ArticlePublisher preview available

A Stimulus‐Responsive Ternary Heterojunction Boosting Oxidative Stress, Cuproptosis for Melanoma Therapy

Wiley
Small
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Cuproptosis, a recently discovered copper‐dependent cell death, presents significant potential for the development of copper‐based nanoparticles to induce cuproptosis in cancer therapy. Herein, a unique ternary heterojunction, denoted as HACT, composed of core–shell Au@Cu2O nanocubes with surface‐deposited Titanium Dioxide quantum dots and modified with hyaluronic acid is introduced. Compared to core–shell AC NCs, the TiO2/Au@Cu2O exhibits improved energy structure optimization, successfully separating electron‐hole pairs for redox use. This optimization results in a more rapid generation of singlet oxygen and hydroxyl radicals triggering oxidative stress under ultrasound radiation. Furthermore, the HACT NCs initiate cuproptosis by Fenton‐like reaction and acidic environment, leading to the sequential release of cupric and cuprous ions. This accumulation of copper induces the aggregation of lipoylated proteins and reduces iron‐sulfur proteins, ultimately initiating cuproptosis. More importantly, HACT NCs show a tendency to selectively target cancer cells, thereby granting them a degree of biosecurity. This report introduces a ternary heterojunction capable of triggering both cuproptosis and oxidative stress‐related combination therapy in a stimulus‐responsive manner. It can energize efforts to develop effective melanoma treatment strategies using Cu‐based nanoparticles through rational design.
This content is subject to copyright. Terms and conditions apply.
RESEARCH ARTICLE
www.small-journal.com
A Stimulus-Responsive Ternary Heterojunction Boosting
Oxidative Stress, Cuproptosis for Melanoma Therapy
Yuqi Huang, Cheng Chen, Huarong Tan, Shuqing Dong, Yiping Ren, Minghao Chao,
Hanrong Yan, Xiang Yan, Guan Jiang,* and Fenglei Gao*
Cuproptosis, a recently discovered copper-dependent cell death, presents
significant potential for the development of copper-based nanoparticles to
induce cuproptosis in cancer therapy. Herein, a unique ternary heterojunction,
denoted as HACT, composed of core–shell Au@Cu2O nanocubes with
surface-deposited Titanium Dioxide quantum dots and modified with
hyaluronic acid is introduced. Compared to core–shell AC NCs, the
TiO2/Au@Cu2O exhibits improved energy structure optimization,
successfully separating electron-hole pairs for redox use. This optimization
results in a more rapid generation of singlet oxygen and hydroxyl radicals
triggering oxidative stress under ultrasound radiation. Furthermore, the HACT
NCs initiate cuproptosis by Fenton-like reaction and acidic environment,
leading to the sequential release of cupric and cuprous ions. This
accumulation of copper induces the aggregation of lipoylated proteins and
reduces iron-sulfur proteins, ultimately initiating cuproptosis. More
importantly, HACT NCs show a tendency to selectively target cancer cells,
thereby granting them a degree of biosecurity. This report introduces a ternary
heterojunction capable of triggering both cuproptosis and oxidative
stress-related combination therapy in a stimulus-responsive manner. It can
energize efforts to develop effective melanoma treatment strategies using
Cu-based nanoparticles through rational design.
1. Introduction
Melanoma, the deadliest skin cancer, originates from
melanocytes’ malignant transformation. While advances in
biological understanding and new therapies targeting driver
genes and immune checkpoints have improved prognosis,
Y. Huang, C. Chen, H. Tan, S. Dong, Y. Ren, M. Chao, H. Yan, X. Yan,
F. Gao
Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
Xuzhou Medical University
Xuzhou, Jiangsu , P. R. China
E-mail: flgao@xzhmu.edu.cn
Y. Huang, C. Chen, H. Tan,S. Dong, Y.Ren, M. Chao, X. Yan, G. Jiang, F. Gao
Department of Dermatology
Aliated Hospital of Xuzhou Medical University
Xuzhou, Jiangsu , P. R. China
E-mail: dr.jiangguan@xzhmu.edu.cn
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/./smll.
