome and analysis of its expression variation in
cancers. Metallomics. 2017;9(2):112–123.
61. Solomon EI, Sundaram UM, Machonkin TE. Multicopper
oxidases and oxygenases. Chem Rev. 1996;96(7):
2563–2606.
62. Que EL, Domaille DW, Chang CJ. Metals in neurobiology:
Probing their chemistry and biology with molecular imaging.
Chem Rev. 2008;108(5):1517–1549.
63. Guan D, Zhao L, Shi X, Ma X, Chen Z. Copper in cancer:
From pathogenesis to therapy. Biomed Pharmacother.
2023;163:Article 114791.
64. Stepien M, Jenab M, Freisling H, Becker N-P, Czuban M,
Tjønneland A, Olsen A, Overvad K, Boutron-Ruault M-C,
Mancini FR, et al. Pre-diagnostic copper and zinc biomarkers
and colorectal cancer risk in the european prospective
investigation into cancer and nutrition cohort. Carcinogenesis.
2017;38(7):699–707.
65. Basu S, Singh MK, Singh TB, Bhartiya SK, Singh SP,
Shukla VK. Heavy and trace metals in carcinoma of the
gallbladder. World J Surg. 2013;37(11):2641–2646.
66. Baltaci AK, Dundar TK, Aksoy F, Mogulkoc R. Changes
in the serum levels of trace elements before and aer the
operation in thyroid cancer patients. Biol Trace Elem Res.
2017;175(1):57–64.
67. Li Y, Liang J, Chen Y, Wang Y. e mechanism of
copper homeostasis and its role in disease. iLABMED.
2023;1(2):109–120.
68. Finney L, Vogt S, Fukai T, Glesne D. Copper and
angiogenesis: Unravelling a relationship key to cancer
progression. Clin Exp Pharmacol Physiol. 2009;36(1):88–94.
69. Li J, Zhang YP, Kirsner RS. Angiogenesis in wound repair:
Angiogenic growth factors and the extracellular matrix.
Microsc Res Tech. 2003;60(1):107–114.
70. Ribatti D, Nico B, Floris C, Mangieri D, Piras F, Ennas MG,
Vacca A, Sirigu P. Microvascular density, vascular endothelial
growth factor immunoreactivity in tumor cells, vessel
diameter and intussusceptive microvascular growth in
primary melanoma. Oncol Rep. 2005;14(1):81–84.
71. Nico B, Crivellato E, Guidolin D, Annese T, Longo V,
Finato N, Vacca A, Ribatti D. Intussusceptive microvascular
growth in human glioma. Clin Exp Med. 2010;10(2):93–98.
72. Patan S, Munn LL, Jain RK. Intussusceptive microvascular
growth in a human colon adenocarcinoma xenogra: A
novel mechanism of tumor angiogenesis. Microvasc Res.
1996;51(2):260–272.
73. Djonov V, Högger K, Sedlacek R, Laissue J, Draeger A.
MMP-19: Cellular localization of a novel metalloproteinase
within normal breast tissue and mammary gland tumours.
J Pathol. 2001;195(2):147–155.
74. Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X,
Cao K, Deng H, He Y, et al. e role of microenvironment in
tumor angiogenesis. J Exp Clin Cancer Res. 2020;39:204.
75. Folkman J. Role of angiogenesis in tumor growth and
metastasis. Semin Oncol. 2002;29(6 Suppl 16):15–18.
76. Nasulewicz A, Mazur A, Opolski A. Role of copper in tumour
angiogenesis—Clinical implications. J Trace Elem Med Biol.
2004;18(1):1–8.
77. Gérard C, Bordeleau L-J, Barralet J, Doillon CJ. e
stimulation of angiogenesis and collagen deposition by
copper. Biomaterials. 2010;31(5):824–831.
78. Hu GF. Copper stimulates proliferation of human endothelial
cells under culture. J Cell Biochem. 1998;69(3):326–335.
79. Soncin F, Guitton J-D, Cartwright T, Badet J. Interaction
of human angiogenin with copper modulates angiogenin
binding to endothelial cells. Biochem Biophys Res Commun.
1997;236(3):604–610.
80. Lowndes SA, Harris AL. e role of copper in tumour
angiogenesis. J Mammary Gland Biol Neoplasia.
