1
|
Liu Z, Tang Z, Yin Y, Wan M, Zhan J, Ren L. A Microneedle Patch Delivers Mitochondria- and Lysosomes- Dual Targeting Prodrug-Like Photosensitizers with Regulated Photoactivity for Precise Photodynamic Therapy. Adv Healthc Mater 2025:e2403954. [PMID: 39967393 DOI: 10.1002/adhm.202403954] [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: 10/11/2024] [Revised: 01/09/2025] [Indexed: 02/20/2025]
Abstract
Antitumor photodynamic therapy (PDT) faces huge challenges as selectivity and phototoxic damage, requiring delivery photosensitizers (PSs) to specifically accumulate in tumors even in organelle, and avoid the phototoxic damage during delivery. Herein, a microneedle patch (AIE-mito-TPP@MN) containing mitochondria- and lysosomes- dual targeting prodrug-like PSs (AIE-mito-TPP/AlPcSNa4) that is self-assembled by mitochondria-targeted aggregation-induced-emission molecule (AIE-mito-TPP) and lysosome-targeted aluminum phthalocyanine tetrasulfonate (AlPcSNa4), is developed to achieve cancer-cell-organelle-specific targeting delivery for precise PDT with high selectivity and low phototoxic damage. AIE-mito-TPP/AlPcSNa4 displays prodrug-like activity via the regulated photoactivity to reduce the phototoxic damage caused by the "always on" PSs. Meanwhile, AIE-mito-TPP/AlPcSNa4@MN can insert into the epidermis to achieve rapid AIE-mito-TPP/AlPcSNa4 delivery in tumor lesion, and enhance selective accumulation in tumor cells. The higher lysosomal acidity in tumor cells facilitates AIE-mito-TPP/AlPcSNa4 disassembly and promotes targeting. Under light irradiation, AIE-mito-TPP/AlPcSNa4@MN impairs mitochondrial and lysosomal function to induce deeper tumor cells apoptosis at a low dose (≈6 µg), presenting greater therapeutic efficacy than AIE-mito-TPP@MN, AlPcSNa4@MN, or intravenous injection. Moreover, AIE-mito-TPP/AlPcSNa4@MN presents good biocompatibility as lower accumulation and targeting in normal cells, as well as the regulated photoactivity of prodrug-like PSs. Therefore, the dual organelle-targeting microneedle possesses great potential for precise PDT with high selectivity.
Collapse
Affiliation(s)
- Zheng Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | - Zhimin Tang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | - Ying Yin
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | - Miaojian Wan
- Department of Dermatology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jiezhao Zhan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| | - Li Ren
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, China
| |
Collapse
|
2
|
Wang T, Liu H, Li M, Ji Z, Zhang X, Wang N, Chen Y, Sun J, Liu F. Microneedle-based nanodrugs for tumor immunotherapy. J Control Release 2025; 380:539-562. [PMID: 39923854 DOI: 10.1016/j.jconrel.2025.02.003] [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: 10/13/2024] [Revised: 01/08/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
Microneedles have emerged as a promising and effective method for delivering therapeutic drugs and immunobiologics to treat various diseases. It is widely recognized that immune therapy has limited efficacy in solid tumors due to physical barriers and the immunosuppressive tumor microenvironment. Microneedle-based nanodrugs (NDMNs) offer a novel approach to overcome these limitations. These tiny needles are designed to load a variety of inorganic and organic nanoparticles, antigen vaccines, gene drugs, oncolytic viruses, and more. Utilizing microneedle arrays, NDMNs can effectively penetrate the skin barrier, delivering drugs precisely to the tumor site or immunoactive regions within the skin. Additionally, by designing and optimizing the microneedle structure, shape, and functionality, NDMNs enable precise drug release and efficient penetration, thereby enhancing the efficacy of tumor immunotherapy. In this review, we comprehensively discuss the pivotal role of NDMNs in cancer immunotherapy, summarizing innovative microneedle design strategies, mechanisms of immune activation, and delivery strategies of various nanodrugs. Furthermore, we explore the current clinical realities, limitations, and future prospects of NDMNs in tumor immunotherapy.
Collapse
Affiliation(s)
- Tianye Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Department of General Surgery, The First Hospital of Dalian Medical University, Dalian 116000, China
| | - Hongyu Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Meng Li
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Zao Ji
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Xinyuan Zhang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Nan Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China
| | - Ying Chen
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang 110001, China; Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China.
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems Ministry of Education, Shenyang 110016, China.
