1
|
Zhang Y, Li L, Jiang X, Liu H, Guo X, Wu H, Huang X, Zhou L, Liu C, Shen XC. Injectable dual-network hyaluronic acid nanocomposite hydrogel for prevention of postoperative breast cancer recurrence and wound healing. Int J Biol Macromol 2025; 291:139125. [PMID: 39725096 DOI: 10.1016/j.ijbiomac.2024.139125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/10/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
High locoregional recurrence rates and potential wound infections remain a significant challenge for postoperative breast cancer patients. Herein, we developed a dual-network hyaluronic acid (HA) nanocomposite hydrogel composed of herring sperm DNA (hsDNA) bridged methacrylated HA (HAMA) and FeMg-LDH-ppsa nanohybrid chelated catechol-modified HA (HADA) for the prevention of breast cancer recurrent, anti-infection, and promoting wound healing. Dynamic reversible hsDNA cross-linking combined with metal-catechol chelating renders the hydrogel injectability, rapid self-healing ability, and enhanced mechanical properties. FeMg-LDH-ppsa nanohybrids obtained by in situ polymerization of aniline derivatives in the FeMg-LDH interlayer exhibited excellent photothermal effect. Upon near-infrared (NIR) irradiation, the photothermal effect mediated by FeMg-LDH-ppsa can unwind the hsDNA duplex, enabling the controlled release of preloaded DOX for synergistic photothermal-chemotherapy antitumor. Meanwhile, the catechol-metal (Fe3+/Mg2+) moieties in the hydrogel enhanced tissue adhesion and exhibited intrinsic antimicrobial and bioactive properties, which in combination with the NIR-assisted photothermal effect, significantly accelerated infected wound healing through sterilizing microorganisms, alleviating inflammation, re-epithelialization, and angiogenesis. Overall, this multifunctional hydrogel represented a promising candidate in postoperative wound management for simultaneous tumor elimination, antiinfection, and wound repair.
Collapse
Affiliation(s)
- Yu Zhang
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Lixia Li
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Xiaohe Jiang
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Haimeng Liu
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Xiaolu Guo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541001, PR China
| | - Hui Wu
- College of Material Engineering, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, PR China
| | - Xiaohua Huang
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Li Zhou
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Chanjuan Liu
- Guangxi Colleges and Universities Key Laboratory of Natural and Biomedical Polymer Materials, Guangxi Key Laboratory of Optical and Electronic Materials and Devices, College of Materials Science and Engineering, Guilin University of Technology, Guilin 541004, PR China.
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541001, PR China
| |
Collapse
|
2
|
Liu Y, Zhou Y, Li Y, Kang W, Zhang Y, Xia X, Wang W. Notch Pathway Deactivation Sensitizes Breast Cancer Stem Cells toward Chemotherapy Using NIR Light-Responsive Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2025; 17:430-444. [PMID: 39689353 DOI: 10.1021/acsami.4c16278] [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: 12/19/2024]
Abstract
Chemotherapy remains a major therapeutic approach to cancer treatment. However, its effectiveness can be compromised by the heterogeneity of a solid tumor, in which different cancer cell populations display varied responses to chemotherapy. Such an intratumor heterogeneous structure is maintained by the cancer stem-like cells (CSCs) with inherent capacities for self-renewal and differentiation, giving rise to diverse cell populations. To address this, we proposed a combinational strategy in which tumor lesion-targeted Notch signaling regulation was achieved to disrupt CSC-mediated cancer heterogeneity, thereby sensitizing solid tumors toward paclitaxel (PTX). Specifically, gamma-secretase inhibitor LY-411,575 was co-delivered with PTX using a near-infrared (NIR) light-controlled drug delivery system to realize targeted ablation of both differentiated cancer cells and undifferentiated CSCs. By enabling precise regulation of the Notch pathway at the tumor site through NIR light, we observed significantly elevated efficacy of chemotherapy and notable prevention of postsurgical tumor relapse while minimizing systemic side effects. The devised strategy shows promise in addressing the nonspecific inhibition of stemness across various organs, a challenge that hampers the clinical translation of gamma-secretase inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Yuwei Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Yang Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Yunong Li
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Weirong Kang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Yaming Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Xiaojiao Xia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
3
|
Zhong Z, Gan L, Feng Z, Wang W, Pan X, Wu C, Huang Y. Hydrogel local drug delivery systems for postsurgical management of tumors: Status Quo and perspectives. Mater Today Bio 2024; 29:101308. [PMID: 39525397 PMCID: PMC11550774 DOI: 10.1016/j.mtbio.2024.101308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Surgery is one of the primary treatments for solid tumors. However, the incomplete resection of tumor cells and the immunosuppressive microenvironment make the issue of postsurgical tumor recurrence a great challenge. Furthermore, a wide range of requirements, including ensuring effective hemostasis, implementing prophylactic measures against infection, and promoting wound healing, were also raised in the postsurgical management of tumors. To fulfill these demands, multiple hydrogel local drug delivery systems (HLDDS) were developed recently. These HLDDS are expected to offer numerous advantages in the postsurgical management of tumors, such as achieving high local drug concentrations at the lesion, efficient delivery to surgical microcavities, mitigating systemic side effects, and addressing the diverse demand. Thus, in this review, a detailed discussion of the diverse demands of postsurgical management of tumors is provided. And the current publication trend on HLDDS in the postsurgical management of tumors is analyzed and discussed. Then, the applications of different types of HLDDS, in-situ HLDDS and non-in-situ HLDDS, in postsurgical management of tumors were introduced and summarized. Besides, the current problems and future perspectives are discussed. The review is expected to provide an overview of HLDDS in postsurgical management of tumors and promote their clinical application.
