1
|
Yan R, Cheng X, Song Y, Wang H, Zhang R, Jin Y, Li X, Chen Y, Xiang H. Cuproptosis nanoprodrug-initiated self-promoted cascade reactions for postoperative tumor therapy. Biomaterials 2025; 318:123176. [PMID: 39954313 DOI: 10.1016/j.biomaterials.2025.123176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Cancer metastasis and recurrence remain a regular cause of postoperative death in patients, implying that extra consolidation treatment strategies are needed. Here, a cuproptosis nanoprodrug, termed as Lipo@CP@DQ NPs, is developed to initiate self-promoted cascade reactions to achieve the combinational effect of cuproptosis, in situ chemotherapy, and oxidative stress amplification for effectively suppressing tumor recurrence and metastasis after postoperative treatment. Lipo@CP@DQ NPs are fabricated by loading copper peroxides (Cu2O2, CP) and hydrogen peroxide (H2O2)-repsonsive prodrug DQ into liposomal nanoparticles. Lipo@CP@DQ NPs rapidly dissociate in the acidic tumor microenvironment to release copper ions, H2O2, and prodrug DQ. Subsequently, the excessive accumulation of Cu ions induces cuproptosis and produces highly cytotoxic hydroxyl radicals (•OH). Meanwhile, the self-supplied H2O2 catalyzes the decomposition of DQ to diethyldithiocarbamate (DTC), which is chelated with self-supplied Cu ions to form the anticancer compound, Cu(DTC)2. The another decomposition product, quinone methide (QM), acts as a glutathione (GSH) scavenger for oxidative stress amplification. The synergistic effect of Lipo@CP@DQ NPs-mediated cuproptosis, in situ chemotherapy, and oxidative stress amplification effectively inhibits the growth and postoperative recurrence of triple-negative breast cancer. This work furnishes a strategy for developing cuproptosis-based nanomedicines for effective antitumor treatment after surgery.
Collapse
Affiliation(s)
- Ruiqi Yan
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xuan Cheng
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yujing Song
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Haiyue Wang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Run Zhang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yiqi Jin
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xingguang Li
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China.
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Huijing Xiang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
2
|
Zhang Z, Zhao Q, Xu Q, Deng Q, Hua A, Wang X, Yang X, Li Z. A mitochondria-interfering nanocomplex cooperates with photodynamic therapy to boost antitumor immunity. Biomaterials 2025; 317:123094. [PMID: 39799701 DOI: 10.1016/j.biomaterials.2025.123094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Immunotherapeutics against triple-negative breast cancer (TNBC) hold great promise. In this work, we provide a combination therapy for simultaneous increasing tumor immunogenicity and down-regulating programmed cell death ligand 1 (PD-L1) to boost antitumor immunity in TNBC. We prepare bis (diethyldithiocarbamate)-copper/indocyanine green nanoparticles (CuET/ICG NPs) simply in aqueous with one-pot method. CuET/ICG NPs interfere mitochondria, reduce oxygen consumption, and alleviate tumor hypoxia to potentiate photodynamic therapy (PDT) for amplifying immunogenic cell death (ICD). Meanwhile, mitochondria dysfunction leads to energy stress and activates AMPK pathway. As a result, CuET/ICG NPs downregulates membrane PD-L1 (mPD-L1) on both 4T1 cancer cells and cancer stem cells (CSCs) through AMP-activated protein kinase (AMPK)-mediated pathway in hypoxia. Cooperatively, the combinational therapy activates antitumor immunity and triggers long lasting immune memory response to resist tumor re-challenge. Our study represents an attempt that conquers tumor immunosuppressive microenvironment with simple biomedical materials and multimodality treatments.
Collapse
Affiliation(s)
- Zhijie Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingfu Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingqing Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingyuan Deng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Ao Hua
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xing Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| | - Zifu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| |
Collapse
|
3
|
Mao L, Lu J, Wen X, Song Z, Sun C, Zhao Y, Huang F, Chen S, Jiang D, Che W, Zhong C, Yu C, Li K, Lu X, Shi J. Cuproptosis: mechanisms and nanotherapeutic strategies in cancer and beyond. Chem Soc Rev 2025. [PMID: 40433941 DOI: 10.1039/d5cs00083a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Cuproptosis, a novel form of copper (Cu)-dependent programmed cell death, is induced by directly binding Cu species to lipoylated components of the tricarboxylic acid (TCA) cycle. Since its discovery in 2022, cuproptosis has been closely linked to the field of materials science, offering a biological basis and bright prospects for the use of Cu-based nanomaterials in various disease treatments. Owing to the unique physicochemical properties of nanomaterials, Cu delivery nanosystems can specifically increase Cu levels at disease sites, inducing cuproptosis to achieve disease treatment while minimizing the undesirable release of Cu in normal tissues. This innovative nanomaterial-mediated cuproptosis, termed as "nanocuproptosis", positions at the intersection of chemistry, materials science, pharmaceutical science, and clinical medicine. This review aims to comprehensively summarize and discuss recent advancements in cuproptosis across various diseases, with a particular focus on cancer. It delves into the biochemical basis of nanomaterial-mediated cuproptosis, the rational design for cuproptosis inducers, strategies for enhancing therapeutic specificity, and cuproptosis-centric synergistic cancer therapeutics. Beyond oncology, this review also explores the expanded applications of cuproptosis, such as antibacterial, wound healing, and bone tissue engineering, highlighting its great potential to open innovative therapeutic strategies. Furthermore, the clinical potential of cuproptosis is assessed from basic, preclinical to clinical research. Finally, this review addresses current challenges, proposes potential solutions, and discusses the future prospects of this burgeoning field, highlighting cuproptosis nanomedicine as a highly promising alternative to current clinical therapeutics.
Collapse
Affiliation(s)
- Lijie Mao
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Ji Lu
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xinyu Wen
- Shanghai Skin Disease Hospital, Institute of Dermatology, School of Medicine, Tongji University, Shanghai 200443, China
| | - Zhiyi Song
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cai Sun
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuanru Zhao
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Fang Huang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Si Chen
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Dongyang Jiang
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
| | - Cheng Zhong
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Ke Li
- School of Materials Science and Engineering, Hainan University, Haikou 570228, China.
| | - Xiangyu Lu
- Department of Nephrology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| | - Jianlin Shi
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200092, China
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China.
| |
Collapse
|
4
|
Okatsu K, Kawaguchi T, Watanabe K, Taguchi Y, Takeuchi R, Okamoto A, Iwasa Y, Tomita T, Saeki Y, Sato Y, Narumi T, Fukai S. Adaptor-Specific Peptide Inhibitors of the Ubiquitin-Chain-Dependent Unfolding Activity of the Human p97(VCP)-UFD1-NPL4 Complex. J Med Chem 2025. [PMID: 40421687 DOI: 10.1021/acs.jmedchem.5c00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
The AAA-ATPase p97, a key component of the ubiquitin-proteasome system (UPS), collaborates with its cofactor, the UFD1-NPL4 (UN) heterodimer, to unfold ubiquitinated substrates leading to proteasomal degradation. In this study, we report the development of novel peptide inhibitors that specifically target the p97-UN complex. These inhibitors are designed based on the NPL4-binding motif (NBM) of UFD1 and disrupt the interaction between p97 and the UN heterodimer. Our results demonstrate that these peptides effectively inhibit the unfolding activity of p97-UN, suggesting their potential as a therapeutic strategy for diseases associated with UPS dysfunction, such as cancer and neurodegenerative disorders. This work provides the first mechanistic insights into the inhibition of p97-UN by high-affinity peptide inhibitors and introduces promising candidates for drug development targeting the stable p97-UN complex in cells.
Collapse
Affiliation(s)
- Kei Okatsu
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Takaya Kawaguchi
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Kohei Watanabe
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Yoshinori Taguchi
- Graduate School of Medical Photonics, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Reon Takeuchi
- Graduate School of Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Akinori Okamoto
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Yasuyuki Iwasa
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Takuya Tomita
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yasushi Saeki
- Division of Protein Metabolism, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yusuke Sato
- Center for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Tetsuo Narumi
- Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
- Graduate School of Medical Photonics, Shizuoka University, Hamamatsu 432-8561, Japan
- Graduate School of Science and Technology, Shizuoka University, Hamamatsu 432-8561, Japan
| | - Shuya Fukai
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
5
|
Sedlacek J. Impact of proteostasis workload on sensitivity to proteasome inhibitors in multiple myeloma. Clin Exp Med 2025; 25:176. [PMID: 40418254 DOI: 10.1007/s10238-025-01713-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Accepted: 05/01/2025] [Indexed: 05/27/2025]
Abstract
Genomic alterations and enormous monoclonal immunoglobulin production cause multiple myeloma to heavily depend on proteostasis mechanisms, including protein folding and degradation. These findings support the use of proteasome inhibitors for treating multiple myeloma and mantle cell lymphoma. Myeloma treatment has evolved, especially with the availability of new drugs, such as proteasome inhibitors, into therapeutic strategies for both frontline and relapsed/refractory disease settings. However, proteasome inhibitors are generally not effective enough to cure most patients. Natural resistance and eventual acquired resistance led to relapsed/refractory disease and poor prognosis. Advances in the understanding of cellular proteostasis and the development of innovative drugs that also target other proteostasis network components offer opportunities to exploit the intrinsic vulnerability of myeloma cells. This review outlines recent findings on the molecular mechanisms regulating cellular proteostasis pathways, as well as resistance, sensitivity, and escape strategies developed against proteasome inhibitors and provides a rationale and examples for novel combinations of proteasome inhibitors with FDA-approved drugs and investigational drugs targeting the NRF1 (NFE2L1)-mediated proteasome bounce-back response, redox homeostasis, heat shock response, unfolding protein response, autophagy, and VCP/p97 to increase proteotoxic stress, which can improve the efficacy of antimyeloma therapy based on proteasome inhibitors.
Collapse
Affiliation(s)
- Jindrich Sedlacek
- Department of Genetics and Microbiology, Charles University and Research Center BIOCEV, Průmyslová 595, 252 50, Vestec, Czech Republic.
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610, Prague, Czech Republic.
| |
Collapse
|
6
|
Wang H, Chen L, Mao Z, Liu S, Huang R, He R, Zhang Y, Wei J. Boosting Energy Deprivation via Synchronous Interventions of Oxidative Phosphorylation and Glycolysis for Cancer Therapy with 1,8-Naphthyridine-Piperazine-Dithiocarbamate Ruthenium(II) Polypyridyl Complexes. J Med Chem 2025; 68:10203-10215. [PMID: 40353767 DOI: 10.1021/acs.jmedchem.5c00384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Bioenergetic therapy targeting mitochondrial bioenergy is a promising therapeutic strategy for cancer. However, its clinical efficacy is limited by the metabolic adaptability of tumor cells, as they can switch between glycolytic and oxidative phosphorylation metabolic phenotypes to maintain energy homeostasis. In this study, we discovered 1,8-naphthyridine-piperazine-dithiocarbamate ruthenium(II) polypyridyl complexes (RuL1) that enhanced energy deprivation by inhibiting the activity of mitochondrial complex I and III, thereby disrupting oxidative phosphorylation. Simultaneously, RuL1 inhibits glycolysis while unexpectedly activating antitumor immunity. This dual metabolic-immunological targeting resulted in enhanced anticancer activity against MGC-803 cells. To the best of our knowledge, RuL1 is the first ruthenium polypyridyl complex reported to achieve high anticancer activity through dual metabolic inhibition.
Collapse
Affiliation(s)
- Huiling Wang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
| | - Lei Chen
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
| | - Zhichen Mao
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
| | - Shuangqiang Liu
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
| | - Rizhen Huang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
| | - Ruijie He
- Guangxi Zhuang Autonomous Region and Chinese Academy of Sciences, Guangxi Key Laboratory of Plant Functional Phytochemicals and Sustainable Utilization, Guangxi Institute of Botany, Guilin 541006, China
| | - Ye Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
| | - Jianhua Wei
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guangxi Engineering Research Center for Pharmaceutical Molecular Screening and Druggability Evaluation, School of Pharmacy, Guilin Medical University, Guilin 541199 Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin 541001 Guangxi, China
| |
Collapse
|
7
|
Kirupakaran A, van den Boom J, Blueggel M, Beuck C, Niemeyer F, Hayduk M, Balszuweit J, Bayer P, Voskuhl J, Meyer H, Schrader T. Molecular Tweezers Block the Functional Pore of a Protein Machine. J Am Chem Soc 2025; 147:16836-16849. [PMID: 40354241 PMCID: PMC12100656 DOI: 10.1021/jacs.4c15288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 05/14/2025]
Abstract
We present symmetric multivalent tweezers as the first class of supramolecular elements designed to cover and functionally block a protein pore. As a model, we chose the enzyme p97, a hexameric AAA-ATPase that unfolds or segregates substrate proteins by threading them through a pore and channel at the center of the symmetric p97 hexamer fueled by ATP hydrolysis. In a rational design approach, we developed a new class of p97 inhibitors, guided by molecular modeling. These dock onto lysine residues at the entry of the pore via appropriately positioned molecular tweezers. Ligand binding was accompanied by induction of fluorescence of the built-in binding sensitive luminophores which served as a sensor for affinity determination. We further confirmed specific interaction with p97 as well as concomitant inhibition of ATPase activity and protein substrate unfolding using an array of biophysical methods and state-of-the art biochemical assays. Specific binding was also validated by mutagenesis, demonstrating that inhibition of p97 function was mediated by blocking the pore entrance. Especially C3-symmetric multivalent tweezers potently inhibited ATPase activity and protein substrate processing consistent with the symmetry of the docking site. Our data independently confirm substrate threading as a mechanism for protein unfolding by p97 and highlight multivalent tweezers as a supramolecular strategy to target pores in various proteins. Since p97 and related protein machines are vital for protein quality control and cell survival, the new pore binders may open a new approach to combat diseases and be employed in drug discovery.
