1
|
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
|
2
|
Zhang R, Tan Y, Xu K, Huang N, Wang J, Liu M, Wang L. Cuproplasia and cuproptosis in hepatocellular carcinoma: mechanisms, relationship and potential role in tumor microenvironment and treatment. Cancer Cell Int 2025; 25:137. [PMID: 40205387 PMCID: PMC11983883 DOI: 10.1186/s12935-025-03683-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 02/08/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the main phenotype of liver cancer with a poor prognosis. Copper is vital in liver function, and HCC cells rely on it for growth and metastasis, leading to cuproplasia. Excessive copper can induce cell death, termed cuproptosis. Tumor microenvironment (TME) is pivotal in HCC, especially in immunotherapy, and copper is closely related to the TME pathogenesis. However, how these two mechanisms contribute to the TME is intriguing. MAIN BODY We conducted the latest progress literature on cuproplasia and cuproptosis in HCC, and summarized their specific roles in TME and treatment strategies. The mechanisms of cuproplasia and cuproptosis and their relationship and role in TME have been deeply summarized. Cuproplasia fosters TME formation, angiogenesis, and metastasis, whereas cuproptosis may alleviate mitochondrial dysfunction and hypoxic conditions in the TME. Inhibiting cuproplasia and enhancing cuproptosis in HCC are essential for achieving therapeutic efficacy in HCC. CONCLUSION An in-depth analysis of cuproplasia and cuproptosis mechanisms within the TME of HCC unveils their opposing nature and their impact on copper regulation. Grasping the equilibrium between these two factors is crucial for a deeper understanding of HCC mechanisms to shed light on novel directions in treating HCC.
Collapse
Affiliation(s)
- Ruoyu Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China
| | - Yunfei Tan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ke Xu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China
| | - Ning Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 Panjiayuan Nanli, Chaoyang District, P.O. Box 2258, 100021, Beijing, People's Republic of China.
| | - Liming Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli Area, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
3
|
Chrzan N, Hartman ML. Copper in melanoma: At the crossroad of protumorigenic and anticancer roles. Redox Biol 2025; 81:103552. [PMID: 39970778 PMCID: PMC11880738 DOI: 10.1016/j.redox.2025.103552] [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: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025] Open
Abstract
Copper is an essential micronutrient that is a cofactor for various enzymes involved in multiple cellular processes. Melanoma patients have high serum copper levels, and elevated copper concentrations are found in melanoma tumors. Copper influences the activity of several melanoma-related proteins involved in cell survival, proliferation, pigmentation, angiogenesis, and metastasis. Targeting these processes with copper chelators has shown efficacy in reducing tumor growth and overcoming drug resistance. In contrast, excessive copper can also have detrimental effects when imported into melanoma cells. Multiple distinct cellular effects of copper overload, including the induction of different types of cell death, have been reported. Cuproptosis, a novel type of copper-dependent cell death, has been recently described and is associated with the metabolic phenotype. Melanoma cells can switch between glycolysis and oxidative phosphorylation, which are crucial for tumor growth and drug resistance. In this respect, metabolic plasticity might be exploited for the use of copper-delivery strategies, including repurposing of disulfiram, which is approved for the treatment of noncancer patients. In addition, the development of nanomedicines can improve the targeted delivery of copper to melanoma cells and enable the use of these drugs alone or in combination as copper has been shown to complement targeted therapy and immunotherapy in melanoma cells. However, further research is needed to explore the specific mechanisms of both copper restriction and excess copper-induced processes and determine effective biomarkers for predicting treatment sensitivity in melanoma patients. In this review, we discuss the dual role of copper in melanoma biology.
Collapse
Affiliation(s)
- Natalia Chrzan
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| | - Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland.
| |
Collapse
|
4
|
Rodriguez R, Müller S, Colombeau L, Solier S, Sindikubwabo F, Cañeque T. Metal Ion Signaling in Biomedicine. Chem Rev 2025; 125:660-744. [PMID: 39746035 PMCID: PMC11758815 DOI: 10.1021/acs.chemrev.4c00577] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/10/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Complex multicellular organisms are composed of distinct tissues involving specialized cells that can perform specific functions, making such life forms possible. Species are defined by their genomes, and differences between individuals within a given species directly result from variations in their genetic codes. While genetic alterations can give rise to disease-causing acquisitions of distinct cell identities, it is now well-established that biochemical imbalances within a cell can also lead to cellular dysfunction and diseases. Specifically, nongenetic chemical events orchestrate cell metabolism and transcriptional programs that govern functional cell identity. Thus, imbalances in cell signaling, which broadly defines the conversion of extracellular signals into intracellular biochemical changes, can also contribute to the acquisition of diseased cell states. Metal ions exhibit unique chemical properties that can be exploited by the cell. For instance, metal ions maintain the ionic balance within the cell, coordinate amino acid residues or nucleobases altering folding and function of biomolecules, or directly catalyze specific chemical reactions. Thus, metals are essential cell signaling effectors in normal physiology and disease. Deciphering metal ion signaling is a challenging endeavor that can illuminate pathways to be targeted for therapeutic intervention. Here, we review key cellular processes where metal ions play essential roles and describe how targeting metal ion signaling pathways has been instrumental to dissecting the biochemistry of the cell and how this has led to the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Raphaël Rodriguez
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sebastian Müller
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Ludovic Colombeau
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Stéphanie Solier
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
- Université
Paris-Saclay, UVSQ, 78180 Montigny-le-Bretonneux, France
| | | | - Tatiana Cañeque
- Institut
Curie, CNRS, INSERM, PSL Research University, 75005 Paris, France
| |
Collapse
|
5
|
Subramaniam J, Aditi A, Arumugam K, Sri S, Bharathidevi SR, Ramkumar KM. Copper Dyshomeostasis and Diabetic Complications: Chelation Strategies for Management. Mini Rev Med Chem 2025; 25:277-292. [PMID: 39328144 DOI: 10.2174/0113895575308206240911104945] [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: 04/09/2024] [Revised: 07/02/2024] [Accepted: 08/05/2024] [Indexed: 09/28/2024]
Abstract
Cuproptosis, an emerging concept in the field of diabetes research, presents a novel and promising perspective for the effective management of diabetes mellitus and its associated complications. Diabetes, characterized by chronic hyperglycemia, poses a substantial global health burden, with an increasing prevalence worldwide. Despite significant progress in our understanding of this complex metabolic disorder, optimal therapeutic strategies still remain elusive. The advent of cuproptosis, a term coined to describe copper-induced cellular cell death and its pivotal role in diabetes pathogenesis, opens new avenues for innovative interventions. Copper, an indispensable trace element, plays a pivotal role in a myriad of vital biological processes, encompassing energy production, bolstering antioxidant defenses, and altered cellular signaling. However, in the context of diabetes, this copper homeostasis is perturbed, driven by a combination of genetic predisposition, dietary patterns, and environmental factors. Excessive copper levels act as catalysts for oxidative stress, sparking intricate intracellular signaling cascades that further exacerbate metabolic dysfunction. In this review, we aim to explore the interrelationship between copper and diabetes comprehensively, shedding light on the intricate mechanisms underpinning cuproptosis. By unraveling the roles of copper transporters, copper-dependent enzymes, and cuproptotic signaling pathways, we seek to elucidate potential therapeutic strategies that harness the power of copper modulation in diabetes management. This insight sets the stage for a targeted approach to challenge the complex hurdles posed by diabetes, potentially transforming our therapeutic strategies in the ongoing fight against this pervasive global health concern.