2005;10(4):299–310.
81. Urso E, Maa M. Behind the link between copper and
angiogenesis: Established mechanisms and an overview on
the role of vascular copper transport systems. J Vasc Res.
2016;52(3):172–196.
82. Li Y. Copper homeostasis: Emerging target for cancer
treatment. IUBMB Life. 2020;72(9):1900–1908.
83. Juarez JC, Betancourt O Jr, Pirie-Shepherd SR, Guan X,
Price ML, Shaw DE, Mazar AP, Doñate F. Copper binding by
tetrathiomolybdate attenuates angiogenesis and tumor cell
proliferation through the inhibition of superoxide dismutase 1.
Clin Cancer Res. 2006;12(16):4974–4982.
84. Qiu L, Ding X, Zhang Z, Kang YJ. Copper is required for
cobalt-induced transcriptional activity of hypoxia-inducible
factor-1. J Pharmacol Exp er. 2012;342(2):561–567.
85. Denoyer D, Masaldan S, La Fontaine S, Cater MA. Targeting
copper in cancer therapy:‘copper that cancer’. Metallomics.
2015;7(11):1459–1476.
86. Park KC, Fouani L, Jansson PJ, Wooi D, Sahni S, Lane DJ,
Palanimuthu D, Lok HC, Kovačević Z, Huang ML, et al.
Copper and conquer: Copper complexes of di-2-
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 18
pyridylketone thiosemicarbazones as novel anti-cancer
therapeutics. Metallomics. 2016;8(9):874–886.
87. Ungar-Waron H, Gluckman A, Spira E, Waron M, Trainin Z.
Ceruloplasmin as a marker of neoplastic activity in rabbits
bearing the vx-2 carcinoma. Cancer Res. 1978;38(5):
1296–1299.
88. Badet J, Soncin F, Guitton J-D, Lamare O, Cartwright T,
Barritault D. Specic binding of angiogenin to calf
pulmonary artery endothelial cells. Proc Natl Acad Sci USA.
1989;86(21):8427–8431.
89. Das A, Ash D, Fouda AY, Sudhahar V, Kim Y-M, Hou Y,
Hudson FZ, Stanseld BK, Caldwell RB, McMenamin M,
et al. Cysteine oxidation of copper transporter ctr1 drives
VEGFR2 signalling and angiogenesis. Nat Cell Biol.
2022;24(1):35–50.
90. Konieczkowski DJ, Johannessen CM, Garraway LA. A
convergence-based framework for cancer drug resistance.
Cancer Cell. 2018;33(5):801–815.
91. Reygaert WC. An overview of the antimicrobial resistance
mechanisms of bacteria. AIMS Microbiol. 2018;4(3):482–501.
92. Longley D, Johnston P. Molecular mechanisms of drug
resistance. J Pathol. 2005;205(2):275–292.
93. Ponte-Sucre A, Gamarro F, Dujardin J-C, Barrett MP,
López-Vélez R, García-Hernández R, Pountain AW,
Mwenechanya R, Papadopoulou B. Drug resistance and
treatment failure in leishmaniasis: A 21st century challenge.
PLOS Negl Trop Dis. 2017;11(12):Article e0006052.
94. Furukawa T, Komatsu M, Ikeda R, Tsujikawa K,
Akiyama S-i. Copper transport systems are involved in
multidrug resistance and drug transport. Curr Med Chem.
2008;15(30):3268–3278.
95. Majumder S, Chatterjee S, Pal S, Biswas J, Eerth T,
Choudhuri SK. e role of copper in drug-resistant murine
and human tumors. Biometals. 2009;22:377–384.
96. Jin J, Ma M, Shi S, Wang J, Xiao P, Yu H-F, Zhang C, Guo Q,
Yu Z, Lou Z, et al. Copper enhances genotoxic drug resistance
via ATOX1 activated DNA damage repair. Cancer Lett.
2022;536:Article 215651.
97. Srinivasan P, Wu X, Basu M, Rossi C, Sandler AD. PD-L1
checkpoint inhibition and anti-CTLA-4 whole tumor cell
vaccination counter adaptive immune resistance: A mouse
neuroblastoma model that mimics human disease. PLOS
Med. 2018;15(1):Article e1002497.