Collapse
Affiliation(s)
- Abbna Kirupakaran
- Faculty
of Chemistry, University of Duisburg-Essen, 45141Essen, Germany
| | | | - Mike Blueggel
- Faculty
of Biology, University of Duisburg-Essen, 45141Essen, Germany
| | - Christine Beuck
- Faculty
of Biology, University of Duisburg-Essen, 45141Essen, Germany
| | - Felix Niemeyer
- Faculty
of Chemistry, University of Duisburg-Essen, 45141Essen, Germany
| | - Matthias Hayduk
- Faculty
of Chemistry, University of Duisburg-Essen, 45141Essen, Germany
| | - Jan Balszuweit
- Faculty
of Chemistry, University of Duisburg-Essen, 45141Essen, Germany
| | - Peter Bayer
- Faculty
of Biology, University of Duisburg-Essen, 45141Essen, Germany
| | - Jens Voskuhl
- Faculty
of Chemistry, University of Duisburg-Essen, 45141Essen, Germany
| | - Hemmo Meyer
- Faculty
of Biology, University of Duisburg-Essen, 45141Essen, Germany
| | - Thomas Schrader
- Faculty
of Chemistry, University of Duisburg-Essen, 45141Essen, Germany
| |
Collapse
|
8
|
Li Z, Yu K, Cao Y, Yuan H, Wu L, Xiong L, Tang Y, Liang B. Breaking the Chains of Therapeutic Blockade: Pyroptosis-Induced Photothermal-Chemotherapy with Targeted Nanoprobes in Triple-Negative Breast Cancer. Biomater Res 2025; 29:0200. [PMID: 40376201 PMCID: PMC12079191 DOI: 10.34133/bmr.0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/10/2025] [Accepted: 04/12/2025] [Indexed: 05/18/2025] Open
Abstract
There is an important clinical need and social significance, especially for young patients, to explore a new breast-conserving strategy that is not dependent on biomarkers for anti-triple-negative breast cancer. Disulfiram, historically employed for the treatment of chronic alcoholism, has recently emerged as a promising antitumor agent in combination with Cu2+. However, reported disulfiram-Cu2+ codelivery regimens often suffer from instability as well as inadequate drug metabolism, which is detrimental to the production and action of the antitumor active ingredient copper(II) bis(diethyldithiocarbamate). To address this obstacle, this study tested nanosystems ICG-CuET@PLGA-CS-HA (IC@PCH) nanoparticles (NPs) carrying the chemotherapeutic agent copper(II) bis(diethyldithiocarbamate) and photosensitizer indocyanine green for the efficient delivery of antitumor drugs. Benefiting from the involvement of hyaluronic acid, the prepared IC@PCH NPs not only targeted CD44 on the surface of tumor cells but also showed a longer in vivo circulation time. The in vitro and in vivo results demonstrated that IC@PCH NP-mediated photothermal-chemotherapy treatment led to pyroptosis via the NLRP3/caspase-1 classical pathway, which had a significant therapeutic effect on triple-negative breast cancer. In addition, targeting IC@PCH NPs allows photoacoustic-magnetic resonance-fluorescence trimodal imaging, which is capable of detecting more insidious cancer foci and opens up new avenues for precise cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Zuying Li
- Department of Ultrasound of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy,
Children’s Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
- Department of Ultrasound & Chongqing Key Laboratory of Ultrasound Molecular Imaging,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Kexiao Yu
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital,
The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing 400021, P. R. China
| | - Youde Cao
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center,
Chongqing Medical University, Chongqing 400016, P. R. China
- Department of Pathology,
The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P. R. China
| | - Hui Yuan
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center,
Chongqing Medical University, Chongqing 400016, P. R. China
- Department of Pathology,
The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P. R. China
| | - Lingcheng Wu
- Department of Ultrasound & Chongqing Key Laboratory of Ultrasound Molecular Imaging,
The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center,
Chongqing Medical University, Chongqing 400016, P. R. China
| | - Linyan Xiong
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center,
Chongqing Medical University, Chongqing 400016, P. R. China
- Department of Pathology,
The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P. R. China
| | - Yi Tang
- Department of Ultrasound of Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy,
Children’s Hospital of Chongqing Medical University, Chongqing 400014, P. R. China
| | - Bing Liang
- Department of Pathology from College of Basic Medicine, and Molecular Medicine Diagnostic & Testing Center, and Department of Clinical Pathology Laboratory of Pathology Diagnostic Center,
Chongqing Medical University, Chongqing 400016, P. R. China
- Department of Pathology,
The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, P. R. China
| |
Collapse
|
9
|
Guo Z, Chen D, Yao L, Sun Y, Li D, Le J, Dian Y, Zeng F, Chen X, Deng G. The molecular mechanism and therapeutic landscape of copper and cuproptosis in cancer. Signal Transduct Target Ther 2025; 10:149. [PMID: 40341098 PMCID: PMC12062509 DOI: 10.1038/s41392-025-02192-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/13/2024] [Accepted: 02/17/2025] [Indexed: 05/10/2025] Open
Abstract
Copper, an essential micronutrient, plays significant roles in numerous biological functions. Recent studies have identified imbalances in copper homeostasis across various cancers, along with the emergence of cuproptosis, a novel copper-dependent form of cell death that is crucial for tumor suppression and therapeutic resistance. As a result, manipulating copper levels has garnered increasing interest as an innovative approach to cancer therapy. In this review, we first delineate copper homeostasis at both cellular and systemic levels, clarifying copper's protumorigenic and antitumorigenic functions in cancer. We then outline the key milestones and molecular mechanisms of cuproptosis, including both mitochondria-dependent and independent pathways. Next, we explore the roles of cuproptosis in cancer biology, as well as the interactions mediated by cuproptosis between cancer cells and the immune system. We also summarize emerging therapeutic opportunities targeting copper and discuss the clinical associations of cuproptosis-related genes. Finally, we examine potential biomarkers for cuproptosis and put forward the existing challenges and future prospects for leveraging cuproptosis in cancer therapy. Overall, this review enhances our understanding of the molecular mechanisms and therapeutic landscape of copper and cuproptosis in cancer, highlighting the potential of copper- or cuproptosis-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Danyao Chen
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Furong Laboratory, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China.
- Furong Laboratory, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
10
|
Xu L, Cao X, Deng Y, Zhang B, Li X, Liu W, Ren W, Tang X, Kong X, Zhang D. Cuproptosis-related genes and agents: implications in tumor drug resistance and future perspectives. Front Pharmacol 2025; 16:1559236. [PMID: 40406488 PMCID: PMC12095339 DOI: 10.3389/fphar.2025.1559236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/23/2025] [Indexed: 05/26/2025] Open
Abstract
In the field of tumor treatment, drug resistance remains a significant challenge requiring urgent intervention. Recent developments in cell death research have highlighted cuproptosis, a mechanism of cell death induced by copper, as a promising avenue for understanding tumor biology and addressing drug resistance. Cuproptosis is initiated by the dysregulation of copper homeostasis, which in turn triggers mitochondrial metabolic disruptions and induces proteotoxic stress. This process specifically entails the accumulation of lipoylated proteins and the depletion of iron-sulfur cluster proteins within the context of the tricarboxylic acid cycle. Simultaneously, it is accompanied by the activation of distinct signaling pathways that collectively lead to cell death. Emerging evidence highlights the critical role of cuproptosis in addressing tumor drug resistance. However, the core molecular mechanisms of cuproptosis, regulation of the tumor microenvironment, and clinical translation pathways still require further exploration. This review examines the intersection of cuproptosis and tumor drug resistance, detailing the essential roles of cuproptosis-related genes and exploring the therapeutic potential of copper ionophores, chelators, and nanodelivery systems. These mechanisms offer promise for overcoming resistance and advancing tumor precision medicine. By elucidating the molecular mechanisms underlying cuproptosis, this study aims to identify novel therapeutic strategies and targets, thereby paving the way for the development of innovative anti-cancer drugs.
Collapse
Affiliation(s)
- Lingwen Xu
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xiaolan Cao
- Department of Radiotherapy, Shandong Second Provincial General Hospital, Jinan, Shandong, China
| | - Yuxiao Deng
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Bin Zhang
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xinzhi Li
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Wentao Liu
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Wenjie Ren
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xuan Tang
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Xiangyu Kong
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| | - Daizhou Zhang
- Institute of Chemical Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
- Shandong Provincial Key Laboratory of Carbohydrate and Glycoconjugate Drugs, Shandong Academy of Pharmaceutical Sciences, Jinan, China
| |
Collapse
|
11
|
Körner M, Müller P, Das H, Kraus F, Pfeuffer T, Spielhaupter S, Oeljeklaus S, Schülein-Völk C, Harper JW, Warscheid B, Buchberger A. p97/VCP is required for piecemeal autophagy of aggresomes. Nat Commun 2025; 16:4243. [PMID: 40335532 PMCID: PMC12059050 DOI: 10.1038/s41467-025-59556-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Metazoan cells adapt to the exhaustion of protein quality control (PQC) systems by sequestering aggregation-prone proteins in large, pericentriolar structures termed aggresomes. Defects in both aggresome formation and clearance affect proteostasis and have been linked to neurodegenerative diseases, but aggresome clearance pathways are still underexplored. Here we show that aggresomes comprising endogenous proteins are cleared via selective autophagy requiring the cargo receptor TAX1BP1. TAX1BP1 proximitomes reveal the presence of various PQC systems at aggresomes, including Hsp70 chaperones, the 26S proteasome, and the ubiquitin-selective unfoldase p97/VCP. While Hsp70 and p97/VCP with its cofactors UFD1-NPL4 and FAF1 play key roles in aggresome disassembly, the 26S proteasome is dispensable. We identify aggresomal client proteins that are degraded via different routes, in part in a p97/VCP-dependent manner via aggrephagy. Upon acute inhibition of p97/VCP, aggresomes fail to disintegrate and cannot be incorporated into autophagosomes despite the presence of factors critical for aggrephagosome formation, including p62/SQSTM1, TAX1BP1, and WIPI2. We conclude that the p97/VCP-mediated removal of ubiquitylated aggresomal clients is essential for the disintegration and subsequent piecemeal autophagy of aggresomes.
Collapse
Affiliation(s)
- Maria Körner
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Paul Müller
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Hirak Das
- Biocenter, Chair of Biochemistry II, University of Würzburg, Würzburg, Germany
| | - Felix Kraus
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Timo Pfeuffer
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Sven Spielhaupter
- Biocenter, Chair of Biochemistry I, University of Würzburg, Würzburg, Germany
| | - Silke Oeljeklaus
- Biocenter, Chair of Biochemistry II, University of Würzburg, Würzburg, Germany
| | | | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Bettina Warscheid
- Biocenter, Chair of Biochemistry II, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
12
|
Paun RA, Li L, Mouncef A, Radzioch D, Tabrizian M. Liposome-Polymer Nanoparticles Loaded with Copper Diethyldithiocarbamate and 6-Bromo-Indirubin-3'-Oxime Enable the Treatment of Refractive Melanoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2409012. [PMID: 40317886 DOI: 10.1002/smll.202409012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/26/2025] [Indexed: 05/07/2025]
Abstract
Despite significant advances in cancer immunotherapy, many patients fail to respond to current treatments, outlining the need to develop novel therapeutic modalities. Therapeutic resistance in cancer cells is mediated by significant genomic instability due to their oncogenic transformation and evolutionary pressures inside the tumor microenvironment (TME). However, these cellular and molecular adaptations can result in a significant increase in the baseline endoplasmic reticulum (ER) stress in TME-resident cells. This can be taken advantage of as a therapeutic strategy by using the metal chelate copper diethyldithiocarbamate (CuET), a potent inhibitor of the p97-UFD1-NPL4 protein complex to induce cytotoxicity and exacerbate ER stress in cancer cells. Here, CuET is combined with the anti-inflammatory drug 6-bromo-indirubin-3'-oxime (BIO), a potent GSK3 inhibitor, to modulate the aberrant inflammatory response inside the TME. However, both CuET and BIO are highly hydrophobic and exhibit poor bioavailability, requiring the development of an appropriate carrier. Herein, it is demonstrated that CuET and BIO can be efficiently loaded into liposomes that are stabilized by poly(vinylpyrrolidone). The liposome-loaded drug combination resulted in a significant decrease of 47% and 76% in the tumor burden of syngeneic B16F10 and YUMM1.7 mouse models, respectively, without any major acute toxicity.