Collapse
Affiliation(s)
- Ziqiao Zhong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Lu Gan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Ziyi Feng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Chuanbin Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| | - Ying Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, 511443, PR China
| |
Collapse
|
4
|
Wang L, Guo H, Zhang W, Li X, Su Z, Huang X. Injectable hydrogels for Fenton-like Mn 2+/Fe 2+ delivery with enhanced chemodynamic therapy prevent osteosarcoma recurrence and promote wound healing after excision surgery. Mater Today Bio 2024; 29:101297. [PMID: 39493811 PMCID: PMC11530760 DOI: 10.1016/j.mtbio.2024.101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
Local recurrence of osteosarcoma and wound healing after excision surgery are major challenges in clinical research. The present anti-tumor treatments could inhibit normal tissues, resulting in difficulties in surgical wound healing. In this study, we constructed an injectable hydrogel as a platform to co-deliver MnO2 nanoparticles and ferrocene Fc, termed as (MnO2/Fc)@PLGA for osteosarcoma treatment and wound healing after excision. By simple local injection, the hydrogel could form a protective barrier on the surgical wound after osteosarcoma excision, which could promote wound healing and steady release of MnO2/Fc nanoparticles. The released MnO2/Fc might undergo the Fenton reaction through Mn2+/Fe2+ to inhibit osteosarcoma cells with chemodynamic therapy (CDT). Furthermore, MnO2 could catalyze endogenous H2O2 to produce O2, which eliminates the adverse effects of H2O2 and remodels the hypoxic state in the local lesions. The increased O2 facilitated surgical wound healing and anti-tumor effects by regulating the hypoxia inducible factor-1 functions. In conclusion, (MnO2/Fc)@PLGA hydrogel could effectively prevent local recurrence of osteosarcoma and promote wound healing after excision surgery, thereby providing a novel strategy for tumor treatment and tissue repair.
Collapse
Affiliation(s)
- Lutong Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Haoyu Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xingyin Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ziliang Su
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
5
|
Wang Z, Zhai B, Sun J, Zhang X, Zou J, Shi Y, Guo D. Recent advances of injectable in situ-forming hydrogels for preventing postoperative tumor recurrence. Drug Deliv 2024; 31:2400476. [PMID: 39252545 PMCID: PMC11389645 DOI: 10.1080/10717544.2024.2400476] [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: 04/12/2024] [Revised: 07/17/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The unavoidable residual tumor tissue from surgery and the strong aggressiveness of tumor cells pose challenges to the postoperative treatment of tumor patients, accompanied by in situ tumor recurrence and decreased quality of life. Therefore, there is an urgent need to explore appropriate postoperative therapeutic strategies to remove residual tumor cells after surgery to inhibit tumor recurrence and metastasis after surgery. In recent years, with the rapid development of biomedical materials, the study of local delivery systems as postoperative delivery of therapeutic agents has gradually attracted the attention of researchers. Injectable in situ-forming hydrogel is a locally administered agent injected in situ as a solution that can be loaded with various therapeutic agents and rapidly gels to form a semi-solid gel at the treatment site. This type of hydrogel tightly fills the surgical site and covers irregular excision surfaces. In this paper, we review the recent advances in the application of injectable in situ-forming hydrogels in postoperative therapy, focusing on the matrix materials of this type of hydrogel and its application in the postoperative treatment of different types of tumors, as well as discussing the challenges and prospects of its clinical application.
Collapse
Affiliation(s)
- Zhanpeng Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Bingtao Zhai
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Jing Sun
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Xiaofei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Junbo Zou
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Yajun Shi
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Dongyan Guo
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| |
Collapse
|
6
|
Giles C, Lee J. Inflammation drives tumor growth in an immunocompetent implantable metastasis model. RESEARCH SQUARE 2024:rs.3.rs-4719290. [PMID: 39149496 PMCID: PMC11326373 DOI: 10.21203/rs.3.rs-4719290/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Nearly 90% of cancer deaths are due to metastasis. Conventional cancer therapeutics including chemotherapy, surgery, and radiotherapy, are effective in treating primary tumors, but may aggravate disseminated tumor cells (DTCs) into regaining a proliferative state. Models isolating the post dissemination environment are needed to address the potential risks of these therapies, however modeling post dissemination environments is challenging. Often, host organisms become moribund due to primary tumor mass before native metastatic niches can evolve. Implantable tissue engineered niches have been used to attract circulating tumor cells independent of the primary tumor. Here, we serially transplant such tissue engineered niches with recruited DTCs in order to isolate the post dissemination environment. After transplantaion, 69% of scaffolds developed overt post-dissemination cancer growth, however 100% of scaffolds did not grow to a life-threatening critical size within twelve weeks. Adjuvant chemotherapy, while initially effective, did not prevent long-term DTC growth in scaffolds. Subjecting these transplanted niches to surgical resection via biopsy punch enhanced CD31, MMP9, Ly6G, and tumor burden compared to control scaffolds. Biopsy punching was able to rescue tumor incidence from prior chemotherapy. This model of serial transplantation of engineered DTC niches is a highly controllable and flexible method of establishing and systematically investigating the post-dissemination niche.
