1
|
Ali MS, Gupta G, Alsayari A, Wahab S, Kesharwani P. Biotinylated nanoparticles: A new frontier in nanomedicine and targeted cancer therapy. BIOMATERIALS ADVANCES 2025; 176:214366. [PMID: 40479769 DOI: 10.1016/j.bioadv.2025.214366] [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: 03/13/2025] [Revised: 05/11/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025]
Abstract
The development of targeted drug delivery systems has become a cornerstone of modern cancer therapy which provides a pathway to maximize treatment effectiveness while reducing side effects. Among the plethora of innovative strategies, biotinylated nanoparticles have evolved as a hopeful tool due to their ability to exploit the elevated expression of biotin receptors on cancerous cells. The design, synthesis, and functionalization of biotinylated nanoparticles for cancer treatment are thoroughly examined in this review article. By leveraging biotin's high affinity for biotin receptors, these nanoparticles achieve selective cancerous cell targeting, leading to enhanced drug bioavailability and cellular uptake. The discussion extends to the underlying mechanisms of drug release, receptor-mediated endocytosis, and strategies for achieving endosomal escape or pH-sensitive drug activation. Furthermore, the article also emphasizes how biotinylation in combination therapy allows for synergistic effects with immunomodulators, nucleic acids, and chemotherapeutic drugs. Preclinical studies are examined to underscore the translational potential of these systems. The review concludes by addressing current challenges, including scalability, and potential immunogenicity, while proposing future directions for optimizing biotinylated nanoparticles as a transformative approach in cancer treatment.
Collapse
Affiliation(s)
- Mohd Shoab Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India; School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
2
|
Liang G, Yang L, Xiao L, Tang L, Cheng S, Sun Z, Zhang Y, Yang F, Zhang GJ. Nanozymes enhancing field-effect transistor nanosensor for in-situ monitoring intracellular hydrogen peroxide release. Biosens Bioelectron 2025; 286:117643. [PMID: 40460459 DOI: 10.1016/j.bios.2025.117643] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 05/29/2025] [Indexed: 06/11/2025]
Abstract
Field-effect transistor (FET) nanodevices are widely recognized as highly sensitive sensors for continuous detection of bioactive molecules like hydrogen peroxide (H2O2). However, accurate and real-time monitoring of H2O2 poses challenges due to its instability and low concentration in organisms. To address these challenges, we construct an enhanced FET by one-step interfacing with nanozymes that possess natural enzyme-like catalytic properties and exceptional stability. Specifically, reduced graphene oxide (RGO) is drop-casted onto the fabricated FET channel, after which the nanozymes are assembled with RGO through π-π stacking interactions. The nanozyme-functionalized FET sensor is able to realize continuous H2O2 monitoring, featuring a broad linear detection range (1 pM-10 nM), an ultralow detection limit (0.5 pM), high selectivity, and rapid response. Moreover, the sensor enables real-time monitoring of intracellular H2O2 release from cells cultured within the FET channel, demonstrating significant potential for distinguishing between cancer cells and normal cells. In addition, the sensor successfully tracks the dynamic regulation of intracellular H2O2 efflux under drug stimulation. This platform combines high sensitivity with excellent biocompatibility, making it highly promising for applications in cell metabolism research, disease diagnosis, and drug efficacy evaluation and screening.
Collapse
Affiliation(s)
- Guowu Liang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Luyao Yang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Ling Xiao
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Lina Tang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Shibo Cheng
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Zhongyue Sun
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Yulin Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, 16 Huangjia Lake West Road, Wuhan, 430065, PR China.
| | - Fan Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, Pharmaceutical College, State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning, 530021, PR China.
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, 16 Huangjia Lake West Road, Wuhan, 430065, PR China.
| |
Collapse
|
3
|
Wang L, Yang H, Huang W, Ran G, He X, Bradley M, Qian S. Tumor-specific cathepsin B-triggered fluorescence imaging and prodrug activation. Eur J Med Chem 2025; 292:117661. [PMID: 40286448 DOI: 10.1016/j.ejmech.2025.117661] [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: 02/27/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Bioorthogonal activation chemistries have great potential in the development of novel drug treatments due to their versatility, tunability, and the ability to generate therapies with improved spatial targeting. The upregulation of Cathepsin B is highly correlated with the development of cancers, however, few fluorescent probes or prodrugs-based on Cathepsin B activity have demonstrated high tumor selectivity, since Cathepsin B is expressed in a variety of normal tissues. In this study, we report a strain-promoted azide-alkyne cycloaddition-activation strategy whereby a para-azido safety-catch linker is triggered by the tumor locating Biotin-TCO (trans-cyclooctene) conjugate, with subsequent tumor-specific Cathepsin B-triggered activation, generating a fluorescent reporter/cytotoxic drug, with high tumor selectivity. Our results suggest that this dual AND-Gate strategy of orthogonal Biotin AND Cathepsin B action would be advantageous for tumor-specific fluorescence labelling, fluorescence-guided surgery and targeted treatment.
Collapse
Affiliation(s)
- Luyang Wang
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Houchi Yang
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Wanyun Huang
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Guojun Ran
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China
| | - Xiaolong He
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China.
| | - Mark Bradley
- Precision Healthcare University Research Institute, Queen Mary University of London, Empire House London E1 1HH, UK
| | - Shan Qian
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Sichuan Engineering Research Center for Molecular Targeted Diagnostic & Therapeutic Drugs, Xihua University, Chengdu 610039, PR China.
| |
Collapse
|
4
|
Acharya A, Roy N, Newaskar V, Rai A, Ghosh A, Nagpure M, Giri SK, Sahni G, Guchhait SK. Topoisomerase II-targeting anticancer clinical candidates and drugs: A critical analysis, unravelling molecular medicinal insights and promising research roadmap. Eur J Med Chem 2025; 291:117611. [PMID: 40249970 DOI: 10.1016/j.ejmech.2025.117611] [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: 02/28/2025] [Revised: 03/25/2025] [Accepted: 04/05/2025] [Indexed: 04/20/2025]
Abstract
In recent years, the USFDA-approved drug molecules are being frequently analyzed to provide perspectives and strategies for novel therapeutic discovery and development. Some of the remarkable analyses include physicochemical properties of drugs relevant to oral bioavailability, frequent presence of drug relevant-structural motifs, natural products as sources of new drugs, and synthetic approaches to new drugs. In this review article, for the first time, we present a structure-function analysis of human topoisomerase II (hTopo II) inhibitors those are currently clinically used or under clinical trials for anticancer treatment. The case studies and a critical molecular medicinal insight for their therapeutic development have been presented. The review illustrates various key aspects: the hTopo II inhibitors' molecular modulations, common pharmacophores, interactions at molecular level crucial for inhibition of enzyme at its various stages of catalytic function, and network polypharmacology of Topo II with different targets. Numerous toxicophore motifs have been identified, which provide important alerts while designing and discovering novel therapeutic agents. A range of innovative approaches including property-focused strategies, ADCs, and Click Activated Protodrugs Against Cancer (CAPAC) that have addressed challenges faced in the hTopo II-based therapeutic development have been discussed. The analysis with perspectives represents a valuable educational and research resource that will encourage hTopo II-inhibition and its network polypharmacology based drug discovery studies.
Collapse
Affiliation(s)
- Ayan Acharya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Nibedita Roy
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Vaishnavi Newaskar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Abhishek Rai
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Abhrajyoti Ghosh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Mithilesh Nagpure
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Santosh Kumar Giri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Gautam Sahni
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India
| | - Sankar K Guchhait
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, SAS Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
5
|
Bangolo A, Amoozgar B, Habibi M, Simms E, Nagesh VK, Wadhwani S, Wadhwani N, Auda A, Elias D, Mansour C, Abbott R, Jebara N, Zhang L, Gill S, Ahmed K, Ip A, Goy A, Cho C. Exploring the gut microbiome’s influence on cancer-associated anemia: Mechanisms, clinical challenges, and innovative therapies. World J Gastrointest Pharmacol Ther 2025; 16:105375. [DOI: 10.4292/wjgpt.v16.i2.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Anemia is a prevalent and challenging complication in patients with hematologic and solid malignancies, which stems from the direct effects of malignancy, treatment-induced toxicities, and systemic inflammation. It affects patients’ survival, functional status, and quality of life profoundly. Recent literature has highlighted the emerging role of the gut microbiome in the pathogenesis of cancer-associated anemia. The gut microbiota, through its intricate interplay with iron metabolism, inflammatory pathways, and immune modulation, may either exacerbate or ameliorate anemia depending on its composition, and functional integrity. Dysbiosis, characterized by disruption in the gut microbial ecosystem, is very common in cancer patients. This microbial imbalance is implicated in anemia causation through diminished iron absorption, persistent low-grade inflammation, and suppression of erythropoiesis.
AIM To consolidate current evidence regarding the interplay between gut microbiome and anemia in the setting of malignancies. It aims to provide a detailed exploration of the mechanistic links between dysbiosis and anemia, identifies unique challenges associated with various cancer types, and evaluates the efficacy of microbiome-focused therapies. Through this integrative approach, the review seeks to establish a foundation for innovative clinical strategies aimed at mitigating anemia and improving patient outcomes in oncology.
METHODS A literature search was performed using multiple databases, including Google Scholar, PubMed, Scopus, and Web of Science, using a combination of keywords and Boolean operators to refine results. Keywords included “cancer-associated anemia”, “gut microbiome”, “intestinal microbiota”, “iron metabolism”, “gut dysbiosis”, “short-chain fatty acids”, “hematopoiesis”, “probiotics”, “prebiotics”, and “fecal microbiota transplantation”. Articles published in English between 2000 and December 2024 were included, with a focus on contemporary and relevant findings.
RESULTS Therapeutic strategies aimed at restoration of gut microbial homeostasis, such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation (FMT), can inhibit anemia-causing pathways by enhancing microbial diversity, suppressing detrimental flora, reducing systemic inflammation and optimizing nutrient absorption.
CONCLUSION Gut dysbiosis causes anemia and impairs response to chemotherapy in cancer patients. Microbiome-centered interventions, such as probiotics, prebiotics, dietary modifications, and FMT, have shown efficacy in restoring microbial balance, reducing inflammation, and enhancing nutrient bioavailability. Emerging approaches, including engineered probiotics and bacteriophage therapies, are promising precision-based, customizable solutions for various microbiome compositions and imbalances. Future research should focus on integrating microbiome-targeted strategies with established anemia therapies.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Maryam Habibi
- Department of Research, Tulane National Primate Research Center, Covington, LA 70433, United States
| | - Elizabeth Simms
- Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, United States
| | - Vignesh K Nagesh
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Shruti Wadhwani
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Nikita Wadhwani
- Department of Internal Medicine, Hackensack Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Auda Auda
- Department of Family Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Daniel Elias
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Charlene Mansour
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, United States
| | - Robert Abbott
- Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Nisrene Jebara
- Columbia University School of Nursing, New York, NY 10032, United States
| | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Sarvarinder Gill
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Kareem Ahmed
- Department of Medicine, University of Washington, Seattle, WA 98195, United States
| | - Andrew Ip
- Division of Lymphoma, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Andre Goy
- Division of Lymphoma, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| | - Christina Cho
- Division of Stem Cell Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack, NJ 07601, United States
| |
Collapse
|
6
|
Xiang G, Shi T, Nwaele CO, Xiao H, Liu Y, Wang Q, Zhang J, Zheng Y. Inhibition of the Sp1/PI3K/AKT signaling pathway exacerbates doxorubicin-induced cardiomyopathy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119960. [PMID: 40246177 DOI: 10.1016/j.bbamcr.2025.119960] [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: 12/26/2024] [Revised: 04/06/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
OBJECTIVE This study aimed to investigate the interaction and underlying mechanisms between specificity protein 1 (Sp1) and the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signaling pathway in the context of doxorubicin-induced cardiomyopathy (DIC). METHODS A rat model of DIC was established by intraperitoneal injection of doxorubicin (1 mg/kg) twice a week for eight weeks. Cardiac function was evaluated using echocardiography, and myocardial histopathology was assessed by hematoxylin-eosin (HE) staining. In vitro, H9c2 cardiomyocytes were treated with doxorubicin (2 μmol/L) to induce cardiotoxicity, followed by co-treatment with the Sp1 inhibitor plicamycin or the PI3K/AKT inhibitor LY294002. Cell viability was measured by the CCK-8 assay. Oxidative stress markers, including reactive oxygen species (ROS) and lactate dehydrogenase (LDH), were quantified using flow cytometry and colorimetric assays. Apoptosis was detected via TUNEL staining, and protein expression of Sp1, PI3K, AKT, and Caspase-3 was analyzed by Western blotting. RESULTS Doxorubicin treatment significantly impaired cardiac function in rats, as evidenced by an increase in both left ventricular internal diameters during diastole (LVIDd) and systole (LVIDs), along with decreased ejection fraction (EF) and fractional shortening (FS) (p < 0.01). Myocardial HE staining in doxorubicin-treated rats revealed disorganized cardiomyocyte structures, edema, and cellular necrosis. In vitro, doxorubicin exposure led to reduced H9c2 cell viability, elevated ROS and LDH levels, and increased apoptosis rates (p < 0.01). Western blotting demonstrated that doxorubicin significantly downregulated the expression of Sp1, PI3K, and AKT while upregulating Caspase-3. Inhibition of Sp1 or PI3K/AKT exacerbated these effects, resulting in further cardiac dysfunction, oxidative stress, and apoptosis. Moreover, Sp1 inhibition led to decreased PI3K/AKT pathway activation, while PI3K/AKT inhibition reciprocally suppressed Sp1 expression, indicating a bidirectional regulatory relationship. CONCLUSION Doxorubicin induces cardiotoxicity by promoting oxidative stress and apoptosis through the downregulation of the Sp1/PI3K/AKT signaling pathway. Inhibition of this pathway exacerbates cardiac injury, suggesting that targeting Sp1 and PI3K/AKT may offer novel therapeutic strategies for the prevention and treatment of DIC.