Collapse
Affiliation(s)
- Radu A Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC, H3A 2B6, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Ling Li
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC, H3A 2B6, Canada
| | - Adam Mouncef
- Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Danuta Radzioch
- Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Medicine, Faculty of Medicine and Health Sciences, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
- Department of Human Genetics, Faculty of Medicine and Health Sciences, 3640 University Street, Montreal, QC, H3A 0C7, Canada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, 3775 University Street, Montreal, QC, H3A 2B6, Canada
- Faculty of Dentistry and Oral Health Sciences, McGill University, McGill University, 2001 McGill College Ave, Montreal, QC, H3A 1G1, Canada
| |
Collapse
|
13
|
Lu K, Wijaya CS, Yao Q, Jin H, Feng L. Cuproplasia and cuproptosis, two sides of the coin. Cancer Commun (Lond) 2025; 45:505-524. [PMID: 39865459 PMCID: PMC12067395 DOI: 10.1002/cac2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/03/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025] Open
Abstract
Copper is an essential micronutrient in the human body, mainly acting as a crucial cofactor required for a wide range of physiological processes across nearly all cell types. Recent advances revealed that tumor cells seize copper to fulfill their rapid proliferation, metastasis, immune evasion, and so on by reprogramming the copper regulatory network, defined as cuproplasia. Thus, targeting copper chelation to reduce copper levels has been considered a rational tumor therapy strategy. However, overloaded copper ions could be toxic, which leads to the aggregation of lipoylated mitochondrial proteins and the depletion of iron-sulfur clusters, ultimately resulting in cell death, termed cuproptosis. Upon its discovery, cuproptosis has attracted great interest from oncologists, and targeting cuproptosis by copper ionophores exhibits as a potential anti-tumor therapy. In this review, we present the underlying mechanisms involved in cuproplasia and cuproptosis. Additionally, we sum up the chemicals targeting either cuproplasia or cuproptosis for cancer therapy. Further attention should be paid to distinguishing cancer patients who are suitable for targeting cuproplasia or cuproptosis.
Collapse
Affiliation(s)
- Kaizhong Lu
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Chandra Sugiarto Wijaya
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Qinghua Yao
- Department of OncologyThe Second Affiliated Hospital of Zhejiang Chinese Medical UniversityXinhua Hospital of Zhejiang ProvinceHangzhouZhejiangP. R. China
- Key Laboratory for Research on the Pathogenesis of Inflammation‐Cancer Transformation in Intestinal DiseasesZhejiang Engineering Research Center of Intelligent Equipment of Chronic Chinese and Western MedicineHangzhouZhejiangP. R. China
| | - Hongchuan Jin
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| | - Lifeng Feng
- Department of Medical OncologyZhejiang Key Laboratory of Multi‐omics Precision Diagnosis and Treatment of Liver DiseasesCancer Center of Zhejiang UniversitySir Run Run Shaw HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiangP. R. China
| |
Collapse
|
14
|
Shan D, Song J, Ren Y, Zhang Y, Ba Y, Luo P, Cheng Q, Xu H, Weng S, Zuo A, Liu S, Han X, Deng J, Liu Z. Copper in cancer: friend or foe? Metabolism, dysregulation, and therapeutic opportunities. Cancer Commun (Lond) 2025; 45:577-607. [PMID: 39945125 PMCID: PMC12067407 DOI: 10.1002/cac2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 05/13/2025] Open
Abstract
Copper, one of the essential nutrients for the human body, acts as an electron relay in multiple pathways due to its redox properties. Both deficiencies and excesses of copper lead to cellular fragility. Therefore, it can manifest pro- and anti-cancer properties in tumors. Therefore, it is crucial to clarify the copper activity within the cell. We have thoughtfully summarized the metabolic activities of copper from a macro and micro perspective. Cuproptosis, as well as other forms of cell death, is directly or indirectly interfered with by Cu2+, causing cancer cell death. Meanwhile, we did pan-cancer analysis of cuproptosis-related genes to further clarify the roles of these genes. In addition, copper has been found to be involved in multiple pathways within the metastasis of cancer cells. Given the complexity of copper's role, we are compelled to ask: is copper a friend or a foe? Up to now, copper has been used in various clinical applications, including protocols for measurement of copper concentration and bioimaging of radioactive 64Cu. But therapeutically it is still a continuation of the old medicine, and new possibilities need to be explored, such as the use of nanomaterials. Some studies have also shown that copper has considerable interventional power in metabolic cancers, which provides the great applications potential of copper therapy in specific cancer types. This paper reviews the dual roles played by cuproptosis in cancer from the new perspectives of oxidative stress, cell death, and tumor metastasis, and points out the value of its application in specific cancer types, summarizes the value of its testing and imaging from the perspective of clinical application as well as the current feasible options for the new use of the old drugs, and emphasizes the prospects for the application of nano-copper.
Collapse
Affiliation(s)
- Dan Shan
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- University Hospital GalwayNational University of Ireland GalwayGalwayIreland
- Department of Biobehavioral SciencesColumbia UniversityNew YorkUSA
| | - Jinling Song
- Division of PulmonologyDepartment of PediatricsThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Peng Luo
- The Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Quan Cheng
- Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaHunanP. R. China
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanP. R. China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer ResearchComprehensive Cancer Centre, Kings College LondonLondonUK
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanP. R. China
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanP. R. China
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| |
Collapse
|
15
|
Dai W, Zhai Y, Yang F, Chen W, Liu C, Tian Y, Huang F, Cai M, Zheng L, Cheng W, Chen W, Zhang J. Different concentrations of 2-Undecanone triggers repellent and nematicidal responses in Caenorhabditis elegans. Sci Rep 2025; 15:14186. [PMID: 40269070 PMCID: PMC12019385 DOI: 10.1038/s41598-025-95332-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
The compound 2-undecanone is widely distributed in the natural environment and exhibits a dual-action mechanism against nematodes. It demonstrates repellency and contact toxicity against both Caenorhabditis elegans and Meloidogyne incognita. However, research on the dual-function mechanism of 2-undecanone remains relatively limited. In this study, using chemotaxis experiments, we found that 2-undecanone (at concentrations of 1-5 mg/mL) signal is detected through AWB olfactory sensory neurons in nematode, and then transduced through the cGMP pathway to induce repellent behavior. Moreover, we observed that 2-undecanone (at concentrations of 0.06-0.08 mg/mL) induces intracellular calcium accumulation and causes lysosomal membrane rupture. We further identified Hsp70 A and V-ATPase A as the targets of 2-undecanone responsible for its contact killing effect. Furthermore, 2-undecanone was found to alter sphingomyelin metabolism in both wild-type C. elegans and hsp-1 mutants. This alteration led to decreased acid sphingomyelinase activity, reduced ceramide levels, and increased sphingomyelin levels. These results indicated that 2-undecanone has dual functions and two target receptors Hsp70 A and V-ATPase A against nematodes, and one AWB olfactory neuron repelled nematodes. The dual-action mode of 2-undecanone can decrease the nematodes' tolerance to the compound appropriately, extending its efficacy duration. Understanding the dual function mechanism of action of 2-undecanone will aid in devising innovative approaches for managing nematode control.
Collapse
Affiliation(s)
- Wei Dai
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yile Zhai
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Fan Yang
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wen Chen
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chen Liu
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yaru Tian
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Feng Huang
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Minmin Cai
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Longyu Zheng
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wanli Cheng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, 430062, Hubei, China
| | - Weidong Chen
- Department of Plant Pathology, Washington State University, 99164-6430, Pullman, USA
| | - Jibin Zhang
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
16
|
Li Y, Han L, Hu H. Research progress on cuproptosis and copper related anti-tumor therapy. Discov Oncol 2025; 16:584. [PMID: 40257639 PMCID: PMC12011693 DOI: 10.1007/s12672-025-02335-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
Copper is a trace element which is essential for biological organisms, and its homeostatic balance is important for living organisms to maintain the normal function. When the copper homeostasis is disordered, the cellular function and structure will be disrupted. Excess copper cause oxidative stress and DNA damage in cells, thereby inducing regulated cell death such as apoptosis and necroptosis. Excess copper in mitochondria can bind to lipoylated proteins in the tricarboxylic acid (TCA) cycle and cause them to aggregate, resulting in proteotoxic stress and eliciting a novel cell death modality: cuproptosis. Cancer cells have a greater demand for copper compared to normal tissue, and high levels of copper ions are closely associated with tumour proliferation and metastasis. The anti-tumor mechanisms of copper include the production of oxidative stress, inhibition of the ubiquitin-proteasome system, suppression of angiogenesis, and induction of copper-dependent cell death. Targeting copper is one of the current directions in oncology research, including the use of copper ion carriers to increase intracellular copper levels to induce oxidative stress and cuproptosis, as well as the use of copper ion chelators to reduce copper bioavailability. However, copper complexes have certain toxicity, so their biosafety needs to be improved. Emerging nanotechnology is expected to solve this problem by utilizing copper-based nanomaterials (Cu-based NMs) to deliver copper ions and a variety of drugs with different functions, thereby improving the anti-tumor efficacy and reducing the side effects. Therefore, a thorough understanding of copper metabolic processes and the mechanism of cuproptosis will greatly benefit anti-tumor therapy. This review summarizes the processes of copper metabolism and the mechanism of cuproptosis. In addition, we discuss the current anti-tumor paradigms related to copper, we also discuss current nanotherapeutic approaches to copper mortality and provide prospective insights into the future copper-mediated cancer therapy.
Collapse
Affiliation(s)
- Yichen Li
- School of Medicine, Southeast University, No. 87, Dingjiaqiao, Hunan Road, Gulou District, Nanjing, 210009, China
| | - Lifei Han
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China
| | - Haolin Hu
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China.
| |
Collapse
|
17
|
Pei X, Zheng F, Li Y, Li X, Lin Z, Han X, Tian Z, Cao K, Ren D, Li C. Disulfiram activation of prostaglandin E2 synthesis: a novel antifibrotic mechanism in pulmonary fibrosis. J Pharmacol Exp Ther 2025; 392:103588. [PMID: 40359874 DOI: 10.1016/j.jpet.2025.103588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/02/2025] [Accepted: 04/13/2025] [Indexed: 05/15/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the pathological replacement of alveolar structures with thickened, inelastic fibrous tissue, which significantly hinders gas exchange in the lungs. Disulfiram (DSF), a Food and Drug Administration-approved drug for alcohol dependence, has shown potential in various diseases. This study investigates the effects of DSF on IPF and its mechanisms, focusing on the cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) pathway. Utilizing primary diseased human lung fibroblast-IPF cells and A549 cells induced with transforming growth factor-beta 1 to model epithelial-mesenchymal transition (EMT), we employed a battery of in vitro assays to assess cellular viability, migratory capacity, and the expression of fibrosis-related genes and proteins. To further substantiate our in vitro findings, a bleomycin-induced mouse model of IPF was treated with DSF, and subjected to a comprehensive evaluation of pulmonary function, histological examination, hydroxyproline assay, and western blot analysis to quantify the extent of fibrosis. DSF reduced cell viability and migration in fibrotic cell models. It increased COX-2 and PGE2 levels, regulated EMT, and extracellular matrix collagen deposition. In vivo, DSF improved pulmonary function and reduced EMT and extracellular matrix accumulation in mice. The COX-2/PGE2 axis was identified as a critical mediator of DSF's effects. DSF exhibits antifibrotic properties in IPF by modulating the COX-2/PGE2 signaling pathway. This study provides a novel therapeutic strategy for IPF and highlights the potential of repurposing DSF for clinical use in this context. SIGNIFICANCE STATEMENT: Disulfiram shows promise in treating idiopathic pulmonary fibrosis by targeting the cyclooxygenase-2/prostaglandin E2 pathway, offering a new therapeutic strategy and highlighting its potential for repurposing in this context.