98. Percival SS. Copper and immunity. Am J Clin Nutr. 1998;67
(5 Suppl):1064S–1068S.
99. Zhou B, Guo L, Zhang B, Liu S, Zhang K, Yan J, Zhang W,
Yu M, Chen Z, Xu Y, et al. Disulram combined with copper
induces immunosuppression via pd-l1 stabilization in
hepatocellular carcinoma. Am J Cancer Res. 2019;9(11):
2442–2455.
100. Voli F, Lerra L, Kimpton K, Saletta F, Cirillo G, Kavallaris M,
Vittorio O. Harnessing copper in cancer to enhance anti-
tumor immune response. Ann Oncol. 2018;29(Suppl 10):
Article x35.
101. Ackermann CJ, Adderley H, Ortega-Franco A, Khan A,
Reck M, Califano R. First-line immune checkpoint inhibition
for advanced non-small-cell lung cancer: State of the art and
future directions. Drugs. 2020;80:1783–1797.
102. Chaer CL, Weinberg RA. A perspective on cancer cell
metastasis. Science. 2011;331(6024):1559–1564.
103. Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ,
del Barrio MG, Portillo F, Nieto MA. e transcription factor
snail controls epithelial–mesenchymal transitions by repressing
e-cadherin expression. Nat Cell Biol. 2000;2(2):
76–83.
104. Mani SA, Yang J, Brooks M, Schwaninger G, Zhou A,
Miura N, Kutok JL, Hartwell K, Richardson AL, Weinberg RA.
Mesenchyme forkhead 1 (FOXC2) plays a key role in
metastasis and is associated with aggressive basal-like breast
cancers. Proc Natl Acad Sci USA. 2007;104(24):
10069–10074.
105. Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA,
Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA.
Twist, a master regulator of morphogenesis, plays an essential
role in tumor metastasis. Cell. 2004;117(7):927–939.
106. Shanbhag V, Jasmer-McDonald K, Zhu S, Martin AL,
Gudekar N, Khan A, Ladomersky E, Singh K, Weisman GA,
Petris MJ. ATP7A delivers copper to the lysyl oxidase family
of enzymes and promotes tumorigenesis and metastasis. Proc
Natl Acad Sci USA. 2019;116(14):6836–6841.
107. Kamiya T. Copper biology in health and disease: Copper in
the tumor microenvironment and tumor metastasis. J Clin
Biochem Nutr. 2022;71(1):22–28.
108. O'Leary BR, Fath MA, Bellizzi AM, Hrabe JE, Button AM,
Allen BG, Case AJ, Altekruse S, Wagner BA, Buettner GR, et al.
Loss of SOD3 (ecSOD) expression promotes an aggressive
phenotype in human pancreatic ductal adenocarcinoma.
Clin Cancer Res. 2015;21(7):1741–1751.
109. Laukkanen MO. Extracellular superoxide dismutase: Growth
promoter or tumor suppressor? Oxid Med Cell Longev.
2016;2016:3612589.
110. Maller O, Drain AP, Barrett AS, Borgquist S, Ruell B,
Zakharevich I, Pham TT, Gruosso T, Kuasne H, Lakins JN,
et al. Tumour-associated macrophages drive stromal cell-
dependent collagen crosslinking and stiening to promote
breast cancer aggression. Nat Mater. 2021;20(4):548–559.
111. Xiao Q, Ge G. Lysyl oxidase, extracellular matrix remodeling
and cancer metastasis. Cancer Microenviron. 2012;5(3):
261–273.
112. Erler JT, Bennewith KL, Nicolau M, Dornhöfer N, Kong C,
Le Q-T, Chi J-TA, Jerey SS, Giaccia AJ. Lysyl oxidase
is essential for hypoxia-induced metastasis. Nature.
2006;440(7088):1222–1226.
113. Kirschmann DA, Seor EA, Fong SF, Nieva DR, Sullivan CM,
Edwards EM, Sommer P, Csiszar K, Hendrix MJ. A molecular
role for lysyl oxidase in breast cancer invasion. Cancer Res.
2002;62(15):4478–4483.
114. Baker A-M, Bird D, Lang G, Cox TR, Erler JT. Lysyl oxidase
enzymatic function increases stiness to drive colorectal
cancer progression through fak. Oncogene. 2013;32(14):
1863–1868.