Collapse
Affiliation(s)
- Guojian Xiang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Tingting Shi
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China; Fuzhou Jian Jia Rehabilitation Hospital, Fuzhou 350007, Fujian, China
| | | | - Huazhen Xiao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Yucheng Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Qingfeng Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China
| | - Jiancheng Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology in South Branch, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China.
| | - Yonghong Zheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, Fujian, China; Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; Department of Cardiology, Fuzhou University Affiliated Provincial Hospital, Fuzhou 350001, Fujian, China; College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, Fujian, China.
| |
Collapse
|
7
|
Swiech O, Boguszewska-Czubara A. Enhancing cardiac safety: Liposomal ciprofloxacin mitigates anthracycline-induced cardiotoxicity without compromising anticancer efficacy. Chem Biol Interact 2025; 414:111496. [PMID: 40157626 DOI: 10.1016/j.cbi.2025.111496] [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: 07/17/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Anthracyclines, such as doxorubicin and epirubicin (EPI), are integral in the treatment of solid tumors and hematological malignancies but are associated with cardiotoxicity, potentially leading to heart failure. The underlying mechanisms involve the generation of reactive oxygen species (ROS), alterations in iron metabolism, and the inhibition of topoisomerase 2β (Top2β), resulting in mitochondrial dysfunction and cell death. Fluoroquinolones, including ciprofloxacin (CPX), enhance the efficacy of anthracyclines by inhibiting topoisomerase II and inducing apoptosis, thereby indicating a promising combination therapy. This study investigated the impact of environmental pH on the cardiotoxicity and myocardial accumulation of anthracyclines, as well as the cardioprotective and synergistic potential of CPX when co-administered with epirubicin. The findings revealed that only the zwitterionic form of CPX, either free or encapsulated in liposomes, offers significant cardioprotection without compromising the anticancer activity of EPI. Remarkably, the combination of liposomal CPX and EPI completely attenuates EPI's cardiotoxicity. These results suggest that initiating treatment with liposomal CPX prior to EPI administration may optimize cardioprotection while maintaining therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Olga Swiech
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland; BS Biotechna S.A, Szlak 77/222, Cracow, Poland.
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Chodzki 4a, 20-093, Lublin, Poland
| |
Collapse
|
8
|
Ruan Y, Zhang L, Zhang L, Zhu K. Therapeutic Approaches Targeting Ferroptosis in Cardiomyopathy. Cardiovasc Drugs Ther 2025; 39:595-613. [PMID: 37930587 DOI: 10.1007/s10557-023-07514-4] [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] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
The term cardiomyopathy refers to a group of heart diseases that cause severe heart failure over time. Cardiomyopathies have been proven to be associated with ferroptosis, a non-apoptotic form of cell death. It has been shown that some small molecule drugs and active ingredients of herbal medicine can regulate ferroptosis, thereby alleviating the development of cardiomyopathy. This article reviews recent discoveries about ferroptosis, its role in the pathogenesis of cardiomyopathy, and the therapeutic options for treating ferroptosis-associated cardiomyopathy. The article aims to provide insights into the basic mechanisms of ferroptosis and its treatment to prevent cardiomyopathy and related diseases.
Collapse
Affiliation(s)
- Yanqian Ruan
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Ling Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Lina Zhang
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Keyang Zhu
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.
| |
Collapse
|
9
|
Mohamed SMM, Aljohani AKB, El-Morsy A, Al Haidari RA, Alamri MAA, Maqnas YA, Alsibaee AM, Mostafa MAH, Keshek DE, Mohamed MG, Bayoumi SAL, Salama SA, El-Adl K. Synthesis and characterization of sodium alginate/poly(N-vinylpyrrolidone) nano-carrier loaded with rebaudioside A and/or stevioside for anticancer drug delivery. Int J Biol Macromol 2025; 316:144778. [PMID: 40449772 DOI: 10.1016/j.ijbiomac.2025.144778] [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: 01/02/2025] [Revised: 05/20/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
Stevioside and rebaudioside A revealed anticancer effects against diversity of cancers, such as colon, breast and liver cancers. Rebaudioside A can trigger apoptosis in cancer cells via activation of caspase-dependent pathway. In this study sodium alginate/poly(N-vinylpyrrolidone) as nano-carriers loaded with natural products rebaudioside A (R) and/or stevioside (S) were assessed for anticancer activities. The nanogel of structure R improved cytotoxicity against MCF-7, HepG2, HCT116 and A549 cancers by 60.29 %, 53.45 %, 72.86 % and 62.13 %, correspondingly. Additionally, the nanogel of structure S improved cytotoxicity against MCF-7, HepG2, HCT116 and A549 cancers by 63.96 %, 53.41 %, 70.59 % and 52.88 %, respectively. Furthermore, the nanogel for mixture of R/S improved cytotoxicity against MCF-7, HepG2, HCT116 and A549 cancers by 78.86 %, 54.75 %, 74.10 % and 56.53 % correspondingly. Also, cytotoxic activities of structures R, S and R/S and their nanogels exhibited low toxicity on VERO cells with IC50 = 30.90-46.50 μM and high selectivity against cancer cells. Moreover, R/S (nanogel), R (nanogel) and S (nanogel) demonstrated the uppermost binding affinities with DNA at reduced IC50 values of 31.50, 32.60, and 33.90 μM, respectively. In addition, they inhibited Topo-II activity with remarkably low IC50 value of 0.95, 1.00, and 1.10 μM, respectively.
Collapse
Affiliation(s)
- Shaymaa M M Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ahmad K B Aljohani
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ahmed El-Morsy
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Rwaida A Al Haidari
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | | | - Yahya A Maqnas
- College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Aishah M Alsibaee
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Mahmoud A H Mostafa
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Al Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Doaa E Keshek
- Department of Biology, Al-Jumum College University, Umm Al-Qura University, P.O Box7388, Makkah 21955, Saudi Arabia; Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Centre, Giza, Egypt
| | - May G Mohamed
- Department of Pharmacy Practice, College of Pharmacy, Gulf Medical University, United Arab Emirates
| | - Soad A L Bayoumi
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Khaled El-Adl
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt; Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11884, Cairo, Egypt.
| |
Collapse
|
10
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [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: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
11
|
Johnson OD, Paul S, Gutiérrez JA, Russell WK, Ward MC. DNA-damage-associated protein co-expression network in cardiomyocytes informs on tolerance to genetic variation and disease. iScience 2025; 28:112474. [PMID: 40469117 PMCID: PMC12135479 DOI: 10.1016/j.isci.2025.112474] [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: 09/05/2024] [Revised: 12/08/2024] [Accepted: 04/15/2025] [Indexed: 06/11/2025] Open
Abstract
Cardiovascular disease (CVD) is associated with genetic variants and environmental factors. A consequence of multiple risk factors is DNA damage. To examine how DNA damage influences the cardiomyocyte proteome and its relationship to CVD risk, we treated human induced pluripotent stem cell (iPSC)-derived cardiomyocytes with the DNA-damaging agent doxorubicin (DOX). A network constructed from 4,178 proteins reveals 12 co-expressed modules with 403 hub proteins. Five modules correlate with DOX and associate with RNA processing, chromatin regulation, and metabolism. DOX-correlated hub proteins are depleted for proteins that vary in expression across individuals due to genetic variation but are enriched for proteins encoded by loss-of-function intolerant genes. While not enriched for known CVD risk proteins, DOX-correlated hub proteins are enriched for the physical protein interactors of CVD risk proteins. These data demonstrate that protein connectivity in DNA-damage-associated modules influences the tolerance to genetic variation and supports the use of dynamic networks to explore complex traits.
Collapse
Affiliation(s)
- Omar Darrel Johnson
- Biochemistry, Cellular and Molecular Biology Graduate Program, University of Texas Medical Branch, Galveston, TX 77555, USA
- MD-PhD Combined Degree Program, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Sayan Paul
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - José Angel Gutiérrez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William Kent Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Michelle Claire Ward
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
12
|
Wang F, Wang K, Fang B, Geng S, Li Y, Qian H, Lin Y, Yu Z. Hollow mesoporous Prussian blue nanozymes alleviate doxorubicin-induced cardiotoxicity by restraining oxidative stress associated with Nrf2 signaling. J Colloid Interface Sci 2025; 686:1074-1088. [PMID: 39933346 DOI: 10.1016/j.jcis.2025.02.033] [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: 12/02/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
Doxorubicin-induced cardiomyopathy (DIC) is a toxic side effect that cannot be ignored during chemotherapy for malignant tumors. In this work, we synthesized a novel nano-chemotherapeutic drug based on Prussian blue nanozyme to alleviate DIC. Hollow mesoporous Prussian blue (HmPB) nanoparticles were used as a carrier to load doxorubicin (DOX) through electrostatic adsorption and obtain a novel chemotherapy drug, HmPB(DOX). In vivo and in vitro chemotherapy efficacy and acute toxicity evaluation experiments were conducted. The results suggest that HmPB(DOX) exhibits pH-responsive characteristics and minimizes the release of DOX from within HmPB(DOX) in cardiomyocytes. However, in the acidic tumor microenvironment, the release of DOX from HmPB(DOX) is notably enhanced. More importantly, HmPB(DOX) possesses excellent antioxidant enzyme activity, effectively clearing DOX-induced reactive oxygen species (ROS) and alleviating oxidative stress in cardiomyocytes. Doxorubicin is pivotal in the chemotherapy of malignant tumors. This study presents novel insights for mitigating the toxic and side effects of DOX, offering new strategies to enhance tolerance to chemotherapy.
Collapse
Affiliation(s)
- Fang Wang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, PR China
| | - Ke Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China; School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China
| | - BaoRu Fang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China; School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China
| | - SiQi Geng
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China; School of Life and Environmental Sciences, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China
| | - Ying Li
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China
| | - HuiFeng Qian
- Department of Clinical Laboratory, Shaoxing Second Hospital, Shaoxing City, Zhejiang Province 312000, PR China
| | - YiNuo Lin
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, PR China.
| | - ZhangSen Yu
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing City, Zhejiang Province 312000, PR China.
| |
Collapse
|
13
|
Cho CJ, Kang S, Pedebos C, Khalid S, Brea RJ, Devaraj NK. Diacylation of Peptides Enables the Construction of Functional Vesicles for Drug-Carrying Liposomes. Angew Chem Int Ed Engl 2025; 64:e202421932. [PMID: 39776211 DOI: 10.1002/anie.202421932] [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: 11/11/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 01/11/2025]
Abstract
Membrane-forming phospholipids are generated in cells by enzymatic diacylation of non-amphiphilic polar head groups. Analogous non-enzymatic processes may have been relevant at the origin of life and could have practical utility in membrane synthesis. However, aqueous head group diacylation is challenging in the absence of enzymes. The use of charged peptides instead of canonical phospholipid head groups offers advantages with respect to ease of acylation and chemical diversity. Here we demonstrate that native chemical ligation (NCL) enables in situ synthesis of diacylated lipopeptides (D-ALPs), which spontaneously self-assemble into micron-sized vesicles resembling cellular membranes. Diacylation occurs between non-amphiphilic peptides possessing an N-terminal cysteine, and acyl thioesters. Peptide head groups endow unique membrane functions, which is demonstrated by incorporation of an arginine-glycine-aspartic acid (RGD) motif, resulting in vesicle targeting to αvβ3 integrin-overexpressing cancer cells. The biocompatibility and functional group programmability of D-ALPs supports their broad utility as membrane mimetics.
