1
|
Wang Y, Jiang N, Wu S, Lin G, Lu W, Shang B, Zhu X, Han W, Li J, Chen Y. An injectable hydrogel for hemostasis and tumor suppression in intraoperative breast cancer. BIOMATERIALS ADVANCES 2025; 172:214219. [PMID: 39987716 DOI: 10.1016/j.bioadv.2025.214219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/14/2025] [Accepted: 02/02/2025] [Indexed: 02/25/2025]
Abstract
In the period between surgery and systemic therapy for breast cancer, residual tumor cells may proliferate, leading to tumor recurrence. Additionally, intraoperative wound bleeding may cause surgical failure or the spread of tumor cells. This study introduces an innovative injectable hydrogel composed of oxidized hyaluronic acid (OHA) loaded 5-fluorouracil (5-FU) and N-carboxyethyl chitosan (CEC), designed for intraoperative hemostasis and tumor suppression in intraoperative breast cancer. The CEC/OHA injectable hydrogel was synthesized through a Schiff base reaction between the aldehyde group of OHA and the amino group of CEC, incorporating 5-FU during hydrogel formation. This CEC/OHA injectable hydrogel demonstrated hemostatic effects comparable to gelatin sponges in both an in vivo rat liver hemorrhage model and an in vitro rat tail amputation model. When loaded with 5-FU, the injectable hydrogel effectively inhibited the proliferation of MDA-MB-231 breast cancer cells in vitro, significantly inhibited tumor growth and recurrence in vivo, and did not induce significant damage or inflammatory response in any major organ. This CEC/OHA & 5-FU injectable hydrogel is envisioned as a complementary therapeutic regimen during the intraoperative period in breast cancer surgery to prevent hemostasis and tumor recurrence.
Collapse
Affiliation(s)
- Yue Wang
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Nan Jiang
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Shuhan Wu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Guangshuai Lin
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Wudang Lu
- Xi'an Libang Pharmaceutical Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Bin Shang
- Xi'an Libang Pharmaceutical Co., Ltd, Xi'an, Shaanxi 710075, China
| | - Xulong Zhu
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Wei Han
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China
| | - Jianhui Li
- Department of Surgical Oncology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, China; Department of Surgical Oncology, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710068, China.
| | - Yongmei Chen
- College of Bioresources Chemical and Materials Engineering, National Demonstration Center for Experimental Light Chemistry Engineering Education, Shaanxi University of Science & Technology, Xi'an, Shaanxi 710021, China.
| |
Collapse
|
2
|
Guo L, Fu Z, Li H, Wei R, Guo J, Wang H, Qi J. Smart hydrogel: A new platform for cancer therapy. Adv Colloid Interface Sci 2025; 340:103470. [PMID: 40086017 DOI: 10.1016/j.cis.2025.103470] [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: 07/25/2024] [Revised: 01/17/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Cancer is a significant contributor to mortality worldwide, posing a significant threat to human life and health. The unique bioactivity, ability to precisely control drug release, and minimally invasive properties of hydrogels are indispensable attributes that facilitate optimal performance in cancer therapy. However, conventional hydrogels lack the ability to dynamically respond to changes in the surrounding environment, withstand drastic changes in the microenvironment, and trigger drug release on demand. Therefore, this review focuses on smart-responsive hydrogels that are capable of adapting and responding to external stimuli. We comprehensively summarize the raw materials, preparation, and cross-linking mechanisms of smart hydrogels derived from natural and synthetic materials, elucidate the response principles of various smart-responsive hydrogels according to different stimulation sources. Further, we systematically illustrate the important role played by hydrogels in modern cancer therapies within the context of therapeutic principles. Meanwhile, the smart hydrogel that uses machine learning to design precise drug delivery has shown great prospects in cancer therapy. Finally, we present the outlook on future developments and make suggestions for future related work. It is anticipated that this review will promote the practical application of smart hydrogels in cancer therapy and contribute to the advancement of medical treatment.
Collapse
Affiliation(s)
- Li Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ziming Fu
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Haoran Li
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Ruibo Wei
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China
| | - Jing Guo
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Haiwang Wang
- Key Laboratory of Dielectric and Electrolyte Functional Material Hebei Province, Northeastern University at Qinhuangdao, Qinhuangdao 066004, China.
| | - Jian Qi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Wen Q, Li J, Deng H, Wang B, Huang J, Dai J, Lu Y, Zeng F, Chen Y, Zhao L, Fu S. Hyaluronic acid modified metal-organic frameworks loading cisplatin achieve combined chemodynamic therapy and chemotherapy for lung cancer. Int J Biol Macromol 2025; 300:140238. [PMID: 39863194 DOI: 10.1016/j.ijbiomac.2025.140238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
As one of the most commonly used chemotherapeutic agents in clinical practice, cisplatin is unable to selectively accumulate in tumor tissue due to its lack of targeting ability, leading to increased systemic toxicities. Additionally, the effectiveness of monotherapy is greatly limited. Therefore, the development of new cisplatin-based drug delivery systems is essential to improve the effectiveness of tumor treatment. In this study, an iron-based metal-organic framework (MOF) was synthesized to encapsulate cisplatin, and then coated with hyaluronic acid (HA) to create a MOF-based nanoplatform called MPt@HA NPs. This novel nanoplatform achieved the combination of chemodynamic therapy (CDT) with targeted chemotherapy for the treatment of lung cancer. The results showed that MPt@HA NPs have stronger cytotoxicity compared to conventional doses of cisplatin due to the generation of reactive oxygen species (ROS) through the Fenton reaction and DNA damage caused by cisplatin. Therefore, MPt@HA NPs effectively inhibit the tumor growth and prolong the median survival of tumor-bearing mice. Therefore, the MOF-based nanoplatform MPt@HA NPs may present a new option for multi-modal therapy of solid tumors.
Collapse
Affiliation(s)
- Qian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Jianmei Li
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Hongjun Deng
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Biqiong Wang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Jingrong Huang
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Jie Dai
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Yun Lu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Yue Chen
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, PR China.
| | - Shaozhi Fu
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
4
|
Raj M, Meena A, Seth R, Mathur A, Luqman S. An update on nanoformulations with FDA approved drugs for female reproductive cancer. J Microencapsul 2025:1-34. [PMID: 40114400 DOI: 10.1080/02652048.2025.2474457] [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: 10/15/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Female reproductive cancers, including ovarian, cervical, breast, gestational trophoblastic and endometrial cancer, present significant challenges in therapy and patient prognosis. Conventional chemotherapy often lacks selectivity, leading to systemic toxicity and reduced treatment efficacy. Nanotechnology has emerged as a promising approach to improve drug delivery and therapeutic outcomes. Encapsulation of FDA-approved drugs within nanocarriers such as liposomes, polymeric nanoparticles, and lipid nanoparticles enables controlled drug release, reduces off-target effects, and enhances drug accumulation at tumor sites. This targeted delivery minimizes damage to healthy tissues and improves patient survival rates. Additionally, nanoformulations facilitate combination therapy, overcoming drug resistance and maximizing therapeutic efficacy. Despite promising results, challenges like scalability, reproducibility, and regulatory approvals hinder widespread clinical applications. Developing personalized nanoformulations tailored to individual patient profiles offers potential for precision cancer therapy. This study explores the role of nanoformulations in enhancing the therapeutic potential of FDA-approved drugs for treating female reproductive cancers.