115. Baker A-M, Cox TR, Bird D, Lang G, Murray GI, Sun X-F,
Southall SM, Wilson JR, Erler JT. e role of lysyl oxidase
in SRC-dependent proliferation and metastasis of colorectal
cance r. J Natl Cancer Inst. 2011;103(5):407–424.
116. Lapointe J, Li C, Higgins JP, Van De Rijn M, Bair E,
Montgomery K, Ferrari M, Egevad L, Rayford W,
Bergerheim U, et al. Gene expression proling identies
clinically relevant subtypes of prostate cancer. Proc Natl Acad
Sci USA. 2004;101(3):811–816.
117. MacDonald G, Nalvarte I, Smirnova T, Vecchi M, Aceto N,
Doelemeyer A, Frei A, Lienhard S, Wycko J, Hess D, et al.
Memo is a copper-dependent redox protein with an essential
role in migration and metastasis. Sci Signal. 2014;7(329):ra56.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 19
118. Marone R, Hess D, Dankort D, Muller WJ, Hynes NE,
Badache A. Memo mediates ErbB2-driven cell motility. Nat
Cell Biol. 2004;6(6):515–522.
119. Johnston KA, Lopez KM. Lysyl oxidase in cancer inhibition
and metastasis. Cancer Lett. 2018;417:174–181.
120. Cox TR, Gartland A, Erler JT. Lysyl oxidase, a targetable
secreted molecule involved in cancer metastasis. Cancer Res.
2016;76(2):188–192.
121. Qu Y, Wang J, Sim M-S, Liu B, Giuliano A, Barsoum J, Cui X.
Elesclomol, counteracted by Akt survival signaling, enhances
the apoptotic eect of chemotherapy drugs in breast cancer
cells. Breast Cancer Res Treat. 2010;121(2):311–321.
122. Crowley LC, Waterhouse NJ. Detecting cleaved caspase-3 in
apoptotic cells by ow cytometry. Cold Spring Harb Protoc.
2016;(11):prot087312.
123. Nagai M, Vo NH, Ogawa LS, Chimmanamada D, Inoue T,
Chu J, Beaudette-Zlatanova BC, Lu R, Blackman RK,
Barsoum J, et al. e oncology drug elesclomol selectively
transports copper to the mitochondria to induce oxidative
stress in cancer cells. Free Radic Biol Med. 2012;52(10):
2142–2150.
124. Rakshit A, Khatua K, Shanbhag V, Comba P, Datta A. Cu2+
selective chelators relieve copper-induced oxidative stress
invivo. Chem Sci. 2018;9(41):7916–7930.
125. Sepand M-R, Aliomrani M, Hasani-Nourian Y, Khalhori M-R,
Farzaei M-H, Sanadgol N. Mechanisms and pathogenesis
underlying environmental chemical-induced necroptosis.
Environ Sci Pollut Res Int. 2020;27(30):37488–37501.
126. Chen J, Zhang R, Tao C, Huang X, Chen Z, Li X, Zhou J,
Zeng Q, Zhao B, Yuan M, et al. CuS–NiS2 nanomaterials
for MRI guided phototherapy of gastric carcinoma
via triggering mitochondria-mediated apoptosis and
MLKL/CAPG-mediated necroptosis. Nanotoxicology.
2020;14(6):774–787.
127. Nirmala JG, Lopus M. Cell death mechanisms in eukaryotes.
Cell Biol Toxicol. 2020;36(2):145–164.
128. Dong J, Wang X, Xu C, Gao M, Wang S, Zhang J, Tong H,
Wang L, Han Y, Cheng N, et al. Inhibiting NLRP3
inammasome activation prevents copper-induced
neuropathology in a murine model of Wilson’s disease.
Cell Death Dis. 2021;12(1):87.
129. Tang D, Chen X, Kroemer G. Cuproptosis: A copper-
triggered modality of mitochondrial cell death. Cell Res.
2022;32(5):417–418.
130. Tsvetkov P, Detappe A, Cai K, Keys HR, Brune Z, Ying W,
iru P, Reidy M, Kugener G, Rossen J, et al. Mitochondrial
metabolism promotes adaptation to proteotoxic stress.
Nat Chem Biol. 2019;15(7):681–689.