DOI: 10.1002/smll.202401147
challenges like low response rates and re-
sistance to current therapies still hinder
further progress in melanoma treatment.[]
Therefore, it is crucial to develop ad-
vanced therapeutic strategies that can de-
liver targeted, controllable and multi-level
damage to melanoma. Since its discovery
in , various strategies have been de-
veloped to induce or enhance cupropto-
sis for cancer treatment. The creation of
nano-systems to trigger cuproptosis eec-
tively bypasses the limitations of traditional
small-molecule drugs, oering new oppor-
tunities in cancer therapy.[]Additionally,
within the cytoplasm, copper ions initiate
a Fenton-like reaction with hydrogen per-
oxide (HO), generating highly toxic hy-
droxyl radicals (OH) that aid in chemo-
dynamic therapy (CDT) for cancer treat-
ment. Compared to other metal ions in-
volved in Fenton-like reactions (such as
iron, cerium, manganese, and cobalt), cop-
per ions are particularly eective, showing
increased cellular toxicity.[]The high toxic-
ity of copper ions significantly limits their
use in biomedical applications. However,
when copper-based nanomaterials are used
in vivo, they do not circulate as free ions, thereby reducing po-
tential damage to normal cells. This eective mitigation allows
safer therapeutic applications. It is highly desirable to design a
single nanocomposite that can simultaneously induce the accu-
mulation of reactive oxygen species (ROS) and cuproptosis, e-
ciently halting the progression of malignant tumors.
The metal oxide semiconductors such as cuprous oxide (CuO)
have been widely investigated as excellent photocatalysts.[]Via
catalytic reactions facilitated by metal oxide semiconductors, elec-
trons (e) and holes (h+) are separated, subsequently engaging in
intricate interactions with HO and oxygen (O) to produce ROS
including superoxide radical (O
)and·OH.[]Nevertheless, ow-
ing to the quick eand h+recombination from the band struc-
ture, conventional semiconductors are limited by the poor quan-
tum yield of ROS.[]This inherent limitation highly hinders fur-
ther biomedical research and clinical application. Accordingly, a
multitude of strategies have been developed to prevent carrier re-
combination. These methods encompass the integration of semi-
conductors with other semiconductors or noble metals like gold
(Au), platinum (Pt), and silver (Ag).[]The mechanism employed
to overcome carrier recombination is coordinated redistribution
Small 2024,20,  ©  Wiley-VCH GmbH
2401147 (1 of 15)
... Ultrasound-triggered generation of singlet oxygen and hydroxyl radicals by NPs effectively eliminated melanoma cells [140] Gallic acid-Fe (III) and PTX-assembled NPs Formation of intracellular ROS by NPs under ultrasound treatment resulted in mitochondrial damage, lipid peroxidation, and apoptosis in melanoma cells [141] Magnetic field-responsive Chitosan biofilm containing iron oxide NPs Hyperthermic conditions generated by NPs under AMF caused a significant elimination of MNT-1 melanoma cells [142] Curcumin and magnetic NPs-loaded thermo-sensitive nanofibers Hyperthermic conditions induced by AMF significantly enhanced curcumin release, leading to a marked reduction in B16F10 cell survival [143] J Nanopart Res ...
Article
Full-text available
Melanoma is a lethal form of skin cancer with a short prognosis, frequent metastases, malignant development, and quick recurrence. Conventional treatment options have multiple shortcomings in treating melanoma, such as poor patient compliance, high cost, frequent side effects, and others. All these limitations demand new and innovative alternatives to address the rising incidence and burgeoning healthcare burden of melanoma. In recent years, stimuli-responsive nanoparticles have emerged as one such alternative. These smart nanoparticles are engineered to release drug molecules in response to specific internal or external stimuli, thereby enhancing the precision of drug delivery and improving the therapeutic outcome against malignancies like melanoma. Additionally, some of these nanoparticles are capable of generating reactive oxygen species and excess heat upon activation by specific triggers, enabling them to kill cancer cells directly. This makes stimuli-responsive nanoparticles one of the most versatile nanocarrier systems in the fight against melanoma. This review highlights various stimuli-responsive nanoparticles, their preparation, and modes of action, as well as summarizes certain research findings, emphasizing their potential applications in melanoma treatment. Finally, it ends with a quick overview of the difficulties and prospects associated with these nanoparticles. Graphical Abstract
Article
Cuproptosis, a newly identified programmed cell death form, is characterized by the excessive copper accumulation in cells, resulting in mitochondria damage and toxic protein stress, ultimately cause cell death. Given...