Collapse
Affiliation(s)
- Mahima Raj
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Richa Seth
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anurag Mathur
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
5
|
Liu X, Wang Y, Wu H, Wang D, Yao H, Ren Z, Cao Y, Cong H, Yu B. Natural polysaccharide hydrogel delivery system remodeling tumor microenvironment to promote postoperative tumor therapy. Int J Biol Macromol 2025; 291:139137. [PMID: 39725109 DOI: 10.1016/j.ijbiomac.2024.139137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
In recent years, postoperative tumor therapy with a suitable approach has been an important issue. Remodeling the tumor microenvironment and accelerating tissue repair can accelerate patients' surgical site recovery, reduce patient pain as well as prevent postoperative tumor recurrence. The shape non-adaptability, cytotoxicity, and non-degradability of some hydrogels still hinder the application of hydrogel-based drug delivery systems in postoperative recovery. Natural polysaccharides (e.g., chitosan, sodium alginate, and hyaluronic acid) are multifunctional compounds with biomimetic advantages to meet the growing demand for nontoxic, targeted therapeutic, and restorative preventive therapies. In this paper, we comprehensively and systematically investigated the synthesis methods, properties, and applications of natural polysaccharide hydrogel (NPH) delivery systems, as well as the mechanisms of remodeling the tumor microenvironment. We aim to provide insights into the design of NPH delivery systems. On this basis, future research directions for NPH delivery systems and their role in remodeling the tumor microenvironment and accelerating postoperative tumor therapy are proposed, and strategies for remodeling the tumor microenvironment using hydrogel delivery systems are discussed, as well as the latest research methods.
Collapse
Affiliation(s)
- Xin Liu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yumei Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Han Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Dayang Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Huanchen Yao
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Zekai Ren
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Yang Cao
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
6
|
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
|
7
|
Gao H, Li H, Shao S, Tan L, Wang Y, Li D, Zhang W, Zhu T, Liu G, Meng X. Self-enhanced PTX@HSA-HA loaded functionalized injectable hydrogel for effective local chemo-photothermal therapy in breast cancer. Carbohydr Polym 2024; 345:122569. [PMID: 39227105 DOI: 10.1016/j.carbpol.2024.122569] [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: 04/06/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024]
Abstract
Breast cancer is a malignant tumor that poses a significant threat to women's health and single therapy fails to play a good oncological therapeutic effect. Synergistic treatment with multiple strategies may make up for the deficiencies and has gained widespread attention. In this study, sulfhydryl-modified hyaluronic acid (HA-SH) was covalently crosslinked with polydopamine (PDA) via a Michael addition reaction to develop an injectable hydrogel, in which PDA can be used not only as a matrix but also as a photothermal agent. After HSA and paclitaxel were spontaneously organized into nanoparticles via hydrophobic interaction, hyaluronic acid with low molecular weight was covalently linked to HSA, thus conferring effectively delivery. This photothermal injectable hydrogel incorporates PTX@HSA-HA nanoparticles, thereby initiating a thermochemotherapeutic response to target malignancy. Our results demonstrated that this injectable hydrogel possesses consistent drug delivery capability in a murine breast cancer model, collaborating with photothermal therapy to effectively suppress tumor growth, represented by low expression of Ki-67 and increasing apoptosis. Photothermal therapy (PTT) can effectively stimulate immune response by increasing IL-6 and TNF-α. Notably, the treatment did not elicit any indications of toxicity. This injectable hydrogel holds significant promise as a multifaceted therapeutic agent that integrates photothermal and chemotherapeutic modalities.
Collapse
Affiliation(s)
- Hang Gao
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hang Li
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuaiqiang Shao
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Lintongqing Tan
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yudie Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Dawei Li
- Shandong Academy of Pharmaceutical Sciences, Shandong Engineering Research Center of New Sustained and Controlled Release Formulations and Drug Targeted Delivery Systems, Jinan 250101, China
| | - Wen Zhang
- Shandong Academy of Pharmaceutical Sciences, Shandong Engineering Research Center of New Sustained and Controlled Release Formulations and Drug Targeted Delivery Systems, Jinan 250101, China
| | - Tao Zhu
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; CanSino Biologics Inc., Tianjin Enterprise Key Laboratory of Respiratory Bacterial Recombination and Conjugated Vaccine, Tianjin 300457, China
| | - Guangchun Liu
- Jecho Biopharmaceuticals Co., Ltd, Tianjin 300467, China
| | - Xin Meng
- Key Laboratory of Industrial Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Shandong Academy of Pharmaceutical Sciences, Shandong Engineering Research Center of New Sustained and Controlled Release Formulations and Drug Targeted Delivery Systems, Jinan 250101, China.
| |
Collapse
|
8
|
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
|
9
|
Peng N, Du Y, Liu J, Li D, Li Y, Deng K, Li L, Jia P, Min J, Lin J. Injectable Polydopamine Nanoparticle-Incorporated Hydrogels for Antiangiogenesis and Stimulating Tumoricidal Immunity to Inhibit Metastasis and Recurrence Postresection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64447-64462. [PMID: 39540317 DOI: 10.1021/acsami.4c10363] [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: 11/16/2024]
Abstract
Surgical resection is still the main means for clinical treatments of breast cancer, but the postoperative immunosuppressive microenvironment and neoangiogenesis of the residual tumors easily lead to tumor metastasis and recurrence, which will further endanger patients' lives. The combination of antiangiogenic therapy and immunotherapy may promote the mutually reinforced cycle of immune reprogramming and vascular normalization to avoid tumor metastasis and recurrence. Herein, we prepared polydopamine nanoparticles for improving tissue adhesion and enriching tumor-associated antigens. This nanoregulator together with regorafenib (REG) was further incorporated into a hydrogel developed from grafting adipic acid dihydrazide onto 2,2,6,6-tetramethylpiperidine-1-oxyl radical oxidized chitin and oxidized hyaluronic acid, which was injectable at the cavity after subcutaneous tumor surgery with good mechanical properties and degradability. The system showed long-term release of REG. After combining with anti-PD-L1, the hydrogel applied to the surgical wound exhibited a reduction in tumor metastasis and recurrence. This effect was achieved by suppressing angiogenesis and enhancing antitumor immunity, characterized by increased levels of effector T lymphocytes and activation of dendritic cells within tumors, spleens, and draining lymph nodes. The injectable hydrogel offers a promising strategy for postoperative management aimed at preventing tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Na Peng
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Yijing Du
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Jinhong Liu
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Danyang Li
- College of Textiles and Clothing, Yancheng Institute of Technology, Yancheng 224051, Jiangsu, P. R. China
| | - Yonghao Li
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, P. R. China
| | - Lewei Li
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Peizhi Jia
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Juan Min
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, P. R. China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| |
Collapse
|
10
|
Nam M, Lee JW, Cha GD. Biomedical Application of Enzymatically Crosslinked Injectable Hydrogels. Gels 2024; 10:640. [PMID: 39451293 PMCID: PMC11507637 DOI: 10.3390/gels10100640] [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: 09/10/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant interest in the biomedical field owing to their tissue-like properties and capability to incorporate various fillers. Among these, injectable hydrogels have been highlighted for their unique advantages, especially their minimally invasive administration mode for implantable use. These injectable hydrogels can be utilized in their pristine forms or as composites by integrating them with therapeutic filler materials. Given their primary application in implantable platforms, enzymatically crosslinked injectable hydrogels have been actively explored due to their excellent biocompatibility and easily controllable mechanical properties for the desired use. This review introduces the crosslinking mechanisms of such hydrogels, focusing on those mediated by horseradish peroxidase (HRP), transglutaminase (TG), and tyrosinase. Furthermore, several parameters and their relationships with the intrinsic properties of hydrogels are investigated. Subsequently, the representative biomedical applications of enzymatically crosslinked-injectable hydrogels are presented, including those for wound healing, preventing post-operative adhesion (POA), and hemostasis. Furthermore, hydrogel composites containing filler materials, such as therapeutic cells, proteins, and drugs, are analyzed. In conclusion, we examine the scientific challenges and directions for future developments in the field of enzymatically crosslinked-injectable hydrogels, focusing on material selection, intrinsic properties, and filler integration.