Collapse
|
7
|
Jing Y, Wang C, Li C, Wei Z, Lei D, Chen A, Li X, He X, Cen L, Sun M, Liu B, Xue B, Li R. Development of a manganese complex hyaluronic acid hydrogel encapsulating stimuli-responsive Gambogic acid nanoparticles for targeted Intratumoral delivery. Int J Biol Macromol 2024; 270:132348. [PMID: 38750838 DOI: 10.1016/j.ijbiomac.2024.132348] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/05/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Gambogic acid is a natural compound with anticancer properties and is effective for many tumors. But its low water solubility and dose-dependent side effects limit its clinical application. This study aims to develop a novel drug delivery system for intratumoral delivery of gambogic acid. In our experimental study, we propose a new method for encapsulating gambogic acid nanoparticles using a manganese composite hyaluronic acid hydrogel as a carrier, designed for targeted drug delivery to tumors. The hydrogel delivery system is synthesized through the coordination of hyaluronic acid-dopamine (HA-DOPA) and manganese ions. The incorporation of manganese ions serves three purposes:1.To form cross-linked hydrogels, thereby improving the mechanical properties of HA-DOPA.2.To monitor the retention of hydrogels in vivo in real-time using magnetic resonance imaging (MRI).3.To activate the body's immune response. The experimental results show that the designed hydrogel has good biosafety, in vivo sustained release effect and imaging tracking ability. In the mouse CT26 model, the hydrogel drug-loaded group can better inhibit tumor growth. Further immunological analysis shows that the drug-loaded hydrogel group can stimulate the body's immune response, thereby better achieving anti-tumor effects. These findings indicate the potential of the developed manganese composite hyaluronic acid hydrogel as an effective and safe platform for intratumoral drug delivery. The amalgamation of biocompatibility, controlled drug release, and imaging prowess positions this system as a promising candidate for tumor treatment.
Collapse
Affiliation(s)
- Yuanhao Jing
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Chun Wang
- Department of Pain, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Chunhua Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Zijian Wei
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Dan Lei
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Anni Chen
- Nanjing International Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Xiang Li
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xiaowen He
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Lanqi Cen
- Department of Oncology, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, China. 210000
| | - Mengna Sun
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China; The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Bin Xue
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing 210008, China.
| | - Rutian Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing 210008, China; The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School & Clinical Cancer Institute of Nanjing University, Nanjing, China.
| |
Collapse
|
8
|
Gui J, Zhu Y, Chen X, Gong T, Zhang Z, Yu R, Fu Y. Systemic platelet inhibition with localized chemotherapy by an injectable ROS-scavenging gel against postsurgical breast cancer recurrence and metastasis. Acta Biomater 2024; 177:388-399. [PMID: 38307476 DOI: 10.1016/j.actbio.2024.01.037] [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: 11/24/2023] [Revised: 01/13/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Abstract
Early solid tumors benefit from surgical resection, but residual stubborn microtumors, pro-inflammatory microenvironment and activated platelets at the postoperative wound site are prone to recurrence and metastasis, resulting in poor prognosis. Here, we developed a dual-pronged strategy consisting of (i) in-situ forming ROS-scavenging gels loaded with anticancer drugs at the postoperative wound site to improve the tumor microenvironment and inhibit the recurrence of residual microtumors after orthotopic surgery, and (ii) systemic administration of clopidegrol via albumin nanoparticles for inhibiting activated platelets in the circulation thus inhibiting tumor remote migration. In a mouse model of postoperative recurrence and metastasis of orthotopic 4T1 breast cancer, the dual-pronged strategy greatly inhibited postoperative orthotopic tumor recurrence and reduced lung metastasis. This work provides an effective strategy for the postoperative intervention and treatment of solid tumors to inhibit postoperative tumor recurrence and metastasis, which has the potential to improve the prognosis and survival of patients with postoperative solid tumors. STATEMENT OF SIGNIFICANCE: Early-stage solid tumors benefit from surgical resection. However, the presence of residual microtumors, pro-inflammatory tumor microenvironment, and activated platelets at the postoperative wound site lead to recurrence and metastasis, ultimately resulting in poor prognosis. Here, we have devised a dual-pronged approach that includes (i) in-situ forming ROS-scavenging gels loaded with anticancer drugs (TM@Gel) at the wound site after surgery to enhance the tumor microenvironment (TME) and hinder the reappearance of residual microtumors, and (ii) systemic administration of clopidegrol through albumin nanoparticles (HHP) for inhibiting activated platelets in the circulation thus impeding tumor distant migration. This work provides a viable option for postoperative intervention and treatment of solid tumors to suppress postoperative tumor recurrence and metastasis.