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang 110001, China; Phase I Clinical Trails Center, The First Hospital, China Medical University, Shenyang 110001, China.
| |
Collapse
|
3
|
Zhang H, Feng L, Wang Y, Bi Z, Wei Z, Sun Y, Shi M, Chen T, Wang L, Zhang S. Spontaneously Photocatalytic Nanoplatform for Sensitive Diagnosis and Penetrated Therapy of Cancer. Anal Chem 2025; 97:2809-2818. [PMID: 39873428 DOI: 10.1021/acs.analchem.4c05185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
In this study, a sensitive diagnosis and spontaneously photocatalytic therapy of cancer based on chemiluminescence (CL) and nanozyme was studied. Briefly, carbon nitride-supported copper nanoparticles (CuCNs) loaded with luminol (CuCN-L) were utilized to develop a microneedle patch (CuCN-L/MN). The CuCN-L probe could target overexpressed H2O2 in the TME and actively emit CL to achieve cancer cell imaging for diagnosis. In addition, 425 nm blue-violet CL could photocatalyze CuCN to reduce carbon dioxide (CO2) to carbon monoxide (CO) for gas therapy (GT). Meanwhile, CuCN, acting as a photosensitizer (PS), could be activated by CL to produce reactive oxygen species (ROS), which realized photodynamic therapy (PDT). Furthermore, the use of a degradable MN based on hyaluronic acid (HA) promotes painless local administration as well as deep administration of GT and PDT. This technology of optical sensor-mediated anticancer treatment will have great potential to provide new inspirations for cancer diagnosis and therapy based on a self-excited nanoplatform.
Collapse
Affiliation(s)
- Huairong Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Lu Feng
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yuqi Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zhiru Bi
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zizhen Wei
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Yongbiao Sun
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
| | - Minghui Shi
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
| | - Tingting Chen
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Linjing Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, College of Medicine, Linyi University, Linyi 276005, China
| | - Shusheng Zhang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| |
Collapse
|
4
|
Hulimane Shivaswamy R, Binulal P, Benoy A, Lakshmiramanan K, Bhaskar N, Pandya HJ. Microneedles as a Promising Technology for Disease Monitoring and Drug Delivery: A Review. ACS MATERIALS AU 2025; 5:115-140. [PMID: 39802146 PMCID: PMC11718548 DOI: 10.1021/acsmaterialsau.4c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025]
Abstract
The delivery of molecules, such as DNA, RNA, peptides, and certain hydrophilic drugs, across the epidermal barrier poses a significant obstacle. Microneedle technology has emerged as a prominent area of focus in biomedical research because of its ability to deliver a wide range of biomolecules, vaccines, medicines, and other substances through the skin. Microneedles (MNs) form microchannels by disrupting the skin's structure, which compromises its barrier function, and facilitating the easy penetration of drugs into the skin. These devices enhance the administration of many therapeutic substances to the skin, enhancing their stability. Transcutaneous delivery of medications using a microneedle patch offers advantages over conventional drug administration methods. Microneedles containing active substances can be stimulated by different internal and external factors to result in the regulated release of the substances. To achieve efficient drug administration to the desired location, it is necessary to consider the design of needles with appropriate optimized characteristics. The choice of materials for developing and manufacturing these devices is vital in determining the pharmacodynamics and pharmacokinetics of drug delivery. This article provides the most recent update and overview of the numerous microneedle systems that utilize different activators to stimulate the release of active components from the microneedles. Further, it discusses the materials utilized for producing microneedles and the design strategies important in managing the release of drugs. An explanation of the commonly employed fabrication techniques in biomedical applications and electronics, particularly for integrated microneedle drug delivery systems, is discussed. To successfully implement microneedle technology in clinical settings, it is essential to comprehensively assess several factors, such as biocompatibility, drug stability, safety, and production cost. Finally, an in-depth review of these criteria and the difficulties and potential future direction of microneedles in delivering drugs and monitoring diseases is explored.
Collapse
Affiliation(s)
| | - Pranav Binulal
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Aloysious Benoy
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Kaushik Lakshmiramanan
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Nitu Bhaskar
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Hardik Jeetendra Pandya
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| |
Collapse
|
5
|
Cao J, Wu B, Yuan P, Liu Y, Hu C. Advances in Research of Hydrogel Microneedle-Based Delivery Systems for Disease Treatment. Pharmaceutics 2024; 16:1571. [PMID: 39771550 PMCID: PMC11676655 DOI: 10.3390/pharmaceutics16121571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Microneedles (MNs), composed of multiple micron-scale needle-like structures attached to a base, offer a minimally invasive approach for transdermal drug delivery by penetrating the stratum corneum and delivering therapeutic agents directly to the epidermis or dermis. Hydrogel microneedles (HMNs) stand out among various MN types due to their excellent biocompatibility, high drug-loading capacity, and tunable drug-release properties. This review systematically examines the matrix materials and fabrication methods of HMN systems, highlighting advancements in natural and synthetic polymers, and explores their applications in treating conditions such as wound healing, hair loss, cardiovascular diseases, and cancer. Furthermore, the potential of HMNs for disease diagnostics is discussed. The review identifies key challenges, including limited mechanical strength, drug-loading efficiency, and lack of standardization, while proposing strategies to overcome these issues. With the integration of intelligent design and enhanced control over drug dosage and safety, HMNs are poised to revolutionize transdermal drug delivery and expand their applications in personalized medicine.