Collapse
Affiliation(s)
| | | | - Gi Doo Cha
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea; (M.N.); (J.W.L.)
| |
Collapse
|
11
|
Zang C, Tian Y, Tang Y, Tang M, Yang D, Chen F, Ghaffarlou M, Tu Y, Ashrafizadeh M, Li Y. Hydrogel-based platforms for site-specific doxorubicin release in cancer therapy. J Transl Med 2024; 22:879. [PMID: 39350207 PMCID: PMC11440768 DOI: 10.1186/s12967-024-05490-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/05/2024] [Indexed: 10/04/2024] Open
Abstract
Hydrogels are promising candidates for the delivery of therapeutics in the treatment of human cancers. Regarding to the biocomaptiiblity, high drug and encapsulation efficacy and adjustable physico-chemical features, the hydrogels have been widely utilized for the delivery of chemotherapy drugs. Doxorubicin (DOX) is one of the most common chemotherapy drugs used in cancer therapy through impairing topoisomerase II function and increasing oxidative damage. However, the tumor cells have developed resistance into DOX-mediated cytotoxic impacts, requiring the delivery systems to increase internalization and anti-cancer activity of this drug. The hydrogels can deliver DOX in a sustained manner to maximize its anti-cancer activity, improving cancer elimination and reduction in side effects and drug resistance. The natural-based hydrogels such as chitosan, alginate and gelatin hydrogels have shown favourable biocompatibility and degradability in DOX delivery for tumor suppression. The hydrogels are able to co-deliver DOX with other drugs or genes to enhance drug sensitivity and mediate polychemotherapy, synergistically suppressing cancer progression. The incorporation of nanoparticles in the structure of hydrogels can improve the sustained release of DOX and enhancing intracellular internalization, accelerating DOX's cytotoxicity. Furthermore, the stimuli-responsive hydrogels including pH-, redox- and thermo-sensitive platforms are able to improve the specific release of DOX at the tumor site. The DOX-loaded hydrogels can be further employed in the clinic for the treatment of cancer patients and improving efficacy of chemotherapy.
Collapse
Affiliation(s)
- Chunbao Zang
- Department of Radiation Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Yu Tian
- Research Center, The Huizhou Central People's Hospital, Guangdong Medical University, No. 41 Eling North Road, Huizhou, Guangdong, China
- School of Public Health, Benedictine University, Lisle, USA
| | - Yujing Tang
- Department of General Surgery, Southwest Jiaotong University Affiliated Chengdu Third People's Hospital, Chengdu, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Dingyi Yang
- Department of Radiation Oncology, Chonging University Cancer Hospital; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Fangfang Chen
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Mohammadreza Ghaffarlou
- Bioengineering Division, Institute of Science and Engineering, Hacettepe University, Ankara, 06800, Turkey
| | - Yanyang Tu
- Research Center, The Huizhou Central People's Hospital, Guangdong Medical University, No. 41 Eling North Road, Huizhou, Guangdong, China.
| | - Milad Ashrafizadeh
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, PR China.
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China.
| | - Yan Li
- Department of Gastrointestinal Surgery, Changzhou Cancer Hospital, No.1 Huaide North Road, Changzhou, Chin, China.
| |
Collapse
|
12
|
Li Z, Zhan J, Zheng Y, Luo Y, Yu X, Chen H. Regulation of tumor antigens-Dependent immunotherapy via the hybrid M1 macrophage/tumor lysates Hydrogel. Heliyon 2024; 10:e37521. [PMID: 39309839 PMCID: PMC11414488 DOI: 10.1016/j.heliyon.2024.e37521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/25/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Tumor treatment poses a significant obstacle in contemporary healthcare. Using components derived from a patient's own cellular and tissue materials to prepare hydrogels and other therapeutic systems has become a novel therapeutic approach, drawing considerable interest for their applicability in basic research on cancer immunotherapy. These hydrogels can engage with cellular components directly and offer a supportive scaffold, aiding in the normalization of tumor tissues. Additionally, their superior capability for encapsulating targeted anti-tumor medications amplifies treatment effectiveness. Given their origin from a patient's own cells, these hydrogels circumvent the risks of immune rejection by the body and severe side effects typically associated with foreign substance. In this study, we developed a composite hydrogel constructed by the cellular lysates of autologous tumor cells and M1 macrophages. This combination promoted the M2 macrophages polarization to the M1 phenotype. Subsequently, the polarized M1 macrophages infiltrated into the hydrogel and can directly capture tumor antigens. As antigen-presenting cells, M1 macrophages can stimulate the production of antigen-specific T cells to kill tumor cells. This work proposes a dual-benefit research strategy that not only polarizes M2 macrophages but also enhances immune activation, boosting T cell-mediated tumor-killing effects. This approach offers a new therapeutic option for clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Zeyang Li
- Department of Ultrasonic Imaging, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jiani Zhan
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yinuo Zheng
- Department of Thyroid and Breast Surgery, Oncological Surgery, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, Zhejiang, China
| | - Yingli Luo
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Xiaoming Yu
- Cancer Center, Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Haha Chen
- Department of Thyroid and Breast Surgery, Oncological Surgery, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, Zhejiang, China
| |
Collapse
|
13
|
Xu K, Zhang Y, Cheng H, Chen W, Chen C, Zhang M, Song H, Wang F. Triple-negative breast cancer treatment with core-shell Magnetic@Platinium-Metal organic framework/epirubicin nano-platforms for chemo-photodynamic based combinational therapy: A review. Medicine (Baltimore) 2024; 103:e39845. [PMID: 39331917 PMCID: PMC11441927 DOI: 10.1097/md.0000000000039845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024] Open
Abstract
The combination of chemotherapy and photodynamic therapy (PDT), enabled by core-shell nano-platforms, is a promising method to improve cancer therapy by overcoming hypoxia and boosting drug penetration in breast tumor. Core-shell magnetic (iron oxide: Fe3O4)@platinum-metal organic framework/epirubicin (abbreviated as M@Pt-MOF/EPI) nano-platform is considered an effective cancer therapeutic agent. Relatively small particle size, round shape, and specific response to pH, are the key features of these nanomaterials to be used as promising therapeutic agents. Chemotherapy and photodynamic therapy, when applied in addition to the anticancer effects of nanomaterials, further enhance the therapeutic efficacy. The extensive use, utilization, and efficacy of Core-Shell Magnetic@Platinium-Metal Organic Framework/epirubicin Nano-Platforms for chemo-photodynamic combination therapy in the treatment of several cancers, including triple-negative breast cancer, are examined in this in-depth investigation.
Collapse
Affiliation(s)
- Kangjie Xu
- Department of Central Laboratory, Binhai County People’s Hospital, Yancheng, China
| | - Yanhua Zhang
- Department of Obstetrics and Gynecology, Binhai County People’s Hospital, Yancheng, China
| | - Hui Cheng
- Department of General Surgery, Binhai County People’s Hospital, Yancheng, China
| | - Weipeng Chen
- Department of General Surgery, Binhai County People’s Hospital, Yancheng, China
| | - Cheng Chen
- Department of General Surgery, Binhai County People’s Hospital, Yancheng, China
| | - Minglei Zhang
- Department of Oncology, Binhai County People’s Hospital, Yancheng, China
| | - He Song
- Department of Rehabilitation Medicine, Kanda College of Nanjing Medical University, Lianyungang, China
| | - Feng Wang
- Department of General Surgery, Binhai County People’s Hospital, Yancheng, China
| |
Collapse
|
14
|
Chen X, Jiang T, Li Y, Zhang Y, Chen J, Zhao X, Yang H. Carrageenan-ferrocene-eicosapentaenoic acid composite hydrogel induce ferroptosis and apoptosis for anti-tumor recurrence and metastasis. Int J Biol Macromol 2024; 276:133942. [PMID: 39025181 DOI: 10.1016/j.ijbiomac.2024.133942] [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: 05/19/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
The immune-suppressive microenvironment of solid tumors is a key factor limiting the effectiveness of immunotherapy, which seriously threatens human life and health. Ferroptosis and apoptosis are key cell-death pathways implicated in cancers, which can synergistically activate tumor immune responses. Here, we developed a multifunctional composite hydrogel (CE-Fc-Gel) based on the self-assembly of poloxamer 407, cystamine-linked ιota-carrageenan (CA)-eicosapentaenoic acid (EPA), and ferrocene (Fc). CE-Fc-Gel improved targeting in tumor microenvironment due to its disulfide bonds. Moreover, CE-Fc-Gel promoted lipid peroxidation, enhanced reactive oxygen species (ROS) production, and decreased glutathione peroxidase 4 (GPX4), inducing ferroptosis by the synergistic effect of Fc and EPA. CE-Fc-Gel induced apoptosis and immunogenic cell death (ICD), thereby promoting dendritic cells (DCs) maturation and T cell infiltration. As a result, CE-Fc-Gel significantly inhibited primary and metastatic tumors in vivo. Our findings provide a novel strategy for enhancing tumor immunotherapy by combining apoptosis, ferroptosis, and ICD.