Collapse
Affiliation(s)
- Jiajia Gui
- Key Laboratory of Drug- Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yueting Zhu
- Key Laboratory of Drug- Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Chen
- Key Laboratory of Drug- Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug- Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug- Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ruilian Yu
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Yao Fu
- Key Laboratory of Drug- Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
Li X, Xing D, Bai Y, Du Y, Lang S, Li K, Xiang J, Liu G, Liu S. Injectable hydrogel with antimicrobial and anti-inflammatory properties for postoperative tumor wound care. Biomed Mater 2024; 19:025028. [PMID: 38290161 DOI: 10.1088/1748-605x/ad2408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
Clinically, tumor removal surgery leaves irregularly shaped wounds that are susceptible to bacterial infection and further lead to excessive inflammation. Injectable hydrogel dressings with antimicrobial and anti-inflammatory properties have been recognized as an effective strategy to care for postoperative tumor wounds and prevent recurrence in recent years. In this work, we constructed a hydrogel network by ionic bonding interactions between quaternized chitosan (QCS) and epigallocatechin gallate (EGCG)-Zn complexes which were coordinated by EGCG and zinc ions. Because of the synergistic effect of QCS and EGCG-Zn, the hydrogel exhibited outstanding antimicrobial capacity (>99.9% inhibition), which could prevent infections caused byEscherichia coli and Staphylococcus aureus. In addition, the hydrogel was able to inhibit the growth of mice breast cancer cells (56.81% survival rate within 72 h) and reduce inflammation, which was attributed to the sustained release of EGCG. The results showed that the hydrogel was effective in inhibiting tumor recurrence and accelerating wound closure when applied to the postoperative tumor wounds. This study provided a simple and reliable strategy for postoperative tumor wound care using antimicrobial and anti-inflammatory injectable dressings, confirming their great potential in the field of postoperative wound dressings.
Collapse
Affiliation(s)
- Xinyun Li
- Department of Oncology, Dazhou Integrated Traditional Chinese Medicine and Western Medicine Hospital, Dazhou Second People's Hospital, Dazhou, Sichuan 635000, People's Republic of China
| | - Dandan Xing
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Yangjing Bai
- West China School of Nursing, Sichuan University/Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yangrui Du
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Shiying Lang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Kaijun Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Jun Xiang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Gongyan Liu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Shan Liu
- Department of Endocrinology, Yueyang Central Hospital, Yueyang 414100, People's Republic of China
| |
Collapse
|
10
|
Ebrahimnia M, Alavi S, Vaezi H, Karamat Iradmousa M, Haeri A. Exploring the vast potentials and probable limitations of novel and nanostructured implantable drug delivery systems for cancer treatment. EXCLI JOURNAL 2024; 23:143-179. [PMID: 38487087 PMCID: PMC10938236 DOI: 10.17179/excli2023-6747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024]
Abstract
Conventional cancer chemotherapy regimens, albeit successful to some extent, suffer from some significant drawbacks, such as high-dose requirements, limited bioavailability, low therapeutic indices, emergence of multiple drug resistance, off-target distribution, and adverse effects. The main goal of developing implantable drug delivery systems (IDDS) is to address these challenges and maintain anti-cancer drugs directly at the intended sites of therapeutic action while minimizing inevitable side effects. IDDS possess numerous advantages over conventional drug delivery, including controlled drug release patterns, one-time drug administration, as well as loading and stabilizing poorly water-soluble chemotherapy drugs. Here, we summarized conventional and novel (three-dimensional (3D) printing and microfluidic) preparation techniques of different IDDS, including nanofibers, films, hydrogels, wafers, sponges, and osmotic pumps. These systems could be designed with high biocompatibility and biodegradability features using a wide variety of natural and synthetic polymers. We also reviewed the published data on these systems in cancer therapy with a particular focus on their release behavior. Various release profiles could be attained in IDDS, which enable predictable, adjustable, and sustained drug releases. Furthermore, multi-step or stimuli-responsive drug release could be obtained in these systems. The studies mentioned in this article have proven the effectiveness of IDDS for treating different cancer types with high prevalence, including breast cancer, and aggressive cancer types, such as glioblastoma and liver cancer. Additionally, the challenges in applying IDDS for efficacious cancer therapy and their potential future developments are also discussed. Considering the high potential of IDDS for further advancements, such as programmable release and degradation features, further clinical trials are needed to ensure their efficiency. The overall goal of this review is to expand our understanding of the behavior of commonly investigated IDDS and to identify the barriers that should be addressed in the pursuit of more efficient therapies for cancer. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Maryam Ebrahimnia
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sonia Alavi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Hamed Vaezi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Karamat Iradmousa
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Chen X, Ouyang H, Zhang Y, Chen C, Nan S, Pu X, Gong T, Zhang ZR, Liu R, Fu Y. Antigen-specific T cell activation through targeted delivery of in-situ generated antigen and calcium ionophore to enhance antitumor immunotherapy. J Control Release 2024; 365:544-557. [PMID: 38052255 DOI: 10.1016/j.jconrel.2023.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023]
Abstract
Recent advances in adoptive T-cell therapy have delivered impressive therapeutic outcomes by instigating enduring anti-tumor responses. Nonetheless, achieving specific T-cell activation remains a challenge due to several factors. Some cancer cells evade T-cell recognition due to the scarcity of tumor-specific T cells and deficiencies in antigen processing or major histocompatibility complex (MHC) presentation. Notably underestimated is the impact of waning T-cell receptor (TCR) expression and the constrained formation of immune synapses (IS) between dendritic cells (DCs) and T cells, impairing T-cell activation. Addressing these complexities, we introduce a pioneering approach featuring the deployment of a gel implant. This implant establishes an on-site antigen reservoir, efficiently targets DCs in lymph nodes, and facilitates calcium ion (Ca2+) delivery. Engineered with controlled swelling, poroelasticity, and resilience, the gel is suitable for surgical implantation. Its ample encapsulation capacity accommodates both photosensitizers and nanoparticles. Upon in situ photothermal irradiation, the gel generates tumor-specific antigens. Furthermore, cationic albumin nanoparticles (cNPs) co-loaded with monophosphoryl lipid A (MPLA) and ionomycin are released, guiding antigens to tumor-draining lymph nodes for DCs maturation. This meticulous process fosters the formation of IS thereby amplifying antigen-specific T-cell activation.