Collapse
Affiliation(s)
- Juan Cao
- School of Fashion and Design Art, Sichuan Normal University, Chengdu 610066, China;
| | - Bo Wu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China; (B.W.); (Y.L.)
| | - Ping Yuan
- School of Mechanical Engineering, Chengdu University, Chengdu 610106, China;
| | - Yeqi Liu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China; (B.W.); (Y.L.)
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
6
|
Wu M, Zhao Y, Zhang C, Pu K. Advancing Proteolysis Targeting Chimera (PROTAC) Nanotechnology in Protein Homeostasis Reprograming for Disease Treatment. ACS NANO 2024; 18:28502-28530. [PMID: 39377250 DOI: 10.1021/acsnano.4c09800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) represent a transformative class of therapeutic agents that leverage the intrinsic protein degradation machinery to modulate the hemostasis of key disease-associated proteins selectively. Although several PROTACs have been approved for clinical application, suboptimal therapeutic efficacy and potential adverse side effects remain challenging. Benefiting from the enhanced targeted delivery, reduced systemic toxicity, and improved bioavailability, nanomedicines can be tailored with precision to integrate with PROTACs which hold significant potential to facilitate PROTAC nanomedicines (nano-PROTACs) for clinical translation with enhanced efficacy and reduced side effects. In this review, we provide an overview of the recent progress in the convergence of nanotechnology with PROTAC design, leveraging the inherent properties of nanomaterials, such as lipids, polymers, inorganic nanoparticles, nanohydrogels, proteins, and nucleic acids, for precise PROTAC delivery. Additionally, we discuss the various categories of PROTAC targets and provide insights into their clinical translational potential, alongside the challenges that need to be addressed.
Collapse
Affiliation(s)
- Mengyao Wu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yilan Zhao
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chi Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, 636921, Singapore
| |
Collapse
|
7
|
Yi H, Yu H, Wang L, Wang Y, Ouyang C, Keshta BE. Microneedle transdermal drug delivery as a candidate for the treatment of gouty arthritis: Material structure, design strategies and prospects. Acta Biomater 2024; 187:20-50. [PMID: 39182801 DOI: 10.1016/j.actbio.2024.08.032] [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: 05/20/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Gouty arthritis (GA) is caused by monosodium urate (MSU) crystals deposition. GA is difficult to cure because of its complex disease mechanism and the tendency to reoccur. GA patients require long-term uric acid-lowering and anti-inflammatory treatments. In the past ten years, as a painless, convenient and well-tolerated new drug transdermal delivery method, microneedles (MNs) administration has been continuously developed, which can realize various drug release modes to deal with various complex diseases. Compared with the traditional administration methods (oral and injection), MNs are more conducive to the long-term independent treatment of GA patients because of their safe, efficient and controllable drug delivery ability. In this review, the pathological mechanism of GA and common therapeutic drugs for GA are summarized. After that, MNs drug delivery mechanisms were summarized: dissolution release mechanism, swelling release mechanism and channel-assisted release mechanism. According to drug delivery patterns of MNs, the mechanisms and applications of rapid-release MNs, long-acting MNs, intelligent-release MNs and multiple-release MNs were reviewed. Additionally, existing problems and future trends of MNs in the treatment of GA were also discussed. STATEMENT OF SIGNIFICANCE: Gout is an arthritis caused by metabolic disease "hyperuricemia". Epidemiological studies show that the number of gouty patients is increasing rapidly worldwide. Due to the complex disease mechanism and recurrent nature of gout, gouty patients require long-term therapy. However, traditional drug delivery modes (oral and injectable) have poor adherence, low drug utilization, and lack of local localized targeting. They may lead to adverse effects such as rashes and gastrointestinal reactions. As a painless, convenient and well-tolerated new drug transdermal delivery method, microneedles have been continuously developed, which can realize various drug release modes to deal with gouty arthritis. In this review, the material structure, design strategy and future outlook of microneedles for treating gouty arthritis will be reviewed.
