1
|
Zhang J, Jin K, Feng Y, Lu D, Chen M, Wang H, Jin C, Wang D, Li Z, Wang Y. Injectable Self-Healing and Anti-Dissolving Low-Molecular-Weight Hydrogels Enabled by Ionic Cross-Linking for Cell Encapsulation. ACS Macro Lett 2025; 14:20-25. [PMID: 39690898 DOI: 10.1021/acsmacrolett.4c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Injectable behavior is often observed in polymer-based hydrogels yet is rarely achieved in low-molecular-weight hydrogels (LMWHs), the realization of which may boost the development of new soft materials for biomedical applications. Here, we report on injectable self-healing and antidissolving LMWHs that are formed through a simple ionic cross-linking strategy, showing a fundamental application for the encapsulation of living cells. The LMWHs are formed by simply mixing Ca2+ with negatively charged supramolecular polymers. Surprisingly, the resultant hydrogels are capable of rapidly self-healing within seconds after damage, showing an unexpected injectable function. When the hydrogel is injected into an aqueous medium, continuous macroscopic hydrogel fibers can be produced. Interestingly, the hydrogel can remain intact in the aqueous medium, showing impressive antidissolving behavior which is less observed in other LMWHs. Furthermore, the hydrogel is demonstrated to be nontoxic and can be used as a cytocompatible scaffold for living cells. This work may open an avenue toward injectable and antidissolving LMWHs for the ever-expanding list of applications in biotherapy and bioprinting.
Collapse
Affiliation(s)
- Jiahao Zhang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyu Jin
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yifei Feng
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Da Lu
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Mai Chen
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hucheng Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Jin
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Dengyu Wang
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhiling Li
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yiming Wang
- School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory for Intelligent Sensing and Detection Technology, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
2
|
Xing Y, Hu Y, Wang H, Diao Y, Yue H. An ionic liquid-based delivery system of small interfering RNA targeting Bcl-2 for melanoma therapy. Biomater Sci 2025; 13:466-476. [PMID: 39611237 DOI: 10.1039/d4bm01159g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Melanoma, characterized by rapid tumour progression and a strong tendency to metastasize, poses significant challenges in clinical treatment. Given the vital role of B-cell lymphoma 2 (Bcl-2) protein overexpression in inhibiting apoptosis in tumour cells, the suppression of Bcl-2 has emerged as a promising anticancer therapy. Here, we have developed a straightforward and effective delivery system that combines small interfering RNA (siRNA) targeting Bcl-2 (siBcl-2) with ionic liquids (ILs) for treating melanoma. The unique properties of ILs including structural tunability, inherent charge, and chemical stability have garnered significant attention in the biomedical fields; however, their application in siRNA delivery remains nascent. Rather than the weak function of free siBcl-2, our delivery system (1-hexyl-3-methylimidazolium-siBcl-2, designated as C6-siBcl-2) demonstrated an outstanding capacity to improve the cellular uptake and lysosomal escape, resulting in robust apoptosis and cytotoxicity in melanoma cells. In addition to exhibiting superior gene silencing activity in vitro, such events were also evident in mice bearing melanoma tumours. In particular, this IL-based delivery system showed advantages in suppressing tumour growth, preventing metastasis, and enhancing the survival time of mice with melanoma tumours. Therefore, our study offered a novel and powerful nanoplatform that integrated ILs and RNA interference therapy, presenting new strategies for cancer treatment.
Collapse
Affiliation(s)
- Yuyuan Xing
- Beijing Key Laboratory of Ionic Liquids Clean Process, CAS Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yanhui Hu
- Beijing Key Laboratory of Ionic Liquids Clean Process, CAS Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongyan Wang
- Beijing Key Laboratory of Ionic Liquids Clean Process, CAS Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yanyan Diao
- Beijing Key Laboratory of Ionic Liquids Clean Process, CAS Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- School of Chemical & Environmental Engineering, China University of Mining and Technology (Beijing), Beijing 100083, P. R. China
| | - Hua Yue
- Beijing Key Laboratory of Ionic Liquids Clean Process, CAS Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P. R. China.