Collapse
Affiliation(s)
- Jahnavi Subramaniam
- Department of Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, 603 203, Tamil Nadu, India
| | - Aarya Aditi
- Department of Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, 603 203, Tamil Nadu, India
| | - Kishore Arumugam
- RS Mehta Jain Department of Biochemistry & Cell Biology, KBIRVO Block, Vision Research Foundation, Chennai, 600006, India
| | - Sathya Sri
- Department of Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, 603 203, Tamil Nadu, India
| | | | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRMIST, Kattankulathur, 603 203, Tamil Nadu, India
| |
Collapse
|
6
|
Lin Y, Yuan M, Wang G. Copper homeostasis and cuproptosis in gynecological disorders: Pathogenic insights and therapeutic implications. J Trace Elem Med Biol 2024; 84:127436. [PMID: 38547725 DOI: 10.1016/j.jtemb.2024.127436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 05/27/2024]
Abstract
This review comprehensively explores the complex role of copper homeostasis in female reproductive system diseases. As an essential trace element, copper plays a crucial role in various biological functions. Its dysregulation is increasingly recognized as a pivotal factor in the pathogenesis of gynecological disorders. We investigate how copper impacts these diseases, focusing on aspects like oxidative stress, inflammatory responses, immune function, estrogen levels, and angiogenesis. The review highlights significant changes in copper levels in diseases such as cervical, ovarian, endometrial cancer, and endometriosis, underscoring their potential roles in disease mechanisms and therapeutic exploration. The recent discovery of 'cuproptosis,' a novel cell death mechanism induced by copper ions, offers a fresh molecular perspective in understanding these diseases. The review also examines genes associated with cuproptosis, particularly those related to drug resistance, suggesting new strategies to enhance traditional therapy effectiveness. Additionally, we critically evaluate current therapeutic approaches targeting copper homeostasis, including copper ionophores, chelators, and nanoparticles, emphasizing their emerging potential in gynecological disease treatment. This article aims to provide a comprehensive overview of copper's role in female reproductive health, setting the stage for future research to elucidate its mechanisms and develop targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ying Lin
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China; Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China; Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Disease, Jinan, Shandong Province China; Gynecology Laboratory, Shandong Provincial Hospital, Jinan Shandong Province, China; Gynecology Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong Province, China
| | - Ming Yuan
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China; Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Disease, Jinan, Shandong Province China; Gynecology Laboratory, Shandong Provincial Hospital, Jinan Shandong Province, China; Gynecology Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong Province, China
| | - Guoyun Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China; Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Disease, Jinan, Shandong Province China; Gynecology Laboratory, Shandong Provincial Hospital, Jinan Shandong Province, China; Gynecology Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan Shandong Province, China.
| |
Collapse
|
7
|
Kalishwaralal K, Azeez Nazeer A, Induja DK, Keerthana CK, Shifana SC, Anto RJ. Enhanced extracellular vesicles mediated uttroside B (Utt-B) delivery to Hepatocellular carcinoma cell: Pharmacokinetics based on PBPK modelling. Biochem Biophys Res Commun 2024; 703:149648. [PMID: 38368675 DOI: 10.1016/j.bbrc.2024.149648] [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: 01/23/2024] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 02/20/2024]
Abstract
Our prior investigation has confirmed that the anti-hepatocellular carcinoma activity of the plant saponin, specifically Uttroside B (Utt-B), derived from the leaves of Solanum nigrum Linn. This study concentrated on formulating a novel biocompatible nanocarrier utilizing Extracellular vesicles (EVs) to enhance the delivery of plant saponin into cells. The physicochemical attributes of Extracellular Vesicles/UttrosideB (EVs/Utt-B) were comprehensively characterized through techniques such as Transmission Electron Microscopy (TEM) and Fourier-transform infrared spectroscopy (FTIR). Despite the promising therapeutic potential of this uttroside B, mechanistic know-how about its entry into cells is still in its infancy. Our research sheds light on the extracellular vesicle-mediated mechanism facilitating the entry of the saponin into cells, a phenomenon confirmed through the use of by confocal microscopy. We further analysed drug-releasing kinetics and simulated the Pharmacokinetics by PBPK modelling. The simulated pharmacokinetics revealed the bioavailability of Uttroside-B in oral administration against intravenous administration.