Collapse
Affiliation(s)
- Hong Yi
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China; Zhejiang-Russia Joint Laboratory of Photo-Electron-Megnetic Functional Materials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China; Zhejiang-Russia Joint Laboratory of Photo-Electron-Megnetic Functional Materials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China.
| | - Yu Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Chenguang Ouyang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Basem E Keshta
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| |
Collapse
|
8
|
Venturi A, Di Bona S, Desantis J, Eleuteri M, Bartalucci M, Baroni M, Benedetti P, Goracci L, Cruciani G. Between Theory and Practice: Computational/Experimental Integrated Approaches to Understand the Solubility and Lipophilicity of PROTACs. J Med Chem 2024; 67:16355-16380. [PMID: 39271471 DOI: 10.1021/acs.jmedchem.4c01235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Emerging drug candidates more often fall in the beyond-rule-of-five chemical space. Among them, proteolysis targeting chimeras (PROTACs) have gained great attention in the past decade. Although physicochemical properties of small molecules accomplishing Lipinski's rule-of-five can now be easily predicted through models generated by large data collections, for PROTACs the knowledge is still limited and heterogeneous, hampering their prediction. Here, the kinetic solubility and the coefficient of distribution at pH 7.4 (LogD7.4) of 44 PROTACs, designed and synthesized to cover a wide chemical space, were measured. Their generally low solubility and high lipophilicity required an optimization of the experimental methods. Concerning the LogD7.4, several in silico prediction tools were tested, which were quite accurate for classical small molecules but provided dissimilar outcomes for PROTACs. Finally, in silico models for the prediction of PROTACs' kinetic solubility and LogD7.4 were proposed by combining in-house generated experimental data with 3D description of PROTACs' structures.
Collapse
Affiliation(s)
- Andrea Venturi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| | - Stefano Di Bona
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| | - Jenny Desantis
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| | - Michela Eleuteri
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| | - Matteo Bartalucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| | - Massimo Baroni
- Kinetic Business Centre, Molecular Discovery Ltd., Elstree, Borehamwood, Hertfordshire WD6 4PJ, United Kingdom
| | - Paolo Benedetti
- Kinetic Business Centre, Molecular Discovery Ltd., Elstree, Borehamwood, Hertfordshire WD6 4PJ, United Kingdom
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| | - Gabriele Cruciani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via dell' Elce di Sotto 8, Perugia 06123, Italy
| |
Collapse
|
9
|
Chu H, Xue J, Yang Y, Zheng H, Luo D, Li Z. Advances of Smart Stimulus-Responsive Microneedles in Cancer Treatment. SMALL METHODS 2024; 8:e2301455. [PMID: 38148309 DOI: 10.1002/smtd.202301455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 12/09/2023] [Indexed: 12/28/2023]
Abstract
Microneedles (MNs) have emerged as a highly promising technology for delivering drugs via the skin. They provide several benefits, including high drug bioavailability, non-invasiveness, painlessness, and high safety. Traditional strategies for intravenous delivery of anti-tumor drugs have risks of systemic toxicity and easy development of drug resistance, while MN technology facilitates precise delivery and on-demand release of drugs in local tissues. In addition, by further combining with stimulus-responsive materials, the construction of smart stimulus-responsive MNs can be achieved, which can respond to specific physical/chemical stimuli from the internal or external environment, thereby further improving the accuracy of tumor treatment and reducing toxicity to surrounding tissues/cells. This review systematically summarizes the classification, materials, and reaction mechanisms of stimulus-responsive MNs, outlines the benefits and challenges of various types of MNs, and details their application and latest progress in cancer treatment. Finally, the development prospects of smart MNs in tumor treatment are also discussed, bringing inspiration for future precision treatment of tumors.
Collapse
Affiliation(s)
- Huaqing Chu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Jiangtao Xue
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuan Yang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dan Luo
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| | - Zhou Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 101400, China
| |
Collapse
|
10
|
Ma T, Wang X, Wang Y, Hao Y, Yang X, Yan X, Huang Q, Li Z, Cong B, Li D. Curcumin analogue AC17-loaded dissolvable microneedles activate FOXO3 and enhance localized drug delivery for oral squamous cell carcinoma treatment. Int J Pharm 2024; 661:124385. [PMID: 38925237 DOI: 10.1016/j.ijpharm.2024.124385] [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: 03/09/2024] [Revised: 06/06/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Curcumin, a polyphenol extracted from turmeric, is a potential alternative for the treatment of oral squamous cell carcinoma (OSCC) due to its remarkable anticancer activity and low systemic toxicity. To further enhance the anticancer activity and bioavailability of curcumin, we synthesized a curcumin analogue, AC17, by modifying the benzene ring and methylene group of curcumin. A soluble hyaluronic acid microneedle patch (AC17@HAMN) was developed to ensure accurate and safe delivery of AC17 to tumor tissues. The inhibitory effect of AC17 on OSCC cells was stronger than that of curcumin and some common analogues. Transcriptome sequencing showed that the target genes of AC17 were mainly concentrated in apoptosis, cell cycle and cell senescence pathways. Among them, AC17 induces cell cycle arrest and inhibits cell proliferation mainly by activating FOXO3 signaling. With good penetration and dissolution properties, microneedles can deliver AC17 directly to the tumor site and show good anti-tumor effect. Moreover, AC17@HAMN showed good biosafety. In summary, AC17@HAMN offers high efficiency, minimal invasiveness, and few adverse reactions. This microneedle patch holds great promise for potential clinical applications, especially for the treatment of OSCC.