Collapse
Affiliation(s)
- Xiangyan Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China; State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Tianze Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Yantao Li
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yifei Zhang
- State Key Laboratory Base of Eco-Chemical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jianqi Chen
- Department of Pharmacy, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266042, China; Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Xia Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.
| | - Hai Yang
- Department of Pharmacy, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266042, China.
| |
Collapse
|
15
|
Singhal R, Sarangi MK, Rath G. Injectable Hydrogels: A Paradigm Tailored with Design, Characterization, and Multifaceted Approaches. Macromol Biosci 2024; 24:e2400049. [PMID: 38577905 DOI: 10.1002/mabi.202400049] [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: 02/05/2024] [Revised: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Biomaterials denoting self-healing and versatile structural integrity are highly curious in the biomedicine segment. The injectable and/or printable 3D printing technology is explored in a few decades back, which can alter their dimensions temporarily under shear stress, showing potential healing/recovery tendency with patient-specific intervention toward the development of personalized medicine. Thus, self-healing injectable hydrogels (IHs) are stunning toward developing a paradigm for tissue regeneration. This review comprises the designing of IHs, rheological characterization and stability, several benchmark consequences for self-healing IHs, their translation into tissue regeneration of specific types, applications of IHs in biomedical such as anticancer and immunomodulation, wound healing and tissue/bone regeneration, antimicrobial potentials, drugs, gene and vaccine delivery, ocular delivery, 3D printing, cosmeceuticals, and photothermal therapy as well as in other allied avenues like agriculture, aerospace, electronic/electrical industries, coating approaches, patents associated with therapeutic/nontherapeutic avenues, and numerous futuristic challenges and solutions.
Collapse
Affiliation(s)
- Rishika Singhal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Manoj Kumar Sarangi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, 751030, India
| |
Collapse
|
16
|
Patra P, Upadhyay TK, Alshammari N, Saeed M, Kesari KK. Alginate-Chitosan Biodegradable and Biocompatible Based Hydrogel for Breast Cancer Immunotherapy and Diagnosis: A Comprehensive Review. ACS APPLIED BIO MATERIALS 2024; 7:3515-3534. [PMID: 38787337 PMCID: PMC11190989 DOI: 10.1021/acsabm.3c00984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 05/25/2024]
Abstract
Breast cancer is the most common type of cancer and the second leading cause of cancer-related mortality in females. There are many side effects due to chemotherapy and traditional surgery, like fatigue, loss of appetite, skin irritation, and drug resistance to cancer cells. Immunotherapy has become a hopeful approach toward cancer treatment, generating long-lasting immune responses in malignant tumor patients. Recently, hydrogel has received more attention toward cancer therapy due to its specific characteristics, such as decreased toxicity, fewer side effects, and better biocompatibility drug delivery to the particular tumor location. Researchers globally reported various investigations on hydrogel research for tumor diagnosis. The hydrogel-based multilayer platform with controlled nanostructure has received more attention for its antitumor effect. Chitosan and alginate play a leading role in the formation of the cross-link in a hydrogel. Also, they help in the stability of the hydrogel. This review discusses the properties, preparation, biocompatibility, and bioavailability of various research and clinical approaches of the multipolymer hydrogel made of alginate and chitosan for breast cancer treatment. With a focus on cases of breast cancer and the recovery rate, there is a need to find out the role of hydrogel in drug delivery for breast cancer treatment.
Collapse
Affiliation(s)
- Pratikshya Patra
- Department
of Biotechnology, Parul Institute of Applied Sciences and Animal Cell
Culture and Immunobiochemistry Lab, Research and Development Cell, Parul University, Vadodara, Gujarat 391760, India
| | - Tarun Kumar Upadhyay
- Department
of Biotechnology, Parul Institute of Applied Sciences and Animal Cell
Culture and Immunobiochemistry Lab, Research and Development Cell, Parul University, Vadodara, Gujarat 391760, India
| | - Nawaf Alshammari
- Department
of Biology, College of Science, University
of Hail, Hail 53962, Saudi Arabia
| | - Mohd Saeed
- Department
of Biology, College of Science, University
of Hail, Hail 53962, Saudi Arabia
| | - Kavindra Kumar Kesari
- Department
of Applied Physics, School of Science, Aalto
University, Espoo FI-00076, Finland
- Centre
of Research Impact and Outcome, Chitkara
University, Rajpura 140417, Punjab, India
| |
Collapse
|
17
|
Xiao M, Wang L, Tang Q, Yang Q, Yang X, Zhu G, Lei L, Li S. Postoperative tumor treatment strategies: From basic research to clinical therapy. VIEW 2024; 5. [DOI: 10.1002/viw.20230117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/15/2024] [Indexed: 07/04/2024] Open
Abstract
AbstractDespite progression in advanced treatments for malignant tumors, surgery remains the primary treatment intervention, which removes a large portion of firm tumor tissues; however, the postoperative phase poses a possible risk for provincial tumor recurrence and metastasis. Consequently, the prevention of tumor recurrence and metastasis has attracted research attention. In this review, we summarized the postoperative treatment strategies for various tumors from both basic research and clinical perspectives. We delineated the underlying factors contributing to the recurrence of malignant tumors with a substantial prevalence rate, related molecular mechanisms of tumor recurrence post‐surgery, and related means of monitoring recurrence and metastasis after surgery. Furthermore, we described relevant therapeutic approaches for postoperative tumor recurrence, including chemotherapy, radiation therapy, immunotherapy, targeted therapy, and photodynamic therapy. This review focused on the emerging technologies used for postoperative tumor treatment in recent years in terms of functional classification, including the prevention of postoperative tumor recurrence, functional reconstruction, and monitoring of recurrence. Finally, we discussed the future development and deficiencies of postoperative tumor therapy. To understand postoperative treatment strategies for tumors from clinical treatment and basic research and further guide the research directions for postoperative tumors.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Lin Wang
- Department of Otorhinolaryngology Head and Neck Surgery Binzhou People's Hospital Binzhou China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| | - Lanjie Lei
- Institute of Translational Medicine Zhejiang Shuren University Hangzhou China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery The Second Xiangya Hospital Central South University Changsha China
| |
Collapse
|
18
|
Huang S, Zhou C, Song C, Zhu X, Miao M, Li C, Duan S, Hu Y. In situ injectable hydrogel encapsulating Mn/NO-based immune nano-activator for prevention of postoperative tumor recurrence. Asian J Pharm Sci 2024; 19:100901. [PMID: 38645467 PMCID: PMC11031726 DOI: 10.1016/j.ajps.2024.100901] [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: 07/10/2023] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 04/23/2024] Open
Abstract
Postoperative tumor recurrence remains a predominant cause of treatment failure. In this study, we developed an in situ injectable hydrogel, termed MPB-NO@DOX + ATRA gel, which was locally formed within the tumor resection cavity. The MPB-NO@DOX + ATRA gel was fabricated by mixing a thrombin solution, a fibrinogen solution containing all-trans retinoic acid (ATRA), and a Mn/NO-based immune nano-activator termed MPB-NO@DOX. ATRA promoted the differentiation of cancer stem cells, inhibited cancer cell migration, and affected the polarization of tumor-associated macrophages. The outer MnO2 shell disintegrated due to its reaction with glutathione and hydrogen peroxide in the cytoplasm to release Mn2+ and produce O2, resulting in the release of doxorubicin (DOX). The released DOX entered the nucleus and destroyed DNA, and the fragmented DNA cooperated with Mn2+ to activate the cGAS-STING pathway and stimulate an anti-tumor immune response. In addition, when MPB-NO@DOX was exposed to 808 nm laser irradiation, the Fe-NO bond was broken to release NO, which downregulated the expression of PD-L1 on the surface of tumor cells and reversed the immunosuppressive tumor microenvironment. In conclusion, the MPB-NO@DOX + ATRA gel exhibited excellent anti-tumor efficacy. The results of this study demonstrated the great potential of in situ injectable hydrogels in preventing postoperative tumor recurrence.