Collapse
Affiliation(s)
- Xiaolin Pei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Fangxu Zheng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Yin Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xiaobo Li
- Beijing Tide Pharmaceutical Co, Ltd, Beijing, China; School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhoujun Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xiao Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Zhenhuan Tian
- Department of Thoracic surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing, China
| | - Ke Cao
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Dunqiang Ren
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Chenggang Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.
| |
Collapse
|
18
|
Wang Y, Du R, Gao R, Guo C, Qi J, Zhang Y, Zhu Q, Deng Q, Hu Z, Wang H, Hong B. Disulfiram potentiates cisplatin-induced apoptosis in small cell lung cancer via the inhibition of cystathionine β-synthase and H 2S. Am J Cancer Res 2025; 15:1647-1661. [PMID: 40371164 PMCID: PMC12070080 DOI: 10.62347/qjhb2816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/03/2025] [Indexed: 05/16/2025] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant neuroendocrine tumor. Platinum-based chemo-resistance is the major issue for the treatment of SCLC. The objective of the study is to identify the drugs that enhance anti-tumor activity of cisplatin (CDDP) in SCLC. Firstly, by a high-throughput drug screening, we found that disulfiram (DSF), a FDA-approved drug that is used to treat alcohol addiction, was able to sensitize CDDP-induced apoptosis in SCLC. RNA-seq analysis revealed that cystathionine β-synthase (CBS) was a potential target of combination treatment of DSF and CDDP in SCLC. CDDP treatment induced CBS expression, while the elevation of CBS expression was down-regulated by DSF and CDDP co-treatment in SCLC. Importantly, the down-regulation of CBS by siRNA silence increased CDDP-induced cellular apoptosis in SCLC. Furthermore, the study found that DSF combined with CDDP decreased the H2S level, and increased the level of ROS. The elevation of H2S level reduced the growth inhibition of SCLC cells by DSF and CDDP co-treatment. Finally, in nude mice bearing SCLC xenografts, DSF and CDDP co-treatment exhibited remarkable anti-tumor activity against SCLC tumors, evidenced by the significant reduction of tumor size, tumor weight and Ki-67 expression as compared with single treatment alone. Therefore, the study indicated that DSF could be re-purposed to potentiate CDDP-induced anti-tumor activity in SCLC, which are worth immediate assessment for SCLC in clinical settings.
Collapse
Affiliation(s)
- Yongguang Wang
- School of Basic Medical Sciences, Anhui Medical UniversityHefei 230032, Anhui, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
| | - Ruiping Du
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
- University of Science and Technology of ChinaHefei 230026, Anhui, China
| | - Rong Gao
- School of Basic Medical Sciences, Anhui Medical UniversityHefei 230032, Anhui, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
| | - Chang Guo
- School of Basic Medical Sciences, Anhui Medical UniversityHefei 230032, Anhui, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
| | - Jian Qi
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
- University of Science and Technology of ChinaHefei 230026, Anhui, China
| | - Yani Zhang
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
- University of Science and Technology of ChinaHefei 230026, Anhui, China
| | - Qizhi Zhu
- University of Science and Technology of ChinaHefei 230026, Anhui, China
| | - Qingmei Deng
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
| | - Zongtao Hu
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
| | - Hongzhi Wang
- School of Basic Medical Sciences, Anhui Medical UniversityHefei 230032, Anhui, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
- University of Science and Technology of ChinaHefei 230026, Anhui, China
| | - Bo Hong
- School of Basic Medical Sciences, Anhui Medical UniversityHefei 230032, Anhui, China
- Hefei Cancer Hospital of CAS, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS)Hefei 230031, Anhui, China
- University of Science and Technology of ChinaHefei 230026, Anhui, China
| |
Collapse
|
19
|
Lu BS, Liu KL, Yin YW, Zhang YP, Qi JC, Zhao CM, Niu YL, Guo PY, Li W. A novel feedback regulation loop of METTL11A-MAFG-NPL4 promotes bladder cancer cell proliferation and tumor progression. FASEB J 2025; 39:e70466. [PMID: 40171788 DOI: 10.1096/fj.202402830r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
Abnormal regulation of gene expression results in the malignant proliferation of bladder cancer (BC) cells. We previously demonstrated that NPL4 upregulation promotes BC progression; however, its regulatory and functional mechanisms on downstream genes in BC remain unknown. Transcriptome sequencing and reverse transcription-quantitative polymerase chain reaction were used to identify and confirm METTL11A as a downstream gene of NPL4. Protein interactions were detected through co-immunoprecipitation assays. Cell growth and tumor progression were assessed in vitro and in vivo using colony formation and MTS assays as well as xenograft animal models. Chromatin immunoprecipitation and luciferase activity assays were performed to investigate gene transcription regulation. We identified METTL11A as a downstream gene of NPL4, with its upregulation linked to poor outcomes in BC patients. METTL11A facilitates NPL4-regulated BC cell proliferation by promoting cyclin D1 expression. METTL11A enhances MAFG expression and contributes to METTL11A-mediated cell proliferation. Mechanistically, METTL11A interacts with MAFG, preventing its degradation through K6 methylation modification. MAFG and NRF2 bind to the promoter region of NPL4, promoting its transcription. Thus, the METTL11A-MAFG-NPL4 axis forms a positive feedback loop, promoting BC cell proliferation and tumor progression. Targeted inhibition of this regulatory loop could offer a novel therapeutic approach for BC.
Collapse
Affiliation(s)
- Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan-Ping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin-Chun Qi
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chen-Ming Zhao
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ya-Lin Niu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ping-Ying Guo
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Anwer MS, Abdel-Rasol MA, El-Sayed WM. Emerging therapeutic strategies in glioblastsoma: drug repurposing, mechanisms of resistance, precision medicine, and technological innovations. Clin Exp Med 2025; 25:117. [PMID: 40223032 PMCID: PMC11994545 DOI: 10.1007/s10238-025-01631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Glioblastoma (GBM) is an aggressive Grade IV brain tumor with a poor prognosis. It results from genetic mutations, epigenetic changes, and factors within the tumor microenvironment (TME). Traditional treatments like surgery, radiotherapy, and chemotherapy provide limited survival benefits due to the tumor's heterogeneity and resistance mechanisms. This review examines novel approaches for treating GBM, focusing on repurposing existing medications such as antipsychotics, antidepressants, and statins for their potential anti-GBM effects. Advances in molecular profiling, including next-generation sequencing, artificial intelligence (AI), and nanotechnology-based drug delivery, are transforming GBM diagnosis and treatment. The TME, particularly GBM stem cells and immune evasion, plays a key role in therapeutic resistance. Integrating multi-omics data and applying precision medicine show promise, especially in combination therapies and immunotherapies, to enhance clinical outcomes. Addressing challenges such as drug resistance, targeting GBM stem cells, and crossing the blood-brain barrier is essential for improving treatment efficacy. While current treatments offer limited benefits, emerging strategies such as immunotherapies, precision medicine, and drug repurposing show significant potential. Technologies like liquid biopsies, AI-powered diagnostics, and nanotechnology could help overcome obstacles like the blood-brain barrier and GBM stem cells. Ongoing research into combination therapies, targeted drug delivery, and personalized treatments is crucial. Collaborative efforts and robust clinical trials are necessary to translate these innovations into effective therapies, offering hope for improved survival and quality of life for GBM patients.
Collapse
Affiliation(s)
- Mohamed S Anwer
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt
| | - Mohammed A Abdel-Rasol
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
21
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025; 16:6160-6187. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
22
|
Ding Y, Chen R, Zhou J, Bao Y, Meng N, Zheng X, Yang S, Lu J, Jiang Z, Liu Y, Xie C, Lu L, Lu W. All-stage targeted nanodiscs for glioma treatment by inducing cuproptosis and apoptosis of cancer cells and cancer stem cells. Asian J Pharm Sci 2025; 20:101010. [PMID: 40182135 PMCID: PMC11964743 DOI: 10.1016/j.ajps.2024.101010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 04/05/2025] Open
Abstract
There remain several intractable challenges for chemotherapy in glioma treatment, including the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), and tumor heterogeneity caused by cancer stem cells (CSCs), which are resistant to conventional chemotherapy. Here, we established a nano strategy to kill glioma cells and CSCs, combining carfilzomib and bis(diethyldithiocarbamate)copper. The synergistic drug combination disturbed cell protein metabolism at different stages and induced apoptosis and cuproptosis. The Y-shaped targeting ligand pHA-VAP-modified nanodiscs were designed to help the chemotherapeutic agents cross the BBB/BBTB and finally accumulate in tumor site. This all-stage targeting and all-stage treatment nanomedicine significantly prolonged the survival in glioma-bearing mice and might inspire the rational design of advanced drug delivery platforms.
Collapse
Affiliation(s)
- Yuan Ding
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Ruohan Chen
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jianfen Zhou
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yanning Bao
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Nana Meng
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Xudong Zheng
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Shengmin Yang
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jiasheng Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zhixuan Jiang
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yu Liu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Cao Xie
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
- Institutes of Integrative Medicine, Fudan University, Shanghai 200032, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China
| |
Collapse
|
23
|
Sun R, Huang Y, Feng H, Zhao N, Wan W, Shen D, Zhong B, Zhang Y, Zhang X, Zhao Q, Zhang L, Liu Y. 1000 fold Ultra-Photosensitized Fluorescent Protein Mimics Toward Photocatalytic Proximity Labeling and Proteomic Profiling Functions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413063. [PMID: 39985251 PMCID: PMC12005797 DOI: 10.1002/advs.202413063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/07/2025] [Indexed: 02/24/2025]
Abstract
Photosensitizing fluorescent proteins (FP) (e.g. KillerRed) have been shown not capable of photo-catalytic protein proximity labeling for downstream proteomic profiling applications. To acquire such a function, FP chromophores are engineered in a 12 × 12 combinatorial matrix of synthetic analoges, achieving up to 1000 fold enhancement of reactive oxygen species (ROS) production compared to the natural FPs. FP chromophores are shown with larger dipole moments exhibit higher ROS yield toward protein labeling. By conjugating the ultra-photosensitized FP chromophore to HaloTag (namely upsFP tag), its photo-catalytic protein proximity labeling function is demonstrated using nucleophilic amino substrates. Through photochemical characterizations, theoretical calculation, and tandem mass spectrometry, a radical-mediated labeling mechanism is revealed with expanded reactivity toward diverse protein residues via a type I photosensitization pathway. Finally, a proteomic profiling application is showcased using the upsFP tag to resolve the dynamic interactome variations upon TAR DNA-binding protein 43 (TDP43) phase separation and suborganellar translocation. Together, this work demonstrates three orders of magnitude ultra-photosensitization of fluorescent protein chromophore enables photocatalytic protein proximity labeling and profiling functions that are impractical for natural fluorescent proteins.
Collapse
Affiliation(s)
- Rui Sun
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanan Huang
- Department of Chemistry and Westlake Laboratory of Life Science and BiomedicineWestlake University600 Dunyu RoadHangzhou310030China
| | - Huan Feng
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Nan Zhao
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Wang Wan
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Di Shen
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Bowen Zhong
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Yukui Zhang
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Xin Zhang
- Department of Chemistry and Westlake Laboratory of Life Science and BiomedicineWestlake University600 Dunyu RoadHangzhou310030China
| | - Qun Zhao
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Lihua Zhang
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Yu Liu
- State Key Laboratory of Medical ProteomicsNational Chromatographic R. & A. CenterCAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| |
Collapse
|
24
|
Said Suliman A, Rehmani S, Small B, Butcher K, Khoder M, Kannappan V, Wang W, Elhissi A, Najlah M. PEGylated Liposomes of Disulfiram and Paclitaxel: A Promising Chemotherapeutic Combination Against Chemoresistant Breast Cancer. Pharmaceuticals (Basel) 2025; 18:487. [PMID: 40283924 PMCID: PMC12030275 DOI: 10.3390/ph18040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Steric stabilization of liposomes using PEGylation has been used widely in pharmaceutical research to overcome the limitations of conventional liposomes and to extend circulation time. PEGylation tended to improve the physicochemical stability and reverse the chemoresistance in multidrug-resistant (MDR) breast cancer cell lines. In this study, PEGylated formulations of disulfiram (DS) and paclitaxel (PAC) were developed using the ethanol-based proliposome technology. Methods: PEGylated liposomal formulations of disulfiram (DS) and paclitaxel (PAC) were developed using the ethanol-based proliposome approach combined with high-pressure homogenization (HPH). The liposomes were characterized for particle size, polydispersity index (PDI), zeta potential, drug loading efficiency (DLE%), and drug entrapment efficiency (DEE%). Cytotoxicity studies were performed on sensitive (MCF7, MDA-MB-231) and chemoresistant (MDA-MB-231PAC10) breast cancer cell lines using the MTT assay to assess the anti-ancer potential of the formulations. Synergistic cytotoxic effects of DS and PAC co-delivery were also evaluated. Results: There was no significant difference in drug loading (DLE%) and drug entrapment efficiency (EE%) between conventional liposomes and the developed PEGylated vesicles. DS demonstrated higher loading in liposomes than PAC, and a greater cytotoxic effect on both sensitive (MCF7 and MDA-MB-231) and chemoresistant (MDA-MB-231PAC10) human breast cancer cell lines. For both DS- and PAC-loaded liposomes, PEGylation did not compromise the cytotoxic effect on both sensitive and chemoresistant cells. Interestingly, the combination of DS- and PAC-loaded PEGylated liposomes had significantly higher cytotoxic effect and lower IC50 than that of each drug alone. Conclusions: Overall, PEGylated liposomal formulation of DS and PAC acted synergistically to reverse the multidrug resistance in breast cancer cells and could serve as a promising system for delivery of PAC and DS simultaneously in one formulation using an alcohol-based proliposome formulation.