- College of Chemical and Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, P. R. China
| |
Collapse
|
3
|
Qian Y, Ding J, Zhao R, Song Y, Yoo J, Moon H, Koo S, Kim JS, Shen J. Intrinsic immunomodulatory hydrogels for chronic inflammation. Chem Soc Rev 2025; 54:33-61. [PMID: 39499495 DOI: 10.1039/d4cs00450g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
The immune system plays a pivotal role in maintaining physiological homeostasis and influencing disease processes. Dysregulated immune responses drive chronic inflammation, which in turn results in a range of diseases that are among the leading causes of death globally. Traditional immune interventions, which aim to regulate either insufficient or excessive inflammation, frequently entail lifelong comorbidities and the risk of severe side effects. In this context, intrinsic immunomodulatory hydrogels, designed to precisely control the local immune microenvironment, have recently attracted increasing attention. In particular, these advanced hydrogels not only function as delivery mechanisms but also actively engage in immune modulation, optimizing interactions with the immune system for enhanced tissue repair, thereby providing a sophisticated strategy for managing chronic inflammation. In this tutorial review, we outline key elements of chronic inflammation and subsequently explore the strategic design principles of intrinsic immunomodulatory hydrogels based on these elements. Finally, we examine the challenges and prospects of such immunomodulatory hydrogels, which are expected to inspire further preclinical research and clinical translation in addressing chronic inflammation.
Collapse
Affiliation(s)
- Yuna Qian
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| | - Jiayi Ding
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Institute of Imaging Diagnosis and Minimally Invasive Intervention, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Rui Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Yang Song
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610065, China
| | - Jiyoung Yoo
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Huiyeon Moon
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Seyoung Koo
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jong Seung Kim
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| |
Collapse
|
4
|
Zhao M, Zhou Q, Ge Z. Supramolecular Assemblies via Host-Guest Interactions for Tumor Immunotherapy. Chemistry 2025; 31:e202403508. [PMID: 39448542 DOI: 10.1002/chem.202403508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cancer immunotherapy has emerged as one of the most promising modalities for cancer treatment providing hopes of cancer patients with the significant advantages over traditional antitumor therapy methods. Supramolecular assemblies based on host-guest interactions have been widely explored in the field of cancer immunotherapy as the delivery systems. A variety of supramolecular materials show unique features for efficient drug encapsulation, targeting delivery and release, which are favorable to activate antitumor immune responses especially through combination of different treatment strategies. In this review article, we summarize the research progresses of supramolecular assemblies via host-guest interactions for tumor immunotherapy. The construction of various drug delivery systems including hydrogels, liposomes, and polymeric nanoparticles, the drug encapsulation and delivery, as well as advantages and disadvantages are discussed. The perspectives related to future developments in this field are also described.
Collapse
Affiliation(s)
- Meng Zhao
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, China
| |
Collapse
|
5
|
Zhang H, Ji M, Wang Y, Jiang M, Lv Z, Li G, Wang L, Zheng Z. Intrinsic PD-L1 Degradation Induced by a Novel Self-Assembling Hexapeptide for Enhanced Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410145. [PMID: 39530653 PMCID: PMC11727121 DOI: 10.1002/advs.202410145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Programmed death-ligand 1 (PD-L1) is a critical immune checkpoint protein that facilitates tumor immune evasion. While antibody-based PD-1/PD-L1 inhibitors have shown promise, their limitations necessitate the development of alternative therapeutic strategies. This work addresses these challenges by developing a hexapeptide, KFM (Lys-Phe-Met-Phe-Met-Lys), capable of both directly downregulating PD-L1 and self-assembling into a ROS-responsive supramolecular hydrogel. This dual functionality allows Gel KFM to function as a localized drug delivery system and a PD-L1 inhibitor. Loading the hydrogel with mitoxantrone (MTX) and metformin (MET) further enhances the therapeutic effect by combining chemotherapy with PD-L1 downregulation. In vitro and in vivo studies demonstrate significant tumor growth inhibition, increased CD8+ T cell infiltration, and reduced intratumoral PD-L1 expression following peritumoral administration. Mechanistically, KFM promotes PD-L1 degradation via a ubiquitin-dependent pathway. This "carrier-free" delivery system expands the role of supramolecular hydrogels beyond passive carriers to active immunotherapeutic agents, offering a promising new strategy for cancer therapy.