Collapse
Affiliation(s)
- Christy J Cho
- Department of Chemistry and Biochemistry, University of California, San Diego La Jolla, CA 92093, USA
| | - Sangyoon Kang
- Department of Chemistry and Biochemistry, University of California, San Diego La Jolla, CA 92093, USA
| | - Conrado Pedebos
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, United Kingdom
- Programa de Pós-Graduação em Biociências (PPGBio), Universidade Federal de Ciências da Saúde de Porto Alegre - UFCSPA, Porto Alegre, 90050-170, Brazil
| | - Syma Khalid
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, United Kingdom
| | - Roberto J Brea
- Bioinspired Nanochemistry (BioNanoChem) Group, CICA -, Centro Interdisciplinar de Química e Bioloxía, Universidade da Coruña, Rúa As Carballeiras, 15071, A Coruña, Spain
| | - Neal K Devaraj
- Department of Chemistry and Biochemistry, University of California, San Diego La Jolla, CA 92093, USA
| |
Collapse
|
14
|
She RL, Liang XY, Hu L, Ma CY, Mu LY, Feng JH, Song JY, Jiang ZY, Li ZX, Qu XQ, Peng BQ, Wu KN, Kong LQ. Effect of chemotherapy and different chemotherapeutic regimens on electrocardiographic parameters in breast cancer women: a retrospective and within-subject longitudinal study. Support Care Cancer 2025; 33:452. [PMID: 40327163 DOI: 10.1007/s00520-025-09499-2] [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: 11/19/2024] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Descriptions of the effect of chemotherapy on all the electrocardiographic parameters in breast cancer women during chemotherapy are limited. METHODS A retrospective and within-subject longitudinal study of the effect of chemotherapeutics and different chemotherapeutic regimens on electrocardiographic parameters was conducted in 948 breast cancer women who had completed chemotherapy with ECG recording at initial diagnosis and before each cycle of chemotherpy. Heart rate (HR), QRS interval, P-R interval, QRS axis, QT interval, QTc interval, and the incidence of QTc interval prolongation were analyzed. Age, body mass index (BMI), history of hypertension, diabetes, and coronary heart disease were also collected for further stratified study. RESULTS Compared to initial diagnosis, changes in HR [74 (67, 82) bpm vs. 79 (73, 87)bpm], P-R interval [148 (136, 160) ms vs. 150 (138, 162) ms], QRS axis [41° (19.25°°, 60°) vs. 33° (15°, 53.75°)], QT interval [376 (358, 394) ms vs. 372 (354, 386) ms], QTc interval [417 (404, 431) ms vs. 426 (414, 436) ms], and incidence of QTc interval prolongation (9.6% vs. 15.8%) were all significant after chemotherapy, P < 0.001. There were statistically differences in HR, QRS axis, QTc interval, and the incidence of QTc interval prolongation between initial diagnosis and prechemotherapy of the last cycle under different age strata (≤ 45 years, 45 ~ 55 years, ≥ 55 years), different BMI range groups (18.5-22.9 kg/m2, 23-24.9 kg/m2, and 25-29.9 kg/m2), and even in patients without history of hypertension, diabetes, or coronary heart disease, respectively, P < 0.05. Resting HR, QRS axis, and QTc interval between each cycle of TEC regimen were different, P < 0.001. Resting HR and QTc interval between different cycles of EC-T(H) regimen were different, P < 0.05. Compared to initial diagnosis, a longer QTc interval occurred from the third to the last cycle of TEC regimen, P < 0.05. Only QTc interval before the fifth cycle of EC-T(H) regimen was statistically different from that at initial diagnosis, P = 0.012. CONCLUSION Chemotherapy affects the ECG parameters of HR, P-R interval, QRS axis, QT interval, QTc interval, and QTc interval prolongation in early-stage breast cancer women during chemotherapy. Electrocardiographic parameters may be affected significantly by TEC regimen than by EC-T(H) or TC regimen. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Rui-Ling She
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xin-Yu Liang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lei Hu
- Information Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chen-Yu Ma
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li-Yuan Mu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jun-Han Feng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jing-Yu Song
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhi-Yu Jiang
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhao-Xing Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiu-Quan Qu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bai-Qing Peng
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Kai-Nan Wu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ling-Quan Kong
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
15
|
Xing S, Liu Y, Xie H, Guo C, Wang X, Lv B, Li X, Shao J, Guo Q, Feng F, Sun H. Discovery of highly potent AKR1Cs pan-inhibitors as chemotherapeutic potentiators to restore breast cancer drug resistance. Eur J Med Chem 2025; 289:117413. [PMID: 40015157 DOI: 10.1016/j.ejmech.2025.117413] [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: 01/07/2025] [Revised: 02/07/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025]
Abstract
The acquired resistance of doxorubicin (DOX) significantly limits their application in breast cancer treatment. In earlier investigations, a pan-inhibitor, S07-2010, exhibiting inhibitory activity against Aldo-Keto Reductase 1C1-1C4 (AKR1C1-1C4) was discovered through virtual screening. In this study, four rounds of structural modifications were conducted, and the optimized compound 29 exhibited potent inhibitory activity against AKR1C1-1C4 (AKR1C1 IC50 = 0.09 μM, AKR1C2 IC50 = 0.28 μM, AKR1C3 IC50 = 0.05 μM, AKR1C4 IC50 = 0.51 μM). Molecular dynamics (MD) simulations revealed that 29 consistently occupied both SP2 and SP3 pockets, which may explain its pan-inhibitory activity. Utilizing highly DOX resistant MCF-7/ADR cells, 29 demonstrated superior potential as a therapeutic agent for re-sensitizing drug-resistant cell lines to chemotherapy both in vitro and in vivo, suggesting that pan-inhibition of AKR1C1-1C4 may serve as a more promising therapeutic strategy for drug-resistant breast cancer. In summary, Compound 29 may be a promising therapeutic adjuvant in the development of novel strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Huanfang Xie
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Can Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xiaolong Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Xinyu Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Jikuan Shao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, People's Republic of China; State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Feng Feng
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, People's Republic of China; Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
16
|
Wu H, Huang Z, Wang J. Neoadjuvant Chemotherapy Efficacy in Breast Cancer: Insights from Magnetic Resonance Imaging Compilation (MAGIC). Acad Radiol 2025:S1076-6332(25)00319-8. [PMID: 40318975 DOI: 10.1016/j.acra.2025.04.020] [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: 01/20/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025]
Abstract
RATIONALE AND OBJECTIVES This study aimed to investigate the role of Magnetic Resonance Imaging Compilation (MAGIC) technology in diagnosing pathologically complete response (pCR) after neoadjuvant chemotherapy (NAC) in breast cancer with mass enhancement (ME) and non-mass enhancement (NME) lesions. MATERIALS AND METHODS A total of 101 breast cancer patients from November 2021 to June 2024 were retrospectively analyzed and divided into the non-pCR group and the pCR group according to the Miller-Payne grade. Magnetic Resonance (MR) parameters before the first NAC and the last preoperative MR examination (post-parameters) were recorded, respectively. To compare the changes in MR parameters before and after NAC in the two types of breast cancer. RESULTS One hundred and one breast cancer patients with 114 lesions. 36.63% (37/101) of patients achieved pCR, and 68.42% (78/114) were ME lesions. The T1, T2, and PD values decreased, and the ADC value increased significantly after NAC in both ME and NME lesions. However, only post-T1, post-T2, and ADC values (excluding PD) effectively differentiated pCR from non-pCR in ME lesions, whereas no significant differences were observed in NME lesions. Notably, MAGIC technology combined with DWI significantly enhanced diagnostic accuracy for pCR in ME lesions (all P values < 0.05), but this approach showed limited utility in NME breast cancer. CONCLUSION For ME breast cancer, MAGIC technology serves as an important tool for clinical decision-making by effectively predicting pCR and, in combination with DWI, improving the accuracy of diagnosis.
Collapse
Affiliation(s)
- Honghong Wu
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310002, China (H.W.).
| | - Zebo Huang
- Department of Radiology, Women's Hospital of Nanjing Medical University, Nanjing Wowen and Children's HealthCare Hospital, Nanjing, Jiangsu 210004, China (Z.H., J.W.).
| | - Jie Wang
- Department of Radiology, Women's Hospital of Nanjing Medical University, Nanjing Wowen and Children's HealthCare Hospital, Nanjing, Jiangsu 210004, China (Z.H., J.W.).
| |
Collapse
|
17
|
Hubiernatorova A, Novak J, Vaskovicova M, Sekac D, Kropyvko S, Hodny Z. Tristetraprolin affects invasion-associated genes expression and cell motility in triple-negative breast cancer model. Cytoskeleton (Hoboken) 2025; 82:311-326. [PMID: 39319680 PMCID: PMC12063522 DOI: 10.1002/cm.21934] [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: 04/26/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Tristetraprolin (TTP) is an RNA-binding protein that negatively regulates its target mRNAs and has been shown to inhibit tumor progression and invasion. Tumor invasion requires precise regulation of cytoskeletal components, and dysregulation of cytoskeleton-associated genes can significantly alter cell motility and invasive capability. Several genes, including SH3PXD2A, SH3PXD2B, CTTN, WIPF1, and WASL, are crucial components of the cytoskeleton reorganization machinery and are essential for adequate cell motility. These genes are also involved in invasion processes, with SH3PXD2A, SH3PXD2B, WIPF1, and CTTN being key components of invadopodia-specialized structures that facilitate invasion. However, the regulation of these genes is not well understood. This study demonstrates that ectopic expression of TTP in MDA-MB-231 cells leads to decreased mRNA levels of CTTN and SH3PXD2A, as well as defects in cell motility and actin filament organization. Additionally, doxorubicin significantly increases TTP expression and reduces the mRNA levels of cytoskeleton-associated genes, enhancing our understanding of how doxorubicin may affect the transcriptional profile of cells. However, doxorubicin affects target mRNAs differently than TTP ectopic expression, suggesting it may not be the primary mechanism of doxorubicin in breast cancer (BC) treatment. High TTP expression is considered as a positive prognostic marker in multiple cancers, including BC. Given that doxorubicin is a commonly used drug for treating triple-negative BC, using TTP as a prognostic marker in this cohort of patients might be limited since it might be challenging to understand if high TTP expression occurred due to the favorable physiological state of the patient or as a consequence of treatment.
Collapse
Affiliation(s)
- Anastasiia Hubiernatorova
- Department of Functional GenomicsInstitute of Molecular Biology and Genetics NAS of UkraineKyivUkraine
- Laboratory of Cell Regeneration and PlasticityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
| | - Josef Novak
- Laboratory of Genome IntegrityInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Michaela Vaskovicova
- Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
- Laboratory of DNA IntegrityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
| | - David Sekac
- Laboratory of Cell Regeneration and PlasticityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
- Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
| | - Serhii Kropyvko
- Department of Functional GenomicsInstitute of Molecular Biology and Genetics NAS of UkraineKyivUkraine
| | - Zdenek Hodny
- Laboratory of Genome IntegrityInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
18
|
Xin W, Yang H, Heng X, Xu T, Zhang K, Zhao Y, Liu Y, Han D, Wu Y, Zhang W, He M, Pu L, Shen Y, Qu X, Sun N, Ye C. Sauchinone preserves cardiac function in doxorubicin-induced cardiomyopathy by inhibiting the NLRP3 inflammasome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156624. [PMID: 40068294 DOI: 10.1016/j.phymed.2025.156624] [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: 10/19/2024] [Revised: 03/02/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Doxorubicin (Dox)-induced cardiomyopathy (DIC) is characterized by severe myocardial damage that can progress to dilated cardiomyopathy and potentially lead to heart failure. No effective prevention or treatment strategies are available for DIC. Sauchinone, a diastereomeric lignan isolated from Saururus chinensis, is known for its notable anti-inflammatory effects. However, a paucity of research on sauchinone in relation to heart disease exists, particularly regarding its role in DIC, which remains unclear. PURPOSE This study aimed to assess the therapeutic potential of sauchinone in alleviating cardiac injury and elucidate its potential molecular mechanism in DIC. METHODS Male C57BL/6J mice were used to construct chronic and acute DIC models in vivo. The mice were administered sauchinone intragastrically concurrently with the first injection of Dox to evaluate the therapeutic effect of sauchinone on DIC. H9c2, a rat cardiomyocyte cell line, was treated with various concentrations of sauchinone in conjunction with Dox to assess the protective effects of sauchinone on cardiomyocyte injury in vitro. RESULTS Supplementation with exogenous sauchinone mitigated Dox-induced cardiac atrophy, cardiac fibrosis, and ventricular remodeling, while preserving cardiac function. Sauchinone reduced Dox-induced abnormal apoptosis both in vitro and in vivo. Additionally, sauchinone restored mitochondrial function and decreased reactive oxygen species levels, which may be attributed to its activation of nuclear factor erythroid 2-related factor 2 (NRF2) signaling, thereby attenuating Dox-induced oxidative damage. Furthermore, sauchinone significantly inhibited the activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome and reduced the cardiac infiltration of inflammatory factors, thereby alleviating oxidative stress and inhibiting the progression of DIC. The NLRP3 agonist nigericin abolished DIC progression, while the NLRP3 antagonist MCC950 further enhanced the beneficial effects of sauchinone on DIC progression both in vivo and in vitro. CONCLUSIONS The key novel finding of the present study is that the use of sauchinone, a diastereomeric lignan isolated from Saururus chinensis, effectively limits the progression of DIC. Specifically, sauchinone not only alleviates Dox-induced chronic cardiac injury but also significantly delays the progression of acute DIC. Mechanistically, inactivation of the NLRP3 inflammasome and NRF2-mediated antioxidant pathways have been identified as two critical signaling pathways regulated by sauchinone, which plays a vital role in blocking the progression of DIC. Sauchinone holds promise as a potential therapeutic approach for DIC or dilated cardiomyopathy.
Collapse
Affiliation(s)
- Wenxu Xin
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Hai Yang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Xinyu Heng
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Tao Xu
- Department of Geriatrics, Sir Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Ke Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Yining Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Yankui Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Deshen Han
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Yueyue Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Wei Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Meiqi He
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Lin Pu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China
| | - Yicong Shen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China
| | - Xiuxia Qu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China.
| | - Ning Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China.
| | - Chao Ye
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, No. 1800, Lihu Avenue, Wuxi, Jiangsu 214122, China; Department of Cardiology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Jiangnan University, Wuxi, Jiangsu 214122, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Wuxi School of Medicine, Jiangnan university, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
19
|
Zhao H, Wu Y, Kim SM. Enhancing doxorubicin's anticancer impact in colorectal cancer by targeting the Akt/Gsk3β/mTOR-SREBP1 signaling axis with an HDAC inhibitor. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:321-335. [PMID: 40254556 PMCID: PMC12012316 DOI: 10.4196/kjpp.24.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/05/2024] [Accepted: 11/19/2024] [Indexed: 04/22/2025]
Abstract
Colorectal cancer ranks third in global incidence and is the second leading cause of cancer-related mortality. Doxorubicin, an anthracycline chemotherapeutic drug, is integral to current cancer treatment protocols. However, toxicity and resistance to doxorubicin poses a significant challenge to effective therapy. Panobinostat has emerged as a critical agent in colorectal cancer treatment due to its potential to overcome doxorubicin resistance and enhance the efficacy of existing therapeutic protocols. This study aimed to evaluate the capability of panobinostat to surmount doxorubicin toxicity and resistance in colorectal cancer. Specifically, we assessed the efficacy of panobinostat in enhancing the therapeutic response to doxorubicin in colorectal cancer cells and explored the potential synergistic effects of their combined treatment. Our results demonstrate that the combination treatment significantly reduces cell viability and colony-forming ability in colorectal cancer cells compared to individual treatments. The combination induces significant apoptosis, as evidenced by increased levels of cleaved PARP and cleaved caspase-9, while also resulting in a greater reduction in p-Akt/p-GSK-3β/mTOR expression, along with substantial decreases in c-Myc and SREBP-1 levels, compared to monotherapies. Consistent with the in vitro experimental results, the combination treatment significantly inhibited tumor formation in colorectal cancer xenograft nude mice compared to the groups treated with either agent alone. In conclusion, our research suggests that the panobinostat effectively enhances the effect of doxorubicin and combination of two drugs significantly reduced colorectal cancer tumor growth by targeting the Akt/GSK-3β/mTOR signaling pathway, indicating a synergistic therapeutic potential of these two drugs in colorectal cancer treatment.
