1
|
Zhang H, Xu X, Cao Y, Chen Z, Liu W, Lu X, Li C. Unlocking the Power of Photothermal Agents: A Universal Platform for Smart Immune NIR-Agonists for Precise Cancer Therapy. Angew Chem Int Ed Engl 2025; 64:e202424830. [PMID: 39907354 DOI: 10.1002/anie.202424830] [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: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/06/2025]
Abstract
Selective ablation of tumor cells allows safe eradication, thereby minimizing off-target damage, while specifically inducing immunogenic cell death (ICD) rather than commonly non-immunogenic apoptosis of tumor cells enables activation of anti-tumor immune response against residual cancer cells, including metastatic lesions. Herein, we present a general strategy leveraging a novel photothermal agent (PTA) that concomitantly enables precise tumor killing and activation of anti-tumor immunity. The unique PTA scaffold exhibits unexpected inherent endoplasmic reticulum (ER)-targeting capability and potent near-infrared (NIR) photothermal activity, inducing NIR-controlled immunogenic pyroptosis in various tumor cell lines via targeting ER stress in an oxygen-independent manner. Moreover, both ER-targeting and NIR-activity of our scaffold can be modulated on demand by chemical caging/uncaging, allowing quick activation with diverse biological and bioorthogonal molecular triggers. The potency of this universal platform is demonstrated via its application to develop a membrane protein-activatable NIR-agonist that selectively activates ICD in tumor sites while priming anti-tumor immunity, minimizing off-target effects and enhancing efficacy against mouse breast tumors. This versatile approach could lead to customization of various personalized and effective immune NIR-agonists for specific photoimmunotherapy applicable to diverse solid tumors.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Xiaona Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Yahui Cao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Zihui Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Weiqing Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Xinyi Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Changhua Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
2
|
Alexander C, Guo Z, Glover PB, Faulkner S, Pikramenou Z. Luminescent Lanthanides in Biorelated Applications: From Molecules to Nanoparticles and Diagnostic Probes to Therapeutics. Chem Rev 2025; 125:2269-2370. [PMID: 39960048 PMCID: PMC11869165 DOI: 10.1021/acs.chemrev.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Lanthanides are particularly effective in their clinical applications in magnetic resonance imaging and diagnostic assays. They have open-shell 4f electrons that give rise to characteristic narrow, line-like emission which is unique from other fluorescent probes in biological systems. Lanthanide luminescence signal offers selection of detection pathways based on the choice of the ion from the visible to the near-infrared with long luminescence lifetimes that lend themselves to time-resolved measurements for optical multiplexing detection schemes and novel bioimaging applications. The delivery of lanthanide agents in cells allows localized bioresponsive activity for novel therapies. Detection in the near-infrared region of the spectrum coupled with technological advances in microscopies opens new avenues for deep-tissue imaging and surgical interventions. This review focuses on the different ways in which lanthanide luminescence can be exploited in nucleic acid and enzyme detection, anion recognition, cellular imaging, tissue imaging, and photoinduced therapeutic applications. We have focused on the hierarchy of designs that include luminescent lanthanides as probes in biology considering coordination complexes, multimetallic lanthanide systems to metal-organic frameworks and nanoparticles highlighting the different strategies in downshifting, and upconversion revealing some of the opportunities and challenges that offer potential for further development in the field.
Collapse
Affiliation(s)
- Carlson Alexander
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
- Department
of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Zhilin Guo
- Department
of Materials Science and Engineering, Southern
University of Science and Technology, Shenzhen 518055, China
| | - Peter B. Glover
- Defence
Science and Technology Laboratory (DSTL), Porton Down, Salisbury SP4 0JQ, United
Kingdom
| | - Stephen Faulkner
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Zoe Pikramenou
- School
of Chemistry, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
3
|
Shao X, Zhao X, Wang B, Fan J, Wang J, An H. Tumor microenvironment targeted nano-drug delivery systems for multidrug resistant tumor therapy. Theranostics 2025; 15:1689-1714. [PMID: 39897552 PMCID: PMC11780529 DOI: 10.7150/thno.103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
In recent years, nano-drug delivery systems (Nano-DDS) that target the tumor microenvironment (TME) to overcome multidrug resistance (MDR) have become a research hotspot in the field of cancer therapy. By precisely targeting the TME and regulating its unique pathological features, such as hypoxia, weakly acidic pH, and abnormally expressed proteins, etc., these Nano-DDS enable effective delivery of therapeutic agents and reversal of MDR. This scientific research community is increasing its investment in the development of diversified systems and exploring their anti-drug resistance potential. Therefore, it is particularly important to conduct a comprehensive review of the research progress of TME-targeted Nano-DDS in recent years. After a brief introduction of TME and tumor MDR, the design principle and structure of liposomes, polymer micelles and inorganic nanocarriers are focused on, and their characteristics as TME-targeted nanocarriers are described. It also demonstrates how these systems break through the cancer MDR treatment through various targeting mechanisms, discusses their synthetic innovation, research results and resistance overcoming mechanisms. The review was concluded with deliberations on the key challenges and future outlooks of targeting TME Nano-DDS in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| |
Collapse
|
4
|
Cheng Y, Lu Y. Physical stimuli-responsive polymeric patches for healthcare. Bioact Mater 2025; 43:342-375. [PMID: 39399837 PMCID: PMC11470481 DOI: 10.1016/j.bioactmat.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 10/15/2024] Open
Abstract
Many chronic diseases have become severe public health problems with the development of society. A safe and efficient healthcare method is to utilize physical stimulus-responsive polymer patches, which may respond to physical stimuli, including light, electric current, temperature, magnetic field, mechanical force, and ultrasound. Under certain physical stimuli, these patches have been widely used in therapy for diabetes, cancer, wounds, hair loss, obesity, and heart diseases since they could realize controllable treatment and reduce the risks of side effects. This review sketches the design principles of polymer patches, including composition, properties, and performances. Besides, control methods of using different kinds of physical stimuli were introduced. Then, the fabrication methods and characterization of patches were explored. Furthermore, recent applications of these patches in the biomedical field were demonstrated. Finally, we discussed the challenges and prospects for its clinical translation. We anticipate that physical stimulus-responsive polymer patches will open up new avenues for healthcare by acting as a platform with multiple functions.
Collapse
Affiliation(s)
- Yifan Cheng
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
5
|
Zhang Z, Lu Y, Liu W, Huang Y. Nanomaterial-assisted delivery of CpG oligodeoxynucleotides for boosting cancer immunotherapy. J Control Release 2024; 376:184-199. [PMID: 39368710 DOI: 10.1016/j.jconrel.2024.09.044] [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: 05/09/2024] [Revised: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Cancer immunotherapy aims to improve immunity to not only eliminate the primary tumor but also inhibit metastasis and recurrence. It is considered an extremely promising therapeutic approach that breaks free from the traditional paradigm of oncological treatment. As the medical community learns more about the immune system's mechanisms that "turn off the brake" and "step on the throttle", there is increasingly successful research on immunomodulators. However, there are still more restrictions than countermeasures with immunotherapy related to immunomodulators, such as low responsiveness and immune-related adverse events that cause multiple adverse reactions. Therefore, medical experts and materials scientists attempted to the efficacy of immunomodulatory treatments through various methods, especially nanomaterial-assisted strategies. CpG oligodeoxynucleotides (CpG) not only act as an adjuvant to promote immune responses, but also induce autophagy. In this review, the enhancement of immunotherapy using nanomaterial-based CpG formulations is systematically elaborated, with a focus on the delivery, protection, synergistic promotion of CpG efficacy by nanomaterials, and selection of the timing of treatment. In addition, we also discuss and prospect the existing problems and future directions of research on nanomaterials in auxiliary CpG therapy.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Wenjing Liu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
6
|
Schwartzman JD, McCall M, Ghattas Y, Pugazhendhi AS, Wei F, Ngo C, Ruiz J, Seal S, Coathup MJ. Multifunctional scaffolds for bone repair following age-related biological decline: Promising prospects for smart biomaterial-driven technologies. Biomaterials 2024; 311:122683. [PMID: 38954959 DOI: 10.1016/j.biomaterials.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
The repair of large bone defects due to trauma, disease, and infection can be exceptionally challenging in the elderly. Despite best clinical practice, bone regeneration within contemporary, surgically implanted synthetic scaffolds is often problematic, inconsistent, and insufficient where additional osteobiological support is required to restore bone. Emergent smart multifunctional biomaterials may drive important and dynamic cellular crosstalk that directly targets, signals, stimulates, and promotes an innate bone repair response following age-related biological decline and when in the presence of disease or infection. However, their role remains largely undetermined. By highlighting their mechanism/s and mode/s of action, this review spotlights smart technologies that favorably align in their conceivable ability to directly target and enhance bone repair and thus are highly promising for future discovery for use in the elderly. The four degrees of interactive scaffold smartness are presented, with a focus on bioactive, bioresponsive, and the yet-to-be-developed autonomous scaffold activity. Further, cell- and biomolecular-assisted approaches were excluded, allowing for contemporary examination of the capabilities, demands, vision, and future requisites of next-generation biomaterial-induced technologies only. Data strongly supports that smart scaffolds hold significant promise in the promotion of bone repair in patients with a reduced osteobiological response. Importantly, many techniques have yet to be tested in preclinical models of aging. Thus, greater clarity on their proficiency to counteract the many unresolved challenges within the scope of aging bone is highly warranted and is arguably the next frontier in the field. This review demonstrates that the use of multifunctional smart synthetic scaffolds with an engineered strategy to circumvent the biological insufficiencies associated with aging bone is a viable route for achieving next-generation therapeutic success in the elderly population.
