1
|
Giri A, Hong IS, Kwon TK, Kang JS, Jeong JH, Kweon S, Yook S. Exploring therapeutic and diagnostic potential of cysteine cathepsin as targets for cancer therapy with nanomedicine. Int J Biol Macromol 2025; 315:144324. [PMID: 40398760 DOI: 10.1016/j.ijbiomac.2025.144324] [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: 03/30/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Cysteine cathepsins have been discovered to be substantially expressed in multiple types of cancer. They play a key role in the progression and growth of these cancers, rendering them appealing targets for nanoscale delivery and noninvasive diagnostic imaging. This review explores cathepsins from the papain-like enzyme family (C1) within the cysteine peptidase clan (CA), emphasizing the role of cathepsin-responsive nanoparticles in tumor growth. Furthermore, it also explores how nanotechnology can harness cathepsin activity to enable targeted drug delivery, improve tumor imaging, and reduce systemic toxicity. By examining the molecular mechanisms governing cathepsin function and evaluating different nanocarrier systems, this work aims to enhance our understanding of targeted cancer treatment. Despite significant advances, challenges remain in translating these nanomedicine platforms into clinical use, including improving delivery efficiency, biocompatibility, long-term safety, and addressing issues such as interspecies protease variability and scalable nanomanufacturing. Future advancement, integrating advanced biomaterials, patient-derived organoid models, bispecific immune-protease targeting, CRISPR-based cathepsin editing, and artificial intelligence-driven pharmacokinetic modeling and analysis will be critical to fully realizing the clinical potential of cathepsin targeted nanomedicines. These innovations hold promises for advancing precision oncology by overcoming current limitations and improving patient outcomes.
Collapse
Affiliation(s)
- Anil Giri
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea; Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seho Kweon
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Simmyung Yook
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
2
|
Nan A, Dumitrascu V, Flangea C, Dumitrescu G, Puscasiu D, Vlad T, Popescu R, Vlad C. From Chemical Composition to Antiproliferative Effects Through In Vitro Studies: Honey, an Ancient and Modern Hot Topic Remedy. Nutrients 2025; 17:1595. [PMID: 40362904 PMCID: PMC12074252 DOI: 10.3390/nu17091595] [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: 04/14/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Honey is a natural product which has been used throughout time as a food, spice, and medicine. Its therapeutic use has its origins in direct empirical observations of various beneficial actions in terms of its anti-infectious, anti-inflammatory, and wound-healing effects, to which an antiproliferative effect is added. In the context of malignant transformation, reductions in chronic inflammation, antioxidant action, cell cycle arrest, and apoptosis activation contribute to this antiproliferative effect, achievements attributed mainly to the polyphenols in its composition. A multitude of in vitro studies performed on malignant cell cultures try to elucidate the real mechanism(s) that can scientifically explain this action. In addition, its use as an adjuvant in association with cytostatic therapy demonstrates a promising effect in enhancing its cytotoxic effect, but also in reducing some adverse effects. Highlighting these actions allows for further perspectives to be opened regarding the use of honey for therapeutic and also prophylactic purposes, as a food supplement. Future studies will support the identification of real antiproliferative effects in patients with malignant tumors in terms of actions on the human body as a whole, moving from cell cultures to complex implications.
Collapse
Affiliation(s)
- Alexandru Nan
- Doctoral School “Engineering of Vegetable and Animal Resources”, University of Life Sciences “King Mihai I” from Timişoara, Calea Aradului 119, 300645 Timisoara, Romania;
| | - Victor Dumitrascu
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (V.D.); (C.V.)
| | - Corina Flangea
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (V.D.); (C.V.)
| | - Gabi Dumitrescu
- Faculty of Bioengineering of Animal Resources, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
| | - Daniela Puscasiu
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
- Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Tania Vlad
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
- Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Roxana Popescu
- ANAPATMOL Research Center, “Victor Babes” University of Medicine and Pharmacy, E. Murgu 2, 300041 Timisoara, Romania; (D.P.); (T.V.); (R.P.)
- Department of Cell and Molecular Biology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Cristian Vlad
- Department of Biochemistry and Pharmacology, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timisoara, Romania; (V.D.); (C.V.)
| |
Collapse
|
3
|
Sun Z, Fu H, Zhang R, Wang H, Shen S, Zhao C, Li X, Sun Y, Li Y, Li Y. Advances in chemically modified HSA as a multifunctional carrier for transforming cancer therapy regimens. Int J Biol Macromol 2025; 305:141373. [PMID: 39988174 DOI: 10.1016/j.ijbiomac.2025.141373] [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: 12/11/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Human serum albumin (HSA) is a versatile, biodegradable, biocompatible, non-toxic, and non-immunogenic protein nanocarrier, making it an ideal platform for developing advanced drug delivery systems. These properties have garnered significant attention in utilizing HSA nanoparticles for the safe and efficient delivery of chemotherapeutic agents. HSA-based nanoparticles can be surface-modified with various ligands to enable tumor-targeted drug delivery, enhancing therapeutic specificity and efficacy. Furthermore, the multifunctionality of HSA nanoparticles offers promising strategies to overcome challenges in cancer therapy, including poor bioavailability, off-target toxicity, and drug resistance. This review highlights the structural features of HSA, explores its diverse modifications to improve drug-binding affinity and targeting ability, and discusses its potential as a multifunctional carrier in oncology. By summarizing the latest advances in HSA modification techniques and applications, this review provides a comprehensive perspective on the future of protein-based drug delivery systems in tumor therapy.
Collapse
Affiliation(s)
- Zheng Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hui Fu
- School of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruixuan Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hui Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shiyang Shen
- School of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chengcheng Zhao
- Experimental Teaching and Practical Training Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiuyan Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yujiao Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yunfei Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yingpeng Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
4
|
Alaridhee ZAI, Alqaraguly MB, Formanova S, Kuryazov R, Mahdi MS, Taher WM, Alwan M, Jabir MS, Zankanah FH, Majdi H, Jawad MJ, Hamad AK, Bozorov K. Recent advances in microfluidic-based photoelectrochemical (PEC) sensing platforms for biomedical applications. Mikrochim Acta 2025; 192:297. [PMID: 40229472 DOI: 10.1007/s00604-025-07135-8] [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: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025]
Abstract
Photoelectrochemical (PEC) techniques seamlessly combine electrochemical and spectroscopic principles, offering a powerful platform for the detection of biomarkers and biological molecules in clinical and biomedical settings. This review provides a comprehensive overview of microfluidic PEC probes, emphasizing their potential for ultrasensitive detection through enhanced light absorption and charge transfer processes. Key advantages of microfluidic PEC include real-time monitoring of biological processes, non-invasive detection, and the possibility of multiplexing when integrated with various quantification modalities. However, the practical implementation of PEC faces challenges such as bulky setup, matrix interference, and stability of PEC-active materials. Also, this paper discusses the intricate mechanisms of PEC sensing, highlighting the roles of nanomaterials in enhancing microfluidic PEC systems. Additionally, the limitations inherent in PEC material selection, including stability and bandgap engineering, are critically discussed. Solutions such as doping and the development of composite materials are proposed to address these issues. Through presented examples of PEC applications in biomedical fields, this review elucidates the future potential of PEC-based methods as reliable and effective tools for diagnostic applications. Additionally, this review proposes the most effective probes for future investigations to develop commercial devices.
Collapse
Affiliation(s)
| | | | - Shoira Formanova
- Department of Chemistry and Its Teaching Methods, Tashkent State Pedagogical University, Tashkent, Uzbekistan.
| | | | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Faeza H Zankanah
- College of Health & Medical Technology, Uruk University, Baghdad, Iraq
| | - Hasan Majdi
- Department of Chemical Engineering and Petroleum Industries, Al-Mustaqbal University College, Babylon, 51001, Iraq
| | | | | | - Khurshed Bozorov
- Department of Organic Synthesis and Bioorganic Chemistry, Institute of Biochemistry, Samarkand State University, University Blvd. 15, 140104, Samarkand, Uzbekistan.