Collapse
Affiliation(s)
- Huaxin Zhao
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Yanling Wu
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Soo Mi Kim
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54907, Korea
- Research Institute of Clinical Medicine, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea
| |
Collapse
|
20
|
Liu H, Fan W, Li H, Qiao L, Liu Z, Zhu B, Guo J, Huang K, Tang Y, Wen J, Xue M, Wu Y, Zhao Y, Jiang Y, Liu K, Liang J, Cao M, Li J. Idarubicin versus Epirubicin in Transarterial Chemoembolization for Barcelona Clinic Liver Cancer Stage B Hepatocellular Carcinoma: An Open-label, Randomized, Phase IV Trial. Radiology 2025; 315:e242315. [PMID: 40326873 DOI: 10.1148/radiol.242315] [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: 05/07/2025]
Abstract
Background Transarterial chemoembolization (TACE) is regarded as the first-line treatment for patients with Barcelona Clinic Liver Cancer (BCLC) stage B hepatocellular carcinoma (HCC). However, the optimal chemotherapeutic agent loaded in TACE remains controversial. Purpose To compare the efficacy and safety of idarubicin and epirubicin as loaded drugs in drug-eluting bead (DEB)-TACE in patients with BCLC stage B HCC. Materials and Methods In this open-label, phase IV trial, patients with BCLC stage B HCC were recruited from four centers from August 2020 to October 2022 and randomly assigned (at a one-to-one ratio) to undergo idarubicin DEB-TACE or epirubicin DEB-TACE. The primary end point was progression-free survival (PFS), which was measured from the time of randomization to the time of progression or death from any cause. The efficacy analysis was conducted on an intention-to-treat basis, and only participants who received treatment were included in the safety analysis. Results A total of 239 participants (median age, 57 years; IQR, 50-66 years; 210 male) were randomly assigned to the idarubicin group (n = 120) or the epirubicin group (n = 119). The primary analysis cutoff for PFS was March 1, 2023, with 167 events observed (70%; idarubicin group, 85 events; epirubicin group, 82 events). The median PFS was 10.8 months and 8.7 months in the idarubicin and epirubicin groups, respectively (hazard ratio [HR], 0.61; 95% CI: 0.44, 0.84; P = .002). The HR for median overall survival (OS) was 0.53 (95% CI: 0.31, 0.88), with OS rates of 81.5% and 77.3% at 12 months and 71.8% and 54.0% at 24 months for the idarubicin and epirubicin groups, respectively. The objective response rates were 70.8% and 57.1% for the idarubicin and epirubicin groups, respectively (P = .03). There was no evidence of a between-group difference in incidence of adverse events, including hematologic toxicity. No treatment-related deaths were observed. Conclusion Idarubicin DEB-TACE increased survival in participants with BCLC stage B HCC, without an increase in the incidence of any adverse events. Clinical trial registration no. ChiCTR2000034758 © RSNA, 2025 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Haikuan Liu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Haiqing Li
- Department of Tumor Minimally Invasive, Taian City Central Hospital, Taian, China
| | - Liangliang Qiao
- Department of Interventional Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhilong Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bowen Zhu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Jian Guo
- Department of Interventional Radiology, Guangdong Province Hospital-Zhuhai Hospital, Zhuhai, China
| | - Kun Huang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Yiyang Tang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Jie Wen
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Miao Xue
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Yanqin Wu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Yue Zhao
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Yang Jiang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| | - Kangshou Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junjie Liang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mingrong Cao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University No.58 Zhongshan 2nd Rd, Guangzhou 510080, China
| |
Collapse
|
21
|
Wang W, Cai Y, Chen Y, Zhu J, Jin J. Comprehensive Analysis of the Multi-Target Binding Mechanism of Doxorubicin: Integrating Protein Microarray Screening, Molecular Docking, and Molecular Dynamics Simulation. Arch Pharm (Weinheim) 2025; 358:e70006. [PMID: 40345153 DOI: 10.1002/ardp.70006] [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: 02/15/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Understanding the mechanisms through which anticancer drugs interact with multiple protein targets is crucial for optimizing drug design and enhancing the efficacy of chemotherapy. This study focuses on doxorubicin, a broad-spectrum anticancer drug recognized for its multi-target mechanisms of action. We initially screened 363 doxorubicin-binding proteins using protein microarrays; of these, 166 proteins with known PDB (Protein Data Bank) structures were selected for molecular docking to evaluate their binding energies. The binding energy distribution and residue enrichment analyses revealed that doxorubicin preferentially binds to specific residues at its binding sites, including serine, glycine, arginine, glutamic acid, lysine, aspartic acid, and leucine. These residues stabilize doxorubicin binding through hydrogen bonds, hydrophobic interactions, and electrostatic interactions. In addition, RUVBL1 (RuvB-like AAA ATPase 1) exhibited the highest integrated score from the protein microarray and molecular docking analyses. Furthermore, PPI (protein-protein interaction) network analysis and centrality calculations identified key proteins with potential regulatory roles, with MAPK1 (mitogen-activated protein kinase 1) exhibiting the highest betweenness centrality in the PPI network. Finally, molecular dynamics simulations of the RUVBL1- and MAPK1-doxorubicin complexes were conducted to evaluate the binding mechanisms. The simulations revealed key binding residues, including Ile56, Lys59, Leu87, Pro296, and Ile326 in RUVBL1 and Asp88, Ile89, Pro93, Phe354, and Ala92 in MAPK1 that mediate stable interactions with doxorubicin. This study presents a comprehensive analytical approach for investigating the interactions between doxorubicin and multiple protein targets, providing a reference framework for understanding the molecular mechanisms of anticancer drugs and for future analyses of similar data sets.
Collapse
Affiliation(s)
- Wentao Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
22
|
Gautam S, Joshi S, Jindal P, Patel P, Pal RR, Kumar M, Gupta GD, Kurmi BD. Recent doxorubicin-conjugates in cancer drug delivery: Exploring conjugation strategies for enhanced efficacy and reduced toxicity. Int J Pharm 2025; 675:125556. [PMID: 40187698 DOI: 10.1016/j.ijpharm.2025.125556] [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: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Doxorubicin is a first-line treatment of cancer that works on the mechanism of DNA intercalation and topoisomerase II poisoning. Since the 20th century, Doxorubicin has been used as a promising drug to treat several types of cancer, both solid or metastatic, including breast, thyroid, bladder, ovarian, or gastric cancer, etc. Even though it shows promising effects on cancer cells, it also shows its effects on healthy cells with cancerous cells, which leads to several severe side effects, such as cardiomyopathy, phlebitis, congestive heart failure (CHF), etc., which limits its usage in chemotherapy. Several research has focused on the targeted delivery of doxorubicin to cancerous cells to reduce side effects and improve efficacy. To optimize its anticancer potential, scientists have recently been developing nano-formulations and investigating various conjugations. The structure of doxorubicin consists of two primary functional groups that can be employed for conjugation with a variety of biomolecules, The first is the primary amine group in a sugar moiety, and the other one is the primary hydroxyl group in the aliphatic chain ring. In this paper, we have mentioned several conjugations of doxorubicin such as antibodies, nanoparticles, polymers, and phytochemical conjugations. Different studies regarding these conjugations are also mentioned, which represent promising strategies to optimize cancer treatment by minimizing side effects.
Collapse
Affiliation(s)
- Shreastha Gautam
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Sachin Joshi
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Priya Jindal
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road Moga-142001, Punjab, India.
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India.
| |
Collapse
|
23
|
Supawat B, Kothan S, Kaewkhao J, Tima S, Tungjai M. Effect of low-dose radiation on the kinetics of pirarubicin and daunorubicin transport in K562 cells and drug resistant K562/
adr
cells. RADIATION EFFECTS AND DEFECTS IN SOLIDS 2025:1-17. [DOI: 10.1080/10420150.2025.2495583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
|
24
|
Shokrzadeh S, Moghim S, Shokrzadeh M, Aghajanshakeri S. An added value of azithromycin: mitigation of doxorubicin-associated oxidative damage and genotoxicity in normal human bronchial epithelium cells. Mutagenesis 2025; 40:126-136. [PMID: 39373202 DOI: 10.1093/mutage/geae024] [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: 06/13/2024] [Accepted: 10/05/2024] [Indexed: 10/08/2024] Open
Abstract
Doxorubicin, a well-known and widely used antineoplastic agent with direct ROS-accumulating activity, has proven effective in treating various cancer types. However, its non-specific cytotoxicity towards non-cancerous cells prompts concerns regarding potential adverse effects. Azithromycin is an antibiotic for treating bacterial infections and an anti-inflammatory agent, particularly beneficial in managing respiratory conditions like bronchitis and sinusitis. Despite azithromycin's well-documented antibacterial properties, its potential cellular/genomic protective effects remain unexplored. As an in vitro model, BEAS-2B cells (normal human bronchial epithelium cells) were employed in this study to assess whether azithromycin possesses any protective properties against doxorubicin-induced cellular toxicity. Cells in pretreatment culture were treated to various amounts of azithromycin (3.125, 6.25, 12.5, 25, and 50 μg/ml) in combination with doxorubicin at IC50 (0.08 μg/ml). Doxorubicin at 0.08 μg/ml highlighted cytotoxicity, oxidative stress, and genotoxicity. Azithromycin at 25 and 50 μg/ml markedly modulated oxidative stress and genomic damage by decreasing the ROS and LPO amounts and suppressing DNA fragmentation in the comet assay parameters. Consequently, azithromycin may be regarded as a cytomodulating, antigenotoxic, and antioxidant agent.
Collapse
Affiliation(s)
| | - Shahrzad Moghim
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Shokrzadeh
- Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shaghayegh Aghajanshakeri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
25
|
Zafar A, Khatoon S, Khan MJ, Abu J, Naeem A. Advancements and limitations in traditional anti-cancer therapies: a comprehensive review of surgery, chemotherapy, radiation therapy, and hormonal therapy. Discov Oncol 2025; 16:607. [PMID: 40272602 PMCID: PMC12021777 DOI: 10.1007/s12672-025-02198-8] [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: 09/21/2024] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Cancer remains a major global health challenge, consistently ranking as the second leading cause of mortality worldwide. Despite significant advancements in research and technology, the need to deepen our understanding of tumor biology and improve therapeutic strategies persists. This review focuses on the progress and challenges of four traditional cancer treatment modalities: surgery, chemotherapy, radiation therapy, and hormonal therapy. Surgery, the primary method for tumor removal, has evolved with the integration of fluorescence-based technology and robotic systems, enhancing precision and minimizing collateral damage. Radiation therapy has progressed with improved focus, intensity control, and 3D technology, refining both diagnosis and treatment. Chemotherapy has advanced from natural extracts to synthesized derivatives with amplified cytotoxicity against cancer cells. Hormonal therapy has emerged as a crucial strategy for hormone-dependent cancers, restraining growth or inducing regression. Despite these advancements, each approach faces ongoing challenges. Surgery struggles with complete tumor removal due to heterogeneity. Chemotherapy contends with drug resistance and side effects. Radiation therapy grapples with precision issues and limited access in some regions. Hormonal therapy faces resistance development and quality of life impacts. This study provides a comprehensive analysis of the evolution of these traditional anti-cancer therapies, offering insights into their progress and highlighting areas for future research. By examining these modalities, we aim to underscore their relevance in the current oncology landscape and identify opportunities for improvement in cancer treatment strategies.