Collapse
Affiliation(s)
| | - Max McCall
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasmine Ghattas
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Fei Wei
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA; Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, USA, Orlando, FL
| | - Melanie J Coathup
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
7
|
Liu J, Yang H, Sang X, Gao T, Zhang Z, Fu S, Yang H, Chang L, Liu X, Liang S, Yuan S, Wei S, Yang Y, Yan X, Zhang X, Mu W, Liu Y, Zhang N. Localized light-triggered release macrophage cytopharmaceuticals containing O-nitrobenzyl group for enhanced solid tumor cell-chemotherapy. Acta Pharm Sin B 2024; 14:5053-5068. [PMID: 39664416 PMCID: PMC11628841 DOI: 10.1016/j.apsb.2024.08.033] [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: 05/28/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 12/13/2024] Open
Abstract
Cytopharmaceutical based on macrophages is a breakthrough in the field of targeted drug delivery. However, it remains a challenge to localize and control drug release while retaining macrophage activity and exerting its immunotherapeutic effect. Herein, a localized light-triggered release macrophage cytopharmaceutical (USIP@M) was proposed, which could utilize the tumor targeting and immunotherapy effects of macrophages to reverse the immune suppression of tumor microenvironment (TME). Amphiphilic block copolymers with ultraviolet (UV)-responsive o-nitrobenzyl groups were synthesized and co-loaded with sorafenib (SF), IMD-0354 (IMD), and upconverting nanoparticles (UCNPs), which were then taken up by macrophages, and the targeted delivery of drugs was realized by using the tumor tropism of macrophages. UCNPs converted near-infrared light with strong penetrability and high safety into UV light, which promoted the photoresponsive depolymerization of block copolymers and production of exosomes from USIP@M, accelerated drug efflux and maintained the activity of macrophages. IMD simultaneously polarized carrier macrophages and tumor-associated macrophages to exert the antitumor effect of macrophages, enhance T cell immunity, and alleviate the immunosuppressive state of TME. Synergistically with the chemotherapeutic effect of SF, it could effectively kill tumors. In conclusion, based on the localized light-triggered release strategy, this study constructed a novel macrophage cytopharmaceutical that could localize and control drug release while retaining the activity of macrophages and exerting its immunotherapeutic effect, which could effectively treat solid tumors.
Collapse
Affiliation(s)
| | | | - Xiao Sang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tong Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zipeng Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huizhen Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lili Chang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaoqing Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shijun Yuan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Suyun Wei
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuxin Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaoxin Yan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinke Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
8
|
Peng X, Liu Y, Peng F, Wang T, Cheng Z, Chen Q, Li M, Xu L, Man Y, Zhang Z, Tan Y, Liu Z. Aptamer-controlled stimuli-responsive drug release. Int J Biol Macromol 2024; 279:135353. [PMID: 39245104 DOI: 10.1016/j.ijbiomac.2024.135353] [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: 06/12/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Aptamers have been widely researched and applied in nanomedicine due to their programmable, activatable, and switchable properties. However, there are few reviews on aptamer-controlled stimuli-responsive drug delivery. This article highlights the mechanisms and advantages of aptamers in the construction of stimuli-responsive drug delivery systems. We summarize the assembly/reconfiguration mechanisms of aptamers in controlled release systems. The assembly and drug release strategies of drug delivery systems are illustrated. Specifically, we focus on the binding mechanisms to the target and the factors that induce/inhibit the binding to the stimuli, such as strand, pH, light, and temperature. The applications of aptamer-based stimuli-responsive drug release are elaborated. The challenges are discussed, and the future directions are proposed.
Collapse
Affiliation(s)
- Xingxing Peng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Feicheng Peng
- Hunan Institute for Drug Control, Changsha 410001, Hunan Province, PR China
| | - Ting Wang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhongyu Cheng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
9
|
Valentino A, Yazdanpanah S, Conte R, Calarco A, Peluso G. Smart Nanocomposite Hydrogels as Next-Generation Therapeutic and Diagnostic Solutions. Gels 2024; 10:689. [PMID: 39590045 PMCID: PMC11594247 DOI: 10.3390/gels10110689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Stimuli-responsive nanocomposite gels combine the unique properties of hydrogels with those of nanoparticles, thus avoiding the suboptimal results of single components and creating versatile, multi-functional platforms for therapeutic and diagnostic applications. These hybrid materials are engineered to respond to various internal and external stimuli, such as temperature, pH, light, magnetic fields, and enzymatic activity, allowing precise control over drug release, tissue regeneration, and biosensing. Their responsiveness to environmental cues permits personalized medicine approaches, providing dynamic control over therapeutic interventions and real-time diagnostic capabilities. This review explores recent advances in stimuli-responsive hybrid gels' synthesis and application, including drug delivery, tissue engineering, and diagnostics. Overall, these platforms have significant clinical potential, and future research is expected to lead to unique solutions to address unmet medical needs.
Collapse
Affiliation(s)
- Anna Valentino
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Sorur Yazdanpanah
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Anna Calarco
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| |
Collapse
|
10
|
Yu ZW, Zheng M, Fan HY, Liang XH, Tang YL. Ultraviolet (UV) radiation: a double-edged sword in cancer development and therapy. MOLECULAR BIOMEDICINE 2024; 5:49. [PMID: 39417901 PMCID: PMC11486887 DOI: 10.1186/s43556-024-00209-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
It has long been widely acknowledged that ultraviolet (UV) light is an environment risk factor that can lead to cancer, particularly skin cancer. However, it is worth noting that UV radiation holds potential for cancer treatment as a relatively high-energy electromagnetic wave. With the help of nanomaterials, the role of UV radiation has caught increasing attention in cancer treatment. In this review, we briefly summarized types of UV-induced cancers, including malignant melanoma, squamous cell carcinoma, basal cell carcinoma, Merkel cell carcinoma. Importantly, we discussed the primary mechanisms underlying UV carcinogenesis, including mutations by DNA damage, immunosuppression, inflammation and epigenetic alterations. Historically limited by its shallow penetration depth, the introduction of nanomaterials has dramatically transformed the utilization of UV light in cancer treatment. The direct effect of UV light itself generally leads to the suppression of cancer cell growth and the initiation of apoptosis and ferroptosis. It can also be utilized to activate photosensitizers for reactive oxygen species (ROS) production, sensitize radiotherapy and achieve controlled drug release. Finally, we comprehensively weigh the significant risks and limitations associated with the therapeutic use of UV radiation. And the contradictory effect of UV exposure in promoting and inhibiting tumor has been discussed. This review provides clues for potential clinical therapy as well as future study directions in the UV radiation field. The precise delivery and control of UV light or nanomaterials and the wavelength as well as dose effects of UV light are needed for a thorough understanding of UV radiation.
Collapse
Affiliation(s)
- Zhen-Wei Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No.14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Hua-Yang Fan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No.14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No.14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, No.14, Sec.3, Renminnan Road, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
11
|
Zhou M, Li G, Yu J, Zhou Q, Wang K, Kang J, Wang T, Li P, Wei H. Interfacial delivery of carbon monoxide via smart titanium implant coating for enhanced soft tissue integration with switchable antibacterial and immunomodulatory properties. Bioact Mater 2024; 40:318-333. [PMID: 38978805 PMCID: PMC11228469 DOI: 10.1016/j.bioactmat.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Soft tissue integration around titanium (Ti) implants is weaker than that around natural teeth, compromising long-term success of Ti implants. Carbon monoxide (CO) possesses distinctive therapeutic properties, rendering it as a highly promising candidate for enhancing STI. However, achieving controlled CO generation at the STI interface remains challenging. Herein, a controlled CO-releasing dual-function coating was constructed on Ti surfaces. Under near-infrared (NIR) irradiation, the designed surface could actively accelerate CO generation for antibiosis against both aerobic and anaerobic bacteria. More importantly, in the absence of NIR, the slow release of CO induces macrophage polarization from pro-inflammatory phenotype towards pro-regenerative phenotype. In a rat implantation model with induced infection, the designed surface effectively controlled the bacterial infection, alleviates accompanying inflammation and modulated immune microenvironment, leading to enhanced STI. Single-cell sequencing revealed that the coating alters the cytokine profile within the soft tissue, thereby influencing cellular functions. Differentially expressed genes in macrophages are highly enriched in the PIK3-Akt pathway. Furthermore, the cellular communication between fibroblasts and macrophages was significantly enhanced through the CXCL12/CXCL14/CXCR4 and CSF1-CSF1R ligand-receptor pair. These findings indicate that our coating showed an appealing prospect for enhancing STI around Ti implants, which would ultimately contribute to the improved long-term success of Ti implants.
Collapse
Affiliation(s)
- Minghao Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Gangfeng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE) & Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, PR China
| | - Jingwei Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Qian Zhou
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE) & Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, PR China
| | - Kun Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE) & Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, PR China
| | - Jiaxin Kang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, PR China
| | - Tengjiao Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE) & Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, PR China
- School of Flexible Electronics, Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou, 450046, PR China
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing, 401135, PR China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Institute of Flexible Electronics (IFE) & Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, PR China
- School of Flexible Electronics, Henan Institute of Flexible Electronics (HIFE), Henan University, 379 Mingli Road, Zhengzhou, 450046, PR China
| | - Hongbo Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, PR China
| |
Collapse
|
12
|
Bao X, Sun M, Meng L, Zhang H, Yi X, Zhang P. Applications of pyroptosis activators in tumor immunotherapy. Mater Today Bio 2024; 28:101191. [PMID: 39221221 PMCID: PMC11363858 DOI: 10.1016/j.mtbio.2024.101191] [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: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Contemporary progress in tumor immunotherapy has solidified its role as an effective approach in combating cancer. Nonetheless, the prevalent "immune cold" state within the tumor microenvironment poses a substantial barrier to its efficacy. Addressing this, pyroptosis-a gasdermin-mediated programmed cell death characterized by its inflammatory profile-emerges as a crucial mechanism. It catalyzes the release of vast quantities of pro-inflammatory cytokines and immunogens, potentially transforming immunosuppressive "cold" tumors into reactive "hot" ones. Herein, we will initially present an overview of pyroptosis as a distinct form of cell death, along with its molecular mechanisms. Subsequently, we will focus on introducing how pyroptosis activators are utilized in the field of tumor immunotherapy. Insights gained from applications of pyroptosis activators in tumor immunotherapy could lead to the development of safe and efficient pyroptosis activators, significantly enriching the arsenal for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Bao
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Mengmeng Sun
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Lingfei Meng
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Hong Zhang
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| |
Collapse
|
13
|
Mao W, Yoo HS. Inorganic Nanoparticle Functionalization Strategies in Immunotherapeutic Applications. Biomater Res 2024; 28:0086. [PMID: 39323561 PMCID: PMC11423863 DOI: 10.34133/bmr.0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024] Open
Abstract
Nanotechnology has been increasingly utilized in anticancer treatment owing to its ability of engineering functional nanocarriers that enhance therapeutic effectiveness while minimizing adverse effects. Inorganic nanoparticles (INPs) are prevalent nanocarriers to be customized for a wide range of anticancer applications, including theranostics, imaging, targeted drug delivery, and therapeutics, because they are advantageous for their superior biocompatibility, unique optical properties, and capacity of being modified via versatile surface functionalization strategies. In the past decades, the high adaptation of INPs in this emerging immunotherapeutic field makes them good carrier options for tumor immunotherapy and combination immunotherapy. Tumor immunotherapy requires targeted delivery of immunomodulating therapeutics to tumor locations or immunological organs to provoke immune cells and induce tumor-specific immune response while regulating immune homeostasis, particularly switching the tumor immunosuppressive microenvironment. This review explores various INP designs and formulations, and their employment in tumor immunotherapy and combination immunotherapy. We also introduce detailed demonstrations of utilizing surface engineering tactics to create multifunctional INPs. The generated INPs demonstrate the abilities of stimulating and enhancing the immune response, specific targeting, and regulating cancer cells, immune cells, and their resident microenvironment, sometimes along with imaging and tracking capabilities, implying their potential in multitasking immunotherapy. Furthermore, we discuss the promises of INP-based combination immunotherapy in tumor treatments.