131. Li S-R, Bu L-L, Cai L. Cuproptosis: Lipoylated TCA cycle
proteins-mediated novel cell death pathway. Signal Transduct
Target er. 2022;7(1):158.
132. Solmonson A, DeBerardinis RJ. Lipoic acid metabolism
and mitochondrial redox regulation. J Biol Chem.
2018;293(20):7522–7530.
133. Skrott Z, Majera D, Gursky J, Buchtova T, Hajduch M,
Mistrik M, Bartek J. Disulram’s anti-cancer activity reects
targeting npl4, not inhibition of aldehyde dehydrogenase.
Oncogene. 2019;38(40):6711–6722.
134. Pan M, Zheng Q, Yu Y, Ai H, Xie Y, Zeng X, Wang C, Liu L,
Zhao M. Seesaw conformations of npl4 in the human p97
complex and the inhibitory mechanism of a disulram
derivative. Nat Commun. 2021;12(1):121.
135. Dreishpoon M, Bick N, Petrova B, Warui DM, Cameron A,
Booker SJ, Kanarek N, Golub T, Tsvetkov P. FDX1 regulates
cellular protein lipoylation through direct binding to LIAS.
bioRxiv. 2023. https://doi.org/10.1101/2023.02.03.526472
136. Steinbrueck A, Sedgwick AC, Brewster JT, Yan K-C, Shang Y,
Knoll DM, Vargas-Zúñiga GI, He X-P, Tian H, Sessler JL.
Transition metal chelators, pro-chelators, and ionophores as
small molecule cancer chemotherapeutic agents. Chem Soc
Rev. 2020;49(12):3726–3747.
137. Garza NM, Swaminathan AB, Maremanda KP, Zulkii M,
Gohil VM. Mitochondrial copper in human genetic
disorders. Trends Endocrinol Metab. 2023;34(1):21–33.
138. Oliveri V. Selective targeting of cancer cells by copper
ionophores: An overview. Front Mol Biosci. 2022;9:Article 841814.
139. Hersey P, Bastholt L, Chiarion-Sileni V, Cinat G, Dummer R,
Eggermont A, Espinosa E, Hauschild A, Quirt I, Robert C,
et al. Small molecules and targeted therapies in distant
metastatic disease. Ann Oncol. 2009;20(Suppl 6):vi35–vi40.
140. Guo B, Yang F, Zhang L, Zhao Q, Wang W, Yin L, Chen D,
Wang M, Han S, Xiao H, et al. Cuproptosis induced by
ros responsive nanoparticles with elesclomol and copper
combined with αpd-l1 for enhanced cancer immunotherapy.
Adv Mater. 2023;35(22):2212267.
141. Lu Y, Pan Q, Gao W, Pu Y, Luo K, He B, Gu Z. Leveraging
disulram to treat cancer: Mechanisms of action,
delivery strategies, and treatment regimens. Biomaterials.
2022;281:Article 121335.
142. Lu Y, Pan Q, Gao W, Pu Y, He B. Reversal of cisplatin
chemotherapy resistance by glutathione-resistant copper-
based nanomedicine via cuproptosis. J Mater Chem B.
2022;10(33):6296–6306.
143. Zhang S, Zong Y, Chen L, Li Q, Li Z, Meng R. e
immunomodulatory function and antitumor eect of
disulram: Paving the way for novel cancer therapeutics.
Discov Oncol. 2023;14(1):103.
144. Huang P, Wang X, Liang X, Yang J, Zhang C, Kong D, Wang W.
Nano-, micro-, and macroscale drug delivery systems for cancer
immunotherapy. Acta Biomater. 2019;85:1–26.
145. Fu P, Zhang J, Li H, Mak M, Xu W, Tao Z. Extracellular
vesicles as delivery systems at nano-/micro-scale. Adv Drug
Deliv Rev. 2021;179:Article 113910.
146. Fu Q, Zhang X, Song J, Yang H. Plasmonic gold nanoagents
for cancer imaging and therapy. VIEW. 2021;2(5):20200149.
147. Woźniak-Budych MJ, Maciejewska B, Przysiecka Ł,
Wieczorek D, Staszak K, Jenczyk J, Jesionowski T, Jurga S.
Comprehensive study of stability of copper oxide
nanoparticles in complex biological media. J Mol Liq.
2020;319:Article 114086.