Collapse
Affiliation(s)
- Kalimuthu Kalishwaralal
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 95014, Kerala, India.
| | - Abdul Azeez Nazeer
- Laboratory of Pharmaceutical Sciences, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon state, 24341, Republic of Korea
| | - D K Induja
- Chemical Sciences and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, Kerala, India
| | - Chenicheri K Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 95014, Kerala, India
| | - Sadiq C Shifana
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram, 695317, Kerala, India
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 95014, Kerala, India; Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram, 695317, Kerala, India
| |
Collapse
|
8
|
Meraz-Torres F, Niessner H, Plöger S, Riel S, Schörg B, Casadei N, Kneilling M, Schaller M, Flatz L, Macek B, Eigentler T, Rieß O, Garbe C, Amaral T, Sinnberg T. Augmenting MEK inhibitor efficacy in BRAF wild-type melanoma: synergistic effects of disulfiram combination therapy. J Exp Clin Cancer Res 2024; 43:30. [PMID: 38263136 PMCID: PMC10804659 DOI: 10.1186/s13046-023-02941-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND MEK inhibitors (MEKi) were shown to be clinically insufficiently effective in patients suffering from BRAF wild-type (BRAF WT) melanoma, even if the MAPK pathway was constitutively activated due to mutations in NRAS or NF-1. Thus, novel combinations are needed to increase the efficacy and duration of response to MEKi in BRAF WT melanoma. Disulfiram and its metabolite diethyldithiocarbamate are known to have antitumor effects related to cellular stress, and induction of endoplasmic reticulum (ER) stress was found to synergize with MEK inhibitors in NRAS-mutated melanoma cells. Therefore, we investigated the combination of both therapeutics to test their effects on BRAF-WT melanoma cells and compared them with monotherapy using the MEKi trametinib. METHODS The effects of combined therapy with disulfiram or its metabolite diethyldithiocarbamate and the MEKi trametinib were evaluated in a series of BRAF-WT melanoma cell lines by measuring cell viability and apoptosis induction. Cytotoxicity was additionally assessed in 3D spheroids, ex vivo melanoma slice cultures, and in vivo xenograft mouse models. The response of melanoma cells to treatment was studied at the RNA and protein levels to decipher the mode of action. Intracellular and intratumoral copper measurements were performed to investigate the role of copper ions in the antitumor cytotoxicity of disulfiram and its combination with the MEKi. RESULTS Diethyldithiocarbamate enhanced trametinib-induced cytotoxicity and apoptosis induction in 2D and 3D melanoma culture models. Mechanistically, copper-dependent induction of oxidative stress and ER stress led to Janus kinase (JNK)-mediated apoptosis in melanoma cells. This mechanism was also detectable in patient-derived xenograft melanoma models and resulted in a significantly improved therapeutic effect compared to monotherapy with the MEKi trametinib. CONCLUSIONS Disulfiram and its metabolite represent an attractive pharmaceutical approach to induce ER stress in melanoma cells that potentiates the antitumor effect of MEK inhibition and may be an interesting candidate for combination therapy of BRAF WT melanoma.
Collapse
Affiliation(s)
| | - Heike Niessner
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany
| | - Sarah Plöger
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Simon Riel
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Barbara Schörg
- Department of Preclinical Imaging and Radiopharmacy, Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation, University of Tübingen, Tübingen, 72076, Germany
| | - Nicolas Casadei
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Manfred Kneilling
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany
- Department of Preclinical Imaging and Radiopharmacy, Laboratory for Preclinical Imaging and Imaging Technology of the Werner Siemens-Foundation, University of Tübingen, Tübingen, 72076, Germany
| | - Martin Schaller
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Olaf Rieß
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
| | - Teresa Amaral
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany
| | - Tobias Sinnberg
- Department of Dermatology, Tübingen University Hospital, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) Image Guided and Functionally Instructed Tumor Therapies, University Hospital Tübingen, Tübingen, 72076, Germany.
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany.
| |
Collapse
|
9
|
Cazzoli R, Zamborlin A, Ermini ML, Salerno A, Curcio M, Nicoletta FP, Iemma F, Vittorio O, Voliani V, Cirillo G. Evolving approaches in glioma treatment: harnessing the potential of copper metabolism modulation. RSC Adv 2023; 13:34045-34056. [PMID: 38020008 PMCID: PMC10661684 DOI: 10.1039/d3ra06434d] [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: 09/21/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023] Open
Abstract
The key properties and high versatility of metal nanoparticles have shed new perspectives on cancer therapy, with copper nanoparticles gaining great interest because of the ability to couple the intrinsic properties of metal nanoparticles with the biological activities of copper ions in cancer cells. Copper, indeed, is a cofactor involved in different metabolic pathways of many physiological and pathological processes. Literature data report on the use of copper in preclinical protocols for cancer treatment based on chemo-, photothermal-, or copper chelating-therapies. Copper nanoparticles exhibit anticancer activity via multiple routes, mainly involving the targeting of mitochondria, the modulation of oxidative stress, the induction of apoptosis and autophagy, and the modulation of immune response. Moreover, compared to other metal nanoparticles (e.g. gold, silver, palladium, and platinum), copper nanoparticles are rapidly cleared from organs with low systemic toxicity and benefit from the copper's low cost and wide availability. Within this review, we aim to explore the impact of copper in cancer research, focusing on glioma, the most common primary brain tumour. Glioma accounts for about 80% of all malignant brain tumours and shows a poor prognosis with the five-year survival rate being less than 5%. After introducing the glioma pathogenesis and the limitation of current therapeutic strategies, we will discuss the potential impact of copper therapy and present the key results of the most relevant literature to establish a reliable foundation for future development of copper-based approaches.