Collapse
Affiliation(s)
- Shengnan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Chenyang Zhou
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Chengzhi Song
- Center for Quantitative Biology, Peking University, Beijing 100871, China
| | - Xiali Zhu
- School of Pharmaceutical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Chunming Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Shaofeng Duan
- School of Pharmaceutical Sciences, Henan University, Zhengzhou 450046, China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
19
|
Olteanu G, Neacșu SM, Joița FA, Musuc AM, Lupu EC, Ioniță-Mîndrican CB, Lupuliasa D, Mititelu M. Advancements in Regenerative Hydrogels in Skin Wound Treatment: A Comprehensive Review. Int J Mol Sci 2024; 25:3849. [PMID: 38612660 PMCID: PMC11012090 DOI: 10.3390/ijms25073849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
This state-of-the-art review explores the emerging field of regenerative hydrogels and their profound impact on the treatment of skin wounds. Regenerative hydrogels, composed mainly of water-absorbing polymers, have garnered attention in wound healing, particularly for skin wounds. Their unique properties make them well suited for tissue regeneration. Notable benefits include excellent water retention, creating a crucially moist wound environment for optimal healing, and facilitating cell migration, and proliferation. Biocompatibility is a key feature, minimizing adverse reactions and promoting the natural healing process. Acting as a supportive scaffold for cell growth, hydrogels mimic the extracellular matrix, aiding the attachment and proliferation of cells like fibroblasts and keratinocytes. Engineered for controlled drug release, hydrogels enhance wound healing by promoting angiogenesis, reducing inflammation, and preventing infection. The demonstrated acceleration of the wound healing process, particularly beneficial for chronic or impaired healing wounds, adds to their appeal. Easy application and conformity to various wound shapes make hydrogels practical, including in irregular or challenging areas. Scar minimization through tissue regeneration is crucial, especially in cosmetic and functional regions. Hydrogels contribute to pain management by creating a protective barrier, reducing friction, and fostering a soothing environment. Some hydrogels, with inherent antimicrobial properties, aid in infection prevention, which is a crucial aspect of successful wound healing. Their flexibility and ability to conform to wound contours ensure optimal tissue contact, enhancing overall treatment effectiveness. In summary, regenerative hydrogels present a promising approach for improving skin wound healing outcomes across diverse clinical scenarios. This review provides a comprehensive analysis of the benefits, mechanisms, and challenges associated with the use of regenerative hydrogels in the treatment of skin wounds. In this review, the authors likely delve into the application of rational design principles to enhance the efficacy and performance of hydrogels in promoting wound healing. Through an exploration of various methodologies and approaches, this paper is poised to highlight how these principles have been instrumental in refining the design of hydrogels, potentially revolutionizing their therapeutic potential in addressing skin wounds. By synthesizing current knowledge and highlighting potential avenues for future research, this review aims to contribute to the advancement of regenerative medicine and ultimately improve clinical outcomes for patients with skin wounds.
Collapse
Affiliation(s)
- Gabriel Olteanu
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania; (G.O.); (M.M.)
| | - Sorinel Marius Neacșu
- Department of Pharmaceutical Technology and Bio-Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania; (S.M.N.); (D.L.)
| | - Florin Alexandru Joița
- Department of Pharmaceutical Technology and Bio-Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania; (S.M.N.); (D.L.)
| | | | - Elena Carmen Lupu
- Department of Mathematics and Informatics, Faculty of Pharmacy, “Ovidius” University of Constanta, 900001 Constanta, Romania;
| | - Corina-Bianca Ioniță-Mîndrican
- Department of Toxicology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania;
| | - Dumitru Lupuliasa
- Department of Pharmaceutical Technology and Bio-Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania; (S.M.N.); (D.L.)
| | - Magdalena Mititelu
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania; (G.O.); (M.M.)
| |
Collapse
|
20
|
Mozafari N, Jahanbekam S, Ashrafi H, Shahbazi MA, Azadi A. Recent Biomaterial-Assisted Approaches for Immunotherapeutic Inhibition of Cancer Recurrence. ACS Biomater Sci Eng 2024; 10:1207-1234. [PMID: 38416058 DOI: 10.1021/acsbiomaterials.3c01347] [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] [Indexed: 02/29/2024]
Abstract
Biomaterials possess distinctive properties, notably their ability to encapsulate active biological products while providing biocompatible support. The immune system plays a vital role in preventing cancer recurrence, and there is considerable demand for an effective strategy to prevent cancer recurrence, necessitating effective strategies to address this concern. This review elucidates crucial cellular signaling pathways in cancer recurrence. Furthermore, it underscores the potential of biomaterial-based tools in averting or inhibiting cancer recurrence by modulating the immune system. Diverse biomaterials, including hydrogels, particles, films, microneedles, etc., exhibit promising capabilities in mitigating cancer recurrence. These materials are compelling candidates for cancer immunotherapy, offering in situ immunostimulatory activity through transdermal, implantable, and injectable devices. They function by reshaping the tumor microenvironment and impeding tumor growth by reducing immunosuppression. Biomaterials facilitate alterations in biodistribution, release kinetics, and colocalization of immunostimulatory agents, enhancing the safety and efficacy of therapy. Additionally, how the method addresses the limitations of other therapeutic approaches is discussed.
Collapse
Affiliation(s)
- Negin Mozafari
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Sheida Jahanbekam
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Hajar Ashrafi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Amir Azadi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, 71468 64685 Shiraz, Iran
| |
Collapse
|
21
|
Fang Y, Huang S, Hu Q, Zhang J, King JA, Wang Y, Wei Z, Lu J, He Z, Kong X, Yang X, Ji J, Li J, Zhai G, Ye L. Injectable Zwitterionic Physical Hydrogel with Enhanced Chemodynamic Therapy and Tumor Microenvironment Remodeling Properties for Synergistic Anticancer Therapy. ACS NANO 2023; 17:24883-24900. [PMID: 37883579 DOI: 10.1021/acsnano.3c05898] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Surgical resection is the first-line therapy for breast cancer. However, residual tumor cells and the highly immunosuppressive tumor microenvironment (TME) continue to have a serious impact on tumor recurrence and metastasis postresection. Implantation of an in situ hydrogel system postresection has shown to be an effective treatment with great clinical potential. Herein, an injectable zwitterionic hydrogel system was developed for local drug delivery with enhanced immune activation and prevention of tumor recurrence. Driven by electrostatic interactions, poly(sulfobetaine methacrylate) (PSBMA) self-assembles into a hydrogel in saline, achieving low protein adsorption and tunable biodegradability. The chemotherapy drug doxorubicin (DOX) was loaded into copper peroxide nanoparticles (CuO2/DOX), which were coated with macrophage membranes to form tumor-targeting nanoparticles (M/CuO2/DOX). Next, M/CuO2/DOX and the stimulator of interferon genes (STING) agonist 2',3'-cGAMP were coloaded into PSBMA hydrogel (Gel@M/CuO2/DOX/STING). The hydrophilic STING agonist was first released by diffusion from hydrogel to activate the STING pathway and upregulate interferon (IFN) signaling related genes, remodeling the immunosuppressive TME. Then, M/CuO2/DOX targeted the residual tumor cells, combining with DOX-induced DNA damage, immunogenic tumor cell death, and copper death. Hence, this work combines chemodynamic therapy with STING pathway activation in TME, encouraging residual tumor cell death, promoting the maturation of dendritic cells, enhancing tumor-specific CD8+ T cell infiltration, and preventing postoperative recurrence and metastasis.