Collapse
Affiliation(s)
- Wei Mao
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
- Kangwon Radiation Convergence Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
14
|
Jiang C, Chen Y, Li X, Li Y. An intelligent NIR-IIb-responsive lanthanide@metal-organic framework core-shell nanocatalyst for combined deep-tumor therapy. J Mater Chem B 2024; 12:8626-8632. [PMID: 39189804 DOI: 10.1039/d4tb01321b] [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: 08/28/2024]
Abstract
The ground-breaking combination of photodynamic therapy (PDT) and photothermal therapy (PTT) has attracted much attention in medical fields as an effective method for fighting cancer. However, evidence suggests that the therapy efficiency is still limited by shallow light penetration depth and poor photosensitizer loading capacity. Herein, we constructed an upconversion nanoparticle@Zr-based metal-organic framework@indocyanine green molecule (UCNPs@ZrMOF@ICG) nanocomposite to integrate 1532 nm light-triggered PDT and 808 nm light-mediated PTT. NaLnF4 nanoparticles are designed to emit upconversion luminescence (UCL) under 1532 nm laser excitation, which is consistent with the absorption spectra of the ZrMOF. Benefiting from the excellent energy transfer efficiency, the ZrMOF can absorb visible light from the UCNPs and then catalyze O2 into 1O2 for deep tissue PDT. To achieve combination therapy, the clinically approved ICG nanocomposite was introduced as a photothermal agent for PTT under 808 nm laser irradiation, and the photothermal conversion efficiency was calculated to be ∼28%. The designed nanosystems facilitate efficient deep-tissue tumor treatment by integrating PDT with PTT. Ultimately, this study creates a multifunctional nanocomposite by combining 1532 nm light-triggered deep tissue PDT and near-infrared (NIR) light-driven PTT for personalized cancer therapy.
Collapse
Affiliation(s)
- Chaoqun Jiang
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, People's Republic of China
| | - Yu Chen
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, People's Republic of China
| | - Xiaolong Li
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, Taiyuan 030032, China.
| | - Youbin Li
- School of Physics and Electronic Sciences, Hunan Provincial Key Laboratory of Flexible Electronic Materials Genome Engineering, Changsha University of Science and Technology, Changsha 410114, People's Republic of China
| |
Collapse
|
15
|
Xuan J, Wang Z, Huang Y, Liu Y, Han Y, Li M, Xiao M. DNA response element-based smart drug delivery systems for precise drug release. Biomater Sci 2024; 12:3550-3564. [PMID: 38832670 DOI: 10.1039/d4bm00138a] [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: 06/05/2024]
Abstract
Smart drug delivery systems (DDSs) that respond to, interact with, or are actuated by biological signals or pathological abnormalities (e.g., the tumor microenvironment) for controllable drug release are appealing therapeutic platforms for cancer treatment. Owing to their inherent self-assembled nature, nucleic acids have emerged as programmable materials for the development of multifunctional structures. In response to external environmental stimuli, DNA response elements can serve as switches to trigger conformational changes in DNA structures. Their stimulus-responsive properties make them promising candidates for constructing smart DDSs, and advancements in DNA response element-based DDSs in the field of biomedicine have been made. This review summarizes different types of DNA response elements, including DNA aptamers, DNAzymes, disulfide bond-modified DNA, pH-responsive DNA motifs, and photocleavable DNA building blocks, and highlights the advancements in DNA response element-based smart DDSs for precise drug release. Finally, future challenges and perspectives in this field are discussed.
Collapse
Affiliation(s)
- Jinnan Xuan
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China.
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Shaanxi Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Yuting Huang
- Department of Radiotherapy, Chaohu Hospital of Anhui Medical University, 64 Chaohu North Road, Chaohu 238000, P. R. China
| | - Yisi Liu
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
| | - Yuqiang Han
- Hubei Key Laboratory of Photoelectric Materials and Devices, School of Materials Science and Engineering, Hubei Normal University, 11 Cihu Road, Huangshi 435002, P. R. China
| | - Man Li
- Key Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Shaanxi Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P. R. China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China.
| |
Collapse
|
16
|
Du Y, Zhao X, He F, Gong H, Yang J, Wu L, Cui X, Gai S, Yang P, Lin J. A Vacancy-Engineering Ferroelectric Nanomedicine for Cuproptosis/Apoptosis Co-Activated Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403253. [PMID: 38703184 DOI: 10.1002/adma.202403253] [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: 03/03/2024] [Revised: 04/14/2024] [Indexed: 05/06/2024]
Abstract
Low efficacy of immunotherapy due to the poor immunogenicity of most tumors and their insufficient infiltration by immune cells highlights the importance of inducing immunogenic cell death and activating immune system for achieving better treatment outcomes. Herein, ferroelectric Bi2CuO4 nanoparticles with rich copper vacancies (named BCO-VCu) are rationally designed and engineered for ferroelectricity-enhanced apoptosis, cuproptosis, and the subsequently evoked immunotherapy. In this structure, the suppressed recombination of the electron-hole pairs by the vacancies and the band bending by the ferroelectric polarization lead to high catalytic activity, triggering reactive oxygen species bursts and inducing apoptosis. The cell fragments produced by apoptosis serve as antigens to activate T cells. Moreover, due to the generated charge by the ferroelectric catalysis, this nanomedicine can act as "a smart switch" to open the cell membrane, promote nanomaterial endocytosis, and shut down the Cu+ outflow pathway to evoke cuproptosis, and thus a strong immune response is triggered by the reduced content of adenosine triphosphate. Ribonucleic acid transcription tests reveal the pathways related to immune response activation. Thus, this study firstly demonstrates a feasible strategy for enhancing the efficacy of immunotherapy using single ferroelectric semiconductor-induced apoptosis and cuproptosis.
Collapse
Affiliation(s)
- Yaqian Du
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Xudong Zhao
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
- State Key Laboratory of Rare Earth Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Fei He
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Haijiang Gong
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Jiani Yang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150001, P. R. China
| | - Linzhi Wu
- College of Aerospace and Civil Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Xianchang Cui
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Shili Gai
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
17
|
Wang LC, Chen HK, Wang WJ, Hsu FY, Huang HZ, Kuo RT, Li WP, Tian HK, Yeh CS. Boosting Upconversion Efficiency in Optically Inert Shelled Structures with Electroactive Membrane through Electron Donation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404120. [PMID: 38727702 DOI: 10.1002/adma.202404120] [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: 03/20/2024] [Revised: 05/05/2024] [Indexed: 05/21/2024]
Abstract
This study innovatively addresses challenges in enhancing upconversion efficiency in lanthanide-based nanoparticles (UCNPs) by exploiting Shewanella oneidensis MR-1, a microorganism capable of extracellular electron transfer. Electroactive membranes, rich in c-type cytochromes, are extracted from bacteria and integrated into membrane-integrated liposomes (MILs), encapsulating core-shelled UCNPs with an optically inactive shell, forming UCNP@MIL constructs. The electroactive membrane, tailored to donate electrons through the inert shell, independently boosts upconversion emission under near-infrared excitation (980 or 1550 nm), bypassing ligand-sensitized UCNPs. The optically inactive shell restricts energy migration, emphasizing electroactive membrane electron donation. Density functional theory calculations elucidate efficient electron transfer due to the electroactive membrane hemes' highest occupied molecular orbital being higher than the valence band maximum of the optically inactive shell, crucial for enhancing energy transfer to emitter ions. The introduction of a SiO2 insulator coating diminishes light enhancement, underscoring the importance of unimpeded electron transfer. Luminescence enhancement remains resilient to variations in emitter or sensitizing ions, highlighting the robustness of the electron transfer-induced phenomenon. However, altering the inert shell material diminishes enhancement, emphasizing the role of electron transfer. This methodology holds significant promise for diverse biological applications. UCNP@MIL offers an advantage in cellular uptake, which proves beneficial for cell imaging.
Collapse
Affiliation(s)
- Liu-Chun Wang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Hong-Kai Chen
- Department of Chemical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wen-Jyun Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Fang-Yi Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hong-Zhang Huang
- Department of Chemical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Rui-Tong Kuo
- Department of Chemical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Peng Li
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Hong-Kang Tian
- Department of Chemical Engineering, National Cheng Kung University, Tainan, 701, Taiwan
- Hierarchical Green-Energy Materials (Hi-GEM) Research Center, National Cheng Kung University, Tainan, 701, Taiwan
- Program on Smart and Sustainable Manufacturing, Academy of Innovative Semiconductor and Sustainable Manufacturing, National Cheng Kung University, Tainan, 701, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
18
|
Peng J, Li S, Ti H. Sensitize Tumor Immunotherapy: Immunogenic Cell Death Inducing Nanosystems. Int J Nanomedicine 2024; 19:5895-5930. [PMID: 38895146 PMCID: PMC11184231 DOI: 10.2147/ijn.s457782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Low immunogenicity of tumors poses a challenge in the development of effective tumor immunotherapy. However, emerging evidence suggests that certain therapeutic approaches, such as chemotherapy, radiotherapy, and phototherapy, can induce varying degrees of immunogenic cell death (ICD). This ICD phenomenon leads to the release of tumor antigens and the maturation of dendritic cells (DCs), thereby enhancing tumor immunogenicity and promoting immune responses. However, the use of a single conventional ICD inducer often fails to achieve in situ tumor ablation and establish long-term anti-tumor immune responses. Furthermore, the induction of ICD induction varies among different approaches, and the distribution of the therapeutic agent within the body influences the level of ICD and the occurrence of toxic side effects. To address these challenges and further boost tumor immunity, researchers have explored nanosystems as inducers of ICD in combination with tumor immunotherapy. This review examines the mechanisms of ICD and different induction methods, with a specific focus on the relationship between ICD and tumor immunity. The aim is to explore the research advancements utilizing various nanomaterials to enhance the body's anti-tumor effects by inducing ICD. This paper aims to contribute to the development and clinical application of nanomaterial-based ICD inducers in the field of cancer immunotherapy by providing important theoretical guidance and practical references.