Collapse
Affiliation(s)
- Riccardo Cazzoli
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Sydney NSW Australia
| | - Agata Zamborlin
- NEST-Scuola Normale Superiore Piazza San Silvestro 12 - 56127 Pisa Italy
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia Piazza San Silvestro 12 - 56127 Pisa Italy
| | - Maria Laura Ermini
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia Piazza San Silvestro 12 - 56127 Pisa Italy
| | - Antonietta Salerno
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Sydney NSW Australia
| | - Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria 87036 Rende Italy +39 0984493208
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria 87036 Rende Italy +39 0984493208
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria 87036 Rende Italy +39 0984493208
| | - Orazio Vittorio
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Sydney NSW Australia
- School of Biomedical Sciences, University of New South Wales Sydney NSW Australia
| | - Valerio Voliani
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia Piazza San Silvestro 12 - 56127 Pisa Italy
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa Viale Cembrano 4 - 16148 Genoa Italy
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria 87036 Rende Italy +39 0984493208
| |
Collapse
|
10
|
Poursani EM, Mercatelli D, Raninga P, Bell JL, Saletta F, Kohane FV, Neumann DP, Zheng Y, Rouaen JRC, Jue TR, Michniewicz FT, Schadel P, Kasiou E, Tsoli M, Cirillo G, Waters S, Shai-Hee T, Cazzoli R, Brettle M, Slapetova I, Kasherman M, Whan R, Souza-Fonseca-Guimaraes F, Vahdat L, Ziegler D, Lock JG, Giorgi FM, Khanna K, Vittorio O. Copper chelation suppresses epithelial-mesenchymal transition by inhibition of canonical and non-canonical TGF-β signaling pathways in cancer. Cell Biosci 2023; 13:132. [PMID: 37480151 PMCID: PMC10362738 DOI: 10.1186/s13578-023-01083-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Metastatic cancer cells exploit Epithelial-mesenchymal-transition (EMT) to enhance their migration, invasion, and resistance to treatments. Recent studies highlight that elevated levels of copper are implicated in cancer progression and metastasis. Clinical trials using copper chelators are associated with improved patient survival; however, the molecular mechanisms by which copper depletion inhibits tumor progression and metastasis are poorly understood. This remains a major hurdle to the clinical translation of copper chelators. Here, we propose that copper chelation inhibits metastasis by reducing TGF-β levels and EMT signaling. Given that many drugs targeting TGF-β have failed in clinical trials, partly because of severe side effects arising in patients, we hypothesized that copper chelation therapy might be a less toxic alternative to target the TGF-β/EMT axis. RESULTS Our cytokine array and RNA-seq data suggested a link between copper homeostasis, TGF-β and EMT process. To validate this hypothesis, we performed single-cell imaging, protein assays, and in vivo studies. Here, we used the copper chelating agent TEPA to block copper trafficking. Our in vivo study showed a reduction of TGF-β levels and metastasis to the lung in the TNBC mouse model. Mechanistically, TEPA significantly downregulated canonical (TGF-β/SMAD2&3) and non-canonical (TGF-β/PI3K/AKT, TGF-β/RAS/RAF/MEK/ERK, and TGF-β/WNT/β-catenin) TGF-β signaling pathways. Additionally, EMT markers of MMP-9, MMP-14, Vimentin, β-catenin, ZEB1, and p-SMAD2 were downregulated, and EMT transcription factors of SNAI1, ZEB1, and p-SMAD2 accumulated in the cytoplasm after treatment. CONCLUSIONS Our study suggests that copper chelation therapy represents a potentially effective therapeutic approach for targeting TGF-β and inhibiting EMT in a diverse range of cancers.
Collapse
Affiliation(s)
- Ensieh M Poursani
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Daniele Mercatelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Prahlad Raninga
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jessica L Bell
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Federica Saletta
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Felix V Kohane
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Daniel P Neumann
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Ye Zheng
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Jourdin R C Rouaen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Toni Rose Jue
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Filip T Michniewicz
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Piper Schadel
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Erin Kasiou
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Maria Tsoli
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Shafagh Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Tyler Shai-Hee
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Riccardo Cazzoli
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Merryn Brettle
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | - Iveta Slapetova
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | - Maria Kasherman
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | - Renee Whan
- Katharina Gauss Light Microscopy Facility, University of New South Wales, Sydney, NSW, Australia
| | | | | | - David Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - John G Lock
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Federico M Giorgi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - KumKum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Orazio Vittorio
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia.
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
11
|
Jiang Z, Sha G, Zhang W, Zhang Z, Liu T, Wang D, Tang D. The huge potential of targeting copper status in the treatment of colorectal cancer. Clin Transl Oncol 2023; 25:1977-1990. [PMID: 36781599 DOI: 10.1007/s12094-023-03107-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/28/2023] [Indexed: 02/15/2023]
Abstract
Colorectal cancer (CRC) commonly leads to cancer deaths and is often diagnosed at advanced stages. It also faces difficulties due to the poor results of conventional treatments such as surgery, chemotherapy, and radiotherapy. Copper is a mineral nutrient whose intrinsic properties have a two-way effect on the production and treatment of cancer. Copper's redox properties allow it to be used in developing anti-cancer drugs, while its potential toxicity leads to oxidative stress and even cancer. Copper status is closely related to colorectal tumors' proliferation and metastasis. The study of the mechanisms of copper homeostasis, cuproplasia, and cuproptosis due to altered copper status plays a crucial role in developing anticancer drugs. Therefore, targeting alteration of copper status becomes a potential option for treating colorectal cancer. This review summarizes the mechanisms by which altered copper status causes CRC progression and emphasizes the potential of regulating copper status in treating CRC.