Collapse
Affiliation(s)
- Tengyu Ma
- School of Stomatology, Binzhou Medical University, Yantai 264003, China
| | - Xinxin Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Yaozhong Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Yuanping Hao
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China
| | - Xinting Yang
- School of Stomatology, Binzhou Medical University, Yantai 264003, China
| | - Xin Yan
- Department of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Qihang Huang
- Department of Stomatology, Shandong Second Medical University, Weifang 261053, China
| | - Zhuoran Li
- Stomatology Department of Jining Medical University, Jining 272067, China
| | - Beibei Cong
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China.
| | - Dechao Li
- School of Stomatology, Binzhou Medical University, Yantai 264003, China; Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao 266001, China.
| |
Collapse
|
11
|
Fan QQ, Tian H, Cheng JX, Zou JB, Luan F, Qiao JX, Zhang D, Tian Y, Zhai BT, Guo DY. Research progress of sorafenib drug delivery system in the treatment of hepatocellular carcinoma: An update. Biomed Pharmacother 2024; 177:117118. [PMID: 39002440 DOI: 10.1016/j.biopha.2024.117118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors in the contemporary era, representing a significant global health concern. Early HCC patients have mild symptoms or are asymptomatic, which promotes the onset and progression of the disease. Moreover, advanced HCC is insensitive to chemotherapy, making traditional clinical treatment unable to block cancer development. Sorafenib (SFB) is a first-line targeted drug for advanced HCC patients with anti-angiogenesis and anti-tumor cell proliferation effects. However, the efficacy of SFB is constrained by its off-target distribution, rapid metabolism, and multi-drug resistance. In recent years, nanoparticles based on a variety of materials have been demonstrated to enhance the targeting and therapeutic efficacy of SFB against HCC. Concurrently, the advent of joint drug delivery systems has furnished crucial empirical evidence for reversing SFB resistance. This review will summarize the application of nanotechnology in the field of HCC treatment over the past five years. It will focus on the research progress of SFB delivery systems combined with multiple therapeutic modalities in HCC treatment.
Collapse
Affiliation(s)
- Qiang-Qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Huan Tian
- Xi'an Hospital of Traditional Chinese Medicine, 710021, China
| | - Jiang-Xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Jun-Bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Jia-Xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Dan Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China
| | - Bing-Tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| | - Dong-Yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| |
Collapse
|
12
|
Fan L, Tong W, Wei A, Mu X. Progress of proteolysis-targeting chimeras (PROTACs) delivery system in tumor treatment. Int J Biol Macromol 2024; 275:133680. [PMID: 38971291 DOI: 10.1016/j.ijbiomac.2024.133680] [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: 05/03/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) can use the intrinsic protein degradation system in cells to degrade pathogenic target proteins, and are currently a revolutionary frontier of development strategy for tumor treatment with small molecules. However, the poor water solubility, low cellular permeability, and off-target side effects of most PROTACs have prevented them from passing the preclinical research stage of drug development. This requires the use of appropriate delivery systems to overcome these challenging hurdles and ensure precise delivery of PROTACs towards the tumor site. Therefore, the combination of PROTACs and multifunctional delivery systems will open up new research directions for targeted degradation of tumor proteins. In this review, we systematically reviewed the design principles and the most recent advances of various PROTACs delivery systems. Moreover, the constructive strategies for developing multifunctional PROTACs delivery systems were proposed comprehensively. This review aims to deepen the understanding of PROTACs drugs and promote the further development of PROTACs delivery system.