Collapse
Affiliation(s)
- Xue Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hongling Ouyang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yunxiao Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Conglin Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Simin Nan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhi-Rong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Renhe Liu
- Global Health Drug Discovery Institute, Beijing, China.
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
12
|
Zheng X, Zhao D, Jin Y, Liu Y, Liu D. Role of the NLRP3 inflammasome in gynecological disease. Biomed Pharmacother 2023; 166:115393. [PMID: 37660654 DOI: 10.1016/j.biopha.2023.115393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023] Open
Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in the innate immune system and is a three-part macromolecular complex comprising the NLRP3 protein, apoptosis-associated speck-like protein containing a CARD (ASC) and the cysteine protease pro-caspase-1. When the NLRP3 inflammasome is activated, it can produce interleukin (IL)- 1β and IL-18 and eventually lead to inflammatory cell pyroptosis. Related studies have demonstrated that the NLRP3 inflammasome can induce an immune response and is related to the occurrence and development of gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer. NLRP3 inflammasome inhibitors are beneficial for maintaining cellular homeostasis and tissue health and have been found effective in targeting some gynecological diseases. However, excessive inhibitor concentrations have been found to cause adverse effects. Therefore, proper control of NLRP3 inflammasome activity is critical. This paper summarizes the structure and function of the NLRP3 inflammasome and highlights the therapeutic potential of targeting it in gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer The application of NLRP3 inflammasome inhibitors is also discussed.
Collapse
Affiliation(s)
- Xu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Dan Zhao
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Yang Liu
- Acupuncture department,Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| |
Collapse
|
13
|
Feng Y, Zhang Z, Tang W, Dai Y. Gel/hydrogel-based in situ biomaterial platforms for cancer postoperative treatment and recovery. EXPLORATION (BEIJING, CHINA) 2023; 3:20220173. [PMID: 37933278 PMCID: PMC10582614 DOI: 10.1002/exp.20220173] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/03/2023] [Indexed: 11/08/2023]
Abstract
Tumor surgical resection is the major strategy for cancer treatment. Meanwhile, perioperative treatment especially the postoperative adjuvant anticancer strategies play essential roles in satisfying therapeutic results and rapid recovery. Postoperative tumor recurrence, metastasis, bleeding, inter-tissue adhesion, infection, and delayed wound healing are vital risks that could lead to poor prognosis or even treatment failure. Therefore, methods targeting these postoperative complications are in desperate need. In situ biomaterial-based drug delivery platforms are promising candidates for postoperative treatment and recovery, resulting from their excellent properties including good biocompatibility, adaptive shape, limited systemic effect, designable function, and easy drug loading. In this review, we focus on introducing the gel/hydrogel-based in situ biomaterial platforms involving their properties, advantages, and synthesis procedures. Based on the loaded contents in the gel/hydrogel such as anticancer drugs, immunologic agents, cell components, and multifunctional nanoparticles, we further discuss the applications of the in situ platforms for postoperative tumor recurrence and metastasis inhibition. Finally, other functions aiming at fast postoperative recovery were introduced, including hemostasis, antibacterial infection, adhesion prevention, tissue repair, and wound healing. In conclusion, gel/hydrogel is a developing and promising platform for postoperative treatment, exhibiting gratifying therapeutic effects and inconspicuous toxicity to normal tissues, which deserves further research and exploration.
Collapse
Affiliation(s)
- Yuzhao Feng
- Cancer Centre and Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacau SARChina
- MoE Frontiers Science Center for Precision OncologyUniversity of MacauMacau SARChina
| | - Zhan Zhang
- Cancer Centre and Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacau SARChina
- MoE Frontiers Science Center for Precision OncologyUniversity of MacauMacau SARChina
| | - Wei Tang
- Departments of Pharmacy and Diagnostic RadiologyNanomedicine Translational Research ProgramFaculty of Science and Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational MedicineFaculty of Health SciencesUniversity of MacauMacau SARChina
- MoE Frontiers Science Center for Precision OncologyUniversity of MacauMacau SARChina
| |
Collapse
|
14
|
Pandya AK, Vora LK, Umeyor C, Surve D, Patel A, Biswas S, Patel K, Patravale VB. Polymeric in situ forming depots for long-acting drug delivery systems. Adv Drug Deliv Rev 2023; 200:115003. [PMID: 37422267 DOI: 10.1016/j.addr.2023.115003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023]
Abstract
Polymeric in situ forming depots have emerged as highly promising drug delivery systems for long-acting applications. Their effectiveness is attributed to essential characteristics such as biocompatibility, biodegradability, and the ability to form a stable gel or solid upon injection. Moreover, they provide added versatility by complementing existing polymeric drug delivery systems like micro- and nanoparticles. The formulation's low viscosity facilitates manufacturing unit operations and enhances delivery efficiency, as it can be easily administered via hypodermic needles. The release mechanism of drugs from these systems can be predetermined using various functional polymers. To enable unique depot design, numerous strategies involving physiological and chemical stimuli have been explored. Important assessment criteria for in situ forming depots include biocompatibility, gel strength and syringeability, texture, biodegradation, release profile, and sterility. This review focuses on the fabrication approaches, key evaluation parameters, and pharmaceutical applications of in situ forming depots, considering perspectives from academia and industry. Additionally, insights about the future prospects of this technology are discussed.