Collapse
Affiliation(s)
- Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Zhilin Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Tian Liu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, People's Republic of China.
| |
Collapse
|
12
|
Zhang X, Jiang Q, Su Y, Bu L, Sun Z, Wu X, Gao B, Wang L, Lin Y, Xie W, Guo J. AMPK phosphorylates and stabilises copper transporter 1 to synergise metformin and copper chelator for breast cancer therapy. Br J Cancer 2023; 128:1452-1465. [PMID: 36807336 PMCID: PMC10070418 DOI: 10.1038/s41416-022-02127-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Predominant roles of copper and its transporter, copper transporter 1 (CTR1), in tumorigenesis have been explored recently; however, the upstream regulation of CTR1 and combinational intervention of copper chelators in malignancies remain largely unclear. METHODS CRISPR/Cas9-based kinome screening was used to identify the CTR1 upstream kinases. Immunofluorescence assays were utilised to detect CTR1 localisation. In vitro kinase assays and mass spectrometry were performed to detect CTR1 phosphorylation. Ubiquitination assays were performed to validate CTR1 stability. Colony formation, EdU labelling, Annexin V-FITC/PI-based apoptosis assays were carried out to detect the drug effect on cell growth and apoptosis. Xenografted mouse models were employed to investigate drug effects in vivo. RESULTS We identify that CTR1 undergoes AMPK-mediated phosphorylation, which enhances CTR1 stabilisation and membrane translocation by affecting Nedd4l interaction, resulting in increased oncogenic roles in breast cancer. Importantly, activation of AMPK with its agonist metformin markedly enhances CTR1 levels, and leads to the combinational usage of AMPK agonists and copper chelators for breast cancer treatment. CONCLUSIONS Our findings not only reveal the crosstalk between energy response and copper uptake via AMPK-mediated CTR1 phosphorylation and stability but also highlight the strategy to combat breast cancer by a combination of AMPK agonists and copper chelators. SIGNIFICANCE The connection between energy response and copper homoeostasis is linked by AMPK phosphorylating and stabilising CTR1, which provides a promising strategy to combat breast cancer by combining AMPK agonists and copper chelators.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Qiwei Jiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Yaqing Su
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Lang Bu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Zicheng Sun
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Xueji Wu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Bing Gao
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Lei Wang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Ying Lin
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China.
| | - Wei Xie
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China.
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China.
| |
Collapse
|
13
|
Magazenkova DN, Skomorokhova EA, Farroukh MA, Zharkova MS, Jassem ZM, Rekina VE, Shamova OV, Puchkova LV, Ilyechova EY. Influence of Silver Nanoparticles on the Growth of Ascitic and Solid Ehrlich Adenocarcinoma: Focus on Copper Metabolism. Pharmaceutics 2023; 15:pharmaceutics15041099. [PMID: 37111584 PMCID: PMC10145613 DOI: 10.3390/pharmaceutics15041099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/11/2023] [Accepted: 03/23/2023] [Indexed: 04/01/2023] Open
Abstract
The link between copper metabolism and tumor progression motivated us to use copper chelators for suppression of tumor growth. We assume that silver nanoparticles (AgNPs) can be used for lowering bioavailable copper. Our assumption is based on the ability of Ag(I) ions released by AgNPs in biological media and interfere with Cu(I) transport. Intervention of Ag(I) into copper metabolism leads to the replacement of copper by silver in ceruloplasmin and the decrease in bioavailable copper in the bloodstream. To check this assumption, mice with ascitic or solid Ehrlich adenocarcinoma (EAC) were treated with AgNPs using different protocols. Copper status indexes (copper concentration, ceruloplasmin protein level, and oxidase activity) were monitored to assess copper metabolism. The expression of copper-related genes was determined by real-time PCR in the liver and tumors, and copper and silver levels were measured by FAAS. Intraperitoneal AgNPs treatment beginning on the day of tumor inoculation enhanced mice survival, reduced the proliferation of ascitic EAC cells, and suppressed the activity of HIF1α, TNF-α and VEGFa genes. Topical treatment by the AgNPs, which was started together with the implantation of EAC cells in the thigh, also enhanced mice survival, decreased tumor growth, and repressed genes responsible for neovascularization. The advantages of silver-induced copper deficiency over copper chelators are discussed.
Collapse
Affiliation(s)
- Daria N. Magazenkova
- Research Center of Advanced Functional Materials and Laser Communication Systems, Institute of Advanced Data Transfer Systems, ITMO University, 197101 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| | - Ekaterina A. Skomorokhova
- Department of Molecular Genetics, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Mohammad Al Farroukh
- Federal State Budgetary Scientific Institution, Saint Petersburg State University, 199034 St. Petersburg, Russia
| | - Maria S. Zharkova
- Department of General Pathology and Pathophysiology, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Zena M. Jassem
- Research Center of Advanced Functional Materials and Laser Communication Systems, Institute of Advanced Data Transfer Systems, ITMO University, 197101 St. Petersburg, Russia
| | - Valeria E. Rekina
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
| | - Olga V. Shamova
- Department of General Pathology and Pathophysiology, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Ludmila V. Puchkova
- Research Center of Advanced Functional Materials and Laser Communication Systems, Institute of Advanced Data Transfer Systems, ITMO University, 197101 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
- Department of Molecular Genetics, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Ekaterina Y. Ilyechova
- Research Center of Advanced Functional Materials and Laser Communication Systems, Institute of Advanced Data Transfer Systems, ITMO University, 197101 St. Petersburg, Russia
- Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, 195251 St. Petersburg, Russia
- Department of Molecular Genetics, Research Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- Correspondence: ; Tel.: +7-(921)-7605274
| |
Collapse
|
14
|
Tsymbal SA, Refeld AG, Kuchur OA. The p53 Tumor Suppressor and Copper Metabolism: An Unrevealed but Important Link. Mol Biol 2022. [DOI: 10.1134/s0026893322060188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
15
|
Cheng F, Peng G, Lu Y, Wang K, Ju Q, Ju Y, Ouyang M. Relationship between copper and immunity: The potential role of copper in tumor immunity. Front Oncol 2022; 12:1019153. [PMID: 36419894 PMCID: PMC9676660 DOI: 10.3389/fonc.2022.1019153] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Copper is an essential trace element in an organism, and changes in copper levels in vivo often indicate a diseased state. Copper and immunity have been discussed since the last century, with copper deficiency significantly affecting the development and function of the immune system, such as increased host susceptibility to various pathogens, decreased number and impaired function of neutrophils, reduced antibacterial activity of macrophages, decreased proliferation of splenocytes, impaired B cell ability to produce antibodies and impaired function of cytotoxic T lymphocyte and helper T cells. In the past 20 years, some studies have shown that copper ions are related to the development of many tumors, including lung cancer, acute lymphoid leukaemia, multiple myeloma and other tumors, wherein copper ion levels were significantly elevated, and current studies reveal that copper ions are involved in the development, growth and metastasis of tumors through various pathways. Moreover, recent studies have shown that copper ions can regulate the expression of PD-L1, thus, attention should be paid to the important role of copper in tumor immunity. By exploring and studying copper ions and tumor immunity, new insights into tumor immunity could be generated and novel therapeutic approaches to improve the clinical prognosis of patients can be provided.