148. Jagaran K, Singh M. Nanomedicine for COVID-19: Potential
of copper nanoparticles. Biointerface Res Appl Chem.
2020;11(3):10716–10728.
149. Hemmati S, Ahany Kamangar S, Ahmeda A, Zangeneh MM,
Zangeneh A. Application of copper nanoparticles containing
natural compounds in the treatment of bacterial and fungal
diseases. Appl Organomet Chem. 2020;34(4):Article e5465.
150. Tyagi PK, Arya A, Mazumder AM, Tyagi S. Development
of copper nanoparticles and their prospective uses as
antioxidants, antimicrobials, anticancer agents in the
pharmaceutical sector. Precis Nanomed. 2023;6(2):1048–1065.
151. Jagaran K, Singh M. Copolymer-green-synthesized copper
oxide nanoparticles enhance folate-targeting in cervical
cancer cells invitro. Polymers. 2023;15(10):2393.
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 20
152. Zhou J, Yu Q, Song J, Li S, Li XL, Kang B, Chen HY, Xu JJ.
Photothermally triggered copper payload release for
cuproptosis-promoted cancer synergistic therapy. Angew
Chem Int Ed. 2023;62(12):Article e202213922.
153. Jia W, Tian H, Jiang J, Zhou L, Li L, Luo M, Ding N, Nice EC,
Huang C, Zhang H. Brain-targeted HFn-Cu-REGO
nanoplatform for site-specic delivery and manipulation
of autophagy and cuproptosis in glioblastoma. Small.
2023;19(2):2205354.
154. Reichel D, Sagong B, Teh J, Zhang Y, Wagner S, Wang H,
Chung LW, Butte P, Black KL, Yu JS, et al. Near infrared
uorescent nanoplatform for targeted intraoperative resection
and chemotherapeutic treatment of glioblastoma. ACS Nano.
2020;14(7):8392–8408.
155. Hersh DS, Harder BG, Roos A, Peng S, Heath JE, Legesse T,
Kim AJ, Woodworth GF, Tran NL, Winkles JA. e TNF
receptor family member Fn14 is highly expressed in recurrent
glioblastoma and in GBM patient-derived xenogras
with acquired temozolomide resistance. Neuro Oncol.
2018;20(10):1321–1330.
156. Jiang J, Zhang L, Chen H, Lei Y, Zhang T, Wang Y, Jin P, Lan J,
Zhou L, Huang Z, et al. Regorafenib induces lethal autophagy
arrest by stabilizing PSAT1 in glioblastoma. Autophagy.
2020;16(1):106–122.
157. Fan K, Jia X, Zhou M, Wang K, Jo Conde J, He JT, Yan X.
Ferritin nanocarrier traverses the blood brain barrier and
kills glioma. ACS Nano. 2018;12(5):4105–4115.
158. Zhen Z, Tang W, Chen H, Lin X, Todd T, Wang G, Cowger T,
Chen X, Xie J. Rgd-modied apoferritin nanoparticles for
ecient drug delivery to tumors. ACS Nano. 2013;7(6):
4830–4837.
159. Lu X, Gao S, Lin H, Yu L, Han Y, Zhu P, Bao W, Yao H, Chen Y,
Shi J. Bioinspired copper single-atom catalysts for tumor parallel
catalytic therapy. Adv Mater. 2020;32(36):2002246.
160. Goto N, Hara H, Kondo M, Yasuda N, Kamiya T, Okuda K,
Adachi T. Hydrogen sulde increases copper-dependent
neurotoxicity via intracellular copper accumulation.
Metallomics. 2020;12(6):868–875.
161. Ren M, Xu Q, Bai Y, Wang S, Kong F. Construction of a
dual-response uorescent probe for copper (II) ions and
hydrogen sulde (H2S) detection in cells and its application
in exploring the increased copper-dependent cytotoxicity
in present of H2S. Spectrochim Acta A Mol Biomol Spectrosc.
2021;249:Article 119299.
162. Setty SRG, Tenza D, Sviderskaya EV, Bennett DC, Raposo G,
Marks MS. Cell-specic ATP7A transport sustains copper-
dependent tyrosinase activity in melanosomes. Nature.
2008;454(7208):1142–1146.