| |
Collapse
|
5
|
Zhang T, Ouyang Z, Zhang Y, Sun H, Kong L, Xu Q, Qu J, Sun Y. Marine Natural Products in Inflammation-Related Diseases: Opportunities and Challenges. Med Res Rev 2025. [PMID: 40202793 DOI: 10.1002/med.22109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
In recent decades, the potentiality of marine natural products (MNPs) in the medical field has been increasingly recognized. Natural compounds derived from marine microorganisms, algae, and invertebrates have shown significant promise for treating inflammation-related diseases. In this review, we cover the three primary sources of MNPs and their diverse and unique chemical structures and bioactivities. This review aims to summarize the progress of MNPs in combating inflammation-related diseases. Moreover, we cover the functions and mechanisms of MNPs in diseases, highlighting their functions in regulating inflammatory signaling pathways, cellular stress responses, and gut microbiota, among others. Meanwhile, we focus on key technologies and scientific methods to address the current limitations and challenges in MNPs.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Yueran Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
6
|
Lai Y, Xie B, Zhang W, He W. Pure drug nanomedicines - where we are? Chin J Nat Med 2025; 23:385-409. [PMID: 40274343 DOI: 10.1016/s1875-5364(25)60851-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 04/26/2025]
Abstract
Pure drug nanomedicines (PDNs) encompass active pharmaceutical ingredients (APIs), including macromolecules, biological compounds, and functional components. They overcome research barriers and conversion thresholds associated with nanocarriers, offering advantages such as high drug loading capacity, synergistic treatment effects, and environmentally friendly production methods. This review provides a comprehensive overview of the latest advancements in PDNs, focusing on their essential components, design theories, and manufacturing techniques. The physicochemical properties and in vivo behaviors of PDNs are thoroughly analyzed to gain an in-depth understanding of their systematic characteristics. The review introduces currently approved PDN products and further explores the opportunities and challenges in expanding their depth and breadth of application. Drug nanocrystals, drug-drug cocrystals (DDCs), antibody-drug conjugates (ADCs), and nanobodies represent the successful commercialization and widespread utilization of PDNs across various disease domains. Self-assembled pure drug nanoparticles (SAPDNPs), a next-generation product, still require extensive translational research. Challenges persist in transitioning from laboratory-scale production to mass manufacturing and overcoming the conversion threshold from laboratory findings to clinical applications.
Collapse
Affiliation(s)
- Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wanting Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
7
|
Ma J, Zhu R, Li M, Jiao H, Fan S, Ma X, Xiang G. Proteolysis-targeting chimera-doxorubicin conjugate nanoassemblies for dual treatment of EGFR-TKI sensitive and resistant non-small cell lung cancer. Acta Biomater 2025; 195:421-435. [PMID: 39922514 DOI: 10.1016/j.actbio.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/12/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Proteolysis-targeting chimeras (PROTACs) have emerged as a promising strategy for targeted protein degradation and drug discovery. However, traditional PROTACs face inherent limitations and may also contribute to induce drug resistance. These challenges have driven the development of innovative strategies to overcome these obstacles. In current study, a PROTAC-DOX conjugates (PDCs) nanoassembly strategy was introduced to enhance tumor-targeting capability and overcome the drawbacks of conventional PROTACs. The designed PDC-S nanoparticles (PDC-S NPs) demonstrated potent anti-tumor activity against drug-resistant strains (IC50 = 4.7 µM) and improved in vivo efficacy (TGI = 76 %) against drug-sensitive strains, while minimizing side effects. Additionally, PDC-S NPs have great potential in tumor immunotherapy. This study provides a novel and promising strategy for the development of PROTAC-Drug Conjugates (PDCs). STATEMENT OF SIGNIFICANCE: We developed a PROTAC-DOX conjugates (PDCs) nanoassembly strategy to address the limitations of traditional PROTACs, such as poor solubility, low targeting specificity, and drug resistance. PDC-S NPs were constructed via self-assembly, which simplified preparation and minimized the toxicity typically associated with carrier-assisted delivery systems. The PDC-S NPs showed improved aqueous solubility and cellular uptake, resulting in efficient EGFR degradation in HCC827 cells. In vivo, PDC-S NPs accumulated at tumor sites via the EPR effect, resulting in enhanced anti-tumor potency with reduced side effects. Furthermore, PDC-S NPs induced immunogenic cell death (ICD) and suppressed PD-L1 and VEGF expression, highlighting great potential in tumor immunotherapy.
Collapse
Affiliation(s)
- Junhui Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Ruixue Zhu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Meijing Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui Jiao
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Sijun Fan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xiang Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Pharmacy, Tongren Polytechnic College, Tongren, Guizhou 554300, China.
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; School of Pharmacy, Tongren Polytechnic College, Tongren, Guizhou 554300, China; The Higher Education Edible and Medicinal Fungi Engineering Research Center of Guizhou Province, Tongren, Guizhou 554300, China.
| |
Collapse
|
8
|
Zhang Y, Zhang N, Gong SP, Chen ZS, Cao HL. Nanozyme-based synergistic therapeutic strategies against tumors. Drug Discov Today 2025; 30:104292. [PMID: 39805540 DOI: 10.1016/j.drudis.2025.104292] [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: 11/27/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Cancer remains a global health threat, with traditional treatments limited by adverse effects and drug resistance. Nanozyme-based catalytic therapy with high stability and controllable activity provides targeted and specific in situ tumor treatment to address these challenges. More intriguingly, the tremendous advances in nanotechnology have enabled nanozymes to rival the catalytic activity of natural enzymes, presenting an exciting opportunity for innovating antitumor nanodrugs. This review systematically summarizes the latest progresses in nanozyme-based anticancer catalytic therapy, with a particular focus on various synergistic antitumor strategies, including other functional enzymes, drugs, exogenous stimuli and radiotherapy. These combinations not only enhance the efficacy of cancer treatment and reduce systemic toxicity but also offer insights into the development of potent antitumor nanodrugs.
Collapse
Affiliation(s)
- Ye Zhang
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi'an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi'an Medical University, Xi'an, China
| | - Ning Zhang
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi'an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi'an Medical University, Xi'an, China
| | - Shou-Ping Gong
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi'an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi'an Medical University, Xi'an, China.
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St John's University NY USA.
| | - Hui-Ling Cao
- Xi'an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi'an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
9
|
Lu C, Wei J, Gao C, Sun M, Dong D, Mu Z. Molecular signaling pathways in doxorubicin-induced nephrotoxicity and potential therapeutic agents. Int Immunopharmacol 2025; 144:113373. [PMID: 39566381 DOI: 10.1016/j.intimp.2024.113373] [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: 08/11/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 11/22/2024]
Abstract
Doxorubicin (DOX), an anthracycline chemotherapeutic agent, is extensively utilized in the clinical management of both solid and hematological malignancies. Nevertheless, the clinical application of this treatment is significantly limited by adverse reactions and toxicity that may arise during or after administration. Its cytotoxic effects are multifaceted, with cardiotoxicity being the most prevalent side effect. Furthermore, it has the potential to adversely affect other organs, including the brain, kidneys, liver, and so on. Notably, it has been reported that DOX may cause renal failure in patients and there is currently no effective treatment for DOX-induced kidney damage, which has raised a high concern about DOX-induced nephrotoxicity (DIN). Although the precise molecular mechanisms underlying DIN remain incompletely elucidated, prior research has indicated that reactive oxygen species (ROS) are pivotal in this process, triggering a cascade of detrimental pathways including apoptosis, inflammation, dysregulated autophagic flux, and fibrosis. In light of these mechanisms, decades of research have uncovered several DIN-associated signaling pathways and found multiple potential therapeutic agents targeting them. Thus, this review intends to delineate the DIN associated signaling pathways, including AMPK, JAKs/STATs, TRPC6/RhoA/ROCK1, YAP/TEAD, SIRTs, Wnt/β-catenin, TGF-β/Smad, MAPK, Nrf2/ARE, NF-κB, and PI3K/AKT, and to summarize their potential regulatory agents, which provide a reference for the development of novel medicines against DIN.
Collapse
Affiliation(s)
- Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China; Department of Urology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China.
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China.
| | - Zhongyi Mu
- Department of Urology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
10
|
Kodel HDAC, Alizadeh P, Ebrahimi SN, Machado TOX, Oliveira MBPP, Fathi F, Souto EB. Liposomes and Niosomes: New trends and applications in the delivery of bioactive agents for cancer therapy. Int J Pharm 2025; 668:124994. [PMID: 39586512 DOI: 10.1016/j.ijpharm.2024.124994] [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: 08/21/2024] [Revised: 11/09/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Lipid-based nanocarriers have been in continuous development as strategies to enhance drug delivery efficiency. Liposomes are delivery systems primarily composed of phospholipids and cholesterol (or other suitable stabilizers) that have transformed the pharmaceutical field by improving drug targeting and release control. The success of this technology is strongly attributed to phospholipids, which are components of cell membranes, forming a biocompatible system. Nevertheless, drawbacks related to their production cost and stability under certain conditions led to the development of niosomes by replacing phospholipids with non-ionic surfactants. Both liposomes and niosomes have been widely studied and optimized for the delivery of bioactive agents targeting many diseases, including cancer. They can improve the efficacy of cancer therapy by reducing toxicity and off-target effects. Due to the complexity of this disease, many approaches should be considered, and the composition and physical properties of liposomes and niosomes influence the outcomes. In this review, we discuss the role of liposomes and niosomes in delivering bioactives for cancer therapy, emphasizing their specific characteristics, associated challenges, and the latest advancements aimed at enhancing their effectiveness.