Collapse
Affiliation(s)
- Jianlan Peng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Shiying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- Guangdong Province Precise Medicine and Big Data Engineering Technology Research Center for Traditional Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
19
|
Fu Y, Zhu X, Ren L, Wan J, Wang H. Syringeable Near-Infrared Light-Activated In Situ Immunogenic Hydrogel Boosts the Cancer-Immunity Cycle to Enhance Anticancer Immunity. ACS NANO 2024; 18:14877-14892. [PMID: 38809421 DOI: 10.1021/acsnano.3c08425] [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: 05/30/2024]
Abstract
Effective anticancer immunity depends on properly activating multiple stepwise events in the cancer-immunity cycle. An immunologically "cold" tumor microenvironment (TME) engenders immune evasion and refractoriness to conventional checkpoint blockade immunotherapy. Here, we combine nanoparticle formulations and an in situ formed hydrogel scaffold to treat accessible tumors locally and to stimulate systemic immunity against metastatic tumor lesions. The nanoparticles encapsulate poly(ε-caprolactone)-derived cytotoxic chemotherapy and adjuvant of Toll-like receptor 7/8 through a reactive oxygen species (ROS)-cleavable linker that can be self-activated by the coassembled neighboring photosensitizer following near-infrared (NIR) laser irradiation. Further development results in syringeable, NIR light-responsive, and immunogenic hydrogel (iGEL) that can be implanted peritumorally and deposited into the tumor surgical bed. Upon NIR laser irradiation, the generated ROS induces iGEL degradation and bond cleavage in the polymer-drug conjugates, triggering the immunogenic cell death cascade in cancer cells and spontaneously releasing encapsulated agents to rewire the cancer-immunity cycle. Notably, upon application in multiple preclinical models of melanoma and triple-negative breast cancer, which are aggressive and refractory to conventional immunotherapy, iGEL induces durable remission of established tumors, extends postsurgical tumor-free survival, and inhibits metastatic burden. The result of this study is a locally administrable immunogenic hydrogel for triggering host systemic immunity to improve immunotherapeutic efficacy with minimal off-target side effects.
Collapse
Affiliation(s)
- Yang Fu
- The First Affiliated Hospital; NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
| | - Xiaoxiao Zhu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310016, P. R. China
| | - Lulu Ren
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
| | - Jianqin Wan
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province 250117, P. R. China
| | - Hangxiang Wang
- The First Affiliated Hospital; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, P. R. China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province 250117, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325000, P. R. China
| |
Collapse
|
20
|
Zheng LE, Huang M, Liu Y, Bao Q, Huang Y, Ye Y, Liu M, Sun P. Colorimetric aptasensor based on temporally controllable light-stimulated oxidase-mimicking fluorescein for the sensitive detection of exosomes in mild conditions. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3577-3586. [PMID: 38787692 DOI: 10.1039/d4ay00561a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Analysis of exosomes provides important information for rapid and non-invasive screening of tumors. However, sensitive and convenient detection of exosomes remains technically challenging to date. Herein, a colorimetric aptasensor based on the light-stimulated oxidase-mimicking activity of FITC was constructed for detecting ovarian cancer (OC) exosomes. The aptasensor contained an EpCAM aptamer to capture OC exosomes. Cholesterol and fluorescein (FITC) were used to modify either end of the DNA (DNA anchor). The DNA anchor could combine with exosomes through a hydrophobic reaction between cholesterol and the lipid membrane. FITC oxidized 3,3',5,5'-tetramethylbenzidine (TMB) under a 365 nm LED light source in a temporally controllable manner under mild conditions, causing the solution to change from colorless to blue, and the corresponding UV-vis absorbance increased. Based on this principle, the exosomes were qualitatively analyzed by observing the color change with the naked eye. In parallel, the exosome concentration was also detected using UV-vis spectrophotometry. The linear range was from 2 × 105 to 100 × 105 particles per mL with a limit of detection of 1.77 × 105 particles per mL. The developed aptasensor also exhibited favorable selectivity and could discriminate the exosomes from OC cells and normal cells. Besides, the receiver operating characteristic (ROC) curve demonstrates that it is possible to distinguish between patients with OC and healthy donors (HDs) using exosomes as the biomarker. Our technology may expand the applications of DNA-based detection method-enabled OC diagnostic tools.
Collapse
Affiliation(s)
- Li-E Zheng
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Min Huang
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yiyang Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Qiufang Bao
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yuxiu Huang
- Department of Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
- Department of Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Yuhong Ye
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China
| | - Mengmeng Liu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, China.
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou 350001, Fujian, China
| |
Collapse
|
21
|
Tang X, Zhao S, Luo J, Wang B, Wu X, Deng R, Chang K, Chen M. Smart Stimuli-Responsive Spherical Nucleic Acids: Cutting-Edge Platforms for Biosensing, Bioimaging, and Therapeutics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310732. [PMID: 38299771 DOI: 10.1002/smll.202310732] [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: 11/21/2023] [Revised: 12/27/2023] [Indexed: 02/02/2024]
Abstract
Spherical nucleic acids (SNAs) with exceptional colloidal stability, multiple modularity, and programmability are excellent candidates to address common molecular delivery-related issues. Based on this, the higher targeting accuracy and enhanced controllability of stimuli-responsive SNAs render them precise nanoplatforms with inestimable prospects for diverse biomedical applications. Therefore, tailored diagnosis and treatment with stimuli-responsive SNAs may be a robust strategy to break through the bottlenecks associated with traditional nanocarriers. Various stimuli-responsive SNAs are engineered through the incorporation of multifunctional modifications to meet biomedical demands with the development of nucleic acid functionalization. This review provides a comprehensive overview of prominent research in this area and recent advancements in the utilization of stimuli-responsive SNAs in biosensing, bioimaging, and therapeutics. For each aspect, SNA nanoplatforms that exhibit responsive behavior to both internal stimuli (including sequence, enzyme, redox reactions, and pH) and external stimuli (such as light and temperature) are highlighted. This review is expected to offer inspiration and guidance strategies for the rational design and development of stimuli-responsive SNAs in the field of biomedicine.
Collapse
Affiliation(s)
- Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Shuang Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Jie Luo
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Binpan Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xianlan Wu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Ruijia Deng
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
- College of Pharmacy and Laboratory Medicine, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
22
|
Aqib RM, Umer A, Li J, Liu J, Ding B. Light Responsive DNA Nanomaterials and Their Biomedical Applications. Chem Asian J 2024; 19:e202400226. [PMID: 38514391 DOI: 10.1002/asia.202400226] [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: 02/29/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 03/23/2024]
Abstract
DNA nanomaterials have been widely employed for various biomedical applications. With rapid development of chemical modification of nucleic acid, serials of stimuli-responsive elements are included in the multifunctional DNA nanomaterials. In this review, we summarize the recent advances in light responsive DNA nanomaterials based on photocleavage/photodecage, photoisomerization, and photocrosslinking for efficient bioimaging (including imaging of small molecule, microRNA, and protein) and drug delivery (including delivery of small molecule, nucleic acid, and gene editing system). We also discuss the remaining challenges and future perspectives of the light responsive DNA nanomaterials in biomedical applications.
Collapse
Affiliation(s)
- Raja Muhammad Aqib
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Arsalan Umer
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jialin Li
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Jianbing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
23
|
Chen B, Huang R, Zeng W, Wang W, Min Y. Nanocodelivery of an NIR photothermal agent and an acid-responsive TLR7 agonist prodrug to enhance cancer photothermal immunotherapy and the abscopal effect. Biomaterials 2024; 305:122434. [PMID: 38141501 DOI: 10.1016/j.biomaterials.2023.122434] [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: 10/02/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/25/2023]
Abstract
The immunosuppressive tumor microenvironment (TME) greatly limits the actual outcome of immunotherapy. Therefore, it is urgent to develop appropriate strategies to reshape the TME and ultimately induce a strong immune response. Here, we developed a dual-functional liposome loaded with the photothermal agent IR808 near the infrared region (NIR) and Toll-like-receptor-7 (TLR7) agonist loxoribine prodrug (Lipo@IR808@Loxo) to achieve NIR light-triggered photothermal therapy (PTT) and the targeted delivery of immune adjuvants. Under NIR irradiation, Lipo@IR808@Loxo could greatly improve the efficiency of PTT to directly kill tumor cells and release tumor-associated antigens, which could work together with loaded loxoribine to relieve the immunosuppressive TME, effectively promoting the activation of antigen-presenting cells and subsequent antigen presentation. In this way, Lipo@IR808@Loxo could act as an in situ therapeutic cancer vaccine, eventually inducing a potent antitumor T-cell response. When further combined with immune checkpoint blockade, Lipo@IR808@Loxo-mediated photothermal immunotherapy could not only eliminate the primary tumors but also inhibit the growth of distant tumors, thus enhancing the abscopal effect.
Collapse
Affiliation(s)
- Bo Chen
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei, 230001, China
| | - Ruijie Huang
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Wei Zeng
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei, 230001, China.
| | - Yuanzeng Min
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Department of Bio-X Interdisciplinary Science at Hefei National Laboratory (HFNL) for Physical Science at the Microscale, University of Science and Technology of China, Hefei, 230001, China; School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, 230026, China; Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China; CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
24
|
Du Y, Yang J, He F, Zhao X, Zhou J, Zang P, Liu C, Xie Y, Zhang Y, Yang P. Revealing the Mutually Enhanced Mechanism of Necroptosis and Immunotherapy Induced by Defect Engineering and Piezoelectric Effect. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304322. [PMID: 37824104 DOI: 10.1002/adma.202304322] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/22/2023] [Indexed: 10/13/2023]
Abstract
Owing to low immunogenicity-induced immune escape and short-term circulating immune responses, the efficiency of immunotherapy is unsatisfactory. Therefore, triggering immunogenic cell death and establishing a long-term, mutually reinforced treatment modality are urgent challenges. In this study, ultrathin CaBi2 Nb2 O9 nanosheets with tunable oxygen vacancies (abbreviated as CBNO-OV1) are prepared for synergistic necroptosis and immunotherapy. The optimized vacancy concentration significantly improves the piezoelectric effect under ultrasound irradiation, thereby considerably improving the generation of reactive oxygen species (ROS). Density functional theory shows that oxygen vacancies can improve the efficiency of electron hole separation by suppressing their recombination, thus resulting in enhanced piezocatalytic activity. Moreover, the piezoelectric effect improves the permeability of tumor cell membranes, thus resulting in Ca2+ influx. Additionally, CBNO-OV1 also releases a portion of Ca2+ , which induces necroptosis assisted by explosive ROS. Ribonucleic acid transcription tests suggest the mechanisms associated with immune response activation and necroptosis. More importantly, necroptosis can trigger a significant immune response in vivo, thus activating macrophage M1 polarization through the NF-kappa B pathway and promoting T-cell differentiation.Tumor Necrosis Factor-α differentiated from macrophages conversely promotes necroptosis, thus realizing a mutually enhanced effect. This study demonstrates the feasibility of mutually reinforced necroptosis and immunotherapy for amplifying tumor efficacy.