163. Mendelsohn BA, Yin C, Johnson SL, Wilm TP,
Solnica-Krezel L, Gitlin JD. Atp7a determines a hierarchy of
copper metabolism essential for notochord development.
Cell Metab. 2006;4(2):155–162.
164. Ascenção K, Szabo C. Emerging roles of cystathionine
β-synthase in various forms of cancer. Redox Biol.
2022;53:Article 102331.
165. Phillips CNM, Zatarain JR, Nicholls ME, Porter C, Widen SG,
anki K, Johnson P, Jawad MU, Moyer MP, Randall JW, et al.
Upregulation of cystathionine-β-synthase in colonic epithelia
reprograms metabolism and promotes carcinogenesis. Cancer
Res. 2017;77(21):5741–5754.
166. Szabo C, Coletta C, Chao C, Módis K, Szczesny B,
Papapetropoulos A, Hellmich MR. Tumor-derived hydrogen
sulde, produced by cystathionine-β-synthase, stimulates
bioenergetics, cell proliferation, and angiogenesis in colon
cance r. Proc Natl Acad Sci USA. 2013;110(30):12474–12479.
167. Zhao F, Liang L, Wang H, Wang C, Su D, Ying Y, Li W, Li J,
Zheng J, Qiao L, et al. H2S-activated ion-interference therapy:
A novel tumor targeted therapy based on copper-overload-
mediated cuproptosis and pyroptosis. Adv Funct Mater.
2023;33(38):2300941.
168. Wu D, Wang S, Yu G, Chen X. Cell death mediated by the
pyroptosis pathway with the aid of nanotechnology: Prospects
for cancer therapy. Angew Chem. 2021;133(15):8096–8112.
169. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y,
Cai T, Wang F, Shao F. Cleavage of gsdmd by inammatory
caspases determines pyroptotic cell death. Nature.
2015;526(7575):660–665.
170. Chen B, Yan Y, Yang Y, Cao G, Wang X, Wang Y, Wan F,
Yin Q, Wang Z, Li Y, et al. A pyroptosis nanotuner for cancer
therapy. Nat Nanotechnol. 2022;17(7):788–798.
171. Shao F. Gasdermins: Making pores for pyroptosis. Nat Rev
Immunol. 2021;21(10):620–621.
172. Dagogo-Jack I, Shaw AT. Tumour heterogeneity and
resistance to cancer therapies. Nat Rev Clin Oncol.
2018;15(2):81–94.
173. Lim Z-F, Ma PC. Emerging insights of tumor heterogeneity
and drug resistance mechanisms in lung cancer targeted
therapy. J Hematol Oncol. 2019;12(1):134.
174. Hänggi K, Ruell B. Cell death, therapeutics, and the immune
response in cancer. Trends Cancer. 2023;9(5):381–396.
175. Wang W, Zhang L, Sun Z. Eliciting pyroptosis to fuel cancer
immunotherapy: Mechanisms and strategies. Cancer Biol
Med. 2022;19(7):948–964.
176. Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of
response, resistance, and toxicity to immune checkpoint
blockade. Cell. 2021;184(21):5309–5337.
177. Oliveri V. Biomedical applications of copper ionophores.
Coord Chem Rev. 2020;422:Article 213474.
178. Ning S, Lyu M, Zhu D, Lam JW, Huang Q, Zhang T, Tang BZ.
Type-I AIE photosensitizer loaded biomimetic system
boosting cuproptosis to inhibit breast cancer metastasis and
rechallenge. ACS Nano. 2023;17(11):10206–10217.
179. He G, Pan Y, Zeng F, Qin S, Luan X, Lu Q, Xie C, Hu P,
Gao Y, Yang J, et al. Microuidic synthesis of cuh
nanoparticles for antitumor therapy through hydrogen-
enhanced apoptosis and cuproptosis. ACS Nano.
2024;18(12):9031–9042.
180. Hu F, Huang J, Bing T, Mou W, Li D, Zhang H, Chen Y,
Jin Q, Yu Y, Yang Z. Stimulus-responsive copper complex
nanoparticles induce cuproptosis for augmented cancer
immunotherapy. Adv Sci. 2024;11(13):2309388.
181. Li Y, Liu J, Chen Y, Weichselbaum RR, Lin W. Nanoparticles
synergize ferroptosis and cuproptosis to potentiate cancer
immunotherapy. Adv Sci. 2024;11(23):2310309.