Collapse
Affiliation(s)
- Anjali K Pandya
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India; School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Chukwuebuka Umeyor
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 422001, Anambra State, Nigeria
| | - Dhanashree Surve
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Akanksha Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, Telangana 500078, India
| | - Ketankumar Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Vandana B Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India.
| |
Collapse
|
15
|
Alfair BM, Jabarti AA, Albalawi SS, Khodir AE, Al-Gayyar MM. Arctiin Inhibits Inflammation, Fibrosis, and Tumor Cell Migration in Rats With Ehrlich Solid Carcinoma. Cureus 2023; 15:e44987. [PMID: 37701157 PMCID: PMC10495034 DOI: 10.7759/cureus.44987] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND AND OBJECTIVES ESC or Ehrlich solid carcinoma is a type of tumor originating from a spontaneous mammary adenocarcinoma in mice. It is a highly aggressive and fast-growing carcinoma that can create a solid mass when inserted under the skin. Its solid, undifferentiated form makes it an ideal model for researching cancer biology, tumor immunology, and testing various anti-cancer treatments. Additionally, arctiin has multiple beneficial properties, such as anti-proliferative, anti-oxidative, anti-adipogenic, and anti-bacterial. This study aimed to explore the potential anti-cancer benefits of arctiin in rats with ESC while also analyzing its effects on cell fibrosis markers, tumor cell migration, and inflammasome pathways. METHODS Rats were given a tumor in their left hind limb via an intramuscular injection consisting of 2×106 cells. After eight days, some of the rats received a daily oral dose of 30 mg/kg of arctiin for three weeks. Muscle samples were observed under an electron microscope or stained with hematoxylin/eosin. Additionally, gene expression and protein levels of toll-like receptor 4 (TLR4), NLR family pyrin domain containing 3 (NLRP3), signal transducer and activator of transcription 3 (STAT3), transforming growth factor (TGF)-β, endothelial growth factor (VEGF), and cyclin D1 were assessed in another part of the muscle samples. RESULTS When ESC rats were given arctiin as a treatment, their mean survival time increased and their tumor volume and weight decreased. Additionally, when tumor tissue was examined under an electron microscope, it showed signs of pleomorphic cells, necrosis, nuclear fragmentation, membrane damage with cytoplasmic content spilling, and loss of cellular junction. The stained sections with hematoxylin/eosin showed a dense cellular mass and compressed, degenerated, and atrophied muscle. However, treatment with arctiin improved all these effects. Finally, the expression of TLR4, NLRP3, STAT3, TGF-β, VEGF, and cyclin D1 was significantly reduced with arctiin treatment. CONCLUSIONS Through the use of arctiin, tumor size and weight were effectively reduced, leading to an increase in the average survival time of rats and an improvement in muscle structure. Additional research has shown that arctiin is able to suppress inflammation, fibrosis, and the migration of tumor cells by inhibiting STAT3, TGF-β1, TLR4, NLRP3, VEGF, and cyclin D1.
Collapse
Affiliation(s)
| | | | | | - Ahmed E Khodir
- Pharmacology and Toxicology, Horus University, New Damietta, EGY
| | - Mohammed M Al-Gayyar
- Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
- Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, EGY
| |
Collapse
|
16
|
Wang Y, Wang Q, Wang X, Yao P, Dai Q, Qi X, Yang M, Zhang X, Huang R, Yang J, Wang Q, Xia P, Zhang D, Sun F. Docetaxel-loaded pH/ROS dual-responsive nanoparticles with self-supplied ROS for inhibiting metastasis and enhancing immunotherapy of breast cancer. J Nanobiotechnology 2023; 21:286. [PMID: 37608285 PMCID: PMC10464340 DOI: 10.1186/s12951-023-02013-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/18/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Although stimuli-responsive nanoplatforms were developed to deliver immunogenic cell death (ICD) inducers to enhance cancer immunotherapy, the complete release of ICD inducers into the tumor microenvironment (TME) was limited by the inadequate supplementation of endogenous stimulus (e.g., reactive oxygen species (ROS)). To address this issue, we synthesized a self-responsive nanomaterial with self-supplied ROS, which mainly consists of a ROS responsive moiety HPAP and cinnamaldehyde (CA) as the ROS-generating agent. The endogenous ROS can accelerate the degradation of HPAP in materials to release docetaxel (DTX, an ICD inducer). In intracellular acidic environment, the pH-sensitive acetal was cleaved to release CA. The released CA in turn induces the generation of more ROS through mitochondrial damage, resulting in amplified DTX release. Using this self-cycling and self-responsive nanomaterial as a carrier, DTX-loaded pH/ROS dual-responsive nanoparticles (DTX/FA-CA-Oxi-αCD NPs) were fabricated and evaluated in vitro and in vivo. RESULTS In vitro experiments validated that the NPs could be effectively internalized by FA-overexpressed cells and completely release DTX in acidic and ROS microenvironments to induce ICD effect. These NPs significantly blocked 4T1 cell migration and decreased cell invasion. In vivo experiments demonstrated that the tumor-targeted NPs significantly inhibited tumor growth and blocked tumor metastasis. More importantly, these NPs significantly improved immunotherapy through triggering effector T-cell activation and relieving the immunosuppressive state of the TME. CONCLUSIONS Our results demonstrated that DTX/FA-CA-Oxi-αCD NPs displayed great potential in preventing tumor metastasis, inhibiting tumor growth, and improving the efficacy of anti-PD-1antibody.