Collapse
Affiliation(s)
- Yaqian Du
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Jiani Yang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150001, P. R. China
| | - Fei He
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Xudong Zhao
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Jialing Zhou
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Pengyu Zang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Changlin Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ying Xie
- Key Laboratory of Functional Inorganic Material Chemistry, Ministry of Education, School of Chemistry and Materials Science, Heilongjiang University, Harbin, 150001, P. R. China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, 150001, P. R. China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| |
Collapse
|
25
|
Liu B, Duan H, Liu Z, Liu Y, Chu H. DNA-functionalized metal or metal-containing nanoparticles for biological applications. Dalton Trans 2024; 53:839-850. [PMID: 38108230 DOI: 10.1039/d3dt03614f] [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: 12/19/2023]
Abstract
The conjugation of DNA molecules with metal or metal-containing nanoparticles (M/MC NPs) has resulted in a number of new hybrid materials, enabling a diverse range of novel biological applications in nanomaterial assembly, biosensor development, and drug/gene delivery. In such materials, the molecular recognition, gene therapeutic, and structure-directing functions of DNA molecules are coupled with M/MC NPs. In turn, the M/MC NPs have optical, catalytic, pore structure, or photodynamic/photothermal properties, which are beneficial for sensing, theranostic, and drug loading applications. This review focuses on the different DNA functionalization protocols available for M/MC NPs, including gold NPs, upconversion NPs, metal-organic frameworks, metal oxide NPs and quantum dots. The biological applications of DNA-functionalized M/MC NPs in the treatment or diagnosis of cancers are discussed in detail.
Collapse
Affiliation(s)
- Bei Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Huijuan Duan
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
| | - Zechao Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Yuechen Liu
- College of Science, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China.
| |
Collapse
|
26
|
Chu Y, Zhang W, Yuan B, Xu XQ, Ma Y, Wang Y. Deepened Photodynamic Therapy through Skin Optical Clearing Technology in the Visible Light Window. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:1007-1015. [PMID: 38117735 DOI: 10.1021/acs.langmuir.3c03231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The trade-off that shorter wavelength light facilitates the efficient generation of reactive oxygen species (ROS) from photosensitizer (PS) while facing the drawback of limited penetration depth through skin tissue restricts the further development of photodynamic therapy (PDT). Here, we address this contradiction and achieve visible-light-tailored deep PDT combined with the skin optical clearing technology. With the help of the prepared skin optical clearing gel, the refractive index inhomogeneity between skin tissue components is greatly attenuated, and the light scattering effect within the skin tissue is remarkably reduced. As a consequence, the transmittance of visible light at 600 nm through in vitro porcine skin and in vivo mouse skin after treatment increases from approximately 10 and 40 to 70 and 70%, respectively. Furthermore, in the tumor cell eradication experiment, the local ROS generation efficiency in the experimental group is several times higher than that in the control group owing to improved visible transmittance, which is thus responsible for the complete eradication of tumor cells, even when shaded by skin tissue. The results suggest that this strategy may serve as a valuable supplement to the current deep PDT strategies.
Collapse
Affiliation(s)
- Yanji Chu
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Wenhui Zhang
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Bin Yuan
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Xiao-Qi Xu
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Yingchao Ma
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing 100872, China
| | - Yapei Wang
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, Department of Chemistry, Renmin University of China, Beijing 100872, China
| |
Collapse
|
27
|
Yadav D, Sharma PK, Malviya R, Mishra PS, Surendra AV, Rao GSNK, Rani BR. Stimuli-responsive Biomaterials for Tissue Engineering Applications. Curr Pharm Biotechnol 2024; 25:981-999. [PMID: 37594093 DOI: 10.2174/1389201024666230818121821] [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: 01/24/2023] [Revised: 06/14/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023]
Abstract
The use of ''smart materials,'' or ''stimulus responsive'' materials, has proven useful in a variety of fields, including tissue engineering and medication delivery. Many factors, including temperature, pH, redox state, light, and magnetic fields, are being studied for their potential to affect a material's properties, interactions, structure, and/or dimensions. New tissue engineering and drug delivery methods are made possible by the ability of living systems to respond to both external stimuli and their own internal signals) for example, materials composed of stimuliresponsive polymers that self assemble or undergo phase transitions or morphology transformation. The researcher examines the potential of smart materials as controlled drug release vehicles in tissue engineering, aiming to enable the localized regeneration of injured tissue by delivering precisely dosed drugs at precisely timed intervals.
Collapse
Affiliation(s)
- Deepika Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Prem Shankar Mishra
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | | | - G S N Koteswara Rao
- Shobhaben Pratapbhai Patel School of Pharmacy, NMIMS Deemed University, Mumbai, India
| | - Budha Roja Rani
- Institute of Pharmaceutical Technology, Sri Padmavathi Mahila Visvavidyalayam, Tirupati, A.P., India
| |
Collapse
|
28
|
Yuan F, Zhang C, Luo X, Cheng S, Zhu Y, Xian Y. An erythrocyte membrane-camouflaged fluorescent covalent organic framework for starving/nitric oxide/immunotherapy of triple-negative breast cancer. Chem Sci 2023; 14:14182-14192. [PMID: 38098713 PMCID: PMC10717584 DOI: 10.1039/d3sc02022c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/24/2023] [Indexed: 12/17/2023] Open
Abstract
It is a great challenge to effectively treat triple-negative breast cancer (TNBC) due to lack of therapeutic targets and drug resistance of systemic chemotherapy. Rational design of nanomedicine with good hemocompatibility is urgently desirable for combination therapy of TNBC. Herein, an erythrocyte membrane-camouflaged fluorescent covalent organic framework (COF) loaded with an NO donor (hydroxyurea, Hu), glucose oxidase (GOx) and cytosine-phosphate-guanine oligonucleotides (CPG) (COF@HGC) was developed for imaging-guided starving/nitric oxide (NO)/immunization synergistic treatment of TNBC. The substances of HGC are easily co-loaded onto the COF due to the ordered pore structure and large surface area. And a folic acid-modified erythrocyte membrane (FEM) is coated on the surface of COF@HGC to improve targeted therapy and haemocompatibility. When COF@HGC@FEM is internalized into tumor cells, hemoglobin (Hb) on FEM and GOx loaded on the COF can trigger cascade reactions to kill tumor cells due to the simultaneous production of NO and exhaustion of glucose. Meanwhile, the COF with excellent fluorescence properties can be used as a self-reporter for bioimaging. Furthermore, the CPG can reprogram tumor-associated macrophages from tumor-supportive phenotype to anti-tumor phenotype and enhance immunotherapy. Through the "three-in-one" strategy, the biomimetic nanoplatform can effectively inhibit tumor growth and reprogram the tumor immunosuppression microenvironment in the TNBC mouse model.
Collapse
Affiliation(s)
- Fang Yuan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Xianzhu Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Yingxin Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200241 China
| |
Collapse
|
29
|
Zhu J, Fan J, Xia Y, Wang H, Li Y, Feng Z, Fu C. Potential targets and applications of nanodrug targeting myeloid cells in osteosarcoma for the enhancement of immunotherapy. Front Pharmacol 2023; 14:1271321. [PMID: 37808190 PMCID: PMC10551637 DOI: 10.3389/fphar.2023.1271321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Targeted immunotherapies have emerged as a transformative approach in cancer treatment, offering enhanced specificity to tumor cells, and minimizing damage to healthy tissues. The targeted treatment of the tumor immune system has become clinically applicable, demonstrating significant anti-tumor activity in both early and late-stage malignancies, subsequently enhancing long-term survival rates. The most frequent and significant targeted therapies for the tumor immune system are executed through the utilization of checkpoint inhibitor antibodies and chimeric antigen receptor T cell treatment. However, when using immunotherapeutic drugs or combined treatments for solid tumors like osteosarcoma, challenges arise due to limited efficacy or the induction of severe cytotoxicity. Utilizing nanoparticle drug delivery systems to target tumor-associated macrophages and bone marrow-derived suppressor cells is a promising and attractive immunotherapeutic approach. This is because these bone marrow cells often exert immunosuppressive effects in the tumor microenvironment, promoting tumor progression, metastasis, and the development of drug resistance. Moreover, given the propensity of myeloid cells to engulf nanoparticles and microparticles, they are logical therapeutic targets. Therefore, we have discussed the mechanisms of nanomedicine-based enhancement of immune therapy through targeting myeloid cells in osteosarcoma, and how the related therapeutic strategies well adapt to immunotherapy from perspectives such as promoting immunogenic cell death with nanoparticles, regulating the proportion of various cellular subgroups in tumor-associated macrophages, interaction with myeloid cell receptor ligands, activating immunostimulatory signaling pathways, altering myeloid cell epigenetics, and modulating the intensity of immunostimulation. We also explored the clinical implementations of immunotherapy grounded on nanomedicine.
Collapse
Affiliation(s)
- Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Yuanliang Xia
- Department of Cardiac Surgery, The First Hospital of Jilin University, Changchun, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuehong Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Zijia Feng
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Wang D, Liu J, Duan J, Yi H, Liu J, Song H, Zhang Z, Shi J, Zhang K. Enrichment and sensing tumor cells by embedded immunomodulatory DNA hydrogel to inhibit postoperative tumor recurrence. Nat Commun 2023; 14:4511. [PMID: 37500633 PMCID: PMC10374534 DOI: 10.1038/s41467-023-40085-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 07/10/2023] [Indexed: 07/29/2023] Open
Abstract
Postoperative tumor recurrence and metastases often lead to cancer treatment failure. Here, we develop a local embedded photodynamic immunomodulatory DNA hydrogel for early warning and inhibition of postoperative tumor recurrence. The DNA hydrogel contains PDL1 aptamers that capture and enrich in situ relapsed tumor cells, increasing local ATP concentration to provide a timely warning signal. When a positive signal is detected, local laser irradiation is performed to trigger photodynamic therapy to kill captured tumor cells and release tumor-associated antigens (TAA). In addition, reactive oxygen species break DNA strands in the hydrogel to release encoded PDL1 aptamer and CpG, which together with TAA promote sufficient systemic antitumor immunotherapy. In a murine model where tumor cells are injected at the surgical site to mimic tumor recurrence, we find that the hydrogel system enables timely detection of tumor recurrence by enriching relapsed tumor cells to increase local ATP concentrations. As a result, a significant inhibitory effect of approximately 88.1% on recurrent tumors and effectively suppressing metastasis, offering a promising avenue for timely and effective treatment of postoperative tumor recurrence.