182. Chen K, Zhou A, Zhou X, He J, Xu Y, Ning X. Cellular trojan
horse initiates bimetallic fe-cu mof-mediated synergistic
cuproptosis and ferroptosis against malignancies. Sci Adv.
2024;10(15):eadk3201.
183. Xu Y, Liu SY, Zeng L, Ma H, Zhang Y, Yang H, Liu Y, Fang S,
Zhao J, Xu Y, et al. An enzyme-engineered nonporous copper
(I) coordination polymer nanoplatform for cuproptosis-based
synergistic cancer therapy. Adv Mater. 2022;34(43):2204733.
184. Xu W, Qian J, Hou G, Wang T, Wang J, Wang Y, Yang L,
Cui X, Suo A. A hollow amorphous bimetal organic
Noh et al. 2024 | https://doi.org/10.34133/bmr.0094 21
framework for synergistic cuproptosis/ferroptosis/apoptosis
anticancer therapy via disrupting intracellular redox
homeostasis and copper/iron metabolisms. Adv Funct Mater.
2022;32(40):2205013.
185. Zhong J, Zheng X, Wen Y, Wang S-B, Zhan G, Chen A-Z.
In situ sacricial growth of metastable copper-enriched
nanomedicine for cuproptosis-based synergistic cancer
therapy. Chem Eng J. 2023;474:Article 145795.
186. Huang Q-X, Liang J-L, Chen Q-W, Jin X-K, Niu M-T,
Dong C-Y, Zhang X-Z. Metal-organic framework nanoagent
induces cuproptosis for eective immunotherapy of
malignant glioblastoma. Nano Today. 2023;51:Article 101911.
187. Tian H, Duan J, Li B, Qin S, Nice EC, Zhang W, Lang T,
Zhang H, Huang C. Clinical chemotherapeutic agent
coordinated copper-based nanoadjuvants for eciently
sensitizing cancer chemo-immunotherapy by cuproptosis-
mediated mitochondrial metabolic reprogramming. Adv
Funct Mater. 2023;33(51):2306584.
188. Zhang J, Peng L, Hao Y, Yang H, Zhao W, Mao C.
Biodegradable CuMoO4 nanodots with multienzyme
activities for multimodal treatment of tumor. Adv Healthc
Mater. 2023;12(22):2300167.
189. Lee SY, Seo JH, Kim S, Hwang C, Jeong DI, Park J, Yang M,
Huh JW, Cho HJ. Cuproptosis-inducible chemotherapeutic/
cascade catalytic reactor system for combating with breast
cance r. Small. 2023;19(35):2301402.
190. Zhang J, Han M, Zhang J, Abdalla M, Sun P, Yang Z,
Zhang C, Liu Y, Chen C, Jiang X. Syphilis mimetic
nanoparticles for cuproptosis-based synergistic cancer
therapy via reprogramming copper metabolism. Int J Pharm.
2023;640:Article 123025.
191. Yang Z, Zhao Z, Cheng H, Shen Y, Xie A, Zhu M. In-situ
fabrication of novel au nanoclusters-cu2+@ sodium alginate/
hyaluronic acid nanohybrid gels for cuproptosis enhanced
photothermal/photodynamic/chemodynamic therapy via
tumor microenvironment regulation. J Colloid Interface Sci.
2023;641:215–228.
192. Liang W, Han C, Zhang D, Liu C, Zhu M, Xu F, Fang C,
Zhang S, Liu C, Li Y. Copper-coordinated nanoassemblies
based on photosensitizer-chemo prodrugs and checkpoint
inhibitors for enhanced apoptosis-cuproptosis and
immunotherapy. Acta Biomater. 2024;175:
341–352.
193. Song Y, Zhan G, Zhou SF. Design of near infrared light-
powered copper phyllosilicate nanomotors for cuproptosis-
based synergistic cancer therapy. Adv Funct Mater.
2024;34(18):2314568.
194. He X, Li M, Fan S, Li Y, Fang L, Xiang G, Yang T. Copper
peroxide and cisplatin co-loaded silica nanoparticles-
based trinity strategy for cooperative cuproptosis/chemo/
chemodynamic cancer therapy. Chem Eng J. 2024;481:
Article 148522.