Collapse
Affiliation(s)
- Lianlian Fan
- Department of Pharmacy, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Weifang Tong
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130021, China
| | - Anhui Wei
- Jilin University School of Pharmaceutical Sciences, Changchun 130021, China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
13
|
Wang Z, Pang S, Liu X, Dong Z, Tian Y, Ashrafizadeh M, Rabiee N, Ertas YN, Mao Y. Chitosan- and hyaluronic acid-based nanoarchitectures in phototherapy: Combination cancer chemotherapy, immunotherapy and gene therapy. Int J Biol Macromol 2024; 273:132579. [PMID: 38795895 DOI: 10.1016/j.ijbiomac.2024.132579] [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: 02/01/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Cancer phototherapy has been introduced as a new potential modality for tumor suppression. However, the efficacy of phototherapy has been limited due to a lack of targeted delivery of photosensitizers. Therefore, the application of biocompatible and multifunctional nanoparticles in phototherapy is appreciated. Chitosan (CS) as a cationic polymer and hyaluronic acid (HA) as a CD44-targeting agent are two widely utilized polymers in nanoparticle synthesis and functionalization. The current review focuses on the application of HA and CS nanostructures in cancer phototherapy. These nanocarriers can be used in phototherapy to induce hyperthermia and singlet oxygen generation for tumor ablation. CS and HA can be used for the synthesis of nanostructures, or they can functionalize other kinds of nanostructures used for phototherapy, such as gold nanorods. The HA and CS nanostructures can combine chemotherapy or immunotherapy with phototherapy to augment tumor suppression. Moreover, the CS nanostructures can be functionalized with HA for specific cancer phototherapy. The CS and HA nanostructures promote the cellular uptake of genes and photosensitizers to facilitate gene therapy and phototherapy. Such nanostructures specifically stimulate phototherapy at the tumor site, with particle toxic impacts on normal cells. Moreover, CS and HA nanostructures demonstrate high biocompatibility for further clinical applications.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng 252000, Shandong, PR China
| | - Shuo Pang
- Department of Urinary Surgery, Jinan Third People's Hospital, Jinan, Shandong 250101, PR China
| | - Xiaoli Liu
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zi Dong
- Department of Gastroenterology, Lincang People's Hospital, Lincang, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, United States
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077 India
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Türkiye; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Türkiye; UNAM-National Nanotechnology Research Center, Bilkent University, Ankara 06800, Türkiye.
| | - Ying Mao
- Department of Oncology, Suining Central Hospital, Suining City, Sichuan, China.
| |
Collapse
|
14
|
Zhao Q, Gu N, Li Y, Wu X, Ouyang Q, Deng L, Ma H, Zhu Y, Fang F, Ye H, Wu K. Self-assembled gel microneedle formed by MS deep eutectic solvent as a transdermal delivery system for hyperpigmentation treatment. Mater Today Bio 2024; 26:101090. [PMID: 38800564 PMCID: PMC11127278 DOI: 10.1016/j.mtbio.2024.101090] [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: 02/09/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Hyperpigmentation (HP) is an unfavorable skin disease that typically caused by injury, inflammation, or photoaging and leads to numerous physical and psychological issues in patients. Recently, development and application of natural whitening substances, particularly compound curcumin (CUR), is one of the most prevalent treatments for HP. However, it is still a formidable challenge to improve the percutaneous delivery of CUR due to its inadequate solubility in water and excellent barrier function of skin. To overcome the limitations of conventional delivery and increase the percutaneous absorption of CUR, the efficient delivery of CUR is urgently required. Herein, we developed a new malic acid-sorbitol deep eutectic solvent (MS/DES) gel microneedle loaded with CUR as a transdermal delivery system for HP treatment. The MS/DES gel produces three-dimensional (3D) network structure by self-assembly of hydrogen bond interactions, which conferred the CUR-MS/DES-GMN with sufficient mechanical properties to successfully penetrate skin tissue while also helping to enhance the drug's release rate. The CUR-MS/DES-GMN exhibit high biocompatibility and mechanical property in vivo of mice. The zebrafish experiments also show that CUR-MS/DES gel has significant effect of anti-pigmentation. Therefore, the designed CUR-MS/DES-GMN system provides a novel strategy for HP treatment based on self-assembly of naturally molecules.