Collapse
Affiliation(s)
- Danyu Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingwen Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jie Duan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hua Yi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Haiwei Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore, 138673, Singapore.
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China.
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China.
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China.
| |
Collapse
|
31
|
Kang W, Liu Y, Wang W. Light-responsive nanomedicine for cancer immunotherapy. Acta Pharm Sin B 2023; 13:2346-2368. [PMID: 37425044 PMCID: PMC10326299 DOI: 10.1016/j.apsb.2023.05.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 07/11/2023] Open
Abstract
Immunotherapy emerged as a paradigm shift in cancer treatments, which can effectively inhibit cancer progression by activating the immune system. Remarkable clinical outcomes have been achieved through recent advances in cancer immunotherapy, including checkpoint blockades, adoptive cellular therapy, cancer vaccine, and tumor microenvironment modulation. However, extending the application of immunotherapy in cancer patients has been limited by the low response rate and side effects such as autoimmune toxicities. With great progress being made in nanotechnology, nanomedicine has been exploited to overcome biological barriers for drug delivery. Given the spatiotemporal control, light-responsive nanomedicine is of great interest in designing precise modality for cancer immunotherapy. Herein, we summarized current research utilizing light-responsive nanoplatforms to enhance checkpoint blockade immunotherapy, facilitate targeted delivery of cancer vaccines, activate immune cell functions, and modulate tumor microenvironment. The clinical translation potential of those designs is highlighted and challenges for the next breakthrough in cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Weirong Kang
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, the University of Hong Kong, Hong Kong, China
| | - Yuwei Liu
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, the University of Hong Kong, Hong Kong, China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
32
|
Liu Y, Shi Y, Yu L, Wu Z, Jiang JH. Reversible Acylation of RNA Enables Activatable Biosensing. Anal Chem 2023; 95:6490-6495. [PMID: 37053522 DOI: 10.1021/acs.analchem.3c00723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
There is a high demand to develop chemical tools to control the property and function of RNA. Current methods mainly rely on ultraviolet light-based caging strategies, which may cause phototoxicity in live cell-based experiments. We herein report an endogenous stimulus-responsive RNA acylation approach by introducing boronate ester (BE) groups to 2'-hydroxyls through postsynthetic modification. Treatment with hydrogen peroxide (H2O2) yields a phenol derivative which undergoes a 1,6-eliminaton for the traceless release of 2'-hydroxyl. We demonstrated that the acylation of crRNA enabled conditional regulation of CRISPR/Cas13a activity for activatable detection of target RNA. We also showed that the highly specific acylation of the single RNA in 8-17 DNAzyme allowed reversible control of the catalytic activity of DNAzyme, which was further applied to the cell-selective imaging of metal ions in cancer cells. Thus, our strategy provides a simple, general, and cell-selective method to control RNA activity, affording great potential in the construction of activatable RNA sensors and pre-RNA medicines.
Collapse
Affiliation(s)
- Yining Liu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yang Shi
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Lanxing Yu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Zhenkun Wu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
33
|
Remmers RCPA, Neumann K. Reaching new lights: a review on photo-controlled nanomedicines and their in vivo evaluation. Biomater Sci 2023; 11:1607-1624. [PMID: 36727448 DOI: 10.1039/d2bm01621d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The selective and efficient delivery of bioactive molecules to sites of interest remains a formidable challenge in medicine. In recent years, it has been shown that stimuli-responsive drug delivery systems display several advantages over traditional drug administration such as an improved pharmacokinetic profile and the desirable ability to gain control over release. Light emerged as one of the most powerful stimuli due to its high biocompatibility, spatio-temporal control, and non-invasiveness. On the road to clinical translation, various chemical systems of high complexity have been reported with the aim to improve efficacy, safety, and versatility of drug delivery under complex biological conditions. For future research on the chemical design of such photo-controlled nanomedicines, it is essential to gain an understanding of their in vivo translation and efficiency. Here, we discuss photo-controlled nanomedicines that have been evaluated in vivo and provide an overview of the state-of-the-art that should guide future research design.
Collapse
Affiliation(s)
- Rik C P A Remmers
- Institute for Molecules and Materials, Radboud University, Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands.
| | - Kevin Neumann
- Institute for Molecules and Materials, Radboud University, Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands.
| |
Collapse
|
34
|
Xu D, Li C, Li W, Lin B, Lv R. Recent advances in lanthanide-doped up-conversion probes for theranostics. Front Chem 2023; 11:1036715. [PMID: 36846851 PMCID: PMC9949555 DOI: 10.3389/fchem.2023.1036715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Up-conversion (or anti-Stokes) luminescence refers to the phenomenon whereby materials emit high energy, short-wavelength light upon excitation at longer wavelengths. Lanthanide-doped up-conversion nanoparticles (Ln-UCNPs) are widely used in biomedicine due to their excellent physical and chemical properties such as high penetration depth, low damage threshold and light conversion ability. Here, the latest developments in the synthesis and application of Ln-UCNPs are reviewed. First, methods used to synthesize Ln-UCNPs are introduced, and four strategies for enhancing up-conversion luminescence are analyzed, followed by an overview of the applications in phototherapy, bioimaging and biosensing. Finally, the challenges and future prospects of Ln-UCNPs are summarized.
Collapse
Affiliation(s)
| | | | | | - Bi Lin
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi, China
| | | |
Collapse
|
35
|
Yang C, Wu X, Liu J, Ding B. Stimuli-responsive nucleic acid nanostructures for efficient drug delivery. NANOSCALE 2022; 14:17862-17870. [PMID: 36458678 DOI: 10.1039/d2nr05316k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Based on complementary base pairing, nucleic acid molecules have acted as engineerable building blocks to prepare versatile nanostructures with unique shapes and sizes. Benefiting from excellent programmability and biocompatibility, rationally designed nucleic acid nanostructures have been widely employed in biomedical applications. With the development of the chemical biology of nucleic acids, various stimuli-responsive nucleic acid nanostructures have been constructed by tailored chemical modification with multifunctional components. In this minireview, we summarize the representative and latest research about the employment of stimuli-responsive nucleic acid nanostructures for drug delivery in response to endogenous and exogenous stimuli (redox gradient, pH, nuclease, biomacromolecule, and light). We also discuss the broad prospects and remaining challenges of nucleic acid nanotechnology in biomedical applications.
Collapse
Affiliation(s)
- Changping Yang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- School of Materials Science and Engineering, Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaohui Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianbing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for NanoScience and Technology, 11 BeiYiTiao, ZhongGuanCun, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- School of Materials Science and Engineering, Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
36
|
Huang L, Liang Z, Zhang F, Luo H, Liang R, Han F, Wu Z, Han D, Shen J, Niu L. Upconversion NaYF 4:Yb/Er–TiO 2–Ti 3C 2 Heterostructure-Based Near-Infrared Light-Driven Photoelectrochemical Biosensor for Highly Sensitive and Selective d-Serine Detection. Anal Chem 2022; 94:16246-16253. [DOI: 10.1021/acs.analchem.2c04101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Likun Huang
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Zhishan Liang
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Hui Luo
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Ruilian Liang
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Fangjie Han
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Zhifang Wu
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
| | - Dongxue Han
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
- Guangzhou Provincial Key Laboratory of Psychoactive Substance Monitoring and Safety, Anti-Drug Technology Center of Guangdong Province, Guangzhou 510230, P. R. China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Li Niu
- Guangzhou Key Laboratory of Sensing Materials and Devices, Center for Advanced Analytical Science, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, P. R. China
- State Key Laboratory of Electroanalytical Chemistry, c/o Engineering Laboratory for Modern Analytical Techniques, CAS Center for Excellence in Nanoscience, Changchun Institute of Applied Chemistry, Changchun 130022, P. R. China
- University of Chinese Academy of Sciences, Beijing 100039, P. R. China
| |
Collapse
|
37
|
Roy S, Curry SD, Bagot CC, Mueller EN, Mansouri AM, Park W, Cha JN, Goodwin AP. Enzyme Prodrug Therapy with Photo-Cross-Linkable Anti-EGFR Affibodies Conjugated to Upconverting Nanoparticles. ACS NANO 2022; 16:15873-15883. [PMID: 36129781 PMCID: PMC10197967 DOI: 10.1021/acsnano.2c02558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
In this work, we demonstrate that a photo-cross-linkable conjugate of upconverting nanoparticles and cytosine deaminase can catalyze prodrug conversion specifically at tumor sites in vivo. Non-covalent association of proteins and peptides with cellular surfaces leads to receptor-mediated endocytosis and catabolic degradation. Recently, we showed that covalent attachment of proteins such as affibodies to cell receptors yields extended expression on cell surfaces with preservation of protein function. To adapt this technology for in vivo applications, conjugates were prepared from upconverting nanoparticles and fusion proteins of affibody and cytosine deaminase enzyme (UC-ACD). The affibody allows covalent photo-cross-linking to epidermal growth factor receptors (EGFRs) overexpressed on Caco-2 human colorectal cancer cells under near-infrared (NIR) light. Once bound, the cytosine deaminase portion of the fusion protein converts the prodrug 5-fluorocytosine (5-FC) to the anticancer drug 5-fluorouracil (5-FU). NIR covalent photoconjugation of UC-ACD to Caco-2 cells showed 4-fold higher retention than observed with cells that were not irradiated in vitro. Next, athymic mice expressing Caco-2 tumors showed 5-fold greater UC-ACD accumulation in the tumors than either conjugates without the CD enzyme or UC-ACDs in the absence of NIR excitation. With oral administration of 5-FC prodrug, tumors with photoconjugated UC-ACD yielded 2-fold slower growth than control groups, and median mouse survival increased from 28 days to 35 days. These experiments demonstrate that enzyme-decorated nanoparticles can remain viable after a single covalent photoconjugation in vivo, which can in turn localize prodrug conversion to tumor sites for multiple weeks.