Collapse
Affiliation(s)
- Helena de A C Kodel
- Rede Nordeste de Biotecnologia-RENORBIO, University of Tiradentes, Farolândia, 49010-390, Aracaju, Sergipe, Brazil
| | - Paria Alizadeh
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Samad N Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Tatiane O X Machado
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy of University of Porto, Jorge de Viterbo Ferreira, 4050-313, Porto, Portugal; Department of Agroindustry, Federal Institute of Sertão Pernambucano, Campus Petrolina Zona Rural, PE 647, Km 22, PISNC N4, 56302-970, Petrolina, Pernambuco, Brazil
| | - M Beatriz P P Oliveira
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Faezeh Fathi
- REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Eliana B Souto
- UCD School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland.
| |
Collapse
|
11
|
Ge W, Zhang X, Lin J, Wang Y, Zhang X, Duan Y, Dai X, Zhang J, Zhang Y, Jiang M, Qiang H, Zhao Z, Zhang X, Sun D. Rnd3 protects against doxorubicin-induced cardiotoxicity through inhibition of PANoptosis in a Rock1/Drp1/mitochondrial fission-dependent manner. Cell Death Dis 2025; 16:2. [PMID: 39755713 DOI: 10.1038/s41419-024-07322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Doxorubicin, a representative drug of the anthracycline class, is widely used in cancer treatment. However, Doxorubicin-induced cardiotoxicity (DIC) presents a significant challenge in its clinical application. Mitochondrial dysfunction plays a central role in DIC, primarily through disrupting mitochondrial dynamics. This study aimed to investigate the impact of Rnd3 (a Rho family GTPase 3) on DIC, with a focus on mitochondrial dynamics. Cardiomyocyte-specific Rnd3 transgenic mice (Rnd3-Tg) and Rnd3LSP/LSP mice (N-Tg) were established for in vivo experiments, and adenoviruses harboring Rnd3 (Ad-Rnd3) or negative control (Ad-Control) were injected in the myocardium for in vitro experiments. The DIC model was established using wild-type, N-Tg, and Rnd3-Tg mice, with subsequent intraperitoneal injection of Dox for 4 weeks. The molecular mechanism was explored through RNA sequencing, immunofluorescence staining, co-immunoprecipitation assay, and protein-protein docking. Dox administration induced significant mitochondrial injury and cardiac dysfunction, which was ameliorated by Rnd3 overexpression. Further, the augmentation of Rnd3 expression mitigated mitochondrial fragmentation which is mediated by dynamin-related protein 1 (Drp1), thereby ameliorating the PANoptosis (pyroptosis, apoptosis, and necroptosis) response induced by Dox. Mechanically, the interaction between Rnd3 and Rho-associated kinase 1 (Rock1) may impede Rock1-induced Drp1 phosphorylation at Ser616, thus inhibiting mitochondrial fission and dysfunction. Interestingly, Rock1 knockdown nullified the effects of Rnd3 on cardiomyocytes PANoptosis, as well as Dox-induced cardiac remodeling and dysfunction elicited by Rnd3. Rnd3 enhances cardiac resilience against DIC by stabilizing mitochondrial dynamics and reducing PANoptosis. Our findings suggest that the Rnd3/Rock1/Drp1 signaling pathway represents a novel target for mitigating DIC, and modulating Rnd3 expression could be a strategic approach to safeguarding cardiac function in patients undergoing Dox treatment. The graphical abstract illustrated the cardioprotective role of Rnd3 in DIC. Rnd3 directly binds to Rock1 in cytoplasm and ameliorates mitochondrial fission by inhibiting Drp1 phosphorylation at ser616, thereby alleviating PANoptosis (apoptosis, pyroptosis, and necroptosis) in DIC.
Collapse
Affiliation(s)
- Wen Ge
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yangyang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiao Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu Duan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhijing Zhao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
12
|
Zhang D, Zhao J, Zhang Y, Jiang H, Liu D. Revisiting immune checkpoint inhibitors: new strategies to enhance efficacy and reduce toxicity. Front Immunol 2024; 15:1490129. [PMID: 39720720 PMCID: PMC11666542 DOI: 10.3389/fimmu.2024.1490129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Affiliation(s)
- Dianying Zhang
- Medical Education Department, Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai, China
| | - Jingjing Zhao
- Sleep Medicine Center, Huai’an No.3 People’s Hospital, Huai’an, China
- Huaian Second Clinical College of Xuzhou Medical University, Huaian, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Dan Liu
- Medical Education Department, Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai, China
| |
Collapse
|
13
|
Qi X, Cheng C, Zhang D, Yu Z, Meng X. Exploring the synergy between tumor microenvironment modulation and STING agonists in cancer immunotherapy. Front Immunol 2024; 15:1488345. [PMID: 39712021 PMCID: PMC11659200 DOI: 10.3389/fimmu.2024.1488345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Affiliation(s)
- Xiaoyan Qi
- Zibo Central Hospital, Zibo, China
- Department of Oncology, Zibo Central Hospital, Zibo, China
| | - Cheng Cheng
- Zibo Central Hospital, Zibo, China
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Dawei Zhang
- Zibo Central Hospital, Zibo, China
- Department of Orthopedics, Zibo Central Hospital, Zibo, China
| | - Zongjiang Yu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Xiangwei Meng
- Zibo Central Hospital, Zibo, China
- Department of Drug Clinical Trials, Zibo Central Hospital, Zibo, China
| |
Collapse
|
14
|
Xu W, Jia A, Lei Z, Wang J, Jiang H, Wang S, Wang Q. Stimuli-responsive prodrugs with self-immolative linker for improved cancer therapy. Eur J Med Chem 2024; 279:116928. [PMID: 39362023 DOI: 10.1016/j.ejmech.2024.116928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
Self-immolative prodrugs have gained significant attention as an innovative approach for targeted cancer therapy. These prodrugs are engineered to release the active anticancer agents in response to specific triggers within the tumor microenvironment, thereby improving therapeutic precision while reducing systemic toxicity. This review focuses on the molecular architecture and design principles of self-immolative prodrugs, emphasizing the role of stimuli-responsive linkers and activation mechanisms that enable controlled drug release. Recent advancements in this field include the development of prodrugs that incorporate targeting moieties for enhanced site-specificity. Moreover, the review discusses the incorporation of targeting moieties to achieve site-specific drug delivery, thereby improving the selectivity of treatment. By summarizing key research from the past five years, this review highlights the potential of self-immolative prodrugs to revolutionize cancer treatment strategies and pave the way for their integration into clinical practice.
Collapse
Affiliation(s)
- Wenting Xu
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Zhixian Lei
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China
| | - Jianing Wang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Hongfei Jiang
- School of Pharmacy, Qingdao University, Qingdao, 266071, China.
| | - Shuai Wang
- Department of Radiotherapy, School of Medical Imaging, Affiliated Hospital of Shandong Second Medical University, Shandong Second Medical University, Weifang, Shandong, China.
| | - Qi Wang
- Department of Pediatric Intensive Care Medicine, Hainan Women and Children's Medical Center, Haikou, China.
| |
Collapse
|
15
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [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: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
16
|
Zheng S, Gu Y, Gu Y, Zhao Y, Li L, Wang M, Jiang R, Yu X, Chen T, Li J. Machine learning-enabled virtual screening indicates the anti-tuberculosis activity of aldoxorubicin and quarfloxin with verification by molecular docking, molecular dynamics simulations, and biological evaluations. Brief Bioinform 2024; 26:bbae696. [PMID: 39737570 DOI: 10.1093/bib/bbae696] [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: 07/16/2024] [Revised: 10/16/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025] Open
Abstract
Drug resistance in Mycobacterium tuberculosis (Mtb) is a significant challenge in the control and treatment of tuberculosis, making efforts to combat the spread of this global health burden more difficult. To accelerate anti-tuberculosis drug discovery, repurposing clinically approved or investigational drugs for the treatment of tuberculosis by computational methods has become an attractive strategy. In this study, we developed a virtual screening workflow that combines multiple machine learning and deep learning models, and 11 576 compounds extracted from the DrugBank database were screened against Mtb. Our screening method produced satisfactory predictions on three data-splitting settings, with the top predicted bioactive compounds all known antibacterial or anti-TB drugs. To further identify and evaluate drugs with repurposing potential in TB therapy, 15 screened potential compounds were selected for subsequent computational and experimental evaluations, out of which aldoxorubicin and quarfloxin showed potent inhibition of Mtb strain H37Rv, with minimal inhibitory concentrations of 4.16 and 20.67 μM/mL, respectively. More inspiringly, these two compounds also showed antibacterial activity against multidrug-resistant TB isolates and exhibited strong antimicrobial activity against Mtb. Furthermore, molecular docking, molecular dynamics simulation, and the surface plasmon resonance experiments validated the direct binding of the two compounds to Mtb DNA gyrase. In summary, our effective comprehensive virtual screening workflow successfully repurposed two novel drugs (aldoxorubicin and quarfloxin) as promising anti-Mtb candidates. The verification results provide useful information for the further development and clinical verification of anti-TB drugs.