Collapse
Affiliation(s)
- Yu Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qianmei Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaowen Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Pu Yao
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qing Dai
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xiao Zhang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Rong Huang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jing Yang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qian Wang
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Peiyuan Xia
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Department of Urology, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing, 400038, China.
| | - Fengjun Sun
- Department of Pharmacy, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
17
|
Si X, Ji G, Ma S, Chen H, Shi Z, Zhang Y, Tang Z, Song W, Chen X. Comprehensive evaluation of biopolymer immune implants for peritoneal metastasis carcinoma therapy. J Control Release 2023; 353:289-302. [PMID: 36403683 DOI: 10.1016/j.jconrel.2022.11.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/03/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022]
Abstract
Immunotherapy has been widely used in the treatment of advanced stage cancers with spreading metastases, while the fully activation of immune system often requires sustained and long-acting immune stimulation by immunotherapeutic agents. In previous studies, we designed a biopolymer immune implant by dynamic covalent bonds and achieved sustained release of loaded immunotherapeutic agents, thus stimulated systemic immune activation and elicited immune memory effects. Herein, we further optimized the implants and carried out a comprehensive evaluation of the implants on peritoneal metastasis carcinoma (PMC) therapy. Our results showed that the implants fabricated with 8-arm polyethylene glycol amine (8-arm PEG-NH2) and 40% oxidation degree dextran (ODEX) exhibited a satisfactory degradation time for activating the antitumor immunity. The drug combination of oxaliplatin (OxP) and resiquimod (R848) could be sustainably released from the implants for 18 days. The implants cured 75% of mice with PMC and elicited immune memory effects to resist tumor re-challenge without obvious side effects observed. Mechanism analysis revealed that the implants could serve as an in-situ vaccine to enhance the infiltration of activated dendritic cells (DCs), T cells and natural killer (NK) cells inside the tumor, as well as increase the serum tumor necrosis factor α (TNF-α), interferon-γ (IFN-γ) and interleukin 12 (IL-12) levels. These results strongly support the clinical translation potential of this sustained released biopolymer immune implants for PMC therapy.
Collapse
Affiliation(s)
- Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China
| | - Guofeng Ji
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Xuanwu Hospital, Capital Medical University, Beijing 100010, PR China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China
| | - Hongyu Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Zhiyuan Shi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China; University of Science and Technology of China, Hefei 230026, PR China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China; University of Science and Technology of China, Hefei 230026, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China; University of Science and Technology of China, Hefei 230026, PR China.
| |
Collapse
|
18
|
Wang M, Wang Y, Liu R, Yu R, Gong T, Zhang Z, Fu Y. TLR4 Blockade Using Docosahexaenoic Acid Restores Vulnerability of Drug-Tolerant Tumor Cells and Prevents Breast Cancer Metastasis and Postsurgical Relapse. ACS BIO & MED CHEM AU 2022; 3:97-113. [PMID: 37101603 PMCID: PMC10125315 DOI: 10.1021/acsbiomedchemau.2c00061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
Nonmutational mechanisms were recently discovered leading to reversible drug tolerance. Despite the rapid elimination of a majority of tumor cells, a small subpopulation of "'drug-tolerant"' cells remain viable with lethal drug exposure, which may further lead to resistance or tumor relapse. Several signaling pathways are involved in the local or systemic inflammatory responses contributing to drug-induced phenotypic switch. Here, we report that Toll-like receptor 4 (TLR4)-interacting lipid docosahexaenoic acid (DHA) restores the cytotoxic effect of doxorubicin (DOX) in the lipopolysaccharide-treated breast tumor cell line 4T1, preventing the phenotypic switch to drug-tolerant cells, which significantly reduces primary tumor growth and lung metastasis in both 4T1 orthotopic and experimental metastasis models. Importantly, DHA in combination with DOX delays and inhibits tumor recurrence following surgical removal of the primary tumor. Furthermore, the coencapsulation of DHA and DOX in a nanoemulsion significantly prolongs the survival of mice in the postsurgical 4T1 tumor relapse model with significantly reduced systemic toxicity. The synergistic antitumor, antimetastasis, and antirecurrence effects of DHA + DOX combination are likely mediated by attenuating TLR4 activation, thus sensitizing tumor cells to standard chemotherapy.