Collapse
Affiliation(s)
- Ammar Said Suliman
- Pharmaceutical Research Group, School of Allied Health, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, Bishops Hall Lane, Chelmsford CM1 1SQ, UK
| | - Sahrish Rehmani
- Pharmaceutical Research Group, School of Allied Health, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, Bishops Hall Lane, Chelmsford CM1 1SQ, UK
- GMPriority Pharma Ltd., Priors Way, Coggeshall, Colchester CO6 1TW, UK
| | - Benjamin Small
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Kate Butcher
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Mouhamad Khoder
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston Upon Thames, London KT1 2EE, UK
| | - Vinodh Kannappan
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Weiguang Wang
- Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Abdelbary Elhissi
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Mohammad Najlah
- Pharmaceutical Research Group, School of Allied Health, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, Bishops Hall Lane, Chelmsford CM1 1SQ, UK
| |
Collapse
|
25
|
Czechowicz P, Więch-Walów A, Sławski J, Collawn JF, Bartoszewski R. Old drugs, new challenges: reassigning drugs for cancer therapies. Cell Mol Biol Lett 2025; 30:27. [PMID: 40038587 PMCID: PMC11881393 DOI: 10.1186/s11658-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug's approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs' potential for new cancer therapies.
Collapse
Affiliation(s)
- Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Anna Więch-Walów
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland.
| |
Collapse
|
26
|
Jones PC, Von Hoff DD. Vitamin A Metabolism and Resistance of Hepatic Metastases to Immunotherapy. Mol Cancer Ther 2025; 24:345-353. [PMID: 39363636 PMCID: PMC11876961 DOI: 10.1158/1535-7163.mct-24-0367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/05/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
The liver is an immune-tolerant organ, allowing for organ transplantation with less immune suppression compared with other organs. It also provides fertile soil for tumor metastases, which tend to be more resistant to checkpoint blockade immunotherapy than metastases in other organs. This resistance may result from the sum of incremental evolutionary adaptions in various cell types to prevent overaction to antigens absorbed from the gut into the portal circulation or it might involve a central mechanism. Here, we propose that metabolism of vitamin A, which is highly concentrated in the liver, is a root source of tolerance and resistance of hepatic metastases to checkpoint blockade. Suppression of retinoic acid synthesis from vitamin A with disulfiram may mitigate tolerance and produce enhanced immunotherapy treatment results for patients with liver metastases.
Collapse
Affiliation(s)
| | - Daniel D. Von Hoff
- HonorHealth Research Institute (HHRI), Scottsdale, Arizona
- Translational Genomics Research Institute (TGen) a Part of City of Hope, Phoenix, Arizona
| |
Collapse
|
27
|
Huang S, Liang X, Shi D, Chen X, Ye S, Liu X, Yang Y, Zou Y, Hu H, Wu H. [ 64Cu]Cu(DDC) 2 NPs: A Novel PET Probe for Noninvasive Visualization of NPL4 Expression in Tumors In Vivo. Mol Pharm 2025; 22:1339-1347. [PMID: 39927715 DOI: 10.1021/acs.molpharmaceut.4c01002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Nuclear protein localization 4 (NPL4) plays a key role in the ubiquitination pathway and has emerged as a promising target for cancer therapy. The ditiocarb-copper complex, Cu(DDC)2, an anticancer metabolite derived from the antialcoholism drug disulfiram (DSF), exhibits a high affinity for NPL4. Thus, quantifying NPL4 expression in tumors is crucial for ubiquitination research and for developing NPL4-targeted diagnostic and therapeutic strategies. In this study, we replaced the cold copper ion in Cu(DDC)2 with the positron-emitting isotope copper-64 and developed three methods for visualizing NPL4 in tumors in vivo using positron emission tomography/computed tomography (PET/CT): (1) an in vivo "synthesis-free" method for preparing [64Cu]Cu(DDC)2, (2) an in vitro synthesis method, and (3) a stabilization method using PEG5000-PLA5000 (PP) to enhance [64Cu]Cu(DDC)2's hydrophilicity by preparing [64Cu]Cu(DDC)2 NPs. Micro-PET/CT imaging showed minimal uptake of [64Cu]Cu(DDC)2 in NPL4-positive tumors with the in vivo "synthesis-free" method, resulting in poor lesion visualization. However, in vitro synthesized [64Cu]Cu(DDC)2 and [64Cu]Cu(DDC)2 NPs successfully visualized NPL4-positive U87MG tumors. Compared to [64Cu]Cu(DDC)2, [64Cu]Cu(DDC)2NPs demonstrated significantly higher tumor uptake (7.2 ± 0.7% ID/g vs 3.8 ± 0.6% ID/g at 12 h postinjection, P = 0.001) and tumor-to-muscle (T/M) ratio (7.8 ± 1.2 vs. 3.2 ± 0.7, P = 0.001). Tumor uptake of [64Cu] Cu (DDC)2NPs was consistent with NPL4 expression levels and was inhibited by an excess of Cu(DDC)2. The optimal PP stabilizer concentration was determined to be 0.0005%. This study successfully developed a PET probe, [64Cu]Cu(DDC)2NPs, and established a novel imaging modality for in vivo visualization of NPL4 expression, potentially guiding future NPL4-targeted therapies.
Collapse
Affiliation(s)
- Shun Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Nuclear Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China
| | - Xiang Liang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Nuclear Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Dazhi Shi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaohui Chen
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Shimin Ye
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinran Liu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yali Yang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yijin Zou
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Huiran Hu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hubing Wu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
28
|
Maduka AO, Manohar S, Foster MW, Silva GM. Localized K63 Ubiquitin Signaling Is Regulated by VCP/p97 During Oxidative Stress. Mol Cell Proteomics 2025; 24:100920. [PMID: 39880084 PMCID: PMC11894314 DOI: 10.1016/j.mcpro.2025.100920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Under stress conditions, cells reprogram their molecular machineries to mitigate damage and promote survival. Ubiquitin signaling is globally increased during oxidative stress, controlling protein fate and supporting stress defenses at several subcellular compartments. However, the rules driving subcellular ubiquitin localization to promote concerted response mechanisms remain understudied. Here, we show that K63-linked polyubiquitin chains, known to promote proteasome-independent pathways, accumulate primarily in noncytosolic compartments during oxidative stress induced by sodium arsenite in mammalian cells. Our subcellular ubiquitin proteomic analyses of noncytosolic compartments expanded 2.5-fold the pool of proteins (2,494) and provided a comprehensive number of sites (10,157) known to be ubiquitinated during arsenite stress, suggesting their involvement in a myriad of cellular pathways. Moreover, subcellular proteome analyses revealed proteins that are recruited to noncytosolic compartments under stress, including a significant enrichment of helper ubiquitin-binding adaptors of the ATPase valosin-containing protein (VCP) that processes ubiquitinated substrates for downstream signaling. We further show that VCP recruitment to noncytosolic compartments under arsenite stress occurs in a ubiquitin-dependent manner mediated by its adaptor NPLOC4. Additionally, we show that VCP and NPLOC4 activities are critical to sustain low levels of noncytosolic K63-linked ubiquitin chains, supporting a cyclical model of ubiquitin conjugation and removal that is disrupted by reactive oxygen species. This work deepens our understanding of the role of localized ubiquitin and VCP signaling in the basic mechanisms of stress response and highlights new pathways and molecular players that are essential to reshape the composition and function of the human subcellular proteome under dynamic environments.
Collapse
Affiliation(s)
- Austin O Maduka
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Sandhya Manohar
- Department of Biology, Institute for Biochemistry, ETH Zürich, Zürich, Switzerland
| | - Matthew W Foster
- Proteomics and Metabolomics Core Facility, Duke University, Durham, North Carolina, USA
| | - Gustavo M Silva
- Department of Biology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
29
|
Wu S, Weng J, Pan Y, Wen Z, Zeng J, Lou Y, Tong S, Liao P, Li N, Yu Z, Xia J. Disulfiram/Cu targeting FOXO6 modulates sensitivity of hepatocellular carcinoma to lenvatinib via disrupt choline metabolic. Cell Signal 2025; 127:111563. [PMID: 39694126 DOI: 10.1016/j.cellsig.2024.111563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Disulfiram/Cu(DSF/Cu) has a known pharmacokinetic and safety profile, exerting a strong antitumor effect. Oral tyrosine kinase inhibitors including lenvatinib are approved as first-line therapy for treating advanced unresectable hepatocellular carcinoma (HCC). These patients still have limited survival due to drug resistance. Disulfiram/Cu and lenvatinib are the promising antitumor treatments. In this study, we studied whether Disulfiram/Cu increased lenvatinib sensitivity in HCC cells. Moreover, the potential drug targets of Disulfiram/Cu and associated mechanisms were explored. We mainly investigated Autophagic flux was determined via immunofluorescence analysis and confocal microscopy. p-PI3K, p-AKT, p62, LC3B, FOXO6, and CHKA proteins associated with autophagy were detected by immunoblotting. In addition, antitumour activity of Disulfiram/Cu in combination with lenvatinib was examined in vivo through construction of the nude mouse transplant tumor model. Furthermore, our results show disulfiram/Cu combined with lenvatinib exerted the synergistic impact on treating HCC in vitro. Mechanistically, transcriptome combined with metabolome reveals Disulfiram/Cu targeting FOXO6 induction of autophagy mediated inhibits cell growth in hepatocellular carcinoma by downregulating CHKα for inhibiting AKT pathway activation while blocking choline metabolic reprogramming in HCC. These effects mostly explain the tumor-promoting effect of FOXO6 on HCC. In general, the results illustrate the mechanistic associations between metabolites and tumor cell malignant phenotype, contributing to developing new anti-HCC pharmacological treatments by Inhibiting FOXO6 for disrupting choline metabolic pathway.
Collapse
Affiliation(s)
- Shiyi Wu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jialu Weng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Yating Pan
- Department of Respiratory Medicine, Yongkang First People's Hospital, Yongkang 321300, China
| | - Zhikai Wen
- Department of Liver and Gall Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jing Zeng
- Department of Otorhinolaryngology, Hanshou County Hospital of Traditional Chinese Medicine, Changdei 415900, China
| | - Yunwei Lou
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Songjian Tong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Pan Liao
- The School of Medicine, Nankai University 94 Weijin Road, Tianjin 300071, China
| | - Na Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Zhijie Yu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jinglin Xia
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Liver Cancer Institute, Zhongshan Hospital of Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
30
|
Wang L, Liu Z, Xu Z, Wang W, Yang J, Zhang J, He S, Liang Q, Li T. Repurposing alcohol-abuse drug disulfiram for the treatment of KSHV-infected primary effusion lymphoma by activating antiviral innate immunity. PLoS Pathog 2025; 21:e1012957. [PMID: 40036222 PMCID: PMC11922253 DOI: 10.1371/journal.ppat.1012957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/19/2025] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
Cancer remains a leading cause of global mortality, characterized by high treatment costs, and generally poor prognoses. Developing new anti-cancer drugs requires substantial investment, extended development timelines, and a high failure rate. Therefore, repurposing existing US Food and Drug Administration (FDA)-approved drugs for other diseases as potential anti-cancer therapies offers a faster and more cost-effective approach. Primary effusion lymphoma (PEL) is an aggressive B-cell malignancy linked to Kaposi's sarcoma-associated herpesvirus (KSHV) infection. In this study, we identified that disulfiram (DSF), an FDA-approved medication for alcohol dependence, acts as a potent inhibitor of KSHV-positive PEL. DSF suppresses PEL cell proliferation by inducing apoptosis through the activation of innate antiviral immunity. Remarkably, DSF effectively impedes KSHV reactivation and virion production in both PEL and endothelial cells. Inhibition of TANK binding kinase 1 (TBK1) or interferon regulatory factor 3 (IRF3), essential activators of antiviral innate immunity, reverses DSF's effects on PEL cell survival and KSHV reactivation. Furthermore, DSF treatment significantly hinders the initiation and progression of PEL tumors in a xenograft mouse model, with this effect was notably abolished by TBK1 depletion. Our findings highlighted DSF as a promising therapeutic agent for targeting persistent KSHV infection and treating PEL tumors.