Article
Full-text available
Polyhexamethylene guanidine phosphate (PHMG-p) is a common biocidal disinfectant that is widely used in industry and household products. However, PHMG-p was misused as a humidifier disinfectant (HD) in South Korea, which had fatal health effects. Various health problems including cardiovascular diseases were observed in HD-exposed groups. However, the potential underlying mechanism of HD-associated cardiovascular diseases is poorly understood. Here, we examined the procoagulant activity of platelets caused by PHMG-p and clarified the underlying mechanism. PHMG-p enhanced phosphatidylserine (PS) exposure through alteration of phospholipid transporters, scramblase, and flippase. Intracellular calcium elevation, intracellular ATP depletion, and caspase-3 activation appeared to underlie phospholipid transporter dysregulation caused by PHMG-p, which was mediated by oxidative stress and mitochondrial dysfunction. Notably, antioxidant enzyme catalase and calcium chelator EGTA reversed PHMG-p-induced PS exposure and thrombin generation, confirming the contributive role of oxidative stress and intracellular calcium in the procoagulant effects of PHMG-p. These series of events led to procoagulant activation of platelets, which was revealed as enhanced thrombin generation. Collectively, PHMG-p triggered procoagulant activation of platelets, which may promote prothrombotic risks and cardiovascular diseases. These findings improve our understanding of HD-associated cardiovascular diseases.
Article
Full-text available
Sonodynamic therapy (SDT) is an anti‐cancer therapeutic strategy based on the generation of reactive oxygen species (ROS) upon local ultrasound (US) irradiation of sono‐responsive molecules or nanomaterials that accumulate in the tumor. In this work, the sonodynamic efficiency of sono‐responsive hybrid nanomaterials composed of amorphous titanium dioxide and an amphiphilic poly(ethylene oxide)‐b‐poly(propylene oxide) block copolymer is synthesized, fully characterized, and investigated both in vitro and in vivo. The modular and versatile synthetic pathway enables the control of the nanoparticle size between 30 and 300 nm (dynamic light scattering) and glucosylation of the surface for active targeting of tumors overexpressing glucose transporters. Studies on 2D and 3D rhabdomyosarcoma cell cultures reveal a statistically significant increase in the sonodynamic efficiency of glucosylated hybrid nanoparticles with respect to unmodified ones. Using a xenograft rhabdomyosarcoma murine model, it is demonstrated that by tuning the nanoparticle size and surface features, the tumor accumulation is increased by ten times compared to main off‐target clearance organs such as the liver. Finally, the SDT of rhabdomyosarcoma‐bearing mice is investigated with 50‐nm glucosylated nanoparticles. Findings evidence a dramatic prolongation of the animal survival and tumor volumes 100 times smaller than those treated only with ultrasound or nanoparticles.
Article
Full-text available
The dense stroma of desmoplastic tumor limits nanotherapeutic penetration and hampers the antitumor immune response. Here, we report a denaturation-and-penetration strategy and the use of tin monosulfide nanoparticles (SnSNPs) as nano-sonosensitizers that can overcome the stromal barrier for the management of desmoplastic triple-negative breast cancer (TNBC). SnSNPs possess a narrow bandgap (1.18 eV), allowing for efficient electron (e⁻)-hole (h⁺) pair separation to generate reactive oxygen species under US activation. More importantly, SnSNPs display mild photothermal properties that can in situ denature tumor collagen and facilitate deep penetration into the tumor mass upon near-infrared irradiation. This approach significantly enhances sonodynamic therapy (SDT) by SnSNPs and boosts antitumor immunity. In mouse models of malignant TNBC and hepatocellular carcinoma (HCC), the combination of robust SDT and enhanced cytotoxic T lymphocyte infiltration achieves remarkable anti-tumor efficacy. This study presents an innovative approach to enhance SDT and antitumor immunity using the denaturation-and-penetration strategy, offering a potential combined sono-immunotherapy approach for the cancer nanomedicine field.
Article
Full-text available
Photocatalytic reactions convert solar energy into chemical energy through a clean and green reaction process. Photocatalytic technology based on semiconductor materials provides us with a new idea in energy utilization and environmental governance. It was found that vanadium pentoxide (V2O5) has a narrow band gap, wide response range in the visible region, high oxygen density in the V2O5 lattice, high oxidation state of V5+, small energy requirement, and superior catalytic activity in partial oxidation. Therefore, the utilization rate of sunlight and photocatalytic oxidation can be greatly improved using V2O5 materials. However, the narrow band gap of V2O5 also makes it easier for the photogenerated electrons and holes to recombine in the excited state, and the stored energy is instantly consumed by carrier recombination. Therefore, how to promote the carrier separation of V2O5 and improve the photocatalytic efficiency are the key problems to be solved. Herein, several methods to improve the photocatalytic performance of V2O5 are reviewed, including metallic ion doping, non-metallic ion doping, semiconductor recombination, and noble metal deposition. Finally, it is suggested that future research directions should focus on a variety of modification methods simultaneously to promote photocatalytic efficiency and lower the cost, which will enable V2O5 to have a broad development prospect in the field of photocatalysis.