Collapse
Affiliation(s)
- Shambojit Roy
- Department of Chemical and Biological Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Shane D. Curry
- Department of Chemical and Biological Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Conrad Corbella Bagot
- Department of Electrical, Computer, and Energy Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Evan N. Mueller
- Department of Chemical and Biological Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Abdulrahman M. Mansouri
- Department of Chemical and Biological Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Wounjhang Park
- Department of Electrical, Computer, and Energy Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Jennifer N. Cha
- Department of Chemical and Biological Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
- Materials Science and Engineering Program, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| | - Andrew P. Goodwin
- Department of Chemical and Biological Engineering, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
- Materials Science and Engineering Program, University of Colorado, 596 UCB, Boulder, Colorado 80309, United States
| |
Collapse
|
38
|
Effect of the surface coverage of an alkyl carboxylic acid monolayer on waterborne and cellular uptake behaviors for silicon quantum dots. Sci Rep 2022; 12:17211. [PMID: 36241686 PMCID: PMC9568572 DOI: 10.1038/s41598-022-21698-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/30/2022] [Indexed: 01/06/2023] Open
Abstract
This article reports the development of highly waterborne silicon quantum dots (Si QDs) terminated with a reactive group for grafting of biomolecules. Hydrogen-terminated QDs were prepared by thermal disproportionation of amorphous hydrogen silsesquioxane derived from triethoxysilane followed by hydrofluoric etching. Next, the hydrogenated Si surfaces were exposed to 10-undecenoic acid at different temperatures in Ar atmosphere, yielding the termination of the QDs with a carboxyl group. The thermal hydrosilylation of 10-undecenoic acid yielded the termination of the QDs with a carboxyl group. An increase in molecular coverage of an undecanoic acid (UA) monolayer resulted in both the enhanced increase of zeta-potential in a negative direction for a greater water-dispersity and the increase of absolute quantum yield (QY) of photoluminescence (PL). PLQY improved for ~ 1% to 26% with increasing UA coverage. We assessed the molecular interaction between the UA-SiQDs and HeLa cells by means of cellular uptake experiments using the QDs with different UA coverages. Results showed that the QDs with the highest dispersity in water were not internalized in the cells under confocal fluorescence microscopic observation. In contrast, the QDs with lower coverage of UA monolayer were internalized by endocytosis when incubated with HeLa cells. This contrasting observation opens the possibility of successfully preparing carboxy-capped SiQDs that do not allow cellular uptake but are targeted to specific cells by appropriate conjugation with biomolecules.
Collapse
|
39
|
Wang F, Duan H, Xu W, Sheng G, Sun Z, Chu H. Light-activated nanomaterials for tumor immunotherapy. Front Chem 2022; 10:1031811. [PMID: 36277335 PMCID: PMC9585221 DOI: 10.3389/fchem.2022.1031811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022] Open
Abstract
Tumor immunotherapy mainly relies on activating the immune system to achieve antitumor treatment. However, the present tumor immunotherapy used in the clinic showed low treatment efficacy with high systematic toxicity. To overcome the shortcomings of traditional drugs for immunotherapy, a series of antitumor immunotherapies based on nanomaterials have been developed to enhance the body’s antitumor immune response and reduce systematic toxicity. Due to the noninvasiveness, remote controllability, and high temporal and spatial resolution of light, photocontrolled nanomaterials irradiated by excitation light have been widely used in drug delivery and photocontrolled switching. This review aims to highlight recent advances in antitumor immunotherapy based on photocontrolled nanomaterials. We emphasized the advantages of nanocomposites for antitumor immunotherapy and highlighted the latest progress of antitumor immunotherapy based on photoactivated nanomaterials. Finally, the challenges and future prospects of light-activated nanomaterials in antitumor immunity are discussed.
Collapse
Affiliation(s)
- Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Huijuan Duan
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Weizhe Xu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Gang Sheng
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- *Correspondence: Hongqian Chu,
| |
Collapse
|
40
|
Wang D, Liu J, Duan J, Ma Y, Gao H, Zhang Z, Liu J, Shi J, Zhang K. Photocontrolled Spatiotemporal Delivery of DNA Immunomodulators for Enhancing Membrane-Targeted Tumor Photodynamic Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44183-44198. [PMID: 36165393 DOI: 10.1021/acsami.2c12774] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Immunotherapy is emerging as a paradigm-shifting modality for treatment cancer. However, systemic administration of immunomodulators is usually accompanied by extra-tumor toxicity and adverse immune effects. Precise delivery of immunomodulators with a highly controllable system may provide a solution for this issue. Here, we developed a photocontrolled DNA nanomedicine for localized delivery of DNA immunomodulators to enhance membrane-targeted photodynamic immunotherapy. Specifically, the DNA nanomedicine is composed of long tandemly repeated functional DNA sequences (PDL1 aptamers and CpG) with a photosensitizer (TMPyP4) inserted into the DNA structure, providing high drug-loading capacity. By blocking the surface PDL1 aptamer with a pHLIP-modified cDNA, the DNA nanomedicine does not induce any obvious immune response and can be specifically delivered and anchored to the tumor membrane. Under localized irradiation, photodynamically generated reactive oxygen species (ROS) causes breakage of DNA sequences, which triggers the collapse of the nanostructure and release of internal DNA immunomodulators. Under localized illumination, photodynamically generated ROS can cause DNA sequence breaks, triggering the collapse of nanostructures and the release of internal DNA immunomodulators thus enhancing membrane-targeted photodynamic immunotherapy. We have demonstrated that the developed DNA nanomedicine can drive efficient immune responses in tumor tissue without perceptibly interfering off-tumor immunity, resulting in efficient antitumor treatment while mitigating systemic toxicity.
Collapse
Affiliation(s)
- Danyu Wang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Jingwen Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Jie Duan
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Yanrui Ma
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Hua Gao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
41
|
Cheng X, Zhou J, Yue J, Wei Y, Gao C, Xie X, Huang L. Recent Development in Sensitizers for Lanthanide-Doped Upconversion Luminescence. Chem Rev 2022; 122:15998-16050. [PMID: 36194772 DOI: 10.1021/acs.chemrev.1c00772] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The attractive features of lanthanide-doped upconversion luminescence (UCL), such as high photostability, nonphotobleaching or photoblinking, and large anti-Stokes shift, have shown great potentials in life science, information technology, and energy materials. Therefore, UCL modulation is highly demanded toward expected emission wavelength, lifetime, and relative intensity in order to satisfy stringent requirements raised from a wide variety of areas. Unfortunately, the majority of efforts have been devoted to either simple codoping of multiple activators or variation of hosts, while very little attention has been paid to the critical role that sensitizers have been playing. In fact, different sensitizers possess different excitation wavelengths and different energy transfer pathways (to different activators), which will lead to different UCL features. Thus, rational design of sensitizers shall provide extra opportunities for UCL tuning, particularly from the excitation side. In this review, we specifically focus on advances in sensitizers, including the current status, working mechanisms, design principles, as well as future challenges and endeavor directions.
Collapse
Affiliation(s)
- Xingwen Cheng
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China
| | - Jie Zhou
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China
| | - Jingyi Yue
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China
| | - Yang Wei
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China
| | - Chao Gao
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China
| | - Xiaoji Xie
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China
| | - Ling Huang
- Institute of Advanced Materials, Nanjing Tech University, 30 South Puzhu Road, Nanjing211816, China.,State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources, College of Chemistry, Xinjiang University, Urumqi830046, China
| |
Collapse
|
42
|
Li R, Chen Z, Li J, Dai Z, Yu Y. Nano-drug delivery systems for T cell-based immunotherapy. NANO TODAY 2022; 46:101621. [DOI: 10.1016/j.nantod.2022.101621] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
43
|
Li J, Luo Y, Zeng Z, Cui D, Huang J, Xu C, Li L, Pu K, Zhang R. Precision cancer sono-immunotherapy using deep-tissue activatable semiconducting polymer immunomodulatory nanoparticles. Nat Commun 2022; 13:4032. [PMID: 35821238 PMCID: PMC9276830 DOI: 10.1038/s41467-022-31551-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/22/2022] [Indexed: 12/24/2022] Open
Abstract
Nanomedicine holds promise to enhance cancer immunotherapy; however, its potential to elicit highly specific anti-tumor immunity without compromising immune tolerance has yet to be fully unlocked. This study develops deep-tissue activatable cancer sono-immunotherapy based on the discovery of a semiconducting polymer that generates sonodynamic singlet oxygen (1O2) substantially higher than other sonosensitizers. Conjugation of two immunomodulators via 1O2-cleavable linkers onto this polymer affords semiconducting polymer immunomodulatory nanoparticles (SPINs) whose immunotherapeutic actions are largely inhibited. Under ultrasound irradiation, SPINs generate 1O2 not only to directly debulk tumors and reprogram tumor microenvironment to enhance tumor immunogenicity, but also to remotely release the immunomodulators specifically at tumor site. Such a precision sono-immunotherapy eliminates tumors and prevents relapse in pancreatic mouse tumor model. SPINs show effective antitumor efficacy even in a rabbit tumor model. Moreover, the sonodynamic activation of SPINs confines immunotherapeutic action primarily to tumors, reducing the sign of immune-related adverse events. To improve the specificity of immunotherapy, here the authors report the design of a semiconducting polymer immunomodulatory nanoparticle with sonodynamic process to remotely release immune-modulators for deep-tissue precision cancer sono-immunotherapy.
Collapse
Affiliation(s)
- Jingchao Li
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Yu Luo
- School of Chemical Science and Engineering, Tongji University, 1239 Siping Road, 200092, Shanghai, China
| | - Ziling Zeng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Dong Cui
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jiaguo Huang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Liping Li
- The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, 030032, Taiyuan, China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
| | - Ruiping Zhang
- The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, 030032, Taiyuan, China.
| |
Collapse
|
44
|
Upconversion nanomaterials and delivery systems for smart photonic medicines and healthcare devices. Adv Drug Deliv Rev 2022; 188:114419. [PMID: 35810884 DOI: 10.1016/j.addr.2022.114419] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/24/2022] [Accepted: 07/03/2022] [Indexed: 12/27/2022]
Abstract
In the past decade, upconversion (UC) nanomaterials have been extensively investigated for the applications to photomedicines with their unique features including biocompatibility, near-infrared (NIR) to visible conversion, photostability, controllable emission bands, and facile multi-functionality. These characteristics of UC nanomaterials enable versatile light delivery for deep tissue biophotonic applications. Among various stimuli-responsive delivery systems, the light-responsive delivery process has been greatly advantageous to develop spatiotemporally controllable on-demand "smart" photonic medicines. UC nanomaterials are classified largely to two groups depending on the photon UC pathway and compositions: inorganic lanthanide-doped UC nanoparticles and organic triplet-triplet annihilation UC (TTA-UC) nanomaterials. Here, we review the current-state-of-art inorganic and organic UC nanomaterials for photo-medicinal applications including photothermal therapy (PTT), photodynamic therapy (PDT), photo-triggered chemo and gene therapy, multimodal immunotherapy, NIR mediated neuromodulations, and photochemical tissue bonding (PTB). We also discuss the future research direction of this field and the challenges for further clinical development.