Collapse
Affiliation(s)
- Lijie Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| | - Zhenshan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyu Xu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| | - Wenjing Wang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| | - Jinhong Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| | - Junjie Zhang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| | - Shanping He
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| | - Qiming Liang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China,
| |
Collapse
|
31
|
Wang H, Liang L, Xie Y, Gong H, Fan F, Wen C, Jiang Y, Lei S, Qiu X, Peng H, Ye M, Xiao X, Liu J. Pseudokinase TRIB3 stabilizes SSRP1 via USP10-mediated deubiquitination to promote multiple myeloma progression. Oncogene 2025; 44:694-708. [PMID: 39653795 DOI: 10.1038/s41388-024-03245-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/17/2024] [Accepted: 11/29/2024] [Indexed: 03/05/2025]
Abstract
Multiple myeloma (MM), the world's second most common hematologic malignancy, poses considerable clinical challenges due to its aggressive progression and resistance to therapy. Addressing these challenges requires a detailed understanding of the mechanisms driving MM initiation, progression, and therapeutic resistance. This study identifies the pseudokinase tribble homolog 3 (TRIB3) as a high-risk factor that promotes MM malignancy in vitro and in vivo. Mechanistically, TRIB3 directly interacts with structure-specific recognition protein 1 (SSRP1) and ubiquitin-specific peptidase 10 (USP10), facilitating the formation of a TRIB3/USP10/SSRP1 ternary complex. This complex stabilizes SSRP1 via USP10-mediated deubiquitination, thereby driving MM cell proliferation. Furthermore, a stapled peptide, SP-A, was developed, which effectively disrupts the TRIB3/USP10/SSRP1 complex, leading to a decrease in SSRP1 levels by inhibiting its stabilization through USP10. Notably, SP-A exhibits strong synergistic effects when combined with the proteasome inhibitor bortezomib. Given the critical role of the TRIB3/USP10/SSRP1 complex in MM pathophysiology, it represents a promising therapeutic target for MM treatment. In MM cells, TRIB3, USP10 and SSRP1 form a ternary complex and TRIB3 enhances the deubiquitinating effect of USP10 on SSRP1, leading to malignant progression of MM. In the case of drug intervention, SP-A attenuates the binding of SSRP1 and USP10 by inhibiting protein interactions between TRIB3 and SSRP1 and promoted SSRP1 protein degradation, leading to significant inhibition of MM development. Visual abstract created with Biorender.
Collapse
Affiliation(s)
- Haiqin Wang
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Long Liang
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Yifang Xie
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Han Gong
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Feifan Fan
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Chengcai Wen
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Yu Jiang
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Shiying Lei
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Xili Qiu
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China
| | - Hongling Peng
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China.
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China.
| | - Xiaojuan Xiao
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China.
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital; School of Life Sciences; Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
32
|
Feng Y, Chen H, Chen S, Zhang K, Yun D, Liu D, Zeng J, Yang C, Xie Q. Disulfiram-Loaded PLGA nanoparticles modified with a Phenyl borate chitosan Conjugate enhance hepatic carcinoma treatment. Int J Pharm 2025; 671:125293. [PMID: 39880140 DOI: 10.1016/j.ijpharm.2025.125293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Disulfiram (DSF), which has been traditionally used to treat alcoholism, has been shown to inhibit tumor growth, indicating its potential as an anticancer agent. However, its development and application are hindered by its poor water solubility, instability in physiological environments, and low bioavailability. In this study, phenylboronic acid-chitosan (PBA-CS) grafts were synthesized using the carbodiimide method. PBA-CS-modified DSF PLGA nanoparticles (DSF@PBA-CS-PLGA NPs) were constructed by coating the nanoparticle surfaces with PBA-CS to improve the stability of DSF in physiological environments and enhance its anti-tumor effects. The structures of PBA-CS and the DSF@PBA-CS-PLGA NPs were confirmed using FTIR UVs, DLS, ELS, TEM, 1HNMR, DSC. Our in vitro degradation experiments showed that PBA-CS-PLGA NPs significantly improved the stability of DSF in physiological environments. Cell experiments showed that PBA-CS-PLGA NPs improved drug uptake and strongly inhibited HepG2 cell migration. A mouse tumor model was established using Dutch H22 cells. DSF@PBA-CS-PLGA NPs showed better tumor-targeting ability than DSF@PLGA NPs, with a tumor inhibition rate of more than 60%, and they induced apoptosis and inhibited neovascularization in mouse tumor tissues. Both the in vitro and in vivo experiments indicated that the DSF@PBA-CS-PLGA NPs overcame the limitations of DSF, improving the dissolution rate and stability of the drug, ultimately offering low toxicity, sustained release, and targeted delivery. These findings demonstrated the potential of DSF@PBA-CS-PLGA NPs for hepatic carcinoma therapy.
Collapse
Affiliation(s)
- Yanyi Feng
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Hongyu Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Simiao Chen
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Kaijun Zhang
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Dan Yun
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Dengyuan Liu
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Jinxin Zeng
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Chutong Yang
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China
| | - Qingchun Xie
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006 China; Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou 510006 China; Guangdong Provincial Engineering Center of Topical Precision Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006 China.
| |
Collapse
|
33
|
Cvek B. The rules often neglected in current medicinal chemistry. Expert Opin Drug Discov 2025:1-3. [PMID: 39921519 DOI: 10.1080/17460441.2025.2465370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/17/2025] [Accepted: 02/07/2025] [Indexed: 02/10/2025]
Affiliation(s)
- Boris Cvek
- Olomouc University Social Health Institute (OUSHI), Palacky University, Olomouc, Czech Republic
| |
Collapse
|
34
|
Dumut DC, Hajduch M, Zacharias AM, Duan Q, Frydrych I, Rozankova Z, Popper M, Garic D, Paun RA, Centorame A, Shah J, Mistrik M, Dzubak P, De Sanctis JB, Radzioch D. Diethyldithiocarbamate-copper complex ignites the tumor microenvironment through NKG2D-NKG2DL axis. Front Immunol 2025; 16:1491450. [PMID: 40013140 PMCID: PMC11860975 DOI: 10.3389/fimmu.2025.1491450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/17/2025] [Indexed: 02/28/2025] Open
Abstract
Advanced metastatic colorectal cancer (CRC) with deficient DNA mismatch repair (MMR-d), or immune-hot CRCs, show significantly improved clinical outcomes compared to MMR-proficient (MMR-p), or immune-cold CRCs. While the prior represents about 5% of all CRCs, the latter represent 95% and are characterized by low immunogenicity. This study investigates bis-diethyldithiocarbamate (CuET), a novel anticancer compound, and its impact on the colorectal cancer tumor microenvironment (TME). CuET is shown to convert immunologically inactive tumors into hotbeds of antitumor immune responses, marked by increased lymphocyte infiltration, heightened cytotoxicity of natural killer (NK) and T cells, and enhanced non-self recognition by lymphocytes. The potent anticancer cytotoxicity and in vivo safety and efficacy of CuET are established. In summary, CuET transforms the colorectal cancer TME, bolstering NK and T cell cytotoxicity and refining tumor cell recognition through non-classical activation via the NKG2D/NKG2DL axis. This study unveils a novel mechanism of action for CuET: a potent immunomodulator capable of turning cold tumors hot.
Collapse
Affiliation(s)
- Daciana C. Dumut
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Amanda M. Zacharias
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
| | - Qingling Duan
- Department of Biomedical & Molecular Sciences, Faculty of Health Sciences, Queen’s University, Kingston, ON, Canada
- School of Computing, Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Ivo Frydrych
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
| | - Zuzana Rozankova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Miroslav Popper
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Dusan Garic
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Radu Alexandru Paun
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Amanda Centorame
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Juhi Shah
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Juan B. De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| | - Danuta Radzioch
- Department of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Centre, Infectious Diseases in Global Health Program, Montreal, QC, Canada
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia
- Czech Advanced Technology and Research Institute, Palacky University Olomouc, Olomouc, Czechia
| |
Collapse
|
35
|
Jia J, Lin R, Liu M, Hou M, Yu H, Lu Q, Ma Y, Zhao T, Zhang F, Mady MF, Elzatahry AA, Wang J, Ji Y, Zhao D, Li X. Dual-Ligand Assisted Anisotropic Assembly for the Construction of NIR-II Light-Propelled Mesoporous Nanomotors. J Am Chem Soc 2025; 147:4198-4209. [PMID: 39871601 DOI: 10.1021/jacs.4c14011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
The advent of autonomous nanomotors presents exciting opportunities for nanodrug delivery. However, significant potential remains for enhancing the asymmetry of nanomotors and advancing the development of second near-infrared (NIR-II) light-propelled nanomotors capable of operating within deep tissues. Herein, we developed a dual-ligand assisted anisotropic assembly strategy that enables precise regulation of the interfacial energy between selenium (Se) nanoparticle and periodic mesoporous organosilica (PMO). This strategy facilitates the controllable anisotropic growth of PMO on the Se nanoparticle, leading to the formation of Se&PMO Janus nanohybrids. The exposure ratio of the Se subunit within the Janus nanohybrids can be finely tuned from 0% to 75%. Leveraging the transformability of the Se subunit, a variety of functional MxSe&PMO Janus nanocomposites (MxSe denotes metal selenide) were further derived. As a proof of concept, CuSe&PMO Janus nanohybrids, with NIR-II photothermal properties, were employed as NIR-II light-driven nanomotors. By precisely controlling the exposure ratio of the CuSe subunit within the Janus nanostructure, these CuSe&PMO nanomotors achieved optimal self-propulsion, thus enhancing cellular uptake and promoting deep tumor penetration. Furthermore, the high loading capacity and hydrophobic framework of the PMO subunit enabled the incorporation of hydrophobic disulfiram, thereby significantly boosting the efficacy of synergistic active-motion photothermal therapy.
Collapse
Affiliation(s)
- Jia Jia
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Runfeng Lin
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Minchao Liu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Mengmeng Hou
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Hongyue Yu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Qianqian Lu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Yuzhu Ma
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Tiancong Zhao
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Fan Zhang
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Mohamed F Mady
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, P.O. Box 2713, Doha 2713, Qatar
| | - Ahmed A Elzatahry
- William A. Brookshire Department of Chemical and Biomolecular Engineering, Cullen College of Engineering, University of Houston, Houston, Texas 77204, United States
| | - Jiawen Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Yujin Ji
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Dongyuan Zhao
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| | - Xiaomin Li
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, P. R. China
| |
Collapse
|
36
|
Yuan M, Shi L, Liu Y, Xiang K, Zhang Y, Zhou Y, Wang J, Ji M, Hou P. Disulfiram/copper triggers cGAS-STING innate immunity pathway via ROS-induced DNA damage that potentiates antitumor response to PD-1 checkpoint blockade. Int J Biol Sci 2025; 21:1730-1748. [PMID: 39990655 PMCID: PMC11844283 DOI: 10.7150/ijbs.105575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
Immune checkpoint blockades (ICBs) have emerged as the leading strategy for treating advanced malignancies; however, their clinical efficacy is frequently constrained by primary or acquired resistance. Harnessing innate immune signaling to increase lymphocyte infiltration into tumors has been recognized a promising approach to augment the anti-cancer immune response to ICBs. Disulfiram (DSF), an FDA-approved drug for chronic alcoholism, has shown potent anti-tumor effect, particularly when used in combination with copper (Cu). Here, we demonstrated a combination treatment of DSF and Cu (DSF/Cu) robustly activated cancer cell-intrinsic cGAS-STING-dependent innate immune signaling pathway. Further studies revealed that DSF/Cu caused mitochondrial and nuclear DNA damage and the release of cytosolic dsDNA by inducing excessive reactive oxygen species (ROS) generation, thereby triggering innate immunity and enhancing anti-tumor effects. Moreover, DSF/Cu significantly increased the intratumoral infiltration of CD8+ cytotoxic lymphocytes and natural killer (NK) cells, and potentiated the therapeutic efficacy of PD-1 checkpoint blockade in murine tumor models. Overall, our findings provide a rationale underlying the anti-cancer and immunomodulatory function of DSF/Cu and highlight the potential of repurposing DSF to improve responses to ICBs in cancer patients.