Collapse
Affiliation(s)
- Si Zheng
- Institute for Artificial Intelligence & Department of Computer Science and Technology, Tsinghua University, Haidian District, Beijing 100084, China
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing 100020, China
| | - Yaowen Gu
- Department of Chemistry, New York University, New York, NY 10027, United States
| | - Yuzhen Gu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Tongzhou District, Beijing 101149, China
| | - Yelin Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing 100050, China
| | - Liang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing 100050, China
| | - Min Wang
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing 100020, China
| | - Rui Jiang
- Department of Automation, Tsinghua University, Haidian District, Beijing 100084, China
| | - Xia Yu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Tongzhou District, Beijing 101149, China
| | - Ting Chen
- Institute for Artificial Intelligence & Department of Computer Science and Technology, Tsinghua University, Haidian District, Beijing 100084, China
| | - Jiao Li
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing 100020, China
| |
Collapse
|
17
|
Hao X, Ding M, Chi C, Xu X, Zhang X, Hu M. The potential of kaempferol in digestive system tumors: recent advances and mechanistic insights. Discov Oncol 2024; 15:658. [PMID: 39546109 PMCID: PMC11568081 DOI: 10.1007/s12672-024-01510-2] [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: 08/24/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Digestive system neoplasms are a heterogeneous group of cancers characterized by diverse symptoms, complex diagnosis, and treatment. Prognosis is poor and influenced by multiple factors, making early detection and comprehensive treatment crucial for patient survival. Kaempferol, a flavonoid compound, has attracted attention due to its anti-tumor biological activity, holding promise as a potential drug for treating digestive system neoplasms. Derived from various plants such as cabbage, propolis, and grapefruit, this compound's anti-inflammatory, antioxidant, and other pharmacological effects have been confirmed. Research has found that kaempferol inhibits the occurrence and development of digestive system neoplasms by inducing apoptosis in cancer cells, inhibiting tumor cell proliferation, suppressing tumor metastasis and invasion, and enhancing the effects of other cancer treatment methods. This paper summarizes the role and mechanisms of kaempferol in the study of digestive system neoplasms, providing valuable insights for both scientists and clinical physicians engaged in this field. By detailing the various pathways through which kaempferol exerts its anticancer effects, the paper not only highlights its potential as a therapeutic agent but also opens avenues for further research into its applications.
Collapse
Affiliation(s)
- Xunxing Hao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province, China
| | - Meng Ding
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province, China
| | - Chenyu Chi
- The Emergency and Critical Care Medicine Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaodong Xu
- Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Xiaoyu Zhang
- Department of Acupuncture and Massage College, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Mingzhe Hu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
18
|
Qi Z, Gu J, Qu L, Shi X, He Z, Sun J, Tan L, Sun M. Advancements of engineered live oncolytic biotherapeutics (microbe/virus/cells): Preclinical research and clinical progress. J Control Release 2024; 375:209-235. [PMID: 39244159 DOI: 10.1016/j.jconrel.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The proven efficacy of immunotherapy in fighting tumors has been firmly established, heralding a new era in harnessing both the innate and adaptive immune systems for cancer treatment. Despite its promise, challenges such as inefficient delivery, insufficient tumor penetration, and considerable potential toxicity of immunomodulatory agents have impeded the advancement of immunotherapies. Recent endeavors in the realm of tumor prophylaxis and management have highlighted the use of living biological entities, including bacteria, oncolytic viruses, and immune cells, as a vanguard for an innovative class of live biotherapeutic products (LBPs). These LBPs are gaining recognition for their inherent ability to target tumors. However, these LBPs must contend with significant barriers, including robust immune clearance mechanisms, cytotoxicity and other in vivo adverse effects. Priority must be placed on enhancing their safety and therapeutic indices. This review consolidates the latest preclinical research and clinical progress pertaining to the exploitation of engineered biologics, spanning bacteria, oncolytic viruses, immune cells, and summarizes their integration with combination therapies aimed at circumventing current clinical impasses. Additionally, the prospective utilities and inherent challenges of the biotherapeutics are deliberated, with the objective of accelerating their clinical application in the foreseeable future.
Collapse
Affiliation(s)
- Zhengzhuo Qi
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Junmou Gu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihang Qu
- The 4th People's Hospital of Shenyang, China Medical University, Shenyang, Liaoning, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Lingchen Tan
- School of Life Sciences and Biopharmaceutical, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| |
Collapse
|
19
|
Nazli A, Irshad Khan MZ, Rácz Á, Béni S. Acid-sensitive prodrugs; a promising approach for site-specific and targeted drug release. Eur J Med Chem 2024; 276:116699. [PMID: 39089000 DOI: 10.1016/j.ejmech.2024.116699] [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: 02/26/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/03/2024]
Abstract
Drugs administered through conventional formulations are devoid of targeting and often spread to various undesired sites, leading to sub-lethal concentrations at the site of action and the emergence of undesired effects. Hence, therapeutic agents should be delivered in a controlled manner at target sites. Currently, stimuli-based drug delivery systems have demonstrated a remarkable potential for the site-specific delivery of therapeutic moieties. pH is one of the widely exploited stimuli for drug delivery as several pathogenic conditions such as tumor cells, infectious and inflammatory sites are characterized by a low pH environment. This review article aims to demonstrate various strategies employed in the design of acid-sensitive prodrugs, providing an overview of commercially available acid-sensitive prodrugs. Furthermore, we have compiled the progress made for the development of new acid-sensitive prodrugs currently undergoing clinical trials. These prodrugs include albumin-binding prodrugs (Aldoxorubicin and DK049), polymeric micelle (NC-6300), polymer conjugates (ProLindac™), and an immunoconjugate (IMMU-110). The article encompasses a broad spectrum of studies focused on the development of acid-sensitive prodrugs for anticancer, antibacterial, and anti-inflammatory agents. Finally, the challenges associated with the acid-sensitive prodrug strategy are discussed, along with future directions.
Collapse
Affiliation(s)
- Adila Nazli
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | | | - Ákos Rácz
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | - Szabolcs Béni
- Integrative Health and Environmental Analysis Research Laboratory, Department of Analytical Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary.
| |
Collapse
|
20
|
Wang B, Yu W, Jiang H, Meng X, Tang D, Liu D. Clinical applications of STING agonists in cancer immunotherapy: current progress and future prospects. Front Immunol 2024; 15:1485546. [PMID: 39421752 PMCID: PMC11483357 DOI: 10.3389/fimmu.2024.1485546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The STING (Stimulator of Interferon Genes) pathway is pivotal in activating innate immunity, making it a promising target for cancer immunotherapy. STING agonists have shown potential in enhancing immune responses, particularly in tumors resistant to traditional therapies. This scholarly review examines the diverse categories of STING agonists, encompassing CDN analogues, non-CDN chemotypes, CDN-infused exosomes, engineered bacterial vectors, and hybrid structures of small molecules-nucleic acids. We highlight their mechanisms, clinical trial progress, and therapeutic outcomes. While these agents offer significant promise, challenges such as toxicity, tumor heterogeneity, and delivery methods remain obstacles to their broader clinical use. Ongoing research and innovation are essential to overcoming these hurdles. STING agonists could play a transformative role in cancer treatment, particularly for patients with hard-to-treat malignancies, by harnessing the body's immune system to target and eliminate cancer cells.
Collapse
Affiliation(s)
- Bin Wang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wanpeng Yu
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hongfei Jiang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Medical Education Department, Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai, China
| | - Xiangwei Meng
- Department of Drug Clinical Trials, Zibo Central Hospital, Zibo, China
| | - Dongmei Tang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Department of Anesthesia, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Liu
- Medical Education Department, Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai, China
| |
Collapse
|
21
|
Wang B, Tang D, Cui J, Jiang H, Yu J, Guo Z. RGD-based self-assembling nanodrugs for improved tumor therapy. Front Pharmacol 2024; 15:1477409. [PMID: 39411070 PMCID: PMC11473307 DOI: 10.3389/fphar.2024.1477409] [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: 08/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
RGD-based self-assembling nanodrugs are a promising advancement in targeted cancer therapy, combining the specificity of RGD peptides with the benefits of nanotechnology. These nanodrugs enhance tumor targeting and cellular uptake while reducing off-target effects. RGD peptides facilitate the self-assembly of stable nanostructures, ensuring efficient drug delivery. Despite their potential, challenges such as immunogenicity, stability, tumor heterogeneity, and manufacturing scalability need to be addressed. Future research should focus on improving biocompatibility, advanced targeting strategies, personalized medicine approaches, and innovative manufacturing techniques. Overcoming these challenges will pave the way for the successful clinical translation of RGD-based nanodrugs, offering more effective and safer cancer treatments.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Dongmei Tang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jianqiao Cui
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hongfei Jiang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, Municipal Hospital, Qingdao, China
| | - Zhu Guo
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Chen S, Yu B, DU GT, Huang TY, Zhang N, Fu N. KIF18B: an important role in signaling pathways and a potential resistant target in tumor development. Discov Oncol 2024; 15:430. [PMID: 39259333 PMCID: PMC11390998 DOI: 10.1007/s12672-024-01330-4] [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: 05/22/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024] Open
Abstract
KIF18B is a key member of the kinesin-8 family, involved in regulating various physiological processes such as microtubule length, spindle assembly, and chromosome alignment. This article briefly introduces the structure and physiological functions of KIF18B, examines its role in malignant tumors, and the associated carcinogenic signaling pathways such as PI3K/AKT, Wnt/β-catenin, and mTOR pathways. Research indicates that the upregulation of KIF18B enhances tumor malignancy and resistance to radiotherapy and chemotherapy. KIF18B could become a new target for anticancer drugs, offering significant potential for the treatment of malignant tumors and reducing chemotherapy resistance.