Collapse
Affiliation(s)
- Yuelin Fang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Susu Huang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qiaoying Hu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jicheng Zhang
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Julia A King
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Yanqing Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhijian Wei
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong 250012, China
| | - Jinghui Lu
- Department of Hernia and Abdominal Wall Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhijing He
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinru Kong
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaoye Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Junjie Li
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Guangxi Zhai
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Ye
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
22
|
Luo Z, Wang Y, Li J, Wang J, Yu Y, Zhao Y. Tailoring Hyaluronic Acid Hydrogels for Biomedical Applications. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202306554] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Indexed: 05/04/2025]
Abstract
AbstractHyaluronic acid (HA) is an attractive anionic polysaccharide polymer with inherent pharmacological properties and versatile chemical groups for modification. Due to their water retention ability, biocompatibility, biodegradation, cluster of differentiation‐44 targeting, and highly designable capacity, HA hydrogels have been an emerging biomaterial, showing tailoring performance in terms of chemical modifications and hydrogel forms. Various preparation technologies have been developed for the fabrication of the tailoring HA hydrogels with unique structures and functions. They have been utilized in diverse biomedical applications like drug delivery and tissue engineering scaffolds. Herein, this review comprehensively summarizes the HA derivatives with different molecule weights and functional modifications. Then the various fabrication methods to obtain tailoring hydrogels in the forms of nanogel, nanofiber, microparticle, microneedle patch, injectable hydrogel, and scaffold are reviewed as well. The emphasis is focused on the shining biomedical applications of these tailoring HA hydrogels in anti‐bacteria, anti‐inflammation, wound healing, cancer treatment, regenerative medicine, psoriasis treatment, diagnosis, etc. The potentials and prospects are subsequently given to inspire further investigation, aiming at accelerating product translation from research to clinic.
Collapse
Affiliation(s)
- Zhiqiang Luo
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Yu Wang
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Jinbo Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
| | - Jinglin Wang
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
| | - Yunru Yu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology Nanjing Drum Tower Hospital School of Biological Science and Medical Engineering Southeast University Nanjing 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) Wenzhou Institute University of Chinese Academy of Sciences Wenzhou Zhejiang 325001 China
- Southeast University Shenzhen Research Institute Shenzhen 518071 China
| |
Collapse
|
23
|
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
|
24
|
Liu Y, Ma W, Zhou P, Wen Q, Wen Q, Lu Y, Zhao L, Shi H, Dai J, Li J, Fu S. In situ administration of temperature-sensitive hydrogel composite loading paclitaxel microspheres and cisplatin for the treatment of melanoma. Biomed Pharmacother 2023; 160:114380. [PMID: 36774723 DOI: 10.1016/j.biopha.2023.114380] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Chemotherapy is one of the main therapeutic strategies for the treatment of malignant melanoma. Conventional chemotherapeutic agents often lack targeting abilities, and efficacy is hampered by their high toxic effects to normal tissues and rapid clearance from the circulation. In this study, porous paclitaxel (PTX)-loaded polylactide (PLA) microspheres (PPMSs) were prepared by a modified double-emulsion-solvent evaporation method. In addition, PPMSs and cisplatin (DDP) were co-embedded in a thermosensitive hydrogel to construct a dual-drug co-delivery hydrogel system (PPMSs/DDP@Gel) for in-situ chemotherapy to treat melanoma by means of an intra-tumoral injection. The system allows for the sustained release of two drugs and exhibits good temperature-sensitive properties. In vitro antitumor activity showed that this hydrogel composite can induce B16 cell apoptosis and inhibit its migration. In vivo, anti-tumor studies have shown that the PPMSs/DDP@Gel significantly inhibited tumor growth, prolonged the survival of tumor-bearing mice, and had no obvious toxic side effects on major organs. Furthermore, immunohistochemical analysis revealed that PPMSs/DDP@Gel significantly inhibited tumor cell proliferation and promoted apoptosis of tumor cells. Taken together, the injectable temperature-sensitive PPMSs/DDP@Gel is a promising candidate for the local treatment of melanoma.
Collapse
Affiliation(s)
- Yanlin Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Wenqiong Ma
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Ping Zhou
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Qian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Yun Lu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou 646000, PR China
| | - Huan Shi
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Jie Dai
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Jianmei Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| |
Collapse
|
25
|
Mi D, Li J, Wang R, Li Y, Zou L, Sun C, Yan S, Yang H, Zhao M, Shi S. Postsurgical wound management and prevention of triple-negative breast cancer recurrence with a pryoptosis-inducing, photopolymerizable hydrogel. J Control Release 2023; 356:205-218. [PMID: 36870543 DOI: 10.1016/j.jconrel.2023.02.042] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/25/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Surgical removal remains the predominant treatment strategy for triple-negative breast cancer (TNBC). However, risks that include high locoregional recurrence and remote metastasis threaten patient survival and quality of life after surgery. In this study, a hydrogel based on poly (ethylene glycol) dimethacrylate and sericin methacryloyl was fabricated by photopolymerization to fill the resection cavity and prevent recurrence. The obtained hydrogel exhibited mechanical properties compatible with breast tissue and facilitated postsurgical wound management by promoting tissue regeneration. The DNA methylation inhibitor decitabine (DEC) and poly (lactic-co-glycolic acid)-encapsulated phytochemical gambogic acid (GA) were loaded into the hydrogel. The as-prepared hydrogel promoted fast release of DEC and sustained release of GA, leading to gasdermin E-mediated tumor cell pyroptosis and activating antitumor immune responses. Inducing postsurgical tumor cell pyroptosis inhibited local tumor recurrence and lung metastasis. While the dual-drug-loaded hydrogel system cured less than half of tumor-bearing mice, the cured mice survived for over half a year. These findings indicate that our hydrogel system is an excellent biocompatible platform for postsurgical TNBC therapy.
Collapse
Affiliation(s)
- Dandan Mi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiaojiao Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rujing Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lan Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shenao Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huan Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
26
|
Qian Q, Song J, Chen C, Pu Q, Liu X, Wang H. Recent advances in hydrogels for preventing tumor recurrence. Biomater Sci 2023; 11:2678-2692. [PMID: 36877511 DOI: 10.1039/d3bm00003f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Malignant tumors remain a high-risk disease with high mortality all over the world. Among all the cancer treatments, surgery is the primary approach in the clinical treatment of tumors. However, tumor invasion and metastasis pose challenges for complete tumor resection, accompanied by high recurrence rates and reduced quality of life. Hence, there is an urgent need to explore effective adjuvant therapies to prevent postoperative tumor recurrence and relieve the pain of the patients. Nowadays, the booming local drug delivery systems which can be applied as postoperative adjuvant therapies have aroused people's attention, along with the rapid development in the pharmaceutical and biological materials fields. Hydrogels are a kind of unique carrier with prominent biocompatibility among a variety of biomaterials. Due to their high similarity to human tissues, hydrogels which load drugs/growth factors can prevent rejection reactions and promote wound healing. In addition, hydrogels are able to cover the postoperative site and maintain sustained drug release for the prevention of tumor recurrence. In this review, we survey controlled drug delivery hydrogels such as implantable, injectable and sprayable formulations and summarize the properties required for hydrogels used as postoperative adjuvant therapies. The opportunities and challenges in the design and clinical application of these hydrogels are also elaborated.