Collapse
Affiliation(s)
- Mengmeng Yuan
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Liang Shi
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yan Liu
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ke Xiang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yan Zhang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ye Zhou
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jianling Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Peng Hou
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| |
Collapse
|
37
|
Xiao C, Wang X, Li S, Zhang Z, Li J, Deng Q, Chen X, Yang X, Li Z. A cuproptosis-based nanomedicine suppresses triple negative breast cancers by regulating tumor microenvironment and eliminating cancer stem cells. Biomaterials 2025; 313:122763. [PMID: 39180917 DOI: 10.1016/j.biomaterials.2024.122763] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/04/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
Cuproptosis is a new kind of cell death that depends on delivering copper ions into mitochondria to trigger the aggradation of tricarboxylic acid (TCA) cycle proteins and has been observed in various cancer cells. However, whether cuproptosis occurs in cancer stem cells (CSCs) is unexplored thus far, and CSCs often reside in a hypoxic tumor microenvironment (TME) of triple negative breast cancers (TNBC), which suppresses the expression of the cuproptosis protein FDX1, thereby diminishing anticancer efficacy of cuproptosis. Herein, a ROS-responsive active targeting cuproptosis-based nanomedicine CuET@PHF is developed by stabilizing copper ionophores CuET nanocrystals with polydopamine and hydroxyethyl starch to eradicate CSCs. By taking advantage of the photothermal effects of CuET@PHF, tumor hypoxia is overcome via tumor mechanics normalization, thereby leading to enhanced cuproptosis and immunogenic cell death in 4T1 CSCs. As a result, the integration of CuET@PHF and mild photothermal therapy not only significantly suppresses tumor growth but also effectively inhibits tumor recurrence and distant metastasis by eliminating CSCs and augmenting antitumor immune responses. This study presents the first evidence of cuproptosis in CSCs, reveals that disrupting hypoxia augments cuproptosis cancer therapy, and establishes a paradigm for potent cancer therapy by simultaneously eliminating CSCs and boosting antitumor immunity.
Collapse
Affiliation(s)
- Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xing Wang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Shiyou Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Zhijie Zhang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Jiayuan Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Qingyuan Deng
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xiang Chen
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, PR China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
| |
Collapse
|
38
|
Safari MH, Rahimzadeh P, Alaei E, Alimohammadi M, Esfandiari N, Daneshi S, Malgard N, Farahani N, Taheriazam A, Hashemi M. Targeting ferroptosis in gastrointestinal tumors: Interplay of iron-dependent cell death and autophagy. Mol Cell Probes 2025; 79:102013. [PMID: 39837469 DOI: 10.1016/j.mcp.2025.102013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Ferroptosis is a regulated cell death mechanism distinct from apoptosis, autophagy, and necroptosis, marked by iron accumulation and lipid peroxidation. Since its identification in 2012, it has developed into a potential therapeutic target, especially concerning GI disorders like PC, HCC, GC, and CRC. This interest arises from the distinctive role of ferroptosis in the progression of diseases, presenting a new avenue for treatment where existing therapies fall short. Recent studies emphasize the promise of focusing on ferroptosis to fight GI cancers, showcasing its unique pathophysiological mechanisms compared to other types of cell death. By comprehending how ferroptosis aids in the onset and advancement of GI diseases, scientists aim to discover novel drug targets and treatment approaches. Investigating ferroptosis in gastrointestinal disorders reveals exciting possibilities for novel therapies, potentially revolutionizing cancer treatment and providing renewed hope for individuals affected by these tumors.
Collapse
Affiliation(s)
- Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Negin Esfandiari
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Neda Malgard
- Department of Internal Medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
39
|
Chen Y, Liu L, Li M, Chen X, Li Y, Tao J, Deng Y. Nanoparticle-enabled In Situ drug potency activation for enhanced tumor-specific therapy. Eur J Pharm Sci 2025; 205:106989. [PMID: 39675436 DOI: 10.1016/j.ejps.2024.106989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Cancer treatment faces significant challenges including inadequate tumor specificity, drug resistance, and severe side effects, often resulting in unsatisfactory patient outcomes. Nanomedicines offer a transformative platform for tumor-targeted drug delivery and antitumor potency activation, providing an indispensable strategy for overcoming the severe damage to normal tissues caused by the inherent "always-on" cytotoxicity of conventional therapeutic agents. This review focuses on the emerging concept of "nanoparticle-enabled in situ drug potency activation", where inactive or minimally toxic agents are selectively activated within tumors to enhance the therapeutic efficacy and minimize the adverse effects. We systematically analyzed literature from PubMed and Web of Science databases spanning the last two decades, emphasizing experimental evidence supporting this in situ drug potency activation concept. Key strategies including stimuli-responsive prodrug nanoparticles, metal-induced activation, and bioorthogonal reactions are critically evaluated for their potential to overcome limitations in current cancer therapies. The findings highlight the potential of in situ potency activation as a promising alternative to conventional therapeutics, with far-reaching implications for advancing effective and safe cancer treatments.
Collapse
Affiliation(s)
- Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Lishan Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ming Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xiaolian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yaoqi Li
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jing Tao
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
40
|
Villegas-Vazquez EY, Marín-Carrasco FP, Reyes-Hernández OD, Báez-González AS, Bustamante-Montes LP, Padilla-Benavides T, Quintas-Granados LI, Figueroa-González G. Revolutionizing ovarian cancer therapy by drug repositioning for accelerated and cost-effective treatments. Front Oncol 2025; 14:1514120. [PMID: 39876896 PMCID: PMC11772297 DOI: 10.3389/fonc.2024.1514120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Drug repositioning, the practice of identifying novel applications for existing drugs beyond their originally intended medical indications, stands as a transformative strategy revolutionizing pharmaceutical productivity. In contrast to conventional drug development approaches, this innovative method has proven to be exceptionally effective. This is particularly relevant for cancer therapy, where the demand for groundbreaking treatments continues to grow. This review focuses on drug repositioning for ovarian cancer treatment, showcasing a comprehensive exploration grounded in thorough in vitro experiments across diverse cancer cell lines, which are validated through preclinical in vivo models. These insights not only shed light on the efficacy of these drugs but also expand in potential synergies with other pharmaceutical agents, favoring the development of cost-effective treatments for cancer patients.
Collapse
Affiliation(s)
- Edgar Yebran Villegas-Vazquez
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Francisco Pável Marín-Carrasco
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Octavio Daniel Reyes-Hernández
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Andrea S. Báez-González
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, United States
| | | | | | - Laura Itzel Quintas-Granados
- Colegio de Ciencias y Humanidades, Plantel Cuautepec, Universidad Autónoma de la Ciudad de México, Ciudad de México, Mexico
| | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
41
|
Imam M, Ji J, Zhang Z, Yan S. Targeting the initiator to activate both ferroptosis and cuproptosis for breast cancer treatment: progress and possibility for clinical application. Front Pharmacol 2025; 15:1493188. [PMID: 39867656 PMCID: PMC11757020 DOI: 10.3389/fphar.2024.1493188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/28/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide. Metal metabolism is pivotal for regulating cell fate and drug sensitivity in breast cancer. Iron and copper are essential metal ions critical for maintaining cellular function. The accumulation of iron and copper ions triggers distinct cell death pathways, known as ferroptosis and cuproptosis, respectively. Ferroptosis is characterized by iron-dependent lipid peroxidation, while cuproptosis involves copper-induced oxidative stress. They are increasingly recognized as promising targets for the development of anticancer drugs. Recently, compelling evidence demonstrated that the interplay between ferroptosis and cuproptosis plays a crucial role in regulating breast cancer progression. This review elucidates the converging pathways of ferroptosis and cuproptosis in breast cancer. Moreover, we examined the value of genes associated with ferroptosis and cuproptosis in the clinical diagnosis and treatment of breast cancer, mainly outlining the potential for a co-targeting approach. Lastly, we delve into the current challenges and limitations of this strategy. In general, this review offers an overview of the interaction between ferroptosis and cuproptosis in breast cancer, offering valuable perspectives for further research and clinical treatment.
Collapse
Affiliation(s)
| | | | | | - Shunchao Yan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
42
|
Long J, Yang S, Bian Z, Zhu H, Ma M, Wang X, Li L, Zhang W, Han Y, Gershwin ME, Lian Z, Zhao Z. PD-1 +CD8 + T Cell-Mediated Hepatocyte Pyroptosis Promotes Progression of Murine Autoimmune Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407284. [PMID: 39494472 PMCID: PMC11714232 DOI: 10.1002/advs.202407284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/10/2024] [Indexed: 11/05/2024]
Abstract
The specific mechanisms underlying effector pathways in autoimmune liver disease remain enigmatic and therefore constructing appropriate murine models to investigate disease pathogenesis becomes critical. A spontaneous severe murine model of autoimmune liver disease has been previously established in dnTGFβRII Aire-/- mice, exhibiting disease phenotypes that resemble both human primary biliary cholangitis (PBC) and autoimmune hepatitis (AIH). The data suggests that auto-reactive liver-specific CD8+ T cells are the primary pathogenic cells in liver injury. In this study, these data are advanced through the use of both single-cell sequencing and extensive in vitro analysis. The results identify a specific expanded pathogenic subset of PD-1+CD8+ T cells in the liver, exhibiting strong functional activity and cytotoxicity against target cells. Depletion of PD-1+CD8+ T cells using CAR-T cells effectively alleviates the disease. GSDMD-mediated pyroptosis is found to be aberrantly activated in the livers of model mice, and treatment with a GSDMD-specific inhibitor significantly inhibits disease progression. In vitro experiments reveal that PD-1+CD8+ T cells can induce the pyroptosis of hepatocytes through elevated production of granzyme B and perforin-1. These results provide a novel explanation for the cytotoxic activity of pathogenic liver PD-1+CD8+ T cells in autoimmune liver diseases and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Jie Long
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Si‐Yu Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zhen‐Hua Bian
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou International CampusGuangzhou511442China
| | - Hao‐Xian Zhu
- School of MedicineSouth China University of TechnologyGuangzhou510006China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Min Ma
- School of MedicineSouth China University of TechnologyGuangzhou510006China
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Xiao‐Qing Wang
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Liang Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California DavisDavisCA95616USA
| | - Ying Han
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive DiseasesAir Force Military Medical UniversityXi'an710000China
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California DavisDavisCA95616USA
| | - Zhe‐Xiong Lian
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| | - Zhi‐Bin Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhou510080China
| |
Collapse
|
43
|
Lutsenko S, Roy S, Tsvetkov P. Mammalian copper homeostasis: physiological roles and molecular mechanisms. Physiol Rev 2025; 105:441-491. [PMID: 39172219 PMCID: PMC11918410 DOI: 10.1152/physrev.00011.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024] Open
Abstract
In the past decade, evidence for the numerous roles of copper (Cu) in mammalian physiology has grown exponentially. The discoveries of Cu involvement in cell signaling, autophagy, cell motility, differentiation, and regulated cell death (cuproptosis) have markedly extended the list of already known functions of Cu, such as a cofactor of essential metabolic enzymes, a protein structural component, and a regulator of protein trafficking. Novel and unexpected functions of Cu transporting proteins and enzymes have been identified, and new disorders of Cu homeostasis have been described. Significant progress has been made in the mechanistic studies of two classic disorders of Cu metabolism, Menkes disease and Wilson's disease, which paved the way for novel approaches to their treatment. The discovery of cuproptosis and the role of Cu in cell metastatic growth have markedly increased interest in targeting Cu homeostatic pathways to treat cancer. In this review, we summarize the established concepts in the field of mammalian Cu physiology and discuss how new discoveries of the past decade expand and modify these concepts. The roles of Cu in brain metabolism and in cell functional speciation and a recently discovered regulated cell death have attracted significant attention and are highlighted in this review.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Department of Physiology, Johns Hopkins Medical Institutes, Baltimore, Maryland, United States
| | - Shubhrajit Roy
- Department of Physiology, Johns Hopkins Medical Institutes, Baltimore, Maryland, United States
| | - Peter Tsvetkov
- Department of Pathology, Cancer Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| |
Collapse
|
44
|
Ohno K, Murakami H, Ogo N, Asai A. Imaging phenotype reveals that disulfirams induce protein insolubility in the mitochondrial matrix. Sci Rep 2024; 14:31401. [PMID: 39733149 PMCID: PMC11682119 DOI: 10.1038/s41598-024-82939-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The cell painting assay is useful for understanding cellular phenotypic changes and drug effects. To identify other aspects of well-known chemicals, we screened 258 compounds with the cell painting assay and focused on a mitochondrial punctate phenotype seen with disulfiram. To elucidate the reason for this punctate phenotype, we looked for clues by examining staining steps and gene knockdown as well as examining protein solubility and comparing cell lines. From these results, we found that the punctate phenotype was caused by protein insolubility in the mitochondrial matrix. Interestingly, the punctate phenotype of disulfiram was sensitive to the relative expression of LonP1, a protease in the mitochondrial matrix that regulates proteostasis, suggesting that the punctate phenotype manifests when the protein quality control capacity in the mitochondrial matrix is exceeded. Moreover, we discovered that disulfiram and its derivatives, which have all been reported to increase acetaldehyde in the blood after the in vivo intake of alcohol, induced a punctate phenotype as well. The investigated punctate phenotype not only provides a novel clue for elucidating the common mechanism of action among disulfiram derivatives but is also a novel example of chemical perturbation of proteostasis in the mitochondrial matrix.