Collapse
Affiliation(s)
- Aasma Zafar
- Department of Biosciences, COMSATS University, Islamabad, 45550, Pakistan
| | - Summaiya Khatoon
- College of Health Sciences, QU Health Sector, Qatar University, P.O. Box 2713, Doha, Qatar
| | | | - Junaid Abu
- Aisha bint Hamad al-Attiyah hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Aisha Naeem
- Research and Graduate Studies, QU Health Sector, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
26
|
Kang H, Thomas RG, Kim S, Ju JK, Jeong YY. Anticancer therapeutic effect of magnetic guided cobalt ferrite/doxorubicin-loaded ROS-responsive bilirubin nanoparticles in a colon cancer model. Colloids Surf B Biointerfaces 2025; 248:114487. [PMID: 39756160 DOI: 10.1016/j.colsurfb.2024.114487] [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: 12/29/2023] [Revised: 11/19/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE The aim of this study is to synthesize the cobalt iron oxide (CoFe) and doxorubicin (Dox)-loaded chitosan bilirubin (ChiBil) nanoparticles and to investigate the anticancer therapeutic effect of the synthesized nanoparticles under magnetic guidance in a colon cancer. MATERIALS AND METHODS ChiBil-CoFe-Dox nanoparticles were synthesized by conjugating CoFe and Dox and then loaded onto ChiBil nanoparticles. Synthesis were characterized using thermogravimetric (TGA) analysis, inductive coupled plasma (ICP) analysis, dynamic light scattering (DLS), zeta potential and field emission-transmission electron microscopy (FE-TEM). Cellular uptake and cytotoxicity studies were conducted in vitro. Biodistribution and tumor inhibition study was done in vivo CT-26 colon cancer model. RESULTS The ChiBil-CoFe-Dox nanoparticles were successfully synthesized in this study. The in vitro cytotoxicity study showed that the ChiBil-CoFe-Dox nanoparticle had a toxic effect on cancer cells. The accumulation of ChiBil-CoFe-Dox nanoparticles was enhanced under magnetic guidance, as observed by in vivo. Tumor inhibition study showed that the ChiBil-CoFe-Dox nanoparticle effectively reduced tumor size in vivo mice colon cancer model, especially when combined with magnetic guidance. CONCLUSION This study showed that ChiBil-CoFe-Dox nanoparticle was successfully synthesized and effectively reduced tumor size, especially when combined with magnetic guidance. The in vitro and in vivo results suggested that the ROS stimuli responsive ChiBil-CoFe-Dox nanoparticles may be a potent therapeutic option for treating colon cancer.
Collapse
Affiliation(s)
- Hyo Kang
- Department of Surgery, Chonnam National University Medical School, 42, Jebong-ro, Dong-gu, Gwangju 61469, South Korea
| | - Reju George Thomas
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, South Korea.
| | - Subin Kim
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Jae Kyun Ju
- Department of Surgery, Chonnam National University Medical School, 42, Jebong-ro, Dong-gu, Gwangju 61469, South Korea.
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, South Korea.
| |
Collapse
|
27
|
Aljohani AKB, El-Hddad SSA, Alsulaimany M, Maghrabi NA, Alhammad AM, Aloufi MN, Alahmadi LF, Ali Saeedi T, Neyaz YY, Anwer KE, Abd El-Sattar NEA, El-Adl K. Design, nanogel synthesis, anti-proliferative activity and in silico ADMET profile of pyrazoles and pyrimidines as topo-II inhibitors and DNA intercalators. RSC Adv 2025; 15:10037-10048. [PMID: 40171282 PMCID: PMC11960622 DOI: 10.1039/d5ra00166h] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
Pyrazole derivatives 2 and 3, pyrimidine derivative 4, and their nanogels as drug delivery systems were synthesized, and their cytotoxicity against MCF-7, HCT-116, HepG2 and A549 cells was evaluated. Herein, we focused on the characterization and synthesis of chitosan/polyvinyl alcohol (Cs/PVA) nanogels loaded with derivatives 2, 3 and 4. The stability of the prepared nanogels 2, 3 and 4 was elucidated by zeta potential measurements, which possessed negative values of -9.7, -1.3 and -1.6 mV, respectively. Our compounds and their nanogels were evaluated as topo-II inhibitors and DNA intercalators. The nanogel delivery system enhanced the cytotoxicity of compound 2 against the A549, HCT116, HepG2 and MCF-7 cancer cell lines by 32.06%, 28.96%, 44.32% and 50.00%, respectively. Moreover, the nanogel of compound 3 exhibited enhanced cytotoxicity against the A549, HCT116, HepG2 and MCF-7 cancer cell lines by 33.61%, 30.64%, 44.69% and 47.86%, respectively. Furthermore, the nanogel of compound 4 showed enhanced cytotoxicity against the A549, HCT116, HepG2 and MCF-7 cancer cell lines by 31.82%, 40.12%, 50.00% and 52.61%, respectively. Moreover, derivatives 2, 2 (nanogel), 3, 3 (nanogel), 4 and 4 (nanogel) exhibited good selectivity against cancer cells and reduced toxicity to VERO cells with IC50 values in the range of 48.29-59.70 μM. Furthermore, our derivatives displayed remarkable in silico predicted ADMET profiles.
Collapse
Affiliation(s)
- Ahmed K B Aljohani
- Department of Pharmacognosy & Pharmaceutical Chemistry, College of Pharmacy, Taibah University Medina 42353 Saudi Arabia
| | - Sanadelaslam S A El-Hddad
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Omar Almukhtar University Al Bayda 991 Libya
| | - Marwa Alsulaimany
- Department of Pharmacognosy & Pharmaceutical Chemistry, College of Pharmacy, Taibah University Medina 42353 Saudi Arabia
| | - Nader A Maghrabi
- Pharmacy Student, College of Pharmacy, Taibah University Medina Saudi Arabia
| | - Alaa M Alhammad
- Pharmacy Student, College of Pharmacy, Taibah University Medina Saudi Arabia
| | - Mayar N Aloufi
- Pharmacy Student, College of Pharmacy, Taibah University Medina Saudi Arabia
| | - Luai F Alahmadi
- Pharmacy Student, College of Pharmacy, Taibah University Medina Saudi Arabia
| | - Turkiah Ali Saeedi
- Pharmacy Student, College of Pharmacy, Taibah University Medina Saudi Arabia
| | - Yousef Yakoub Neyaz
- Pharmacy Student, College of Pharmacy, Taibah University Medina Saudi Arabia
| | - Kurls E Anwer
- Department of Chemistry, Faculty of Science, Ain Shams University Abbassia Cairo Egypt
| | - Nour E A Abd El-Sattar
- Department of Chemistry, Faculty of Science, Ain Shams University Abbassia Cairo Egypt
- Basic & Medical Sciences Department, Faculty of Dentistry, Alryada University for Science & Technology Egypt
| | - Khaled El-Adl
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development Cairo Egypt
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University Nasr City 11884 Cairo Egypt
| |
Collapse
|
28
|
Li M, Kou X, Zheng X, Guo X, Qi W, Li C, Chen J. Effects of Anthracyclines on Pericardial Adipose Tissue Assessed by Magnetic Resonance Imaging - An Animal Experiment. Circ J 2025:CJ-24-0794. [PMID: 40128948 DOI: 10.1253/circj.cj-24-0794] [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] [Indexed: 03/26/2025]
Abstract
BACKGROUND Anthracyclines are widely used in cancer treatment, yet their potential for anthracycline-induced cardiotoxicity (AIC) limits their clinical utility. Despite the significant anatomical relevance of pericardial adipose tissue (PeAT) to cardiovascular disease, its response to anthracycline exposure remains poorly understood. METHODS AND RESULTS Male New Zealand White rabbits (n=17) received weekly doxorubicin injections and underwent magnetic resonance imaging (MRI) scans biweekly for 10 weeks. PeAT volumes (total, left paraventricular, right paraventricular) were measured together with ventricular function. Histopathological evaluations were also conducted. A mixed linear model identified the earliest timeframe for detectable changes in PeAT volume and left ventricular function. Total PeAT volume decreased from the 6th week (1.17±0.06, P<0.05) and continued to decrease until the 8th week (0.96±0.06, P<0.05) and left paraventricular adipose tissue volume decreased significantly, but no changes were observed in right paraventricular adipose tissue volume. The volume of PeAT exhibited a positive correlation with left ventricular ejection fraction (LVEF) (r=0.43, P<0.05), which declined below 50% by the 8th week, and a negative correlation with myocardial cell injury scores (r=-0.595, P<0.05). CONCLUSIONS Anthracycline administration led to an early reduction in PeAT volume, particularly in the left paraventricular region, detectable by MRI as early as the 6th week. Changes in PeAT volume preceded alterations in LVEF and were associated with declines in cardiac function and myocardial cell damage.
Collapse
Affiliation(s)
- Mengxi Li
- School of Clinical Medicine, Southwest Medical University
| | - Xingyuan Kou
- Department of Radiology, The Affiliated Hospital, Southwest Medical University
| | - Xue Zheng
- Department of Radiology, The Affiliated Hospital, Southwest Medical University
| | - Xi Guo
- Department of Radiology, The Affiliated Hospital, Southwest Medical University
| | - Wanyin Qi
- Department of Radiology, The Affiliated Hospital, Southwest Medical University
| | - Cao Li
- Department of Radiology, The Affiliated Hospital, Southwest Medical University
| | - Jing Chen
- Department of Radiology, The Affiliated Hospital, Southwest Medical University
| |
Collapse
|
29
|
Camilli M, Maggio L, Tinti L, Torre I, Viscovo M, Viscovo M, Tamburrini G, Lombardo A, Cardinale DM, Minotti G, Rocca B. Cardio-oncology: Emerging Concepts in Cardiovascular Sequelae of Cancer Therapies, Translational Research and Reverse Cardio-oncology. Eur Cardiol 2025; 20:e05. [PMID: 40170756 PMCID: PMC11959581 DOI: 10.15420/ecr.2024.49] [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: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 04/03/2025] Open
Abstract
Cardio-oncology was established with the aim of defining primary and secondary prevention approaches through surveillance and the use of tools to stratify and diminish the cardiovascular risk to cancer patients. This branch of medicine also contributes to establishing a new field in translational medicine for cardiovascular disease by focusing on the interplay between cancer and heart disease. In this first article in the new cardio-oncology section of the journal, we explore the main concepts of emerging anti-cancer therapies and their plausible cardiotoxic effects and we will describe advances and gaps in knowledge, highlighting how cardio-oncology is contributing to translational cardiology. We will speculate on the complex interplay between cancer and heart failure and discuss an emerging concept known as reverse cardio-oncology. We also present the perspective that cardio-oncology represents a promising platform area of research, allowing the discovery of novel pathways involved in cardiovascular disease through the identification of toxicities induced by targeted cancer therapies.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | - Luca Maggio
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Ilaria Torre
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Marcello Viscovo
- Department of Laboratory and Hematology Sciences, Fondazione Policlinico Universitario A Gemelli IRCCSRome, Italy
- Department of Radiology and Hematology Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Marcello Viscovo
- Department of Laboratory and Hematology Sciences, Fondazione Policlinico Universitario A Gemelli IRCCSRome, Italy
- Department of Radiology and Hematology Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Giulia Tamburrini
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | | | - Giorgio Minotti
- Unit of Drug Sciences, Fondazione Policlinico Universitario Campus Bio-MedicoRome, Italy
| | - Bianca Rocca
- Department of Medicine and Surgery, Libera Università MediterraneaBari, Italy
- Department of Safety and Bioethics, Università Cattolica del Sacro CuoreRome, Italy
| |
Collapse
|
30
|
Kondaka K, Rząd K, Maciejewska N, Gabriel I. Anticancer drugs targeting topoisomerase II for antifungal treatment. Sci Rep 2025; 15:9311. [PMID: 40102495 PMCID: PMC11920057 DOI: 10.1038/s41598-025-93863-z] [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: 07/04/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Fungal topoisomerase II (TopoII) has been identified as essential for viability. Thus, our research aimed to investigate the potential of fungal TopoII as a novel target for antifungal chemotherapy. We conducted studies on eleventh antitumor compounds targeting human topoisomerase II, either approved by the U.S. Food and Drug Administration (FDA) or currently under clinical trials to evaluate their potential for use in other therapeutic applications. While most of the compounds we analyzed are potent inhibitors of yeast TopoII, only a few exhibited antifungal activity. Idarubicin emerged as the most potent compound effectively inhibiting the growth of five reference fungal strains as well as clinical Candida glabrata fluconazole-resistant cells. Antifungal activity of this compound corresponded with its very high yeast TopoII inhibitory effectiveness. Additionally, idarubicin ability to be effectively accumulated into fungal cells is crucial for yeast TopoII targeting. Idarubicin, epirubicin, and bisantrene appeared to be even more effective inhibitors of yeast enzyme than its human counterpart. In fungal cells idarubicin exhibited a multifaceted mechanisms of action, including nuclear DNA fragmentation, disruption of mitochondrial network architecture and mitochondrial DNA aggregation as well as oxidative stress induction. Our results indicate that fungal topoisomerase II targeting is worth considering in antifungal treatment and the reported drugs may serve as a starting point for the reinnovation of a new molecule.