Collapse
Affiliation(s)
- Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Jie Song
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Chen Chen
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Qian Pu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Xingcheng Liu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
27
|
Luo P, Fang J, Yang D, Yu L, Chen H, Jiang C, Guo R, Zhu T, Tang S. OP3-4 peptide sustained-release hydrogel inhibits osteoclast formation and promotes vascularization to promote bone regeneration in a rat femoral defect model. Bioeng Transl Med 2023; 8:e10414. [PMID: 36925715 PMCID: PMC10013759 DOI: 10.1002/btm2.10414] [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: 03/15/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022] Open
Abstract
Bone injury caused changes to surrounding tissues, leading to a large number of osteoclasts appeared to clear the damaged bone tissue before bone regeneration. However, overactive osteoclasts will inhibit bone formation. In this study, we prepared methacrylylated gelatin (GelMA)-based hydrogel to co-crosslink with OP3-4 peptide, a receptor activator of NF-κB ligand (RANKL) binding agent, to achieve the slow release of OP3-4 peptide to inhibit the activation of osteoclasts, thus preventing the long-term existence of osteoclasts from affecting bone regeneration, and promoting osteogenic differentiation. Moreover, CXCL9 secreted by osteoblasts will bind to endogenous VEGF and inhibit vascularization, finally hinder bone formation. Thus, anti-CXCL9 antibodies (A-CXCL9) were also loaded in the hydrogel to neutralize excess CXCL9. The hydrogel slow released of OP3-4 cyclic peptide and A-CXCL9 to simultaneously inhibiting osteoclast activation and promoting vascularization, thereby accelerating the healing of femur defect. Further analysis of osteogenic protein expression and signal pathways showed that the hydrogel may be through activating the AKT-RUNX2-ALP pathway and ultimately promote osteogenic differentiation. This dual-acting hydrogel can effectively prevent nonunion caused by low vascularization and provide long-term support for the treatment of bone injury.
Collapse
Affiliation(s)
- Peng Luo
- Department of Sport MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenChina
| | - Jiarui Fang
- Department of Sport MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenChina
| | - Dazhi Yang
- Department of Spine SurgeryHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenChina
| | - Lan Yu
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenChina
| | - Houqing Chen
- Department of Sport MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenChina
| | - Changging Jiang
- Department of Sport MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenChina
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical EngineeringJinan UniversityGuangzhouChina
| | - Tao Zhu
- Department of Respiratory and Critical Care Medicine, and Preclinical Research CenterSuining Central HospitalSichuanChina
| | - Shuo Tang
- Department of Orthopaedics, The Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| |
Collapse
|
28
|
Grieco M, Ursini O, Palamà IE, Gigli G, Moroni L, Cortese B. HYDRHA: Hydrogels of hyaluronic acid. New biomedical approaches in cancer, neurodegenerative diseases, and tissue engineering. Mater Today Bio 2022; 17:100453. [PMID: 36254248 PMCID: PMC9568881 DOI: 10.1016/j.mtbio.2022.100453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 10/30/2022] Open
Abstract
In the last decade, hyaluronic acid (HA) has attracted an ever-growing interest in the biomedical engineering field as a biocompatible, biodegradable, and chemically versatile molecule. In fact, HA is a major component of the extracellular matrix (ECM) and is essential for the maintenance of cellular homeostasis and crosstalk. Innovative experimental strategies in vitro and in vivo using three-dimensional (3D) HA systems have been increasingly reported in studies of diseases, replacement of tissue and organ damage, repairing wounds, and encapsulating stem cells for tissue regeneration. The present work aims to give an overview and comparison of recent work carried out on HA systems showing advantages, limitations, and their complementarity, for a comprehensive characterization of their use. A special attention is paid to the use of HA in three important areas: cancer, diseases of the central nervous system (CNS), and tissue regeneration, discussing the most innovative experimental strategies. Finally, perspectives within and beyond these research fields are discussed.
Collapse
Affiliation(s)
- Maddalena Grieco
- National Research Council-Nanotechnology Institute (CNR Nanotec), 73100, Lecce, Italy
| | - Ornella Ursini
- National Research Council-Nanotechnology Institute (CNR Nanotec), 00185, Rome, Italy
| | - Ilaria Elena Palamà
- National Research Council-Nanotechnology Institute (CNR Nanotec), 73100, Lecce, Italy
| | - Giuseppe Gigli
- National Research Council-Nanotechnology Institute (CNR Nanotec), 73100, Lecce, Italy
- Department of Mathematics and Physics “Ennio De Giorgi” University of Salento, Via Arnesano, 73100, Lecce, Italy
| | - Lorenzo Moroni
- National Research Council-Nanotechnology Institute (CNR Nanotec), 73100, Lecce, Italy
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, the Netherlands
| | - Barbara Cortese
- National Research Council-Nanotechnology Institute (CNR Nanotec), 00185, Rome, Italy
| |
Collapse
|
29
|
Tian S, Lu Y, Gao H, Chen Z, Niu M, Wang C, Liu B. Epirubicin may enhance the inhibition of hepatocellular carcinoma induced by iodine‐125 seeds through downregulating WNT pathway. Asia Pac J Clin Oncol 2022; 19:355-364. [DOI: 10.1111/ajco.13873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Shilin Tian
- Department of Interventional MedicineShandong Cancer Hospital and InstituteShandong First Medical UniversityShandong Academy of Medical Sciences JinanChina
| | - Yue Lu
- Cheeloo College of MedicineShandong University JinanChina
| | - Haifeng Gao
- Department of OncologyDongying People's Hospital DongyingChina
| | - Zitong Chen
- Cheeloo College of MedicineShandong University JinanChina
| | - Min Niu
- Department of Gastrointestinal SurgeryTraditional Chinese Medicine Hospital of Jiyang County JinanChina
| | - Changjun Wang
- Department of RadiologyPeople's Hospital of Jiyang County JinanChina
| | - Bin Liu
- Department of Interventional MedicineThe Second Hospital of Shandong University JinanChina
- Interventional Oncology Institute of Shandong University JinanChina
| |
Collapse
|
30
|
Ma Q, Li Q, Cai X, Zhou P, Wu Z, Wang B, Ma W, Fu S. Injectable hydrogels as drug delivery platform for in-situ treatment of malignant tumor. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Li F, Shao X, Liu D, Jiao X, Yang X, Yang W, Liu X. Vascular Disruptive Hydrogel Platform for Enhanced Chemotherapy and Anti-Angiogenesis through Alleviation of Immune Surveillance. Pharmaceutics 2022; 14:1809. [PMID: 36145556 PMCID: PMC9505154 DOI: 10.3390/pharmaceutics14091809] [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: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Patients undergoing immunotherapy always exhibit a low-response rate due to tumor heterogeneity and immune surveillance in the tumor. Angiogenesis plays an important role in affecting the status of tumor-infiltrated lymphocytes by inducing hypoxia and acidosis microenvironment, suggesting its synergistic potential in immunotherapy. However, the antitumor efficacy of singular anti-angiogenesis therapy often suffers from failure in the clinic due to the compensatory pro-angiogenesis signaling pathway. In this work, classic injectable thermosensitive PLGA-PEG-PLGA copolymer was used to construct a platform to co-deliver CA4P (vascular disruptive agent) and EPI for inducing immunogenic cell death of cancer cells by targeting the tumor immune microenvironment. Investigation of 4T1 tumor-bearing mouse models suggests that local administration of injectable V+E@Gel could significantly inhibit the proliferation of cancer cells and prolong the survival rate of 4T1 tumor-bearing mouse models. Histological analysis further indicates that V+E@Gel could effectively inhibit tumor angiogenesis and metastasis by down-regulating the expression of CD34, CD31, MTA1 and TGF-β. Moreover, due to the sustained release kinetics of V+E@Gel, its local administration relieves the immune surveillance in tumor tissues and thus induces a robust and long-lasting specific antitumor immune response. Overall, this work provides a new treatment strategy through the mediation of the tumor immune microenvironment by vascular disruption to fulfill enhanced chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Fasheng Li
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xinmei Shao