Collapse
Affiliation(s)
- Qi Zhao
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Na Gu
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Yier Li
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Xia Wu
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Qianqian Ouyang
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Luming Deng
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Hui Ma
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Yuzhen Zhu
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Fang Fang
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
| | - Hua Ye
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| | - Kefeng Wu
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, 524003, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang, 524023, China
| |
Collapse
|
15
|
Zhang R, Xie S, Ran J, Li T. Restraining the power of Proteolysis Targeting Chimeras in the cage: A necessary and important refinement for therapeutic safety. J Cell Physiol 2024; 239:e31255. [PMID: 38501341 DOI: 10.1002/jcp.31255] [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: 01/22/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
Proteolysis Targeting Chimeras (PROTACs) represent a significant advancement in therapeutic drug development by leveraging the ubiquitin-proteasome system to enable targeted protein degradation, particularly impacting oncology. This review delves into the various types of PROTACs, such as peptide-based, nucleic acid-based, and small molecule PROTACs, each addressing distinct challenges in protein degradation. It also discusses innovative strategies like bridged PROTACs and conditional switch-activated PROTACs, offering precise targeting of previously "undruggable" proteins. The potential of PROTACs extends beyond oncology, with ongoing research and technological advancements needed to maximize their therapeutic potential. Future progress in this field relies on interdisciplinary collaboration and the integration of advanced computational tools to open new treatment avenues across various diseases.
Collapse
Affiliation(s)
- Renshuai Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Songbo Xie
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jie Ran
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Te Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
16
|
Zuo Y, Sun R, Del Piccolo N, Stevens MM. Microneedle-mediated nanomedicine to enhance therapeutic and diagnostic efficacy. NANO CONVERGENCE 2024; 11:15. [PMID: 38634994 PMCID: PMC11026339 DOI: 10.1186/s40580-024-00421-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
Nanomedicine has been extensively explored for therapeutic and diagnostic applications in recent years, owing to its numerous advantages such as controlled release, targeted delivery, and efficient protection of encapsulated agents. Integration of microneedle technologies with nanomedicine has the potential to address current limitations in nanomedicine for drug delivery including relatively low therapeutic efficacy and poor patient compliance and enable theragnostic uses. In this Review, we first summarize representative types of nanomedicine and describe their broad applications. We then outline the current challenges faced by nanomedicine, with a focus on issues related to physical barriers, biological barriers, and patient compliance. Next, we provide an overview of microneedle systems, including their definition, manufacturing strategies, drug release mechanisms, and current advantages and challenges. We also discuss the use of microneedle-mediated nanomedicine systems for therapeutic and diagnostic applications. Finally, we provide a perspective on the current status and future prospects for microneedle-mediated nanomedicine for biomedical applications.
Collapse
Affiliation(s)
- Yuyang Zuo
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Rujie Sun
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Nuala Del Piccolo
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK.
- Department of Physiology, Anatomy and Genetics, Department of Engineering Science, and Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU, UK.
| |
Collapse
|
17
|
Zhang Q, Liu X, He J. Applications and prospects of microneedles in tumor drug delivery. J Mater Chem B 2024; 12:3336-3355. [PMID: 38501172 DOI: 10.1039/d3tb02646a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
As drug delivery devices, microneedles are used widely in the local administration of various drugs. Such drug-loaded microneedles are minimally invasive, almost painless, and have high drug delivery efficiency. In recent decades, with advancements in microneedle technology, an increasing number of adaptive, engineered, and intelligent microneedles have been designed to meet increasing clinical needs. This article summarizes the types, preparation materials, and preparation methods of microneedles, as well as the latest research progress in the application of microneedles in tumor drug delivery. This article also discusses the current challenges and improvement strategies in the use of microneedles for tumor drug delivery.
Collapse
Affiliation(s)
- Qiang Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
18
|
Cheng X, Henick BS, Cheng K. Anticancer Therapy Targeting Cancer-Derived Extracellular Vesicles. ACS NANO 2024; 18:6748-6765. [PMID: 38393984 DOI: 10.1021/acsnano.3c06462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Extracellular vesicles (EVs) are natural lipid nanoparticles secreted by most types of cells. In malignant cancer, EVs derived from cancer cells contribute to its progression and metastasis by facilitating tumor growth and invasion, interfering with anticancer immunity, and establishing premetastasis niches in distant organs. In recent years, multiple strategies targeting cancer-derived EVs have been proposed to improve cancer patient outcomes, including inhibiting EV generation, disrupting EVs during trafficking, and blocking EV uptake by recipient cells. Developments in EV engineering also show promising results in harnessing cancer-derived EVs as anticancer agents. Here, we summarize the current understanding of the origin and functions of cancer-derived EVs and review the recent progress in anticancer therapy targeting these EVs.