Collapse
Affiliation(s)
- Shicheng Chen
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P. R. China
| | - Bo Yu
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P. R. China
| | - Guo Tu DU
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P. R. China
| | - Tian Yu Huang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P. R. China
| | - Neng Zhang
- Department of Urology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P. R. China.
| | - Ni Fu
- Department of Urology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, P. R. China.
| |
Collapse
|
23
|
Dönmez S, Lapinskaite R, Atalay HN, Tokay E, Kockar F, Rycek L, Özbil M, Tumer TB. Selagibenzophenone B and Its Derivatives: SelB-1, a Dual Topoisomerase I/II Inhibitor Identified through In Vitro and In Silico Analyses. ACS BIO & MED CHEM AU 2024; 4:178-189. [PMID: 39184056 PMCID: PMC11342340 DOI: 10.1021/acsbiomedchemau.4c00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 08/27/2024]
Abstract
The development of multitargeted drugs represents an innovative approach to cancer treatment, aiming to enhance drug effectiveness while minimizing side effects. Herein, we sought to elucidate the inhibitory effect of selagibenzophenone B derivatives on the survival of cancer cells and dual topoisomerase I/II enzyme activity. Results demonstrated that among the compounds, SelB-1 selectively inhibited the proliferation and migration of prostate cancer cells while exhibiting minimal effects on healthy cells. Furthermore, SelB-1 showed a dual inhibitory effect on topoisomerases. Computational analyses mirrored the results from enzyme inhibition assays, demonstrating the compound's strong binding affinity to the catalytic sites of the topoisomerases. To our surprise, SelB-1 did not induce apoptosis in prostate cancer cells; instead, it induced autophagic gene expression and lipid peroxidation while reducing GSH levels, which might be associated with ferroptotic death mechanisms. To summarize, the findings suggest that SelB-1 possesses the potential to serve as a dual topoisomerase inhibitor and can be further developed as a promising candidate for prostate cancer treatment.
Collapse
Affiliation(s)
- Serhat Dönmez
- Graduate
Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Ringaile Lapinskaite
- Department
of Organic Chemistry, Center for Physical
Sciences and Technology (FTMC), Akademijos g. 7, Vilnius LT-08412, Lithuania
- Department
of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 43 Praha 2, Czechia
| | - Hazal Nazlican Atalay
- Graduate
Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Esra Tokay
- Department
of Molecular Biology and Genetics, Faculty of Sciences and Arts, Balikesir University, Balikesir 10145, Turkey
| | - Feray Kockar
- Department
of Molecular Biology and Genetics, Faculty of Sciences and Arts, Balikesir University, Balikesir 10145, Turkey
| | - Lukas Rycek
- Department
of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 43 Praha 2, Czechia
| | - Mehmet Özbil
- Institute
of Biotechnology, Gebze Technical University, Kocaeli 41400, Turkey
| | - Tugba Boyunegmez Tumer
- Department
of Molecular Biology and Genetics, Faculty of Science, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| |
Collapse
|
24
|
Jin Z, Xiao X, Gui L, Lu Q, Zhang J. Determination of doxorubicin in plasma and tissues of mice by UPLC-MS/MS and its application to pharmacokinetic study. Heliyon 2024; 10:e35123. [PMID: 39157405 PMCID: PMC11328074 DOI: 10.1016/j.heliyon.2024.e35123] [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: 03/11/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
A rapid and sensitive ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was established for the simultaneous determination of doxorubicin (DOX) in mouse plasma and tissues, including the heart, liver, spleen, lung, kidney and tumor, and to investigate the pharmacokinetics and distribution in mice. In this study, daunorubicin (DNR) was used as an internal standard, and the mobile phase consisted of ammonium formate 2 mM containing 0.1 % formic acid (A) and acetonitrile (B), the chromatographic column was ACQUITY UPLC BEHTM C18 with a gradient elution at a flow rate of 0.2 mL/min. Electrospray ionization (ESI) in positive ion pattern was utilized for the ion separation of DOX, with the ions used for quantitative analysis being DOX m/z 544.28 → 397.10 and DNR m/z 528.35 → 321.08, respectively. The results showed that a good linear relationship in the calibration curve range of 1-800 ng/mL in mouse plasma and 1-2500 ng/g in tissues (R2 > 0.99) with the limits of quantification of 1 ng/mL in plasma and tissues. The method exhibited good matrix effect and extraction recovery, with the intra-day and inter-day precision of plasma and tissue were less than 10.3 % and 15.4 %, and the relative error (RE) were both less than ±14.8 % and ±18.9 %, respectively. The stability results under different conditions were found to be accurate. It also revealed the distribution of DOX in various tissues of mice, with the concentration ranking as liver > heart > kidney > spleen > lung > tumor. This method was successfully used to the study for the pharmacokinetics in plasma and drug distribution in tissues of BALB/c mice.
Collapse
Affiliation(s)
- Zhilin Jin
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Xue Xiao
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Lili Gui
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Qiao Lu
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| | - Jicai Zhang
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, 442000, China
| |
Collapse
|
25
|
Pudełek M, Ryszawy D, Piwowarczyk K, Lasota S, Madeja Z, Kędracka-Krok S, Czyż J. Metabolic reprogramming of poly(morpho)nuclear giant cells determines glioblastoma recovery from doxorubicin-induced stress. J Transl Med 2024; 22:757. [PMID: 39135106 PMCID: PMC11318163 DOI: 10.1186/s12967-024-05541-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/26/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Multi-drug resistance of poly(morpho)nuclear giant cells (PGCs) determines their cytoprotective and generative potential in cancer ecosystems. However, mechanisms underlying the involvement of PGCs in glioblastoma multiforme (GBM) adaptation to chemotherapeutic regimes remain largely obscure. In particular, metabolic reprogramming of PGCs has not yet been considered in terms of GBM recovery from doxorubicin (DOX)-induced stress. METHODS Long-term proteomic and metabolic cell profiling was applied to trace the phenotypic dynamics of GBM populations subjected to pulse DOX treatment in vitro, with a particular focus on PGC formation and its metabolic background. The links between metabolic reprogramming, drug resistance and drug retention capacity of PGCs were assessed, along with their significance for GBM recovery from DOX-induced stress. RESULTS Pulse DOX treatment triggered the transient formation of PGCs, followed by the appearance of small expanding cell (SEC) clusters. Development of PGCs was accompanied by the mobilization of their metabolic proteome, transient induction of oxidative phosphorylation (OXPHOS), and differential intracellular accumulation of NADH, NADPH, and ATP. The metabolic background of PGC formation was confirmed by the attenuation of GBM recovery from DOX-induced stress following the chemical inhibition of GSK-3β, OXPHOS, and the pentose phosphate pathway. Concurrently, the mobilization of reactive oxygen species (ROS) scavenging systems and fine-tuning of NADPH-dependent ROS production systems in PGCs was observed. These processes were accompanied by perinuclear mobilization of ABCB1 and ABCG2 transporters and DOX retention in the perinuclear PGC compartments. CONCLUSIONS These data demonstrate the cooperative pattern of GBM recovery from DOX-induced stress and the crucial role of metabolic reprogramming of PGCs in this process. Metabolic reprogramming enhances the efficiency of self-defense systems and increases the DOX retention capacity of PGCs, potentially reducing DOX bioavailability in the proximity of SECs. Consequently, the modulation of PGC metabolism is highlighted as a potential target for intervention in glioblastoma treatment.