Collapse
Affiliation(s)
- Kavya Kondaka
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Kamila Rząd
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Natalia Maciejewska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland
| | - Iwona Gabriel
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 11/12 Narutowicza Str, Gdansk, 80-233, Poland.
| |
Collapse
|
31
|
Zhang X, Yin G, Lan M, Zhao H. 2,2,6,6-tetramethylpiperidin-1-oxyl: a new potential targeted ligand based on lipid peroxidation for targeted drug delivery. J Drug Target 2025:1-12. [PMID: 40022629 DOI: 10.1080/1061186x.2025.2474639] [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: 11/10/2024] [Revised: 01/27/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The side effects of chemotherapy drugs have prompted the development of targeted therapies. Distinctive abundance of lipid peroxidation (LPO) in tumour cells represents a potential target for drug delivery. However, LPO-based targeted ligands remain under-exploited. In this work, the targeting of 2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPO), was investigated within a mesoporous silica nanoparticle (MSN) loaded with doxorubicin (DOX) and connected with 4-NH2-TEMPO obtaining DOX/MSN-TEMPO. A cellular uptake assay showed a faster uptake of DOX/MSN-TEMPO than blank group on Hela, L929 and 4T1 cells, revealing TEMPO's active targeting ability for tumour cells. After observing this phenomenon, the fabrication of a basic copolymer module carrying cyanine5.5 (Cy5.5) and TEMPO was reported. In vivo experiments were conducted on mouse MCF-7 tumour models, displaying selective aggregation of nano micelles at the tumour site and thereby verifying the broad applicability of TEMPO. Since the large amounts of LPO lead to the presence of numerous free radicals, whereas TEMPO, as a free radical capture agent, further selectively targets tumour cells. These findings verify the targeting ability of TEMPO for most tumour cells and collectively underscore the potential of TEMPO and analogous capture agents as innovative targeted ligands for drug delivery.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Research Center of Analysis and Test, East China University of Science and Technology, Shanghai, P.R. China
| | - Guohao Yin
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Research Center of Analysis and Test, East China University of Science and Technology, Shanghai, P.R. China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Research Center of Analysis and Test, East China University of Science and Technology, Shanghai, P.R. China
| | - Hongli Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Research Center of Analysis and Test, East China University of Science and Technology, Shanghai, P.R. China
| |
Collapse
|
32
|
Nicol M, Deniau B, Rahli R, Genest M, Polidano E, Assad N, Samuel JL, Mebazaa A, Solal AC, Azibani F. Streptozotocin-induced hyperglycemia unmasks cardiotoxicity induced by doxorubicin. Sci Rep 2025; 15:8104. [PMID: 40057546 PMCID: PMC11890863 DOI: 10.1038/s41598-025-91824-0] [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: 01/25/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
Late-onset cardiotoxicity induced by anthracyclines occurs years to decades after completion of anti-cancer therapy and is associated with increased morbi-mortality of cancer survivors. Chemotherapy at the time of treatment probably causes cardiac damages for which the juvenile heart compensate. Co-morbidities happening in the adulthood such as type 1 diabetes (DT1), affect the heart and thus can unmask chemotherapy induced cardiotoxicity. To prove our hypothesis, we induced hyperglycemia [Streptozotocin treatment (STZ), 50 mg/kg/day for 5 days] in 11 weeks old mice who previously received doxorubicin treatment (Dox, 3 mg/kg) when they were six-weeks old. Interestingly, streptozotocin-induced hyperglycemia in Dox-pretreated mice (Dox-STZ) induced a higher mortality (p < 0.05) and more severe cardiac dysfunction (p < 0.0001) when compared with mice receiving Dox or STZ alone. Apoptosis evaluated by caspase 3 protein expression and Bax/Bcl2 genes expression was higher in Dox-STZ mice compared to STZ or Dox alone. While Dox and STZ independently induced capillary rarefaction, cardiomyocytes atrophy was only induced by STZ. Furthermore, Sirius-red staining of cardiac sections showed higher fibrosis levels (p < 0.0001) in Dox-STZ compared to Dox or STZ alone. All together, these results demonstrate that STZ precipitates and unmask cardiac dysfunction in previously treated Dox animals.
Collapse
Affiliation(s)
- Martin Nicol
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Cardiology Department, Lariboisière Hospital, University of Paris, Paris, France
| | - Benjamin Deniau
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Department of Anesthesiology and Intensive Care, Lariboisière - Saint Louis Hospitals, APHP, Paris, France
| | - Roza Rahli
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Magali Genest
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Evelyne Polidano
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Noma Assad
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Jane-Lise Samuel
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
| | - Alexandre Mebazaa
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Department of Anesthesiology and Intensive Care, Lariboisière - Saint Louis Hospitals, APHP, Paris, France
| | - Alain Cohen Solal
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France
- Cardiology Department, Lariboisière Hospital, University of Paris, Paris, France
| | - Feriel Azibani
- Inserm UMR-S 942 MASCOT, University of Paris Cité, Lariboisière Hospital, Paris, France.
| |
Collapse
|
33
|
Kumar M, Sengar AS, Lye A, Kumar P, Mukherjee S, Kumar D, Das P, Chatterjee S, Stewart A, Maity B. FNDC5/irisin mitigates the cardiotoxic impacts of cancer chemotherapeutics by modulating ROS-dependent and -independent mechanisms. Redox Biol 2025; 80:103527. [PMID: 39923397 PMCID: PMC11850786 DOI: 10.1016/j.redox.2025.103527] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/17/2024] [Accepted: 02/01/2025] [Indexed: 02/11/2025] Open
Abstract
Cardiotoxicity remains a major limiting factor in the clinical implementation of anthracycline chemotherapy. Though the etiology of doxorubicin-dependent heart damage has yet to be fully elucidated, the ability of doxorubicin to damage DNA and trigger oxidative stress have been heavily implicated in the pathogenesis of chemotherapy-associated cardiomyopathy. Here, we demonstrate that fibronectin type III domain-containing protein 5 (FNDC5), the precursor protein for myokine irisin, is depleted in the hearts of human cancer patients or mice exposed to chemotherapeutics. In cardiomyocytes, restoration of FNDC5 expression was sufficient to mitigate reactive oxygen species (ROS) accumulation and apoptosis following doxorubicin exposure, effects dependent on the irisin encoding domain of FNDC5 as well as signaling via the putative irisin integrin receptor. Intriguingly, we identified two parallel signaling cascades impacted by FNDC5 in cardiomyocytes: the ROS-driven intrinsic mitochondrial apoptosis pathway and the ROS-independent Ataxia Telangiectasia and Rad3-Related Protein (ATR)/Checkpoint Kinase 1 (Chk1) pathway. In fact, FNDC5 forms a co-precipitable complex with Chk1 alluding to possible intracellular actions for this canonically membrane-associated protein. Whereas FNDC5 overexpression in murine heart was cardioprotective, introduction of FNDC5-targeted shRNA into the myocardium was sufficient to trigger Bax up-regulation, ATR/Chk1 activation, oxidative stress, cardiac fibrosis, loss of ventricular function, and compromised animal survival. The detrimental impact of FNDC5 depletion on heart function could be mitigated via treatment with a Chk1 inhibitor identifying Chk1 hyperactivity as a causative factor in cardiac disease. Though our data point to the potential clinical utility of FNDC5/irisin-targeted agents in the treatment of chemotherapy-induced cardiotoxicity, we also found significant down regulation in FNDC5 expression in the hearts of aged mice that attenuated the cardioprotective impacts of FNDC5 overexpression following doxorubicin exposure. Together our data underscore the importance of FNDC5/irisin in maintenance of cardiac health over the lifespan.
Collapse
Affiliation(s)
- Manish Kumar
- Centre of Biomedical Research, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Abhishek Singh Sengar
- Centre of Biomedical Research, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Anushree Lye
- Centre of Biomedical Research, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India; Department of Biological Sciences, Bose Institute, EN 80, Sector V, Kolkata, West Bengal, 700091, India
| | - Pranesh Kumar
- Institute of Pharmaceutical Science, University of Lucknow, Uttar Pradesh, 226007, India
| | - Sukhes Mukherjee
- Department of Biochemistry, AIIMS Bhopal, Saketnagar, Bhopal, Madhya Pradesh, India
| | - Dinesh Kumar
- Centre of Biomedical Research, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology, Tamil Nadu, 603203, India
| | - Suvro Chatterjee
- Department of Biotechnology, Burdwan University, West Bengal, 713104, India
| | - Adele Stewart
- Department of Neuroscience & Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Biswanath Maity
- Centre of Biomedical Research, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India; Department of Biological Sciences, Bose Institute, EN 80, Sector V, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
34
|
Li S, Li N, Yin Q, Zhang Z, Hu H, Hao L. cRGD-based MRI imaging-enhanced nanoplatform helps DOX target pancreatic cancer. Sci Rep 2025; 15:7217. [PMID: 40021813 PMCID: PMC11871140 DOI: 10.1038/s41598-025-91549-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025] Open
Abstract
This project aims to construct cRGD functionalized mesoporous silica nanoparticles and cRGD modified mesoporous silica nanoparticles for the diagnosis and treatment of tumors, providing new ideas for targeted therapy of tumors. The mesoporous silica nanoparticles were doped with gadolinium in situ to provide excellent imaging; cRGD was coupled on the particle surface to confer particle targeting; and hyaluronic acid was loaded onto the particles by electrostatic adsorption, thereby improving the biocompatibility of the particles and prolonging their in vivo circulation time.Taking pancreatic cancer as a model, we studied its targeting ability to pancreatic cancer and its phagocytosis to cancer cells; Using methods such as cell growth experiments and flow cytometry, the anti-cancer effect and pro apoptotic effect of the system were studied. In vivo distribution, tumor targeting and therapeutic efficacy of nanoparticles evaluated in a mouse model of pancreatic cancer with loaded tumors.Evaluate the bioavailability and enrichment of nanoparticles in tumor tissue using MRI technology. Evaluate the therapeutic effect and safety through changes in tumor volume, histopathological examination, and prognosis. Characterization of the synthesis results proved that cRGD-HA-DOX-Gd2O3@MSN (cHDG@MSN) was successfully synthesized with a particle size of 230.83 ± 12.36 nm.In vitro drug release experiments of DOX were carried out at different pH values (5.5 and 7.4), where the release was only up to 22.65% at pH 7.4, whereas DOX release was increased up to 78.75% at pH = 5.5.The results confirm the pH responsiveness of this nanocarrier platform.The results of cytotoxicity studies showed that cHDG@MSN itself is not cytotoxic. Biosafety evaluation and hemolysis test results confirmed that the probe is highly biocompatible.Notably, Gd3+ significantly enhanced the T1 contrast of the system according to MR imaging results.The apoptosis rates of SW1990 cells treated with PBS, DOX and cHDG@MSN in flow cytometry were 13.97%, 18.38% and 29.02%, respectively, demonstrating the effectiveness of the nanoprobes at the cellular level. Animal experiments demonstrated the effectiveness of nanoprobes at the pathological level and imaging level.Cells and animals demonstrated that cHDG@MSN effectively inhibited the proliferation of pancreatic cancer cells. This research further verified the pH sensitivity of the constructed compound drug delivery system to achieve accurate diagnosis and treatment of pancreatic cancer tumor cells.
Collapse
Affiliation(s)
- Silong Li
- Medical Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang Province, 161006, People's Republic of China
| | - Na Li
- Department of Imaging Medicine and Nuclear Medicine, School of Clinical Medicine, Jiamusi University, Jiamusi, 154002, People's Republic of China
| | - Qiangqiang Yin
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People's Republic of China
| | - Zhichen Zhang
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People's Republic of China
| | - Haifeng Hu
- Medical Imaging Center, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang, 161000, People's Republic of China
| | - Liguo Hao
- Department of Molecular Imaging, School of Medical Technology, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, People's Republic of China.
| |
Collapse
|
35
|
Huang W, Zhang P, Zhao E, Xiong J, Wang S, Lv Z. Potential application and prospects of ROS-sensitive biomaterials in cancer therapy: a immune microenvironment triggered nanomaterial. Discov Oncol 2025; 16:185. [PMID: 39954216 PMCID: PMC11829892 DOI: 10.1007/s12672-025-01900-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
Reactive Oxygen Species (ROS) is the collective term used for the extremely reactive molecules that are important mediators in physiological processes as well as the development of various disease conditions. Normal cells maintain a delicate equilibrium, known as redox homeostasis, between antioxidants and ROS levels. Any imbalance in the redox homeostasis of the body results in oxidative stress which can result in inflammation, necrosis, apoptosis, cell death, and eventually a disease state. Enhanced ROS levels are a key feature in cancer cells that is being explored for developing reactive oxygen species-sensitive biomaterials. The distinct variation in redox potential between normal cells and tumour cells is one of the major physiological differences between them, that has enabled the development of ROS-sensitive nanomaterials for cancer therapy. ROS-sensitive nanomaterials are sensitive to the physiological variations in the cells, like high levels of hydrogen peroxide and glutathione in the cancer cells. ROS-responsive nanomaterials have the unique property of modulating microenvironmental redox conditions in cancer cells. ROS-sensitive material can work either by scavenging the ROS or by simulating the cellular antioxidants, leading to cancer cell cytotoxicity. These ROS-sensitive nanomaterials can simulate the human body's natural antioxidants like, superoxide dismutase and peroxidase. Thus, ROS-sensitive nanomaterials hold promise as a potential platform for the treatment of cancer. The present review will cover the importance of ROS in cancer, the different types of ROS-sensitive nanomaterials available and their therapeutic application in cancer therapy.
Collapse
Affiliation(s)
- Weiming Huang
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Pengju Zhang
- Department of General Surgery, Jinling Hospital Affiliated Hospital of Medical School, Nanjing University, Nanjing, 200002, Jiangsu, China
| | - Eryong Zhao
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Jian Xiong
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| | - Shaokun Wang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| | - Zi Lv
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
36
|
Jeong SH, Chun SM, Lee H, Kim M, Choi M, Leigh JH. Association between chemotherapy and the risk of developing breast cancer-related lymphedema: a nationwide retrospective cohort study. Support Care Cancer 2025; 33:143. [PMID: 39899036 PMCID: PMC11790788 DOI: 10.1007/s00520-025-09169-3] [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: 07/08/2024] [Accepted: 01/11/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Breast cancer-related lymphedema (BCRL) is a well-known complication of breast cancer treatment, which often includes chemotherapy. This study aimed to investigate the association between chemotherapy and the risk of developing BCRL in patients with new-onset breast cancer. METHODS This nationwide retrospective cohort study utilized data from the Korean National Health Insurance database and the Korea National Cancer Incidence Database (2006-2017). Using 1:1 propensity score matching, 37,202 participants who received chemotherapy and 37,202 who did not receive chemotherapy were included in the analysis. Cox proportional hazard regression models were employed to examine the association between chemotherapy and the risk of developing BCRL. RESULTS Among the 74,404 participants, 11,508 (15.5%) were diagnosed with BCRL during the follow-up period. Compared with patients who did not receive chemotherapy, the risk of BCRL was higher in patients undergoing chemotherapy (hazard ratio [95% confidence interval]: 1.95 [1.87-2.04]). Furthermore, compared to patients who did not receive chemotherapy, the risk of BCRL was confirmed in the taxane (3.38 [3.19-3.58]), antimetabolite (1.79 [1.67-1.91]), and anthracycline (1.49 [1.41-1.56]) chemotherapy groups. CONCLUSION Chemotherapy administration following a diagnosis of breast cancer increases the risk of BCRL. Therefore, vigilant monitoring for BCRL, particularly in patients undergoing chemotherapy with taxanes, antimetabolites, or anthracyclines, is warranted during follow-up.