Article
Full-text available
The rapid development of nanomedicine and nanobiotechnology has allowed the emergence of various therapeutic modalities with excellent therapeutic efficiency and biosafety, among which, the sonodynamic therapy (SDT), a combination of low‐intensity ultrasound and sonosensitizers, is emerging as a promising noninvasive treatment modality for cancer treatment due to its deeper penetration, good patient compliance, and minimal damage to normal tissue. The sonosensitizers are indispensable components in the SDT process because their structure and physicochemical properties are decisive for therapeutic efficacy. Compared to the conventional and mostly studied organic sonosensitizers, inorganic sonosensitizers (noble metal‐based, transition metal‐based, carbon‐based, and silicon‐based sonosensitizers) display excellent stability, controllable morphology, and multifunctionality, which greatly expand their application in SDT. In this review, the possible mechanisms of SDT including the cavitation effect and reactive oxygen species generation are briefly discussed. Then, the recent advances in inorganic sonosensitizers are systematically summarized and their formulations and antitumor effects, particularly highlighting the strategies for optimizing the therapeutic efficiency, are outlined. The challenges and future perspectives for developing state‐of‐the‐art sonosensitizers are also discussed. It is expected that this review will shed some light on future screening of decent inorganic sonosensitizers for SDT.
Article
Full-text available
Activation of cuproptosis pathway has been reported to hold great potential in the application of tumor treatment. But the efficacy of cuproptosis seriously limited by the insufficient accumulation in the tumor sites. Therefore, herein based on the strong stabilization effects of the metalloid element selenium (Se) on copper (Cu), a photothermic Cu2‐xSe nanoparticle encapsulated with bioresponsive dimethyl maleic anhydride (DMMA@Cu2‐xSe) as a copper‐carrier to improve the copper accumulation in tumor is constructed, thus achieving cuproptosis‐driven enhancement of thermotherapy. This nanosystem exhibits the enhancement of tumor cellular uptake by a weak acid‐triggered charge‐switching ability. Next step, the exposed Cu2‐xSe is oxidized and releases divalent copper by high‐level oxide. Then, the abundant copper induces more dihydrolipoamide S‐acetyltransferase oligomerization to down‐regulate FDX1 and tricarboxylic acid cycle‐related proteins, which leads to inhibiting aerobic respiration. Cuproptosis‐induced mitochondrial damage further improves thermotherapy by up‐regulating reactive oxygen species (ROS). In addition, the generated ROS also promotes copper release to strengthen cuproptosis, and eventually improves tumor thermotherapy in turn. In general, DMMA@Cu2‐xSe with sequentially triggered copper‐release, efficient cuproptosis, and appropriate photothermal is a self‐enhanced nanoplatform for cuproptosis‐driven enhancement of thermotherapy.
Article
Full-text available
Background: Hepatocellular carcinoma (HCC) remains a challenging medical problem. Cuproptosis is a novel form of cell death that plays a crucial role in tumorigenesis, angiogenesis, and metastasis. However, it remains unclear whether cuproptosis-related genes (CRGs) influence the outcomes and immune microenvironment of HCC patients. Method: From The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases, we obtained the mRNA expression file and related clinical information of HCC patients. We selected 19 CRGs as candidate genes for this study according to previous literature. We performed a differential expression analysis of the 19 CRGs between malignant and precancerous tissue. Based on the 19 CRGs, we enrolled cluster analysis to identify cuproptosis-related subtypes of HCC patients. A prognostic risk signature was created utilizing univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses. We employed independent and stratification survival analyses to investigate the predictive value of this model. The functional enrichment features, mutation signatures, immune profile, and response to immunotherapy of HCC patients were also investigated according to the two molecular subtypes and the prognostic signature. Results: We found that 17 CRGs significantly differed in HCC versus normal samples. Cluster analysis showed two distinct molecular subtypes of cuproptosis. Cluster 1 is preferentially related to poor prognosis, high activity of immune response signaling, high mutant frequency of TP53, and distinct immune cell infiltration versus cluster 2. Through univariate and LASSO Cox regression analyses, we created a cuproptosis-related prognostic risk signature containing LIPT1, DLAT, MTF1, GLS, and CDKN2A. High-risk HCC patients were shown to have a worse prognosis. The risk signature was proved to be an independent predictor of prognosis in both the TCGA and ICGC datasets, according to multivariate analysis. The signature also performed well in different stratification of clinical features. The immune cells, which included regulatory T cells (Treg), B cells, macrophages, mast cells, NK cells, and aDCs, as well as immune functions containing cytolytic activity, MHC class I, and type II IFN response, were remarkably distinct between the high-risk and low-risk groups. The tumor immune dysfunction and exclusion (TIDE) score suggested that high-risk patients had a higher response rate to immune checkpoint inhibitors than low-risk patients. Conclusion: This research discovered the potential prognostic and immunological significance of cuproptosis in HCC, improved the understanding of cuproptosis, and may deliver new directions for developing more efficacious therapeutic techniques for HCC patients.