Collapse
Affiliation(s)
- Maciej Pudełek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Damian Ryszawy
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Katarzyna Piwowarczyk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Sławomir Lasota
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland
| | - Sylwia Kędracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jarosław Czyż
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, 30-387, Poland.
| |
Collapse
|
26
|
Tu Y, Gong J, Mou J, Jiang H, Zhao H, Gao J. Strategies for the development of stimuli-responsive small molecule prodrugs for cancer treatment. Front Pharmacol 2024; 15:1434137. [PMID: 39144632 PMCID: PMC11322083 DOI: 10.3389/fphar.2024.1434137] [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: 05/17/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Approved anticancer drugs typically face challenges due to their narrow therapeutic window, primarily because of high systemic toxicity and limited selectivity for tumors. Prodrugs are initially inactive drug molecules designed to undergo specific chemical modifications. These modifications render the drugs inactive until they encounter specific conditions or biomarkers in vivo, at which point they are converted into active drug molecules. This thoughtful design significantly improves the efficacy of anticancer drug delivery by enhancing tumor specificity and minimizing off-target effects. Recent advancements in prodrug design have focused on integrating these strategies with delivery systems like liposomes, micelles, and polymerosomes to further improve targeting and reduce side effects. This review outlines strategies for designing stimuli-responsive small molecule prodrugs focused on cancer treatment, emphasizing their chemical structures and the mechanisms controlling drug release. By providing a comprehensive overview, we aim to highlight the potential of these innovative approaches to revolutionize cancer therapy.
Collapse
Affiliation(s)
- Yuxuan Tu
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jianbao Gong
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, China
| | - Jing Mou
- Department of Neonatology, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
| | - Hongfei Jiang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Haibo Zhao
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiake Gao
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Zhang T, Yin K, Niu X, Bai X, Wang Z, Ji M, Yuan B. Development of Bivalent Aptamer-DNA Carrier-Doxorubicin Conjugates for Targeted Killing of Esophageal Squamous Cell Carcinoma Cells. Int J Mol Sci 2024; 25:7959. [PMID: 39063201 PMCID: PMC11276760 DOI: 10.3390/ijms25147959] [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: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Esophageal cancer ranks the seventh in cancer incidence and the sixth in cancer death. Esophageal squamous cell carcinoma (ESCC) accounts for approximately 90% of the total cases of esophageal cancer. Chemotherapy is the most effective drug-based method for treatment of esophageal cancer. However, severe side effects of traditional chemotherapy limit its treatment efficacy. Targeted chemotherapy can deliver chemotherapeutic drugs to cancer cells and specifically kill these cells with reduced side effects. In the work, the bivalent aptamer-DNA carrier (BAD) was designed by using an ESCC cell-specific aptamer as the recognition molecule and a GC base-rich DNA sequence as the drug carrier. With doxorubicin (Dox) as chemotherapeutic drugs, the bivalent aptamer-DNA-Dox conjugate (BADD) was constructed for targeted killing of ESCC cells. Firstly, the truncated A2(35) aptamer with a retained binding ability was obtained through optimization of an intact A2(80) aptamer and was used to fuse with DNA carrier sequences for constructing the BAD through simple DNA hybridization. The results of gel electrophoresis and flow cytometry analysis showed that the BAD was successfully constructed and had a stronger binding affinity than monovalent A2(35). Then, the BAD was loaded with Dox drugs to construct the BADD through noncovalent intercalation. The results of fluorescence spectra and flow cytometry assays showed that the BADD was successfully constructed and can bind to target cells strongly. Confocal imaging further displayed that the BADD can be specifically internalized into target cells and release Dox. The results of CCK-8 assays, Calcein AM/PI staining, and wound healing assays demonstrated that the BADD can specifically kill target cells, but not control cells. Our results demonstrate that the developed BADD can specifically deliver doxorubicin to target ESCC cells and selectively kill these cells, offering a potentially effective strategy for targeted chemotherapy of ESCC.
Collapse
Affiliation(s)
- Tianlu Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Kai Yin
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Xidong Niu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Xue Bai
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Zhaoting Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Mengmeng Ji
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Baoyin Yuan
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
- Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
28
|
Shang B, Yu Z, Wang Z. Recent advances and applications of nitroreductase activable agents for tumor theranostic. Front Pharmacol 2024; 15:1451517. [PMID: 39101150 PMCID: PMC11294179 DOI: 10.3389/fphar.2024.1451517] [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: 06/19/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Nitroreductase activable agents offer a personalized and targeted approach to cancer theranostics by selectively activating prodrugs within the tumor microenvironment. These agents enable non-invasive tumor imaging, image-guided drug delivery, and real-time treatment monitoring. By leveraging the enzymatic action of tumor-specific nitroreductase enzymes, cytotoxic drugs are delivered directly to cancer cells while minimizing systemic toxicity. This review highlights the key features, mechanisms of action, diagnostic applications, therapeutic potentials, and future directions of nitroreductase activable agents for tumor theranostics. Integration with imaging modalities, advanced drug delivery systems, immunotherapy combinations, and theranostic biomarkers shows promise for optimizing treatment outcomes and improving patient survival in oncology. Continued research and innovation in this field are crucial for advancing novel theranostic strategies and enhancing patient care. Nitroreductase activable agents represent a promising avenue for personalized cancer therapy and have the potential to transform cancer diagnosis and treatment approaches.
Collapse
Affiliation(s)
- Baoxin Shang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Zongjiang Yu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Zhengdan Wang
- The Afffliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Kong JC, Zhou F, Shi L, Wei Y, Wu C. A novel nanodrug for the sensitization of photothermal chemotherapy for breast cancer in vitro. RSC Adv 2024; 14:21292-21299. [PMID: 38974230 PMCID: PMC11225340 DOI: 10.1039/d4ra01611d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024] Open
Abstract
Owing to the complexity of tumor treatment, clinical tumor treatment has evolved from a single treatment mode to multiple combined treatment modes. Reducing the tolerance of tumors to heat and the toxicity of chemotherapy drugs to the body, as well as increasing the sensitivity of tumors to photothermal therapy and chemotherapy drugs, are key issues that urgently need to be addressed in the current cancer treatment. In this work, polylactic acid-based drug nanoparticles (PLA@DOX/GA/ICG) were synthesized with good photothermal conversion ability by encapsulating the water-soluble anticancer drug doxorubicin (DOX), photothermal conversion agent indocyanine green (ICG) and liposoluble drug gambogic acid (GA) using a double emulsion method. The preparation process of PLA@DOX/GA/ICG was examined. Gambogic acid entrapped in PLA@DOX/GA/ICG nanoparticles could act as an HSP90 protein inhibitor to achieve bidirectional sensitization to chemotherapy and photothermal therapy under 808 nm laser irradiation for the first time, effectively ablating breast cancer cells in vitro. This nanodrug was expected to be used for the efficient treatment of tumors.
Collapse
Affiliation(s)
- Ji Chuan Kong
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Feng Zhou
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Liting Shi
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Yihui Wei
- Henan Polytechinc University Jiaozuo Henan 45400 China
| | - Chunhong Wu
- Henan Polytechinc University Jiaozuo Henan 45400 China
| |
Collapse
|
30
|
Chen Z, Liu Y, Ma R, Zhang M, Wu X, Pen H, Gui F, Liu Y, Xia H, Hu N, Ai B, Xiong J, Xia H, Li W, Ai F. Protective Effect of Long Noncoding RNA OXCT1-AS1 on Doxorubicin-Induced Apoptosis of Human Myocardial Cells by the Competitive Endogenous RNA Pattern. Arq Bras Cardiol 2024; 121:e20230675. [PMID: 38958296 PMCID: PMC11216341 DOI: 10.36660/abc.20230675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/26/2024] [Accepted: 03/11/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND The anthracycline chemotherapeutic antibiotic doxorubicin (DOX) can induce cumulative cardiotoxicity and lead to cardiac dysfunction. Long non-coding RNAs (lncRNAs) can function as important regulators in DOX-induced myocardial injury. OBJECTIVE This study aims to investigate the functional role and molecular mechanism of lncRNA OXCT1 antisense RNA 1 (OXCT1-AS1) in DOX-induced myocardial cell injury in vitro. METHODS Human cardiomyocytes (AC16) were stimulated with DOX to induce a myocardial cell injury model. OXCT1-AS1, miR-874-3p, and BDH1 expression in AC16 cells were determined by RT-qPCR. AC16 cell viability was measured by XTT assay. Flow cytometry was employed to assess the apoptosis of AC16 cells. Western blotting was used to evaluate protein levels of apoptosis-related markers. Dual-luciferase reporter assay was conducted to verify the binding ability between miR-874-3p and OXCT1-AS1 and between miR-874-3p and BDH1. The value of p<0.05 indicated statistical significance. RESULTS OXCT1-AS1 expression was decreased in DOX-treated AC16 cells. Overexpression of OXCT1-AS1 reversed the reduction of cell viability and promotion of cell apoptosis caused by DOX. OXCT1-AS1 is competitively bound to miR-874-3p to upregulate BDH1. BDH1 overexpression restored AC16 cell viability and suppressed cell apoptosis under DOX stimulation. Knocking down BDH1 reversed OXCT1-AS1-mediated attenuation of AC16 cell apoptosis under DOX treatment. CONCLUSION LncRNA OXCT1-AS1 protects human myocardial cells AC16 from DOX-induced apoptosis via the miR-874-3p/BDH1 axis.