Collapse
Affiliation(s)
- Fu Cheng
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Geng Peng
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Lu
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Kang Wang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinuo Ju
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- Guangdong Country Garden School, Shunde, Foshan, Guangdong, China
| | - Yongle Ju
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Manzhao Ouyang
- Department of Gastrointestinal Surgery, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Ramchandani D, Berisa M, Tavarez DA, Li Z, Miele M, Bai Y, Lee SB, Ban Y, Dephoure N, Hendrickson RC, Cloonan SM, Gao D, Cross JR, Vahdat LT, Mittal V. Copper depletion modulates mitochondrial oxidative phosphorylation to impair triple negative breast cancer metastasis. Nat Commun 2021; 12:7311. [PMID: 34911956 PMCID: PMC8674260 DOI: 10.1038/s41467-021-27559-z] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/05/2021] [Indexed: 12/26/2022] Open
Abstract
Copper serves as a co-factor for a host of metalloenzymes that contribute to malignant progression. The orally bioavailable copper chelating agent tetrathiomolybdate (TM) has been associated with a significant survival benefit in high-risk triple negative breast cancer (TNBC) patients. Despite these promising data, the mechanisms by which copper depletion impacts metastasis are poorly understood and this remains a major barrier to advancing TM to a randomized phase II trial. Here, using two independent TNBC models, we report a discrete subpopulation of highly metastatic SOX2/OCT4+ cells within primary tumors that exhibit elevated intracellular copper levels and a marked sensitivity to TM. Global proteomic and metabolomic profiling identifies TM-mediated inactivation of Complex IV as the primary metabolic defect in the SOX2/OCT4+ cell population. We also identify AMPK/mTORC1 energy sensor as an important downstream pathway and show that AMPK inhibition rescues TM-mediated loss of invasion. Furthermore, loss of the mitochondria-specific copper chaperone, COX17, restricts copper deficiency to mitochondria and phenocopies TM-mediated alterations. These findings identify a copper-metabolism-metastasis axis with potential to enrich patient populations in next-generation therapeutic trials.
Collapse
Affiliation(s)
- Divya Ramchandani
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mirela Berisa
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Diamile A Tavarez
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhuoning Li
- Department of Microchemistry and Proteomics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Matthew Miele
- Department of Microchemistry and Proteomics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yang Bai
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sharrell B Lee
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Yi Ban
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Noah Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ronald C Hendrickson
- Department of Microchemistry and Proteomics, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Suzanne M Cloonan
- Department of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- The School of Medicine and Tallaght University Hospital, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Dingcheng Gao
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Cell and Developmental biology, Weill Cornell Medicine, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Linda T Vahdat
- Department of Medicine, Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, NY, 10065, USA.
- Department of Cell and Developmental biology, Weill Cornell Medicine, New York, NY, 10065, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
17
|
Babak MV, Ahn D. Modulation of Intracellular Copper Levels as the Mechanism of Action of Anticancer Copper Complexes: Clinical Relevance. Biomedicines 2021; 9:biomedicines9080852. [PMID: 34440056 PMCID: PMC8389626 DOI: 10.3390/biomedicines9080852] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Copper (Cu) is a vital element required for cellular growth and development; however, even slight changes in its homeostasis might lead to severe toxicity and deleterious medical conditions. Cancer patients are typically associated with higher Cu content in serum and tumor tissues, indicating increased demand of cancer cells for this micronutrient. Cu is known to readily cycle between the +1 and +2 oxidation state in biological systems. The mechanism of action of Cu complexes is typically based on their redox activity and induction of reactive oxygen species (ROS), leading to deadly oxidative stress. However, there are a number of other biomolecular mechanisms beyond ROS generation that contribute to the activity of anticancer Cu drug candidates. In this review, we discuss how interfering with intracellular Cu balance via either diet modification or addition of inorganic Cu supplements or Cu-modulating compounds affects tumor development, progression, and sensitivity to treatment modalities. We aim to provide the rationale for the use of Cu-depleting and Cu-overloading conditions to generate the best possible patient outcome with minimal toxicity. We also discuss the advantages of the use of pre-formed Cu complexes, such as Cu-(bis)thiosemicarbazones or Cu-N-heterocyclic thiosemicarbazones, in comparison with the in situ formed Cu complexes with metal-binding ligands. In this review, we summarize available clinical and mechanistic data on clinically relevant anticancer drug candidates, including Cu supplements, Cu chelators, Cu ionophores, and Cu complexes.