Collapse
Affiliation(s)
- Sung Hoon Jeong
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, 03080, Seoul, Republic of Korea
- National Traffic Injury Rehabilitation Research Institute, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Gyeonggi-Do, Republic of Korea
- Institute of Health Services Research, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, 03722, Seoul, Republic of Korea
| | - Seong Min Chun
- Department of Physical Medicine and Rehabilitation, Soonchunhyang University Hospital Seoul, Soonchunhyang University College of Medicine, 59 Daesagwan-Ro, 04401, Seoul, Republic of Korea
| | - Hyunji Lee
- National Traffic Injury Rehabilitation Research Institute, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Gyeonggi-Do, Republic of Korea
- Department of Rehabilitation Medicine, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Gyeonggi-Do, Republic of Korea
| | - Miji Kim
- National Traffic Injury Rehabilitation Research Institute, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Gyeonggi-Do, Republic of Korea
- Department of Biostatistics and Computing, Yonsei University Graduate School, 50 Yonsei-Ro, Seodaemun-Gu, 03722, Seoul, Republic of Korea
| | - Mira Choi
- Department of Dermatology, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, 10380, Gyeonggi-Do, Republic of Korea
| | - Ja-Ho Leigh
- Department of Rehabilitation Medicine, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, 03080, Seoul, Republic of Korea.
- National Traffic Injury Rehabilitation Research Institute, National Traffic Injury Rehabilitation Hospital, Yangpyeong, 12564, Gyeonggi-Do, Republic of Korea.
- Institute of Health Policy and Management, Medical Research Center, Seoul National University, Seoul, Korea.
| |
Collapse
|
37
|
Wei D, Zhang F, Li M, Fan Z, Ma J, Ji J, Qiao S, Huang P, Zhang W, Fan K, Li L, Zheng W, Li X, Ren L. CircDUSP16 mediates the effect of triple-negative breast cancer in pirarubicin via the miR-1224-3p/TFDP2 axis. Biochem Pharmacol 2025; 232:116719. [PMID: 39710273 DOI: 10.1016/j.bcp.2024.116719] [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: 08/20/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive molecular subtype of breast cancer characterized by a high recurrence rate, poor prognosis, and elevated mortality. Identifying novel molecular targets is crucial for developing more effective therapeutic strategies against TNBC. Recent studies have highlighted the role of circular RNAs (circRNAs) in the progression of TNBC. In this study, we identified and validated that circDUSP16 (hsa_circ_0003855) is significantly upregulated in TNBC cells, tissues, and plasma exosomes. Functional assays in vitro demonstrated that overexpression of circDUSP16 promoted the proliferation, migration and invasion of TNBC cells, weathers circDUSP16 knockdown exerted the opposite effect. In vivo studies confirmed that circDUSP16 knockdown can inhibit tumor growth. Using bioinformatics analysis, circDUSP16/miR-1224-3p/TFDP2 pathway was predicted and cascaded. Mechanistically, circDUSP16 was shown to promote the progression of TNBC via the miR-1224-3p/TFDP2 axis. Additionally, THP, a commonly used anthracycline chemotherapy drug, was found to downregulate circDUSP16, suggesting that its therapeutic effects on TNBC may be mediated through circDUSP16/miR-1224-3p/TFDP2 pathway. Our findings suggest that circDUSP16 is a promising biomarker and potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Dexian Wei
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Fan Zhang
- Qingdao Municipal Hospital Qingdao, Shandong 266000, China
| | - Min Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Zhimin Fan
- General Surgery Center, Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jiulong Ma
- School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, Shandong 266000, China
| | - Jiahua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Sennan Qiao
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Peng Huang
- School of Agroforestry and Medicine, The Open University of China, Beijing 100000, China
| | - Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Kaiqi Fan
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Lu Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Wentao Zheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China.
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 130021, China.
| |
Collapse
|
38
|
Wang Y, Tang Y, Guo L, Yang X, Wu S, Yue Y, Xu C. Recent advances in zeolitic imidazolate frameworks as drug delivery systems for cancer therapy. Asian J Pharm Sci 2025; 20:101017. [PMID: 39931355 PMCID: PMC11808527 DOI: 10.1016/j.ajps.2025.101017] [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: 04/28/2023] [Revised: 04/16/2024] [Accepted: 11/24/2024] [Indexed: 02/13/2025] Open
Abstract
Biological nanotechnologies based on functional nanoplatforms have synergistically catalyzed the emergence of cancer therapies. As a subtype of metal-organic frameworks (MOFs), zeolitic imidazolate frameworks (ZIFs) have exploded in popularity in the field of biomaterials as excellent protective materials with the advantages of conformational flexibility, thermal and chemical stability, and functional controllability. With these superior properties, the applications of ZIF-based materials in combination with various therapies for cancer treatment have grown rapidly in recent years, showing remarkable achievements and great potential. This review elucidates the recent advancements in the use of ZIFs as drug delivery agents for cancer therapy. The structures, synthesis methods, properties, and various modifiers of ZIFs used in oncotherapy are presented. Recent advances in the application of ZIF-based nanoparticles as single or combination tumor treatments are reviewed. Furthermore, the future prospects, potential limitations, and challenges of the application of ZIF-based nanomaterials in cancer treatment are discussed. We except to fully explore the potential of ZIF-based materials to present a clear outline for their application as an effective cancer treatment to help them achieve early clinical application.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xi Yang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Shanli Wu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| |
Collapse
|
39
|
Guo S, Zhang Y, Wang Y, Guo T, Zhu J, Chang L, Ling W, Westover KD, Zhou Z, Wei X. DCR2-targeted ultrasound nanobubbles loaded with verteporfin promote M2 macrophage polarization to overcome doxorubicin resistance in breast cancer. CHEMICAL ENGINEERING JOURNAL 2025; 505:159277. [DOI: 10.1016/j.cej.2025.159277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
|
40
|
Fabiani I, Chianca M, Cipolla CM, Cardinale DM. Anthracycline-induced cardiomyopathy: risk prediction, prevention and treatment. Nat Rev Cardiol 2025:10.1038/s41569-025-01126-1. [PMID: 39875555 DOI: 10.1038/s41569-025-01126-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Anthracyclines are the cornerstone of treatment for many malignancies. However, anthracycline cardiotoxicity is a considerable concern given that it can compromise the clinical effectiveness of the treatment and patient survival despite early discontinuation of therapy or dose reduction. Patients with cancer receiving anthracycline treatment can have a reduction in their quality of life and likelihood of survival due to cardiotoxicity, irrespective of their oncological prognosis. Increasing knowledge about anthracycline cardiotoxicity has enabled the identification of patients who are candidates for anthracycline regimens and those who might develop anthracycline-induced cardiomyopathy. Anthracycline cardiotoxicity is a unique and evolving phenomenon that begins with myocardial cell damage, progresses to reduced left ventricular ejection fraction, and culminates in symptomatic heart failure if it is not promptly detected and treated. Early risk stratification can be guided by imaging or biomarkers. In this Review, we present a comprehensive and clinically useful approach to cardiomyopathy related to anthracycline therapy, encompassing its epidemiology, definition, mechanisms, novel classifications, risk factors and patient risk stratification, diagnostic approaches (including imaging and biomarkers), treatment guidelines algorithms, and the role of new cardioprotective drugs that are used for the treatment of heart failure.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
| | - Michela Chianca
- Division of Cardiology, Cardiothoracic and Vascular Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
41
|
Huang Y, Chen X, Chen M, Lin Y, Chen B, Gao H, Chen M. Drug-induced heart failure: a real-world pharmacovigilance study using the FDA adverse event reporting system database. Front Pharmacol 2025; 15:1523136. [PMID: 39881876 PMCID: PMC11775474 DOI: 10.3389/fphar.2024.1523136] [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: 11/05/2024] [Accepted: 12/24/2024] [Indexed: 01/31/2025] Open
Abstract
Objective Although there are certain drug categories associated with heart failure (HF), most of the associated risks are unclear. We investigated the top drugs associated with HF and acute HF (AHF) reported in the FDA Adverse Event Reporting System (FAERS). Methods We reviewed publicly available FAERS databases from 2004 to 2023. Using the search terms "cardiac failure" or "cardiac failure acute" and classifying cases by drug name, we processed and analyzed drug reports related to HF or AHF. Results From 2004 to 2023, 17,379,609 adverse drug events were reported by FAERS, of which 240,050 (1.38%) were reported as HF. Among those with HF, the male-to-female ratio was 0.94% and 52.37% were >65 years old; 46.2% were from the United States. There were 5,971 patients with AHF. We identified 38 drugs and 13 drug classes with a potential high risk of causing HF, and 41 drugs and 19 drug classes were associated with AHF. The median onset times of HF and AHF were 83 days (IQR: 11-416) and 49 days (IQR: 8-259), respectively. The Weibull shape parameter (WSP) test showed early failure-type profile characteristics. Conclusion This study highlights key drugs associated with drug-induced HF and AHF, emphasizing the importance of early risk assessment and close monitoring, particularly during the initial stages of treatment. These findings contribute to a better understanding of drug-induced HF and provide a basis for future research on its underlying mechanisms.
Collapse
Affiliation(s)
- Youqi Huang
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiaowen Chen
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Mingyu Chen
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Yuze Lin
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bingqi Chen
- Department of pharmacy, Xiamen Medical College, Xiamen, China
| | - Hongjin Gao
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Min Chen
- Shengli Clinical College of Fujian Medical University, Department of Pharmacy, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| |
Collapse
|
42
|
Yi HZ, Liang SM, Li JJ, Liu H, Liao JX, Liu DY, Zhang QJ, Cai MZ, Sun JS. Collective total synthesis of chartreusin derivatives and bioactivity investigations. Chem Sci 2025; 16:1241-1249. [PMID: 39677934 PMCID: PMC11635980 DOI: 10.1039/d4sc05629a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024] Open
Abstract
Capitalizing on Hauser annulation and Yu glycosylation, the chemical synthesis of chartreusin-type aromatic polycyclic polyketide glycosides has been investigated, culminating in the successful establishment of chemical approaches toward chartreusin derivatives with intricate chemical structures but promising bioactivities. Based on the chemical synthesis strategy, the first and collective chemical syntheses of chartreusin, D329C, and elsamicins A and B have been accomplished. The chemical strategy was featured by two complementary routes to secure chartarin 10-O-monosaccharide glycosides, the key intermediates in chartreusin derivative synthesis, as well as the highly stereoselective construction of the difficult glycosidic linkages. Through the synthetic investigations, viable donors and acceptors of 3-C-methyl-branched sugars were determined for the first time. Moreover, facilitated by the established chemical synthetic strategy, the cytotoxic activities of chartreusin derivatives against human cancer cell lines were assessed and profound antineoplastic effects for chartreusin and elsamicins A and B were recorded. Based on RNA-seq analysis, the underlying working mechanisms against ES-2 cells were investigated, and the appended sugar chain-determined function mechanisms were disclosed.
Collapse
Affiliation(s)
- Hong-Zhou Yi
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
- School of Life Science and Health Engineering, Jiangnan University 1800 Lihu Avenue Wuxi 214122 China
| | - Shu-Min Liang
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
| | - Jing-Jing Li
- Affiliated Hospital of Shandong Secondary Medicinal University 4948 Shengli East Street Weifang 261042 China
| | - Hui Liu
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
| | - Jin-Xi Liao
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
| | - De-Yong Liu
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
| | - Qing-Ju Zhang
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
| | - Ming-Zhong Cai
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
| | - Jian-Song Sun
- National Research Center for Carbohydrate Synthesis, Jiangxi Normal University 99 Ziyang Avenue Nanchang 330022 China
- School of Life Science and Health Engineering, Jiangnan University 1800 Lihu Avenue Wuxi 214122 China
| |
Collapse
|
43
|
Feng W, Wang Q, Tan Y, Qiao J, Liu Q, Yang B, Yang S, Cui L. Early detection of anthracycline-induced cardiotoxicity. Clin Chim Acta 2025; 565:120000. [PMID: 39401650 DOI: 10.1016/j.cca.2024.120000] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Although anthracyclines are important anticancer agents, their use is limited due to various adverse effects, particularly cardiac toxicity. Mechanisms underlying anthracycline-induced cardiotoxicity (AIC) are complex. Given the irreplaceable role of anthracyclines in treatment of malignancies and other serious diseases, early monitoring of AIC is paramount. In recent years, multiple studies have investigated various biomarkers for early detection of AIC. Currently, the two most common are cardiac troponin and B-type natriuretic peptide. In addition, a range of other molecules, including RNAs, myeloperoxidase (MPO), C-reactive protein (CRP), various genes, and others, also play roles in AIC prediction. Unfortunately, current research indicates a need to validate their sensitivity and specificity of these biomarkers especially in large study populations. In this review, we summarize the mechanisms and potential biomarkers of AIC, although some remain preliminary.