Collapse
Affiliation(s)
- Hongxia Zhang
- The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Ming Ji
- The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Yamei Wang
- Tianjin Key Laboratory of Biosensing and Molecular RecognitionResearch Center for Analytical ScienceFrontiers Science Center for New Organic MatterCollege of ChemistryNankai UniversityTianjin300071China
| | - Mengmeng Jiang
- The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Zongyu Lv
- The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Gongyu Li
- Tianjin Key Laboratory of Biosensing and Molecular RecognitionResearch Center for Analytical ScienceFrontiers Science Center for New Organic MatterCollege of ChemistryNankai UniversityTianjin300071China
| | - Lulu Wang
- The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| | - Zhen Zheng
- The Province and Ministry Co‐sponsored Collaborative Innovation Center for Medical EpigeneticsTianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and DiagnosticsSchool of PharmacyTianjin Medical UniversityTianjin300070China
| |
Collapse
|
6
|
Yang Z, Yin G, Sun S, Xu P. Medical applications and prospects of polylactic acid materials. iScience 2024; 27:111512. [PMID: 39759018 PMCID: PMC11699620 DOI: 10.1016/j.isci.2024.111512] [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] [Indexed: 01/07/2025] Open
Abstract
Polylactic acid (PLA) is a biodegradable and bio-based polymer that has gained significant attention as an environmentally friendly alternative to traditional petroleum-based plastics. In clinical treatment, biocompatible and non-toxic PLA materials enhance safety and reduce tissue reactions, while the biodegradability allows it to breakdown over time naturally, avoiding a second surgery. With the emergence of nanotechnology and three-dimensional (3D) printing, medical utilized-PLA has been produced with more structural and biological properties at both micro and macro scales for clinical therapy. This review summarizes current applications of the PLA-based biomaterials in drug delivery systems, orthopedic treatment, tissue regenerative engineering, and surgery and medical devices, providing viewpoints regarding the prospective medical utilization.
Collapse
Affiliation(s)
- Zhenqi Yang
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shuyang Sun
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Ping Xu
- State Key Laboratory of Microbial Metabolism and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
- Asia Pacific Graduate Institute of Shanghai Jiao Tong University, Campus for Research Excellence and Technological Enterprise (CREATE), 1 CREATE Way, Singapore 138602, Singapore
| |
Collapse
|
7
|
Fan M, Zheng J, Huang Y, Lu M, Shang Z, Du M. Nanoparticle-mediated universal CAR-T therapy. Int J Pharm 2024; 666:124779. [PMID: 39349228 DOI: 10.1016/j.ijpharm.2024.124779] [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: 05/22/2024] [Revised: 09/16/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
In recent years, chimeric antigen receptor (CAR)-T cell therapy has been highly successful in treating hematological malignancies, leading to significant advancements in the cancer immunotherapy field. However, the typical CAR-T therapy necessitates the enrichment of patients' own leukocytes for ex vivo production of CAR-T cells, this customized pattern requires a complicated and time-consuming manufacturing procedure, making it costly and less accessible. The off-the-shelf universal CAR-T strategy could reduce manufacturing costs and realize timely drug administration, presenting as an ideal substitute for typical CAR-T therapy. Utilizing nanocarriers for targeted gene delivery is one of the approaches for the realization of universal CAR-T therapy, as biocompatible and versatile nanoparticles could deliver CAR genes to generate CAR-T cells in vivo. Nanoparticle-mediated in situ generation of CAR-T cells possesses multiple advantages, including lowered cost, simplified manufacturing procedure, and shortened administration time, this strategy is anticipated to provide a potentially cost-effective alternative to current autologous CAR-T cell manufacturing, thus facilitating the prevalence and improvement of CAR-T therapy.
Collapse
Affiliation(s)
- Mingliang Fan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jiayu Zheng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yue Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Mingxia Lu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Biomaterials and Tissue Engineering Research Center, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Zhi Shang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Mingwei Du
- Department of Dermatology, Shanghai Key Laboratory of Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
8
|
Liu Y, Tao D, Li M, Luo Z. Biomaterial-Mediated Metabolic Regulation of Ferroptosis for Cancer Immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2010. [PMID: 39492611 DOI: 10.1002/wnan.2010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/18/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Ferroptosis is a lipid peroxidation-driven cell death route and has attracted enormous interest for cancer therapy. Distinct from other forms of regulated cell death, its process is involved with multiple metabolic pathways including lipids, bioenergetics, iron, and so on, which influence cancer cell ferroptosis sensitivity and communication with the immune cells in the tumor microenvironment. Development of novel technologies for harnessing the ferroptosis-associated metabolic regulatory network would profoundly improve our understanding of the immune responses and enhance the efficacy of ferroptosis-dependent immunotherapy. Interestingly, the recent advances in bio-derived material-based therapeutic platforms offer novel opportunities to therapeutically modulate tumor metabolism through the in situ delivery of molecular or material cues, which not only allows the tumor-specific elicitation of ferroptosis but also holds promise to maximize their immunostimulatory impact. In this review, we will first dissect the crosstalk between tumor metabolism and ferroptosis and its impact on the immune regulation in the tumor microenvironment, followed by the comprehensive analysis on the recent progress in biomaterial-based metabolic regulatory strategies for evoking ferroptosis-mediated antitumor immunity. A perspective section is also provided to discuss the challenges in metabolism-regulating biomaterials for ferroptosis-immunotherapy. We envision that this review may provide new insights for improving tumor immunotherapeutic efficacy in the clinic.