Collapse
|
45
|
He M, Wang Q, Feng Y, Gao X, He C, Li J, Zhao W, Tian W, Zhao C. Spatiotemporal Management of the Osteoimmunomodulation of Fibrous Scaffolds by Loading a Novel Amphiphilic Nanomedicine. ACS APPLIED MATERIALS & INTERFACES 2022; 14:13991-14003. [PMID: 35311248 DOI: 10.1021/acsami.1c20809] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Implanted bone scaffolds or their biodegradation products may disturb the sequential functions of distinct macrophage phenotypes and cause improper timing of macrophage activation, resulting in delayed or dysfunctional bone regeneration. Although spatiotemporal manipulation of the immune response has been recognized as a promising strategy to address this issue, developing satisfactory drug delivery systems with the function of proper timing control on the macrophage phenotype transformation from pro-inflammatory M1 to anti-inflammatory M2 phenotype still remains a challenge. Here, we propose an amphiphilic nanomedicine with dual anti-inflammatory functions and inflammation-responsive drug release properties to spatiotemporally manage the osteoimmunomodulation of the bone scaffold. The nanomedicine enables the modified scaffold to manipulate the immune response in a staged manner, not only avoiding the overinhibition of M1 macrophages in the initial phase but also facilitating its polarization to M2 phenotype, as well as exhibiting full-course inhibition on later biodegradation-induced inflammation. The described immunomodulatory manner attempts to conform to the principle of osteoimmunomodulation, consequently resulting in better in vivo osteogenesis compared with traditional drug delivery systems. We anticipate that this strategy might aid the development of advanced immunomodulatory bone biomaterials.
Collapse
Affiliation(s)
- Min He
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Wang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Yunbo Feng
- College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xinghui Gao
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chao He
- College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Jiangge Li
- College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Weidong Tian
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Changsheng Zhao
- College of Polymer Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
46
|
Zhang J, Lin Y, Lin Z, Wei Q, Qian J, Ruan R, Jiang X, Hou L, Song J, Ding J, Yang H. Stimuli-Responsive Nanoparticles for Controlled Drug Delivery in Synergistic Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103444. [PMID: 34927373 PMCID: PMC8844476 DOI: 10.1002/advs.202103444] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/28/2021] [Indexed: 05/10/2023]
Abstract
Cancer immunotherapy has achieved promising clinical progress over the recent years for its potential to treat metastatic tumors and inhibit their recurrences effectively. However, low patient response rates and dose-limiting toxicity remain as major dilemmas for immunotherapy. Stimuli-responsive nanoparticles (srNPs) combined with immunotherapy offer the possibility to amplify anti-tumor immune responses, where the weak acidity, high concentration of glutathione, overexpressions of enzymes, and reactive oxygen species, and external stimuli in tumors act as triggers for controlled drug release. This review highlights the design of srNPs based on tumor microenvironment and/or external stimuli to combine with different anti-tumor drugs, especially the immunoregulatory agents, which eventually realize synergistic immunotherapy of malignant primary or metastatic tumors and acquire a long-term immune memory to prevent tumor recurrence. The authors hope that this review can provide theoretical guidance for the construction and clinical transformation of smart srNPs for controlled drug delivery in synergistic cancer immunotherapy.
Collapse
Affiliation(s)
- Jin Zhang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Yandai Lin
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Zhe Lin
- Ruisi (Fujian) Biomedical Engineering Research Center Co LtdFuzhou350100P. R. China
| | - Qi Wei
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Jiaqi Qian
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Renjie Ruan
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Xiancai Jiang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Linxi Hou
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| |
Collapse
|
47
|
Cha GD, Lee WH, Sunwoo SH, Kang D, Kang T, Cho KW, Kim M, Park OK, Jung D, Lee J, Choi SH, Hyeon T, Kim DH. Multifunctional Injectable Hydrogel for In Vivo Diagnostic and Therapeutic Applications. ACS NANO 2022; 16:554-567. [PMID: 35014797 DOI: 10.1021/acsnano.1c07649] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Injectable hydrogels show high potential for in vivo biomedical applications owing to their distinctive mode of administration into the human body. In this study, we propose a material design strategy for developing a multifunctional injectable hydrogel with good adhesiveness, stretchability, and bioresorbability. Its multifunctionality, whereupon multiple reactions occur simultaneously during its injection into the body without requiring energy stimuli and/or additives, was realized through meticulous engineering of bioresorbable precursors based on hydrogel chemistry. The multifunctional injectable hydrogel can be administered through a minimally invasive procedure, form a conformal adhesive interface with the target tissue, dynamically stretch along with the organ motions with minimal mechanical constraints, and be resorbed in vivo after a specific period. Further, the incorporation of functional nanomaterials into the hydrogel allows for various in vivo diagnostic and therapeutic applications, without compromising the original multifunctionality of the hydrogel. These features are verified through theranostic case studies on representative organs, including the skin, liver, heart, and bladder.
Collapse
Affiliation(s)
- Gi Doo Cha
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Wang Hee Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung-Hyuk Sunwoo
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dayoung Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Taegyu Kang
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung Won Cho
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Minjeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Dongjun Jung
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jongha Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
48
|
Ni Q, Xu F, Wang Y, Li Y, Qing G, Zhang Y, Zhong J, Li J, Liang XJ. Nanomaterials with changeable physicochemical property for boosting cancer immunotherapy. J Control Release 2022; 342:210-227. [PMID: 34998916 DOI: 10.1016/j.jconrel.2022.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022]
Abstract
The past decade has witnessed a great progress in cancer immunotherapy with the sequential approvals of therapeutic cancer vaccine, immune checkpoint inhibitor and chimeric antigen receptor (CAR) T cell therapy. However, some hurdles still remain to the wide implementation of cancer immunotherapy, including low immune response, complex tumor heterogeneity, off-target immunotoxicity, poor solid tumor infiltration, and immune evasion-induced treatment tolerance. Owing to changeable physicochemical properties in response to endogenous or exogenous stimuli, nanomaterials hold the remarkable potential in incorporation of multiple agents, efficient biological barrier penetration, precise immunomodulator delivery, and controllable content release for boosting cancer immunotherapy. Herein, we review the recent advances in nanomaterials with changeable physicochemical property (NCPP) to develop cancer vaccine, remold tumor microenvironment and evoke direct T cell activation. Besides, we provide our outlook on this emerging field at the intersection of NCPP design and cancer immunotherapy.
Collapse
Affiliation(s)
- Qiankun Ni
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Fengfei Xu
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yufei Wang
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yujie Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Guangchao Qing
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxuan Zhang
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zhong
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (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, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
49
|
Zheng B, Fan J, Chen B, Qin X, Wang J, Wang F, Deng R, Liu X. Rare-Earth Doping in Nanostructured Inorganic Materials. Chem Rev 2022; 122:5519-5603. [PMID: 34989556 DOI: 10.1021/acs.chemrev.1c00644] [Citation(s) in RCA: 255] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Impurity doping is a promising method to impart new properties to various materials. Due to their unique optical, magnetic, and electrical properties, rare-earth ions have been extensively explored as active dopants in inorganic crystal lattices since the 18th century. Rare-earth doping can alter the crystallographic phase, morphology, and size, leading to tunable optical responses of doped nanomaterials. Moreover, rare-earth doping can control the ultimate electronic and catalytic performance of doped nanomaterials in a tunable and scalable manner, enabling significant improvements in energy harvesting and conversion. A better understanding of the critical role of rare-earth doping is a prerequisite for the development of an extensive repertoire of functional nanomaterials for practical applications. In this review, we highlight recent advances in rare-earth doping in inorganic nanomaterials and the associated applications in many fields. This review covers the key criteria for rare-earth doping, including basic electronic structures, lattice environments, and doping strategies, as well as fundamental design principles that enhance the electrical, optical, catalytic, and magnetic properties of the material. We also discuss future research directions and challenges in controlling rare-earth doping for new applications.
Collapse
Affiliation(s)
- Bingzhu Zheng
- State Key Laboratory of Silicon Materials, Institute for Composites Science Innovation, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jingyue Fan
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Bing Chen
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Xian Qin
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Juan Wang
- Institute of Environmental Health, MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental & Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Wang
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Renren Deng
- State Key Laboratory of Silicon Materials, Institute for Composites Science Innovation, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
50
|
Liu J, Liew SS, Wang J, Pu K. Bioinspired and Biomimetic Delivery Platforms for Cancer Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2103790. [PMID: 34651344 DOI: 10.1002/adma.202103790] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/30/2021] [Indexed: 06/13/2023]
Abstract
Cancer vaccines aim at eliciting tumor-specific responses for the immune system to identify and eradicate malignant tumor cells while sparing the normal tissues. Furthermore, cancer vaccines can potentially induce long-term immunological memory for antitumor responses, preventing metastasis and cancer recurrence, thus presenting an attractive treatment option in cancer immunotherapy. However, clinical efficacy of cancer vaccines has remained low due to longstanding challenges, such as poor immunogenicity, immunosuppressive tumor microenvironment, tumor heterogeneity, inappropriate immune tolerance, and systemic toxicity. Recently, bioinspired materials and biomimetic technologies have emerged to play a part in reshaping the field of cancer nanomedicine. By mimicking desirable chemical and biological properties in nature, bioinspired engineering of cancer vaccine delivery platforms can effectively transport therapeutic cargos to tumor sites, amplify antigen and adjuvant bioactivities, and enable spatiotemporal control and on-demand immunoactivation. As such, integration of biomimetic designs into delivery platforms for cancer vaccines can enhance efficacy while retaining good safety profiles, which contributes to expediting the clinical translation of cancer vaccines. Recent advances in bioinspired delivery platforms for cancer vaccines, existing obstacles faced, as well as insights and future directions for the field are discussed here.
Collapse
Affiliation(s)
- Jing Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Si Si Liew
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| |
Collapse
|