Collapse
|
18
|
Puchkova LV, Kiseleva IV, Polishchuk EV, Broggini M, Ilyechova EY. The Crossroads between Host Copper Metabolism and Influenza Infection. Int J Mol Sci 2021; 22:ijms22115498. [PMID: 34071094 PMCID: PMC8197124 DOI: 10.3390/ijms22115498] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Three main approaches are used to combat severe viral respiratory infections. The first is preemptive vaccination that blocks infection. Weakened or dead viral particles, as well as genetic constructs carrying viral proteins or information about them, are used as an antigen. However, the viral genome is very evolutionary labile and changes continuously. Second, chemical agents are used during infection and inhibit the function of a number of viral proteins. However, these drugs lose their effectiveness because the virus can rapidly acquire resistance to them. The third is the search for points in the host metabolism the effect on which would suppress the replication of the virus but would not have a significant effect on the metabolism of the host. Here, we consider the possibility of using the copper metabolic system as a target to reduce the severity of influenza infection. This is facilitated by the fact that, in mammals, copper status can be rapidly reduced by silver nanoparticles and restored after their cancellation.
Collapse
Affiliation(s)
- Ludmila V. Puchkova
- International Research Laboratory of Trace Elements Metabolism, ADTS Institute, RC AFMLCS, ITMO University, 197101 St. Petersburg, Russia;
| | - Irina V. Kiseleva
- Department of Virology, Institute of Experimental Medicine, 197376 St. Petersburg, Russia;
| | | | - Massimo Broggini
- Istituto di Ricerche Farmacologiche “Mario Negri”, IRCCS, 20156 Milan, Italy;
| | - Ekaterina Yu. Ilyechova
- International Research Laboratory of Trace Elements Metabolism, ADTS Institute, RC AFMLCS, ITMO University, 197101 St. Petersburg, Russia;
- Department of Molecular Genetics, Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- Correspondence: ; Tel.: +7-921-760-5274
| |
Collapse
|
19
|
Disulfiram as a Therapeutic Agent for Metastatic Malignant Melanoma-Old Myth or New Logos? Cancers (Basel) 2020; 12:cancers12123538. [PMID: 33260923 PMCID: PMC7760689 DOI: 10.3390/cancers12123538] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary In recent years, disulfiram has gained in attention as an anticancer drug due to its broad activity against various cancers, and its mechanisms and molecular targets have been deciphered in vitro and in vivo. One of these cancers is melanoma. Initial data from human studies show some benefit, but do not confirm its broad efficacy as a monotherapy. However, combination approaches could pave the way for exploiting the beneficial effects of disulfiram for cancer patients, including those with melanoma. Abstract New therapeutic concepts such as anti-PD-1-based immunotherapy or targeted therapy with BRAF and MEK inhibitors have significantly improved the survival of melanoma patients. However, about 20% of patients with targeted therapy and up to 50% with immunotherapies do not respond to their first-line treatment or rapidly develop resistance. In addition, there is no approved targeted therapy for certain subgroups, namely BRAF wild-type melanomas, although they often bear aggressive tumor biology. A repurposing of already approved drugs is a promising strategy to fill this gap, as it will result in comparatively low costs, lower risks and time savings. Disulfiram (DSF), the first drug to treat alcoholism, which received approval from the US Food and Drug Administration more than 60 years ago, is such a drug candidate. There is growing evidence that DSF has great potential for the treatment of various human cancers, including melanoma. Several mechanisms of its antitumor activity have been identified, amongst them the inhibition of the ubiquitin-proteasome system, the induction of reactive oxygen species and various death signaling pathways. This article provides an overview of the application of DSF in humans, its molecular mechanisms and targets in cancer therapy with a focus on melanoma. The results of clinical studies and experimental combination approaches of DSF with various cancer therapies are discussed, with the aim of exploring the potential of DSF in melanoma therapy.
Collapse
|
20
|
Baldari S, Di Rocco G, Toietta G. Current Biomedical Use of Copper Chelation Therapy. Int J Mol Sci 2020; 21:1069. [PMID: 32041110 PMCID: PMC7037088 DOI: 10.3390/ijms21031069] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
Copper is an essential microelement that plays an important role in a wide variety of biological processes. Copper concentration has to be finely regulated, as any imbalance in its homeostasis can induce abnormalities. In particular, excess copper plays an important role in the etiopathogenesis of the genetic disease Wilson's syndrome, in neurological and neurodegenerative pathologies such as Alzheimer's and Parkinson's diseases, in idiopathic pulmonary fibrosis, in diabetes, and in several forms of cancer. Copper chelating agents are among the most promising tools to keep copper concentration at physiological levels. In this review, we focus on the most relevant compounds experimentally and clinically evaluated for their ability to counteract copper homeostasis deregulation. In particular, we provide a general overview of the main disorders characterized by a pathological increase in copper levels, summarizing the principal copper chelating therapies adopted in clinical trials.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
- Department of Medical Surgical Sciences and Biotechnologies, University of Rome “La Sapienza”, C.so della Repubblica 79, 04100 Latina, Italy
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy; (S.B.); (G.D.R.)
| |
Collapse
|
21
|
De Luca A, Barile A, Arciello M, Rossi L. Copper homeostasis as target of both consolidated and innovative strategies of anti-tumor therapy. J Trace Elem Med Biol 2019; 55:204-213. [PMID: 31345360 DOI: 10.1016/j.jtemb.2019.06.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Copper was reported to be involved in the onset and progression of cancer. Proteins in charge of copper uptake and distribution, as well as cuproenzymes, are altered in cancer. More recently, proteins involved in signaling cascades, regulating cell proliferation, and anti-apoptotic protein factors were found to interact with copper. Therefore, therapeutic strategies using copper complexing molecules have been proposed for cancer therapy and used in clinical trials. OBJECTIVES This review will focus on novel findings about the involvement of copper and cupro-proteins in cancer dissemination process, epithelium to mesenchymal transition and vascularization. Particularly, implication of well-established (e.g. lysil oxidase) or newly identified copper-binding proteins (e.g. MEMO1), as well as their interplay, will be discussed. Moreover, we will describe recently synthesized copper complexes, including plant-derived ones, and their efficacy in contrasting cancer development. CONCLUSIONS The research on the involvement of copper in cancer is still an open field. Further investigation is required to unveil the mechanisms involved in copper delivery to the novel copper-binding proteins, which may identify other possible gene and protein targets for cancer therapy.