Collapse
Affiliation(s)
- Zhen Chen
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Yijue Liu
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Rui Ma
- Hubei University of MedicineSinopharm Dongfeng General HospitalDepartment of Geriatric MedicineShiyanChinaDepartment of Geriatric Medicine – Sinopharm Dongfeng General Hospital – Hubei University of Medicine, Shiyan – China
| | - Mengli Zhang
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Xian Wu
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Huan Pen
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Feng Gui
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| | - Yafeng Liu
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Hao Xia
- Renmin Hospital of Wuhan UniversityDepartment of CardiologyWuhanChinaDepartment of Cardiology – Renmin Hospital of Wuhan University, Wuhan – China
| | - Niandan Hu
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Bo Ai
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Jun Xiong
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Hongxia Xia
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Wenqiang Li
- Renmin Hospital of Wuhan UniversityDepartment of EmergencyWuhanChinaDepartment of Emergency – Renmin Hospital of Wuhan University, Wuhan – China
| | - Fen Ai
- Huazhong University of Science and TechnologyTongji Medical CollegeThe Central Hospital of WuhanWuhanChinaDepartment of Emergency – The Central Hospital of Wuhan – Tongji Medical College – Huazhong University of Science and Technology, Wuhan – China
| |
Collapse
|
31
|
Wang Y, Chang L, Gao H, Yu C, Gao Y, Peng Q. Nanomaterials-based advanced systems for photothermal / photodynamic therapy of oral cancer. Eur J Med Chem 2024; 272:116508. [PMID: 38761583 DOI: 10.1016/j.ejmech.2024.116508] [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: 01/28/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
The traditional clinical approaches for oral cancer consist of surgery, chemotherapy, radiotherapy, immunotherapy, and so on. However, these treatments often induce side effects and exhibit limited efficacy. Photothermal therapy (PTT) emerges as a promising adjuvant treatment, utilizing photothermal agents (PTAs) to convert light energy into heat for tumor ablation. Another innovative approach, photodynamic therapy (PDT), leverages photosensitizers (PSs) and specific wavelength laser irradiation to generate reactive oxygen species (ROS), offering an effective and non-toxic alternative. The relevant combination therapies have been reported in the field of oral cancer. Simultaneously, the advancement of nanomaterials has propelled the clinical application of PTT and PDT. Therefore, a comprehensive understanding of PTT and PDT is required for better application in oral cancer treatment. Here, we review the use of PTT and PDT in oral cancer, including noble metal materials (e.g., Au nanoparticles), carbon materials (e.g., graphene oxide), organic dye molecules (e.g., indocyanine green), organic molecule-based agents (e.g., porphyrin-analog phthalocyanine) and other inorganic materials (e.g., MXenes), exemplify the advantages and disadvantages of common PTAs and PSs, and summarize the combination therapies of PTT with PDT, PTT/PDT with chemotherapy, PTT with radiotherapy, PTT/PDT with immunotherapy, and PTT/PDT with gene therapy in the treatment of oral cancer. The challenges related to the PTT/PDT combination therapy and potential solutions are also discussed.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lili Chang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hongyu Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Chenhao Yu
- Department of Periodontology, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, No. 22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, China
| | - Yujie Gao
- Department of Stomatology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, 610500, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
32
|
Zhao J, Li X, Ma T, Chang B, Zhang B, Fang J. Glutathione-triggered prodrugs: Design strategies, potential applications, and perspectives. Med Res Rev 2024; 44:1013-1054. [PMID: 38140851 DOI: 10.1002/med.22007] [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: 10/17/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
The burgeoning prodrug strategy offers a promising avenue toward improving the efficacy and specificity of cytotoxic drugs. Elevated intracellular levels of glutathione (GSH) have been regarded as a hallmark of tumor cells and characteristic feature of the tumor microenvironment. Considering the pivotal involvement of elevated GSH in the tumorigenic process, a diverse repertoire of GSH-triggered prodrugs has been developed for cancer therapy, facilitating the attenuation of deleterious side effects associated with conventional chemotherapeutic agents and/or the attainment of more efficacious therapeutic outcomes. These prodrug formulations encompass a spectrum of architectures, spanning from small molecules to polymer-based and organic-inorganic nanomaterial constructs. Although the GSH-triggered prodrugs have been gaining increasing interests, a comprehensive review of the advancements made in the field is still lacking. To fill the existing lacuna, this review undertakes a retrospective analysis of noteworthy research endeavors, based on a categorization of these molecules by their diverse recognition units (i.e., disulfides, diselenides, Michael acceptors, and sulfonamides/sulfonates). This review also focuses on explaining the distinct benefits of employing various chemical architecture strategies in the design of these prodrug agents. Furthermore, we highlight the potential for synergistic functionality by incorporating multiple-targeting conjugates, theranostic entities, and combinational treatment modalities, all of which rely on the GSH-triggering. Overall, an extensive overview of the emerging field is presented in this review, highlighting the obstacles and opportunities that lie ahead. Our overarching goal is to furnish methodological guidance for the development of more efficacious GSH-triggered prodrugs in the future. By assessing the pros and cons of current GSH-triggered prodrugs, we expect that this review will be a handful reference for prodrug design, and would provide a guidance for improving the properties of prodrugs and discovering novel trigger scaffolds for constructing GSH-triggered prodrugs.
Collapse
Affiliation(s)
- Jintao Zhao
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Xinming Li
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Tao Ma
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Bingbing Chang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Baoxin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| | - Jianguo Fang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
33
|
Stepanović A, Terzić Jovanović N, Korać A, Zlatović M, Nikolić I, Opsenica I, Pešić M. Novel hybrid compounds of sclareol and doxorubicin as potential anticancer nanotherapy for glioblastoma. Biomed Pharmacother 2024; 174:116496. [PMID: 38537581 DOI: 10.1016/j.biopha.2024.116496] [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: 12/14/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
Two novel hybrid compounds, CON1 and CON2, have been developed by combining sclareol (SC) and doxorubicin (DOX) into a single molecular entity. These hybrid compounds have a 1:1 molar ratio of covalently linked SC and DOX. They have demonstrated promising anticancer properties, especially in glioblastoma cells, and have also shown potential in treating multidrug-resistant (MDR) cancer cells that express the P-glycoprotein (P-gp) membrane transporter. CON1 and CON2 form nanoparticles, as confirmed by Zetasizer, transmission electron microscopy (TEM), and chemical modeling. TEM also showed that CON1 and CON2 can be found in glioblastoma cells, specifically in the cytoplasm, different organelles, nucleus, and nucleolus. To examine the anticancer properties, the U87 glioblastoma cell line, and its corresponding multidrug-resistant U87-TxR cell line, as well as patient-derived astrocytoma grade 3 cells (ASC), were used, while normal human lung fibroblasts were used to determine the selectivity. CON1 and CON2 exhibited better resistance and selectivity profiles than DOX, showing less cytotoxicity, as evidenced by real-time cell analysis, DNA damage determination, cell death induction, mitochondrial respiration, and mitochondrial membrane depolarization studies. Cell cycle analysis and the β-galactosidase activity assay suggested that glioblastoma cells die by senescence following CON1 treatment. Overall, CON1 and CON2 showed great potential as they have better anticancer features than DOX. They are promising candidates for additional preclinical and clinical studies on glioblastoma.
Collapse
Affiliation(s)
- Ana Stepanović
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, Belgrade 11108, Serbia
| | - Nataša Terzić Jovanović
- University of Belgrade - Institute of Chemistry, Technology and Metallurgy - National Institute of the Republic of Serbia, Njegoševa 12, Belgrade 11000, Serbia
| | - Aleksandra Korać
- University of Belgrade - Faculty of Biology & Center for Electron Microscopy, Studentski trg 16, Belgrade 11158, Serbia
| | - Mario Zlatović
- University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, Belgrade 11158, Serbia
| | - Igor Nikolić
- Clinic for Neurosurgery, Clinical Center of Serbia, Pasterova 2, Belgrade 11000, Serbia; School of Medicine, University of Belgrade, Doktora Subotića 8v, Belgrade 11000, Serbia
| | - Igor Opsenica
- University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, Belgrade 11158, Serbia
| | - Milica Pešić
- Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, Belgrade 11108, Serbia.