Collapse
Affiliation(s)
- Weimin Feng
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qingchen Wang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Yuan Tan
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Jiao Qiao
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Qi Liu
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Boxin Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, No. 49 Huayuan North Road, 100191, China; Institute of Medical Technology, Peking University Health Science Center, No. 38 Xueyuan Road, Beijing, 100191, China.
| |
Collapse
|
44
|
Gabizon AA, Gabizon-Peretz S, Modaresahmadi S, La-Beck NM. Thirty years from FDA approval of pegylated liposomal doxorubicin (Doxil/Caelyx): an updated analysis and future perspective. BMJ ONCOLOGY 2025; 4:e000573. [PMID: 39885941 PMCID: PMC11751825 DOI: 10.1136/bmjonc-2024-000573] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025]
Abstract
In 2025, it will be 30 years since the initial clinical approval of pegylated liposomal doxorubicin (PLD) by the Food and Drug Administration. PLD predated the field of nanomedicine and became a model nanomedicine setting key pharmacological principles (prolonged circulation, slow drug release and the enhanced permeability and retention (EPR) effect) for clinical application of other nano-drugs in cancer therapy. The impressive reduction of cardiotoxicity conferred by PLD is the most valuable clinical asset. While PLD has gained a strong foothold in relapsed ovarian cancer and metastatic breast cancer, it has not been extensively tested in primary (neoadjuvant) and adjuvant therapy and has not fulfilled the expectations from the results in animal models efficacy-wise. This discrepancy may be due to the large dose gap between mice and humans and the apparent variability of the EPR effect in human cancer. PLD is a complex product and we are still in a learning curve regarding a number of factors such as its interaction with the complement system and its immune modulatory properties, as well as its integration in multimodality therapy that may potentiate its value and role in cancer therapy.
Collapse
Affiliation(s)
- Alberto A Gabizon
- The Leah and Jakub Susskind Nano-Oncology Research Laboratory, Helmsley Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
- Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel
| | - Shira Gabizon-Peretz
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Central, Israel
- Tel Aviv University, Faculty of Medicine, Tel Aviv, Israel
| | - Shadan Modaresahmadi
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H Hodge School of Pharmacy, Abilene, Texas, USA
| | - Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H Hodge School of Pharmacy, Abilene, Texas, USA
| |
Collapse
|
45
|
Singuru MMR, Tabrizi MA, Bhattacharyya P, Ali AA, You M. Force-Responsive Delivery of Anticancer Drugs via a DNA Mechanical Nanovehicle. NANO LETTERS 2025; 25:336-342. [PMID: 39719379 PMCID: PMC12046514 DOI: 10.1021/acs.nanolett.4c05076] [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] [Indexed: 12/26/2024]
Abstract
Cellular mechanical dysregulation can lead to diseases and conditions like tumorigenesis. Drug delivery systems that recognize and respond to specific cellular mechanical characteristics are potentially useful for targeted therapy. We report here the creation of a DNA mechanical nanovehicle that is responsive to cell surface receptor-mediated tensile forces, which can then correspondingly deliver an anticancer drug in situ. These DNA mechanical nanovehicles can spontaneously anchor onto cell membranes and enable the real-time visualization of molecular tensions at intercellular junctions. Once a strong intercellular force was detected, a rapid drug release event was followed automatically. Force-triggered targeted cancer cell treatment was demonstrated both in vitro and in a cell mixture. Our results proved a novel cellular force-responsive platform that can be used for highly specific drug delivery, which may potentially lead to smart cancer therapy with enhanced efficacy and safety.
Collapse
Affiliation(s)
| | | | | | - Ahsan Ausaf Ali
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Mingxu You
- Department of Chemistry, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
46
|
Sahu SK, Vyas M, Prabhakar PK. Emerging Role of Natural Topoisomerase Inhibitors as Anticancer agents. Med Chem 2025; 21:195-210. [PMID: 40070141 DOI: 10.2174/0115734064311729240911102646] [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: 02/20/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 05/13/2025]
Abstract
Topoisomerases I and II are the functionally two forms of DNA topoisomerase. In anticancer research, novel anticancer chemotherapeutical capable of blocking topoisomerase enzymes have been discovered. Most commonly, topoisomerase causes replication fork arrest and doublestrand breaks, and this is how a clinically successful topoisomerase-targeting anticancer medicines work. Unfortunately, this novel mechanism of action has been linked to the development of secondary malignancies as well as cardiotoxicity. The specific binding locations and mechanisms of topoisomerase poisons have been identified by studying the structures of topoisomerase-drug-DNA ternary complexes. Recent breakthroughs in science have revealed that isoform-specific human topoisomerase II poison could be created as safer anticancer drug molecules. It may also be able to develop catalytic inhibitors of topoisomerases by focusing on their inactive conformations. In addition to this, the discovery of new bacterial topoisomerase inhibitor molecules and regulatory proteins could lead to the discovery of new human topoisomerase inhibitors. As a result, biologists, organic chemists, and medicinal chemists worldwide have been identifying, designing, synthesizing, and testing a variety of novel topoisomerase-targeting bioactive compounds. This review focused on topoisomerase inhibitors, their mechanisms of action, and different types of topoisomerase inhibitors that have been developed during the last ten years.
Collapse
Affiliation(s)
- Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Manish Vyas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Pranav Kumar Prabhakar
- Research and Development Cell, Parul University, P.O. Limda, Dist. Vadodara, Ta.Waghodia, 391760 Gujarat, India
| |
Collapse
|
47
|
Tang Y, Zhu J, Flesch RCC, Jin T. Effect of proposed asynchronous injection strategy on the combination therapy of magnetic hyperthermia and thermosensitive liposome. J Therm Biol 2025; 127:104040. [PMID: 39765148 DOI: 10.1016/j.jtherbio.2024.104040] [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: 09/14/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 02/24/2025]
Abstract
Magnetic nanoparticles (MNPs) used for magnetic hyperthermia can not only damage tumor cells after elevating to a specific temperature but also provide the temperature required for thermosensitive liposomes (TSL) to release doxorubicin (DOX). MNPs injected into tumor will generate heat under an alternating magnetic field, so the MNPs distribution can determine temperature distribution and further affect the DOX concentration used for tumor therapy. This study proposes an asynchronous injection strategy for this combination therapy in order to improve the DOX concentration value for drug therapy, in which the MNPs are injected into tumor after a certain lagging of TSL injection in order to increase the TSL concentration inside tumor. In addition, the evaluation of treatment effect for this combination therapy is implemented by considering two different MNPs concentration distributions and two biological heat transfer models. The simulation results demonstrate that the treatment effect for combination therapy can be significantly improved after considering the proposed asynchronous injection strategy, which can mainly attribute to the improvement of DOX concentration. The DOX concentration difference during therapy is generally relevant to both the lagging time of different injections and the local temperature distribution due to MNPs concentration distribution.
Collapse
Affiliation(s)
- Yundong Tang
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, 350116, China.
| | - Jiajia Zhu
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, 350116, China
| | - Rodolfo C C Flesch
- Departamento de Automação e Sistemas, Universidade Federal de Santa Catarina, 88040-900, Florianópolis, SC, Brazil
| | - Tao Jin
- College of Electrical Engineering and Automation, Fuzhou University, Fuzhou, 350116, China
| |
Collapse
|
48
|
Karamanolis NN, Kounatidis D, Vallianou NG, Dimitriou K, Tsaroucha E, Tsioulos G, Anastasiou IA, Mavrothalassitis E, Karampela I, Dalamaga M. Unraveling the Anti-Cancer Mechanisms of Antibiotics: Current Insights, Controversies, and Future Perspectives. Antibiotics (Basel) 2024; 14:9. [PMID: 39858295 PMCID: PMC11762948 DOI: 10.3390/antibiotics14010009] [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: 12/01/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Cancer persists as a significant global health challenge, claiming millions of lives annually despite remarkable strides in therapeutic innovation. Challenges such as drug resistance, toxicity, and suboptimal efficacy underscore the need for novel treatment paradigms. In this context, the repurposing of antibiotics as anti-cancer agents has emerged as an attractive prospect for investigation. Diverse classes of antibiotics have exhibited promising anti-cancer properties in both in vitro and in vivo studies. These mechanisms include the induction of apoptosis and cell cycle arrest, generation of reactive oxygen species, and inhibition of key regulators of cell proliferation and migration. Additional effects involve the disruption of angiogenesis and modulation of pivotal processes such as inflammation, immune response, mitochondrial dynamics, ferroptosis, and autophagy. Furthermore, antibiotics have demonstrated the potential to enhance the efficacy of conventional modalities like chemotherapy and radiotherapy, while alleviating treatment-induced toxicities. Nevertheless, the integration of antibiotics into oncological applications remains contentious, with concerns centered on their disruption of gut microbiota, interference with immunotherapeutic strategies, contribution to microbial resistance, and potential association with tumorigenesis. This narrative review explores the mechanisms of antibiotics' anti-cancer activity, addresses controversies about their dual role in cancer biology, and envisions future perspectives that include the development of novel derivatives and innovative frameworks for their incorporation into cancer treatment paradigms.
Collapse
Affiliation(s)
- Nikolaos Nektarios Karamanolis
- Second Department of Internal Medicine, Hippokratio General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.N.K.); (K.D.)
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (I.A.A.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Krystalia Dimitriou
- Second Department of Internal Medicine, Hippokratio General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.N.K.); (K.D.)
| | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Georgios Tsioulos
- Fourth Department of Internal Medicine, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Ioanna A. Anastasiou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (I.A.A.)
| | - Evangelos Mavrothalassitis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (N.G.V.); (E.T.); (E.M.)
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12461 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
49
|
Maud L, Barakat N, Bornot J, Snini SP, Mathieu F. Biocontrol of Mycotoxigenic Fungi by Actinobacteria. J Fungi (Basel) 2024; 11:4. [PMID: 39852424 PMCID: PMC11766079 DOI: 10.3390/jof11010004] [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: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 01/26/2025] Open
Abstract
Actinobacteria are well known for their production of metabolites of interest. They have been previously studied to identify new antibiotics in medical research and for their ability to stimulate plant growth in agronomic research. Actinobacteria represents a real source of potential biocontrol agents (BCAs) today. With the aim of reducing the use of phytosanitary products by 50% with the different Ecophyto plans, a possible application is the fight against mycotoxin-producing fungi in food matrices and crops using BCAs. To deal with this problem, the use of actinobacteria, notably belonging to the Streptomyces genus, or their specialized metabolites seems to be a solution. In this review, we focused on the impact of actinobacteria or their metabolites on the development of mycotoxigenic fungi and mycotoxin production on the one hand, and on the other hand on their ability to detoxify food matrices contaminated by mycotoxins.
Collapse
Affiliation(s)
| | | | | | - Selma P. Snini
- Laboratoire de Génie Chimique, Université de Toulouse, CNRS, INPT, UPS, 31326 Toulouse, France; (L.M.); (N.B.); (J.B.)
| | - Florence Mathieu
- Laboratoire de Génie Chimique, Université de Toulouse, CNRS, INPT, UPS, 31326 Toulouse, France; (L.M.); (N.B.); (J.B.)
| |
Collapse
|
50
|
Vishnubalaji R, Alajez NM. Disrupted Lipid Metabolism, Cytokine Signaling, and Dormancy: Hallmarks of Doxorubicin-Resistant Triple-Negative Breast Cancer Models. Cancers (Basel) 2024; 16:4273. [PMID: 39766172 PMCID: PMC11674486 DOI: 10.3390/cancers16244273] [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: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chemoresistance in triple-negative breast cancer (TNBC) presents a significant clinical hurdle, limiting the efficacy of treatments like doxorubicin. This study aimed to explore the molecular changes associated with doxorubicin resistance and identify potential therapeutic targets to overcome this resistance, thereby improving treatment outcomes for TNBC patients. METHODS Doxorubicin-resistant (DoxR) TNBC models (MDA-MB-231 and BT-549) were generated by exposing cells to increasing concentrations of doxorubicin. RNA sequencing (RNA-Seq) was performed using the Illumina platform, followed by bioinformatics analysis with CLC Genomics Workbench and iDEP. Functional assays assessed proliferation, sphere formation, migration, and cell cycle changes. Protein expression and phosphorylation were confirmed via Western blotting. Pathway and network analyses were conducted using Ingenuity Pathway Analysis (IPA) and STRING, while survival analysis was performed using Kaplan-Meier Plotter database. RESULTS DoxR cells exhibited reduced proliferation, sphere formation, and migration, but showed enhanced tolerance to doxorubicin. Increased CHK2 and p53 phosphorylation indicated cellular dormancy as a resistance mechanism. RNA-Seq analysis revealed upregulation of cytokine signaling and stress-response pathways, while cholesterol and lipid biosynthesis were suppressed. Activation of the IL1β cytokine network was prominent in DoxR cells, and CRISPR-Cas9 screens data identified dependencies on genes involved in rRNA biogenesis and metabolism. A 27-gene signature associated with doxorubicin resistance was linked to worse clinical outcomes in a large breast cancer cohort (HR = 1.76, FDR p < 2.0 × 10-13). CONCLUSIONS This study uncovers potential therapeutic strategies for overcoming TNBC resistance, including dormancy reversal and targeting onco-ribosomal pathways and cytokine signaling networks, to improve the efficacy of doxorubicin-based treatments.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Oncology Research Center (TORC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
| | - Nehad M. Alajez
- Translational Oncology Research Center (TORC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar;
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| |
Collapse
|