Collapse
Affiliation(s)
- Xiao Cheng
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Joint Department of Biomedical EngineeringNorth Carolina State University, Raleigh, North Carolina 27606, United States
| | - Brian S Henick
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
19
|
Zhong J, Zhao R, Wang Y, Su YX, Lan X. Nano-PROTACs: state of the art and perspectives. NANOSCALE 2024; 16:4378-4391. [PMID: 38305466 DOI: 10.1039/d3nr06059d] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
PROteolysis TArgeting Chimeras (PROTACs), as a recently identified technique in the field of new drug development, provide new concepts for disease treatment and are expected to revolutionize drug discovery. With high specificity and flexibility, PROTACs serve as an innovative research tool to target and degrade disease-relevant proteins that are not currently pharmaceutically vulnerable to eliminating their functions by hijacking the ubiquitin-proteasome system. To date, PROTACs still face the challenges of low solubility, poor permeability, off-target effects, and metabolic instability. The combination of nanotechnology and PROTACs has been explored to enhance the in vivo performance of PROTACs regarding overcoming these challenging hurdles. In this review, we summarize the latest advancements in the building-block design of PROTAC prodrug nanoparticles and provide an overview of existing/potential delivery systems and loading approaches for PROTAC drugs. Furthermore, we discuss the current status and prospects of the split-and-mix approach for PROTAC drug optimization. Additionally, the advantages and translational potentials of carrier-free nano-PROTACs and their combinational therapeutic effects are highlighted. This review aims to foster a deeper understanding of this rapidly evolving field and facilitate the progress of nano-PROTACs that will continue to push the boundaries of achieving selectivity and controlled release of PROTAC drugs.
Collapse
Affiliation(s)
- Jie Zhong
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China.
- Discipline of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China.
| | - Ruiqi Zhao
- Discipline of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China.
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China.
| | - Yu-Xiong Su
- Discipline of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR 999077, China.
| | - Xinmiao Lan
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
20
|
Wang G, Kato K, Aoki I, Ichinose S, Inoue D, Tottori S, Nishizawa M. Transdermal drug delivery using a porous microneedle device driven by a hydrogel electroosmotic pump. J Mater Chem B 2024; 12:1490-1494. [PMID: 38234189 DOI: 10.1039/d3tb02208k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Integrating a hydrogel electroosmotic pump with a parylene C-coated porous microneedle (PMN) is developed for transdermal drug delivery applications. The hydrogel pump is fabricated by combining an anionic and a cationic hydrogel to generate enhanced electroosmosis flow (EOF) to drive the transportation of molecules via PMN.
Collapse
Affiliation(s)
- Gaobo Wang
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
| | - Kosuke Kato
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
| | - Izuru Aoki
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
| | - Sae Ichinose
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
| | - Daisuke Inoue
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
| | - Soichiro Tottori
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
| | - Matsuhiko Nishizawa
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-1 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan.
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6-4 Aramaki Aoba, Aoba-ku, Sendai 980-8579, Japan
- Division for the Establishment of Frontier Sciences of the Organization for Advanced Studies, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan
| |
Collapse
|
21
|
Filho D, Guerrero M, Pariguana M, Marican A, Durán-Lara EF. Hydrogel-Based Microneedle as a Drug Delivery System. Pharmaceutics 2023; 15:2444. [PMID: 37896204 PMCID: PMC10609870 DOI: 10.3390/pharmaceutics15102444] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
The skin is considered the largest and most accessible organ in the human body, and allows the use of noninvasive and efficient strategies for drug administration, such as the transdermal drug delivery system (TDDS). TDDSs are systems or patches, with the ability and purpose to deliver effective and therapeutic doses of drugs through the skin. Regarding the specific interaction between hydrogels (HG) and microneedles (MNs), we seek to find out how this combination would be applied in the context of drug delivery, and we detail some possible advantages of the methods used. Depending on the components belonging to the HG matrix, we can obtain some essential characteristics that make the combination of hydrogels-microneedles (HG-MNs) very advantageous, such as the response to external stimuli, among others. Based on multiple characteristics provided by HGMNs that are depicted in this work, it is possible to obtain unique properties that include controlled, sustained, and localized drug release, as well as the possibility of a synergistic association between the components of the formulation and the combination of more than one bioactive component. In conclusion, a system based on HG-MNs can offer many advantages in the biomedical field, bringing to light a new technological and safe system for improving the pharmacokinetics and pharmacodynamics of drugs and new treatment perspectives.
Collapse
Affiliation(s)
- David Filho
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), University of Talca, Talca 3460000, Chile
| | - Marcelo Guerrero
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), University of Talca, Talca 3460000, Chile
| | - Manuel Pariguana
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), University of Talca, Talca 3460000, Chile
| | - Adolfo Marican
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), University of Talca, Talca 3460000, Chile
- Institute of Chemistry of Natural Research, University of Talca, Talca 3460000, Chile
| | - Esteban F Durán-Lara
- Laboratory of Bio & Nano Materials, Drug Delivery and Controlled Release, Department of Microbiology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), University of Talca, Talca 3460000, Chile
| |
Collapse
|