Collapse
Affiliation(s)
- Yingqi Liu
- School of Life Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Dan Tao
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Menghuan Li
- School of Life Sciences, Chongqing University, Chongqing, People's Republic of China
| | - Zhong Luo
- School of Life Sciences, Chongqing University, Chongqing, People's Republic of China
| |
Collapse
|
9
|
Xu F, Ni Q, Gong N, Xia B, Zhang J, Guo W, Hu Z, Li J, Liang XJ. Delivery Systems Developed for Treatment Combinations to Improve Adoptive Cell Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407525. [PMID: 39165065 DOI: 10.1002/adma.202407525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/26/2024] [Indexed: 08/22/2024]
Abstract
Adoptive cell therapy (ACT) has shown great success in the clinic for treating hematologic malignancies. However, solid tumor treatment with ACT monotherapy is still challenging, owing to insufficient expansion and rapid exhaustion of adoptive cells, tumor antigen downregulation/loss, and dense tumor extracellular matrix. Delivery strategies for combination cell therapy have great potential to overcome these hurdles. The delivery of vaccines, immune checkpoint inhibitors, cytokines, chemotherapeutics, and photothermal reagents in combination with adoptive cells, have been shown to improve the expansion/activation, decrease exhaustion, and promote the penetration of adoptive cells in solid tumors. Moreover, the delivery of nucleic acids to engineer immune cells directly in vivo holds promise to overcome many of the hurdles associated with the complex ex vivo cell engineering strategies. Here, these research advance, as well as the opportunities and challenges for integrating delivery technologies into cell therapy s are discussed, and the outlook for these emerging areas are criticlly analyzed.
Collapse
Affiliation(s)
- Fengfei Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Qiankun Ni
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Ningqiang Gong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, 071002, China
| | - Weisheng Guo
- College of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, New Cornerstone Science Institute, Tsinghua University, Beijing, China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
10
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
11
|
Nie S, Qin Y, Ou L, Chen X, Li L. In Situ Reprogramming of Immune Cells Using Synthetic Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310168. [PMID: 38229527 DOI: 10.1002/adma.202310168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/12/2024] [Indexed: 01/18/2024]
Abstract
In the past decade, adoptive cell therapy with chimeric antigen receptor-T (CAR-T) cells has revolutionized cancer treatment. However, the complexity and high costs involved in manufacturing current adoptive cell therapy greatly inhibit its widespread availability and access. To address this, in situ cell therapy, which directly reprograms immune cells inside the body, has recently been developed as a promising alternative. Here, an overview of the recent progress in the development of synthetic nanomaterials is provided to deliver plasmid DNA or mRNA for in situ reprogramming of T cells and macrophages, focusing especially on in situ CAR therapies. Also, the main challenges for in situ immune cell reprogramming are discussed and some approaches to overcome these barriers to fulfill the clinical applications are proposed.
Collapse
Affiliation(s)
- Shihong Nie
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyang Qin
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
12
|
Lin Y, Chen Y, Luo Z, Wu YL. Recent advances in biomaterial designs for assisting CAR-T cell therapy towards potential solid tumor treatment. NANOSCALE 2024; 16:3226-3242. [PMID: 38284230 DOI: 10.1039/d3nr05768b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cells have shown promising outcomes in the treatment of hematologic malignancies. However, CAR-T cell therapy in solid tumor treatment has been significantly hindered, due to the complex manufacturing process, difficulties in proliferation and infiltration, lack of precision, or poor visualization ability. Fortunately, recent reports have shown that functional biomaterial designs such as nanoparticles, polymers, hydrogels, or implantable scaffolds might have potential to address the above challenges. In this review, we aim to summarize the recent advances in the designs of functional biomaterials for assisting CAR-T cell therapy for potential solid tumor treatments. Firstly, by enabling efficient CAR gene delivery in vivo and in vitro, functional biomaterials can streamline the difficult process of CAR-T cell therapy manufacturing. Secondly, they might also serve as carriers for drugs and bioactive molecules, promoting the proliferation and infiltration of CAR-T cells. Furthermore, a number of functional biomaterial designs with immunomodulatory properties might modulate the tumor microenvironment, which could provide a platform for combination therapies or improve the efficacy of CAR-T cell therapy through synergistic therapeutic effects. Last but not least, the current challenges with biomaterials-based CAR-T therapies will also be discussed, which might be helpful for the future design of CAR-T therapy in solid tumor treatment.
Collapse
Affiliation(s)
- Yuting Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Ying Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), Singapore 138634, Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|