Collapse
Affiliation(s)
- Mou Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Yuejing Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Renhe Liu
- The Scripps Research Institute, 10550 North Torrey Pines Road,
La Jolla, San Diego, California92037, United States
| | - Ruilian Yu
- Department of Oncology, Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610072, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| |
Collapse
|
19
|
Wang Y, Wang S, Hu W, Su F, Liu F, Li S. In situ photo‐crosslinked hydrogels prepared from acrylated 4‐arm‐poly(ethylene glycol)‐poly(ε‐caprolactone) block copolymers for local cancer therapy. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Yuandou Wang
- State Key Laboratory Base of Eco‐chemical Engineering, College of Chemical Engineering Qingdao University of Science and Technology Qingdao China
- Institute of High Performance Polymers Qingdao University of Science and Technology Qingdao China
| | - Shuxin Wang
- State Key Laboratory Base of Eco‐chemical Engineering, College of Chemical Engineering Qingdao University of Science and Technology Qingdao China
- Institute of High Performance Polymers Qingdao University of Science and Technology Qingdao China
| | - Wenju Hu
- State Key Laboratory Base of Eco‐chemical Engineering, College of Chemical Engineering Qingdao University of Science and Technology Qingdao China
- Institute of High Performance Polymers Qingdao University of Science and Technology Qingdao China
| | - Feng Su
- State Key Laboratory Base of Eco‐chemical Engineering, College of Chemical Engineering Qingdao University of Science and Technology Qingdao China
- Institute of High Performance Polymers Qingdao University of Science and Technology Qingdao China
| | - Fusheng Liu
- State Key Laboratory Base of Eco‐chemical Engineering, College of Chemical Engineering Qingdao University of Science and Technology Qingdao China
| | - Suming Li
- Institut Européen des Membranes, IEM, UMR 5635 Univ Montpellier, CNRS, ENSCM Montpellier France
| |
Collapse
|
20
|
Lv B, Shen N, Cheng Z, Chen Y, Ding H, Yuan J, Zhao K, Zhang Y. Strategies for Biomaterial-Based Spinal Cord Injury Repair via the TLR4-NF-κB Signaling Pathway. Front Bioeng Biotechnol 2022; 9:813169. [PMID: 35600111 PMCID: PMC9116428 DOI: 10.3389/fbioe.2021.813169] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022] Open
Abstract
The repair and motor functional recovery after spinal cord injury (SCI) has remained a clinical challenge. Injury-induced gliosis and inflammation lead to a physical barrier and an extremely inhibitory microenvironment, which in turn hinders the recovery of SCI. TLR4-NF-κB is a classic implant-related innate immunomodulation signaling pathway and part of numerous biomaterial-based treatment strategies for SCI. Numerous experimental studies have demonstrated that the regulation of TLR4-NF-κB signaling pathway plays an important role in the alleviation of inflammatory responses, the modulation of autophagy, apoptosis and ferroptosis, and the enhancement of anti-oxidative effect post-SCI. An increasing number of novel biomaterials have been fabricated as scaffolds and carriers, loaded with phytochemicals and drugs, to inhibit the progression of SCI through regulation of TLR4-NF-κB. This review summarizes the empirical strategies for the recovery after SCI through individual or composite biomaterials that mediate the TLR4-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bin Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Naiting Shen
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangrong Cheng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhang Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Ding
- Department of Orthopedics, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Jishan Yuan
- Department of Orthopedics, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Kangchen Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yukun Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Wang Y, Niu W, Qu X, Lei B. Bioactive Anti-Inflammatory Thermocatalytic Nanometal-Polyphenol Polypeptide Scaffolds for MRSA-Infection/Tumor Postsurgical Tissue Repair. ACS APPLIED MATERIALS & INTERFACES 2022; 14:4946-4958. [PMID: 35073045 DOI: 10.1021/acsami.1c21082] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Postsurgical tumor recurrence, infection, and tissue defect are still the challenges in clinical medicine. The development of multifunctional biomaterial scaffolds with a microenvironment-responsive tumor-infection therapy-tissue repair is highly desirable. Herein, we report a bioactive, injectable, adhesive, self-healing, antibacterial, and anti-inflammatory metal-polyphenol polypeptide nanocomposite scaffold (PEAPF) with temporal-spatial-controlled inflammation-triggered therapeutic properties for efficient infection and postsurgical tumor therapy and skin repair. PEAPF scaffolds showed sustained and inherent inflammation-triggered Fenton catalysis and mild thermochemical effect for specifically inhibiting tumor recurrence in vitro and in vivo. The PEAPF scaffolds significantly facilitated skin tissue regeneration in MRSA-infected chronic wounds and postsurgical tissue defects after tumor resection. This study presents the multifunctional scaffold-based safe and efficient therapeutic strategy to prevent local tumor recurrence and enhance postsurgical tissue regeneration.
Collapse
Affiliation(s)
- Yidan Wang
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
| | - Wen Niu
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
| | - Xiaoyan Qu
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
| | - Bo Lei
- Frontier Institute of Science and Technology, Instrument Analysis Center, Xi'an Jiaotong University, Xi'an 710054, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710054, China
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710054, China
| |
Collapse
|