- Department of Neurology, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Dehui Liu
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xiaogang Jiao
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xinqi Yang
- Department of Imaging, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Wencai Yang
- Department of Interventional, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| | - Xiaoyan Liu
- Department of Neurology, The Fifth Affiliated Hospital of Jinan University, Jinan University, Heyuan 517000, China
| |
Collapse
|
32
|
Abstract
Biomaterials with the ability to self-heal and recover their structural integrity offer many advantages for applications in biomedicine. The past decade has witnessed the rapid emergence of a new class of self-healing biomaterials commonly termed injectable, or printable in the context of 3D printing. These self-healing injectable biomaterials, mostly hydrogels and other soft condensed matter based on reversible chemistry, are able to temporarily fluidize under shear stress and subsequently recover their original mechanical properties. Self-healing injectable hydrogels offer distinct advantages compared to traditional biomaterials. Most notably, they can be administered in a locally targeted and minimally invasive manner through a narrow syringe without the need for invasive surgery. Their moldability allows for a patient-specific intervention and shows great prospects for personalized medicine. Injected hydrogels can facilitate tissue regeneration in multiple ways owing to their viscoelastic and diffusive nature, ranging from simple mechanical support, spatiotemporally controlled delivery of cells or therapeutics, to local recruitment and modulation of host cells to promote tissue regeneration. Consequently, self-healing injectable hydrogels have been at the forefront of many cutting-edge tissue regeneration strategies. This study provides a critical review of the current state of self-healing injectable hydrogels for tissue regeneration. As key challenges toward further maturation of this exciting research field, we identify (i) the trade-off between the self-healing and injectability of hydrogels vs their physical stability, (ii) the lack of consensus on rheological characterization and quantitative benchmarks for self-healing injectable hydrogels, particularly regarding the capillary flow in syringes, and (iii) practical limitations regarding translation toward therapeutically effective formulations for regeneration of specific tissues. Hence, here we (i) review chemical and physical design strategies for self-healing injectable hydrogels, (ii) provide a practical guide for their rheological analysis, and (iii) showcase their applicability for regeneration of various tissues and 3D printing of complex tissues and organoids.
Collapse
Affiliation(s)
- Pascal Bertsch
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands
| | - Mani Diba
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - David J. Mooney
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - Sander C. G. Leeuwenburgh
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,
| |
Collapse
|
33
|
Guo L, Sun J, Wang C, Wang Y, Wang Y, Li D, Li Y. Epirubicin Enhances the Anti-Cancer Effects of Radioactive 125I Seeds in Hepatocellular Carcinoma via Downregulation of the JAK/STAT1 Pathway. Front Oncol 2022; 12:854023. [PMID: 35692770 PMCID: PMC9184686 DOI: 10.3389/fonc.2022.854023] [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: 01/13/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
The application and promotion of 125I seed implantation technology have increased the safety and effectiveness of the clinical treatment of advanced hepatocellular carcinoma (HCC). Epirubicin (EPI) is a traditional anthracycline chemotherapy agent that has minimal side effects and has been widely used in the clinical treatment of HCC. We hypothesized that EPI would enhance the anti-cancer effects of 125I seeds via the JAK/STAT1 signaling pathway. Thus, we aimed to investigate whether EPI could enhance the radiosensitivity of HCC cells to 125I and determine the underlying molecular mechanism. This basic study was conducted in an animal laboratory at Shandong University. BALB/C male nude mice were used, and all animals were fed and treated according to the standards of the Institutional Animal Care and Use Committee of Shandong University. Both in vitro and in vivo models of 125I irradiation of HCC cells were created. The anti-cancer effects of 125I and the role of EPI in promoting these effects were evaluated using flow cytometry for apoptosis and cell cycle, CCK-8 assay for EPI drug cytotoxicity, and transwell assays for migration and invasion. The potential mediating effect of the JAK/STAT1 pathway was assessed using an isobaric tag for relative and absolute quantitation analysis to identify differentially expressed proteins after 125I treatment. Transfection of HCC cells with STAT1-RNAi were performed to determine the effect of STAT1 downregulation on 125I and EPI treatment effects. The radiosensitivity concentration of EPI promoted 125I-induced anti-cancer effects, including apoptosis, anti-proliferation, and inhibition of migration and invasion. These effects were mediated via the JAK/STAT1 pathway. Downregulation of STAT1 compromised measured anti-cancer effects, which were both confirmed in the in vivo and in vitro models. EPI can promote 125I-induced anti-cancer effects in HCC. The JAK/STAT1 pathway may be a potential target for 125I seed implantation in the treatment of HCC.
Collapse
Affiliation(s)
- Lei Guo
- Department of Vascular Anomalies and Interventional Radiology, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Vascular Anomalies and Interventional Radiology, Jinan Children's Hospital, Jinan, China.,Shandong Provincial Clinical Research Center for Children's Health and Disease, Jinan, China
| | - Jiali Sun
- Department of Vascular Anomalies and Interventional Radiology, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Vascular Anomalies and Interventional Radiology, Jinan Children's Hospital, Jinan, China
| | - Changjun Wang
- Department of Interventional Radiology, Jiyang People's Hospital of Jinan, Jinan, China
| | - Yang Wang
- Department of Interventional Medicine, The Second Hospital, Cheello College of Medicine, Shandong University, Jinan, China.,Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Ya Wang
- Department of Vascular Anomalies and Interventional Radiology, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Vascular Anomalies and Interventional Radiology, Jinan Children's Hospital, Jinan, China
| | - Dong Li
- Department of Interventional Medicine, The Second Hospital, Cheello College of Medicine, Shandong University, Jinan, China.,Institute of Interventional Oncology, Shandong University, Jinan, China
| | - Yuliang Li
- Department of Interventional Medicine, The Second Hospital, Cheello College of Medicine, Shandong University, Jinan, China.,Institute of Interventional Oncology, Shandong University, Jinan, China
| |
Collapse
|
34
|
Zhang H, Zhang M, Zhang X, Gao Y, Ma Y, Chen H, Wan J, Li C, Wang F, Sun X. Enhanced postoperative cancer therapy by iron-based hydrogels. Biomater Res 2022; 26:19. [PMID: 35606838 PMCID: PMC9125885 DOI: 10.1186/s40824-022-00268-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/11/2022] [Indexed: 12/13/2022] Open
Abstract
AbstractSurgical resection is a widely used method for the treatment of solid tumor cancers. However, the inhibition of tumor recurrence and metastasis are the main challenges of postoperative tumor therapy. Traditional intravenous or oral administration have poor chemotherapeutics bioavailability and undesirable systemic toxicity. Polymeric hydrogels with a three-dimensional network structure enable on-site delivery and controlled release of therapeutic drugs with reduced systemic toxicity and have been widely developed for postoperative adjuvant tumor therapy. Among them, because of the simple synthesis, good biocompatibility, biodegradability, injectability, and multifunctionality, iron-based hydrogels have received extensive attention. This review has summarized the general synthesis methods and construction principles of iron-based hydrogels, highlighted the latest progress of iron-based hydrogels in postoperative tumor therapy, including chemotherapy, photothermal therapy, photodynamic therapy, chemo-dynamic therapy, and magnetothermal-chemical combined therapy, etc. In addition, the challenges towards clinical application of iron-based hydrogels have also been discussed. This review is expected to show researchers broad perspectives of novel postoperative tumor therapy strategy and provide new ideas in the design and application of novel iron-based hydrogels to advance this sub field in cancer nanomedicine.
Collapse
|