Collapse
Affiliation(s)
| | - Anna Barile
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Mario Arciello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
22
|
Baldari S, Di Rocco G, Heffern MC, Su TA, Chang CJ, Toietta G. Effects of Copper Chelation on BRAF V600E Positive Colon Carcinoma Cells. Cancers (Basel) 2019; 11:659. [PMID: 31083627 PMCID: PMC6562624 DOI: 10.3390/cancers11050659] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 01/05/2023] Open
Abstract
High affinity copper binding to mitogen-activated protein kinase kinase 1 (MAP2K1, also known as MEK1) allosterically promotes the kinase activity of MEK1/2 on extracellular signal regulated kinases 1 and 2 (ERK1/2). Consequently, copper-dependent activation of the mitogen-activated (MAP) kinase pathway has a role in promoting tumor growth. Conversely, copper chelation may represent a possible therapeutic approach for a specific subset of tumors characterized by activating mutations in the serine/threonine protein kinase V-Raf Murine Sarcoma Viral Oncogene Homolog B1 (BRAF), such as the V600E, occurring within the kinase domain (BRAFV600E). Tetrathiomolybdate (TM) is a specific copper chelating agent currently used for the treatment of Wilson's disease and in preclinical studies for the management of metastatic cancers owing to its anti-angiogenic and anti-inflammatory properties. We evaluated in vitro and in vivo the effects of copper depletion achieved by pharmacological treatment with TM in human colorectal cells bearing the BRAFV600E mutation in comparison with BRAF wild type cells. We provide evidence that selective copper chelation differentially affects proliferation, survival and migration of colon cancer cells bearing the BRAFV600E mutation compared to BRAFwt acting via differential phosphorylation levels of ERK1/2. Moreover, tetrathiomolybdate treatment was also effective in reducing the clonogenic potential of colon cancer BRAFV600E cells resistant to BRAF pharmacological inhibition. In conclusion, these results support further assessment of copper chelation therapy as an adjuvant therapy for inhibiting the progression of colon cancers containing the BRAFV600E mutation.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy.
| | - Marie C Heffern
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| | - Timothy A Su
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
| | - Christopher J Chang
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA.
- Howard Hughes Medical Institute, University of California, Berkeley, CA 94720, USA.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, via E. Chianesi 53, 00144 Rome, Italy.
| |
Collapse
|
23
|
Gaur K, Vázquez-Salgado A, Duran-Camacho G, Dominguez-Martinez I, Benjamín-Rivera J, Fernández-Vega L, Carmona Sarabia L, Cruz García A, Pérez-Deliz F, Méndez Román J, Vega-Cartagena M, Loza-Rosas S, Rodriguez Acevedo X, Tinoco A. Iron and Copper Intracellular Chelation as an Anticancer Drug Strategy. INORGANICS 2018. [DOI: https://doi.org/10.3390/inorganics6040126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A very promising direction in the development of anticancer drugs is inhibiting the molecular pathways that keep cancer cells alive and able to metastasize. Copper and iron are two essential metals that play significant roles in the rapid proliferation of cancer cells and several chelators have been studied to suppress the bioavailability of these metals in the cells. This review discusses the major contributions that Cu and Fe play in the progression and spreading of cancer and evaluates select Cu and Fe chelators that demonstrate great promise as anticancer drugs. Efforts to improve the cellular delivery, efficacy, and tumor responsiveness of these chelators are also presented including a transmetallation strategy for dual targeting of Cu and Fe. To elucidate the effectiveness and specificity of Cu and Fe chelators for treating cancer, analytical tools are described for measuring Cu and Fe levels and for tracking the metals in cells, tissue, and the body.
Collapse
|
24
|
Gaur K, Vázquez-Salgado AM, Duran-Camacho G, Dominguez-Martinez I, Benjamín-Rivera JA, Fernández-Vega L, Sarabia LC, García AC, Pérez-Deliz F, Méndez Román JA, Vega-Cartagena M, Loza-Rosas SA, Acevedo XR, Tinoco AD. Iron and Copper Intracellular Chelation as an Anticancer Drug Strategy. INORGANICS 2018; 6:126. [PMID: 33912613 PMCID: PMC8078164 DOI: 10.3390/inorganics6040126] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A very promising direction in the development of anticancer drugs is inhibiting the molecular pathways that keep cancer cells alive and able to metastasize. Copper and iron are two essential metals that play significant roles in the rapid proliferation of cancer cells and several chelators have been studied to suppress the bioavailability of these metals in the cells. This review discusses the major contributions that Cu and Fe play in the progression and spreading of cancer and evaluates select Cu and Fe chelators that demonstrate great promise as anticancer drugs. Efforts to improve the cellular delivery, efficacy, and tumor responsiveness of these chelators are also presented including a transmetallation strategy for dual targeting of Cu and Fe. To elucidate the effectiveness and specificity of Cu and Fe chelators for treating cancer, analytical tools are described for measuring Cu and Fe levels and for tracking the metals in cells, tissue, and the body.
Collapse
Affiliation(s)
- Kavita Gaur
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Geraldo Duran-Camacho
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Josué A Benjamín-Rivera
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Lauren Fernández-Vega
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Lesly Carmona Sarabia
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Angelys Cruz García
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Felipe Pérez-Deliz
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - José A Méndez Román
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Melissa Vega-Cartagena
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | - Sergio A Loza-Rosas
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| | | | - Arthur D Tinoco
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, Río Piedras, PR 00931, USA
| |
Collapse
|