Article
Full-text available
Background: Acute myeloid leukemia (AML) is a highly heterogeneous cluster of hematologic malignancies. Leukemic stem cells (LSCs) are one of the culprits for the persistence and relapse of AML. The discovery of copper-induced cell death, namely cuproptosis, gives bright insights into the treatment of AML. Analogous to copper ions, long non-coding RNAs (lncRNAs) are not bystanders for AML progression, especially for LSC physiology. Uncovering the involvement of cuproptosis-related lncRNAs in AML will benefit clinical management. Methods: Detection of prognostic relevant cuproptosis-related lncRNAs are carried out by Pearson correlation analysis and univariate Cox analysis with RNA sequencing data of The Cancer Genome Atlas-Acute Myeloid Leukemia (TCGA-LAML) cohort. After the least absolute shrinkage and selection operator (LASSO) regression and the subsequent multivariate Cox analysis, a cuproptosis-related risk score (CuRS) system was derived to weigh the risk of AML patients. Thereafter, AML patients were classified into two groups by their risk property which was validated with principal component analysis (PCA), risk curves, Kaplan-Meier survival analysis, the combined receiver operating characteristic (ROC) curves, and nomogram. Variations in biological pathways and divergences in immune infiltration and immune-related processes between groups were resolved by GSEA and CIBERSORT algorism, respectively. Response to chemotherapies were scrutinized as well. The expression profiles of the candidate lncRNAs were examined by real-time quantitative polymerase chain reaction (RT-qPCR) and the specific mechanisms of lncRNA FAM30A were determined by transcriptomic analysis. Results: We fabricated an efficient prognostic signature named CuRS incorporating 4 lncRNAs (TRAF3IP2-AS1, NBR2, TP53TG1, and FAM30A) relevant to immune environment and chemotherapy responsiveness. The relevance of lncRNA FAM30A with proliferation, migration ability, Daunorubicin resistance and its reciprocal action with AUF1 were demonstrated in an LSC cell line. Transcriptomic analysis suggested correlations between FAM30A and T cell differentiation and signaling, intercellular junction genes. Conclusions: The prognostic signature CuRS can guide prognostic stratification and personalized AML therapy. Analysis of FAM30A offers a foundation for investigating LSC-targeted therapies.
Article
Cuproptosis shows good application prospects in tumor therapy. However, the copper efflux mechanism and highly expressed intracellular reducing substances can inhibit the cuproptosis effects. In this study, a platelet vesicle (PV) coated cuprous oxide nanoparticle (Cu2O)/TBP-2 cuproptosis sensitization system (PTC) was constructed for multiple induction of tumor cuproptosis. PTC was prepared by physical extrusion of AIE photosensitizer (TBP-2), Cu2O, and PV. After the biomimetic modification, PTC can enhance its long-term blood circulation and tumor targeting ability. Subsequently, PTC was rapidly degraded to release copper ions under acid conditions and hydrogen peroxides in tumor cells. Then, under light irradiation, TBP-2 quickly enters the cell membrane and generates hydroxyl radicals to consume glutathione and inhibit copper efflux. Accumulated copper can cause lipoylated protein aggregation and iron-sulfur protein loss, which result in proteotoxic stress and ultimately cuproptosis. PTC treatment can target and induce cuproptosis in tumor cells in vitro and in vivo, significantly inhibit lung metastasis of breast cancer, increase the number of central memory T cells in peripheral blood, and prevent tumor rechallenge. It provides an idea for the design of nanomedicine based on cuproptosis.