Collapse
Affiliation(s)
- Ken Ohno
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan
- Discovery Technology Laboratories, Sohyaku Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Muraoka-Higashi, Fujisawa, 251-8555, Kanagawa, Japan
| | - Hisashi Murakami
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Suruga-ku, Shizuoka, 422-8526, Shizuoka, Japan.
| |
Collapse
|
45
|
Butcher K, Wang Z, Kurusamy S, Zhang Z, Morris MR, Najlah M, McConville C, Kannappan V, Wang W. PLGA-Nano-Encapsulated Disulfiram Inhibits Cancer Stem Cells and Targets Non-Small Cell Lung Cancer In Vitro and In Vivo. Biomolecules 2024; 14:1651. [PMID: 39766358 PMCID: PMC11674892 DOI: 10.3390/biom14121651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer stem cells (CSCs) play a key role in non-small cell lung cancer (NSCLC) chemoresistance and metastasis. In this study, we used two NSCLC cell lines to investigate the regulating effect of hypoxia in the induction and maintenance of CSC traits. Our study demonstrated hypoxia-induced stemness and chemoresistance at levels comparable to those in typical CSC sphere culture. Activation of the NF-κB pathway (by transfection of NF-κB-p65) plays a key role in NSCLC CSCs and chemoresistance. Disulfiram (DS), an anti-alcoholism drug, showed a strong in vitro anti-CSC effect. It blocked cancer cell sphere reformation and clonogenicity, synergistically enhanced the cytotoxicity of four anti-NSCLC drugs (doxorubicin, gemcitabine, oxaliplatin and paclitaxel) and reversed hypoxia-induced resistance. The effect of DS on CSCs is copper-dependent. A very short half-life in the bloodstream is the major limitation for the translation of DS into a cancer treatment. Our team previously developed a poly lactic-co-glycolic acid (PLGA) nanoparticle encapsulated DS (DS-PLGA) with a long half-life in the bloodstream. Intra venous injection of DS-PLGA in combination with the oral application of copper gluconate has strong anticancer efficacy in a metastatic NSCLC mouse model. Further study may be able to translate DS-PLGA into cancer applications.
Collapse
Affiliation(s)
- Kate Butcher
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- Disulfican Ltd., Wolverhampton WV9 5HD, UK
| | - Zhipeng Wang
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Sathishkumar Kurusamy
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- School of Biosciences, Division of Natural Sciences, University of Kent, Canterbury CT2 7NZ, UK
| | - Zaixing Zhang
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of North China University of Science and Technology, Tangshan 063000, China
| | - Mark R. Morris
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Mohammad Najlah
- Faculty of Health, Medicine and Social Care, Anglia Ruskin University, Cambridge CB1 1PT, UK;
| | | | - Vinodh Kannappan
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- Disulfican Ltd., Wolverhampton WV9 5HD, UK
| | - Weiguang Wang
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, UK
- Disulfican Ltd., Wolverhampton WV9 5HD, UK
| |
Collapse
|
46
|
Jiang Q, Tong F, Xu Y, Liu C, Xu Q. Cuproptosis: a promising new target for breast cancer therapy. Cancer Cell Int 2024; 24:414. [PMID: 39702350 DOI: 10.1186/s12935-024-03572-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/10/2024] [Indexed: 12/21/2024] Open
Abstract
Breast cancer (BC) is the leading cause of cancer-related mortality among women globally, affecting approximately one-quarter of all female cancer patients and accounting for one-sixth of cancer-related deaths in women. Despite significant advancements in diagnostic and therapeutic approaches, breast cancer treatment remains challenging due to issues such as recurrence and metastasis. Recently, a novel form of regulated cell death, termed cuproptosis, has been identified. This process disrupts mitochondrial respiration by targeting the copper-dependent cellular pathways. The role of cuproptosis has been extensively investigated in various therapeutic contexts, including chemotherapy, immunotherapy, radiotherapy, and nanotherapy, with the development of novel drugs significantly improving clinical outcomes. This article aims to further elucidate the connection between cuproptosis and breast cancer, focusing on its therapeutic targets, signaling pathways, and potential biomarkers that could enhance treatment strategies. These insights may offer new opportunities for improved patient care and outcomes in breast cancer therapy.
Collapse
Affiliation(s)
- Qianqian Jiang
- Department of Pharmacy, Traditional Chinese Medicine Hospital of Changshan, Quzhou, 324200, P.R. China
| | - Fei Tong
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, P.R. China
| | - Yun Xu
- Department of Pharmacy, Zhejiang Medical&Health Group Hangzhou Hospital, Hangzhou, Zhejiang, 310022, China
| | - Cheng Liu
- Department of Pharmacy, The Secend People's Hospital Of Jiande, Hangzhou, 311604, P.R. China
| | - Qiaoping Xu
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Cancer Center, Afliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
47
|
Hu J, Zhu J, Chen T, Zhao Y, Xu Q, Wang Y. Cuproptosis in cancer therapy: mechanisms, therapeutic application and future prospects. J Mater Chem B 2024; 12:12191-12206. [PMID: 39526989 DOI: 10.1039/d4tb01877j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Cuproptosis is a regulated form of cell death induced by the accumulation of metal ions and is closely linked to aspects of cellular drug resistance, cellular metabolism, and signalling pathways. Due to its crucial role in regulating physiological and pathological processes, cuproptosis has gained increasing significance as a potential target for anticancer drug development. In this review, we introduce the definition of cuproptosis and provide a comprehensive discussion of the mechanisms of cuproptosis. In addition, the methods for the detection of cuproptosis are summarized, and recent advances in cuproptosis in cancer therapy are reviewed, mainly in terms of elesclomol (ES)-mediated cuproptosis and disulfiram (DSF)-mediated cuproptosis, which provided practical value for applications. Finally, the current challenges and future development of cuproptosis-mediated cancer therapy are discussed. In summary, this review highlights recent progress on cuproptosis in cancer therapy, offering novel ideas and strategies for future research and applications.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Tao Chen
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Department of Pharmacy, Wannan Medical College, Wuhu, China
| |
Collapse
|
48
|
Huang D, Yao Y, Lou Y, Kou L, Yao Q, Chen R. Disulfiram and cancer immunotherapy: Advanced nano-delivery systems and potential therapeutic strategies. Int J Pharm X 2024; 8:100307. [PMID: 39678262 PMCID: PMC11638648 DOI: 10.1016/j.ijpx.2024.100307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
The initial focus of the clinical application of disulfiram was its efficacy in treating alcoholism. However, recent research has revealed its potential as an anti-tumor agent and even as an enhancer of cancer immunotherapy. Disulfiram has received safety approval from the FDA, indicating its safety advantages over other substances used for disease treatment. Although clinical trials have been conducted on strategies involving disulfiram or its combination with other anti-tumor drugs, the treatment outcomes have not yielded satisfactory results, thereby emphasizing the significance of addressing drug delivery as a crucial challenge to be resolved. The need to explore advanced nano-delivery systems and the potential immunotherapy enhancement effect of disulfiram in cancer treatment has increased. This review highlights various ways in which disulfiram can combat cancer and importantly, activate immune-related mechanisms. It also discusses obstacles related to delivering disulfiram and provides existing solutions in terms of drug delivery. These drug delivery strategies offer solutions to address various challenges encountered in diverse delivery methods and aim to achieve enhanced therapeutic effects. The focus is on recent advancements in disulfiram delivery strategies and the future potential of disulfiram in immune regulation.
Collapse
Affiliation(s)
- Di Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yinsha Yao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Yifei Lou
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
49
|
Ye W, Lv H, Zhang Q, Zhao J, Zhao X, Zhao G, Yan C, Sun F, Zhao Z, Jia X. A cisplatin and disulphiram co-loaded inclusion complex overcomes drug resistance by inhibiting cancer cell stemness in non-small cell lung cancer. J Drug Target 2024; 32:159-171. [PMID: 38133515 DOI: 10.1080/1061186x.2023.2298844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Introduction: Non-small cell lung cancer (NSCLC) accounting for about 80-85% of all lung cancer cases is one of the fastest-growing malignancies in terms of incidence and mortality worldwide and is commonly treated with cisplatin (DDP). Although treatment may initially be effective, the DDP therapy often leads to the development of chemoresistance and treatment failure. Disulphiram (DSF), an old alcohol-aversion drug, has been revealed to help reverse drug resistance in several cancers. In addition, several studies have shown a close relationship between drug resistance and cancer cell stemness.Methods: In this study, DDP and DSF were embedded in hydroxypropyl-β-cyclodextrin (CD) to prepare a co-loaded inclusion complex of DDP and DSF (DDP-DSF/CD) with enhanced solubility and therapeutic effects. The effects and mechanism of DSF on the DDP resistance from the perspective of cancer cell stemness were determined.Results: Our data show that DDP-DSF/CD increased cytotoxicity and apoptosis of DDP-resistant A549 (A549/DDP) cells, inhibited stem cell transcriptional regulatory genes and drug resistance-associated proteins and reversed the DDP resistance in vitro and in vivo.Discussion: Overall, DDP-DSF/CD could be a promising formulation for the reversal of DDP resistance in NSCLC by inhibiting cancer cell stemness.
Collapse
Affiliation(s)
- Wenhui Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacy, Qilu Hospital (Qingdao), Shandong University, Qingdao, China
| | - Huaiyou Lv
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacy, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Qinxiu Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianxiong Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xin Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Guozhi Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chongzheng Yan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fengqin Sun
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhongxi Zhao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key University Laboratory of Pharmaceutics & Drug Delivery Systems of Shandong Province, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiumei Jia
- Department of Pharmacy, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| |
Collapse
|
50
|
Cheng W, Dai W, Chen W, Xue H, Zhao Z, Jiang Z, Li H, Liu J, Huang F, Cai M, Zheng L, Yu Z, Peng D, Zhang J. Nematodes exposed to furfural acetone exhibit a species-specific vacuolar H +-ATPase response. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117407. [PMID: 39603226 DOI: 10.1016/j.ecoenv.2024.117407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
Furfural acetone (FAc) is widely used as an additive by the food industry, as well as an intermediate in several fine chemical industries. Its nematicidal activity against the free-living model organism Caenorhabditis elegans and the parasitic nematode Meloidogyne incognita are well known, but its molecular mechanism of action remains unclear. To deep this subject, we performed 48-h lethal tests on eight nematode species, encompassing free-living, plant-parasitic, and animal-parasitic nematodes. Our results revealed that FAc possesses broad-spectrum nematicidal activity, with potent effects against parasitic nematodes such as Strongyloides stercoralis and M. incognita. In contrast, it exhibited weak activity against the free-living nematode C. elegans, suggesting its potential as a selective nematicide. Our investigation unveiled that FAc binds to the vacuolar H+-ATPase subunits VHA-12 and VHA-13, accelerating intestinal cell necrosis and leading to the death of C. elegans. It is the first discovery that VHA-12 and VHA-13 can serve as target proteins for triggering nematode cell necrosis. The interaction results indicated that FAc targets proteins VHA-12 and VHA-13 of different nematodes and confers broad-spectrum nematicidal activity. And the Spearman analysis results illustrated that the differential nematicidal activity of FAc against various nematodes is attributed to variations in the sequence and structure of the receptor proteins VHA-12 and VHA-13 among different nematode species. Our results illuminate the molecular mechanism underlying the differential toxicity of FAc to different nematodes, and provide valuable data for the comprehensive risk assessment of FAc release into the environment.
Collapse
Affiliation(s)
- Wanli Cheng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei 430062, China; National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wei Dai
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Wen Chen
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Hua Xue
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Zhengzheng Zhao
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Zhengbing Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei 430062, China
| | - Huanan Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei 430062, China
| | - Jiashu Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei 430062, China
| | - Feng Huang
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Minmin Cai
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Longyu Zheng
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Ziniu Yu
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Donghai Peng
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| | - Jibin Zhang
- National Key Laboratory of Agricultural Microbiology and National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China.
| |
Collapse
|