| |
Collapse
|
34
|
Wiraswati HL, Bashari MH, Alfarafisa NM, Ma’ruf IF, Sholikhah EN, Wahyuningsih TD, Satriyo PB, Mustofa M, Satria D, Damayanti E. Pyrazoline B-Paclitaxel or Doxorubicin Combination Drugs Show Synergistic Activity Against Cancer Cells: In silico Study. Adv Appl Bioinform Chem 2024; 17:33-46. [PMID: 38435441 PMCID: PMC10908341 DOI: 10.2147/aabc.s452281] [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: 11/29/2023] [Accepted: 02/10/2024] [Indexed: 03/05/2024] Open
Abstract
Background Multidrug resistance in various cancer types is a major obstacle in cancer treatment. The concept of a single drug molecular target often causes treatment failure due to the complexity of the cellular processes. Therefore, combination chemotherapy, in which two or more anticancer drugs are co-administered, can overcome this problem because it potentially have synergistic efficacy besides reducing resistance, and drug doses. Previously, we reported that pyrazoline B had promising anticancer activity in both in silico and in vitro studies. To increase the efficacy of this drug, co-administration with established anticancer drugs such as doxorubicin and paclitaxel is necessary. Materials and Methods In this study, we used an in silico approach to predict the synergistic effect of pyrazoline B with paclitaxel or doxorubicin using various computational frameworks and compared the results with those of an established study on the combination of doxorubicin-cyclophosphamide and paclitaxel-ascorbic acid. Results and Discussion Drug interaction analysis showed the combination was safe with no contraindications or side effects. Furthermore, molecular docking studies revealed that doxorubicin-pyrazoline B and doxorubicin-cyclophosphamide may synergistically inhibit cancer cell proliferation by inhibiting the binding of topoisomerase I to the DNA chain. Moreover, the combination of pyrazoline B-paclitaxel may has synergistic activity to cause apoptosis by inhibiting Bcl2 binding to the Bax fragment or inhibiting cell division by inhibiting α-β tubulin disintegration. Paclitaxel-ascorbic acid had a synergistic effect on the inhibition of α-β tubulin disintegration. Conclusion The results show that this combination is promising for further in vitro and in vivo studies.
Collapse
Affiliation(s)
- Hesti Lina Wiraswati
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Oncology and Stem Cells Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Hasan Bashari
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Oncology and Stem Cells Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Oncology and Stem Cells Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Ilma Fauziah Ma’ruf
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency, Bogor, Indonesia
| | - Eti Nurwening Sholikhah
- Department of Pharmacology and Therapy, Faculty of Medicine Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Tutik Dwi Wahyuningsih
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Pamungkas Bagus Satriyo
- Department of Pharmacology and Therapy, Faculty of Medicine Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Mustofa Mustofa
- Department of Pharmacology and Therapy, Faculty of Medicine Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Denny Satria
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Ema Damayanti
- Research Center for Food Technology and Processing, National Research and Innovation Agency, Gunungkidul, Indonesia
| |
Collapse
|
35
|
Agalakova NI. Chloroquine and Chemotherapeutic Compounds in Experimental Cancer Treatment. Int J Mol Sci 2024; 25:945. [PMID: 38256019 PMCID: PMC10815352 DOI: 10.3390/ijms25020945] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Chloroquine (CQ) and its derivate hydroxychloroquine (HCQ), the compounds with recognized ability to suppress autophagy, have been tested in experimental works and in clinical trials as adjuvant therapy for the treatment of tumors of different origin to increase the efficacy of cytotoxic agents. Such a strategy can be effective in overcoming the resistance of cancer cells to standard chemotherapy or anti-angiogenic therapy. This review presents the results of the combined application of CQ/HCQ with conventional chemotherapy drugs (doxorubicin, paclitaxel, platinum-based compounds, gemcitabine, tyrosine kinases and PI3K/Akt/mTOR inhibitors, and other agents) for the treatment of different malignancies obtained in experiments on cultured cancer cells, animal xenografts models, and in a few clinical trials. The effects of such an approach on the viability of cancer cells or tumor growth, as well as autophagy-dependent and -independent molecular mechanisms underlying cellular responses of cancer cells to CQ/HCQ, are summarized. Although the majority of experimental in vitro and in vivo studies have shown that CQ/HCQ can effectively sensitize cancer cells to cytotoxic agents and increase the potential of chemotherapy, the results of clinical trials are often inconsistent. Nevertheless, the pharmacological suppression of autophagy remains a promising tool for increasing the efficacy of standard chemotherapy, and the development of more specific inhibitors is required.
Collapse
Affiliation(s)
- Natalia I Agalakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Avenue, Saint-Petersburg 194223, Russia
| |
Collapse
|
36
|
Kaźmierczak-Siedlecka K, Bulman N, Ulasiński P, Sobocki BK, Połom K, Marano L, Kalinowski L, Skonieczna-Żydecka K. Pharmacomicrobiomics of cell-cycle specific anti-cancer drugs - is it a new perspective for personalized treatment of cancer patients? Gut Microbes 2023; 15:2281017. [PMID: 37985748 PMCID: PMC10730203 DOI: 10.1080/19490976.2023.2281017] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
Intestinal bacteria are equipped with an enzyme apparatus that is involved in the active biotransformation of xenobiotics, including drugs. Pharmacomicrobiomics, a new area of pharmacology, analyses interactions between bacteria and xenobiotics. However, there is another side to the coin. Pharmacotherapeutic agents can significantly modify the microbiota, which consequently affects their efficacy. In this review, we comprehensively gathered scientific evidence on the interplay between anticancer therapies and gut microbes. We also underlined how such interactions might impact the host response to a given therapy. We discuss the possibility of modulating the gut microbiota to increase the effectiveness/decrease the incidence of adverse events during tumor therapy. The anticipation of the future brings new evidence that gut microbiota is a target of interest to increase the efficacy of therapy.
Collapse
Affiliation(s)
- Karolina Kaźmierczak-Siedlecka
- Department of Medical Laboratory Diagnostics – Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
| | - Nikola Bulman
- Department of Medical Laboratory Diagnostics – Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
| | - Paweł Ulasiński
- Unit of Surgery with Unit of Oncological Surgery in Koscierzyna, Kościerzyna, Poland
| | - Bartosz Kamil Sobocki
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdańsk, Poland
| | - Karol Połom
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
| | - Luigi Marano
- Academy of Medical and Social Applied Sciences, Elbląg, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics – Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, Gdańsk, Poland
- BioTechMed Centre/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Gdansk, Poland
| | | |
Collapse
|
37
|
Zhang R, Yu J, Guo Z, Jiang H, Wang C. Camptothecin-based prodrug nanomedicines for cancer therapy. NANOSCALE 2023; 15:17658-17697. [PMID: 37909755 DOI: 10.1039/d3nr04147f] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Camptothecin (CPT) is a cytotoxic alkaloid that attenuates the replication of cancer cells via blocking DNA topoisomerase 1. Despite its encouraging and wide-spectrum antitumour activity, its application is significantly restricted owing to its instability, low solubility, significant toxicity, and acquired tumour cell resistance. This has resulted in the development of many CPT-based therapeutic agents, especially CPT-based nanomedicines, with improved pharmacokinetic and pharmacodynamic profiles. Specifically, smart CPT-based prodrug nanomedicines with stimuli-responsive release capacity have been extensively explored owing to the advantages such as high drug loading, improved stability, and decreased potential toxicity caused by the carrier materials in comparison with normal nanodrugs and traditional delivery systems. In this review, the potential strategies and applications of CPT-based nanoprodrugs for enhanced CPT delivery toward cancer cells are summarized. We appraise in detail the chemical structures and release mechanisms of these nanoprodrugs and guide materials chemists to develop more powerful nanomedicines that have real clinical therapeutic capacities.
Collapse
Affiliation(s)
- Renshuai Zhang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Zhu Guo
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
- The Affiliated Hospital of Qingdao University, Qingdao 266061, China
| | - Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Chao Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| |
Collapse
|
38
|
Zhang R, Zhao X, Jia A, Wang C, Jiang H. Hyaluronic acid-based prodrug nanomedicines for enhanced tumor targeting and therapy: A review. Int J Biol Macromol 2023; 249:125993. [PMID: 37506794 DOI: 10.1016/j.ijbiomac.2023.125993] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Hyaluronic acid (HA) represents a natural polysaccharide which has attracted significant attention owing to its improved tumor targeting capacity, enzyme degradation capacity, and excellent biocompatibility. Its receptors, such as CD44, are overexpressed in diverse cancer cells and are closely related with tumor progress and metastasis. Accordingly, numerous researchers have designed various kinds of HA-based drug delivery platforms for CD44-mediated tumor targeting. Specifically, the HA-based nanoprodrugs possess distinct advantages such as good bioavailability, long circulation time, and controlled drug release and retention ability and have been extensively studied during the past years. In this review, the potential strategies and applications of HA-modified nanoprodrugs for drug molecule delivery in anti-tumor therapy are summarized.
Collapse
Affiliation(s)
- Renshuai Zhang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China
| | - Xiaohua Zhao
- Department of Thoracic surgery, Affiliated Hospital of Weifang Medical University, No.2428, Yuhe road, Kuiwen district, Weifang 261000, China
| | - Ang Jia
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Chao Wang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| | - Hongfei Jiang
- Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China.
| |
Collapse
|