1
|
Konjengbam BD, Meitei HN, Pandey A, Haobam R. Goals and strategies in vaccine development against tuberculosis. Mol Immunol 2025; 183:56-71. [PMID: 40327952 DOI: 10.1016/j.molimm.2025.04.016] [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: 10/25/2023] [Revised: 04/16/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), continues to be a major health problem globally. The emergence of multi-drug-resistant TB and extensively drug-resistant TB has become a severe threat to TB control programs. Currently, the Bacille Calmette-Guerin (BCG) vaccine protects a child from disease dissemination efficiently, but its efficiency wanes in adults. Despite all the limitations of BCG and accelerated TB vaccine research, BCG remains the only approved vaccine available for TB. Anti-TB drug treatment has been successful in combating the disease, but it has various side effects and requires an extended drug treatment period. So, vaccination is the finest outlook that can surpass the above-mentioned limitations. Several vaccine candidates are in the pipeline, and the hope for a potential candidate to either boost the BCG vaccine or replace BCG is underway. This review discusses different approaches to TB vaccine development. It summarizes all the challenges and limitations in vaccine development, and its preclinical and clinical trials. Additionally, DNA vaccines and their vaccination techniques are also discussed. Furthermore, the immunoinformatics approach and nanomaterial-based vaccine delivery with practical and productive endpoints are also discussed. Lastly, the potential prospects are also suggested for further studies, which would help bring positive outcomes.
Collapse
Affiliation(s)
| | | | - Anupama Pandey
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, Manipur 795003, India.
| |
Collapse
|
2
|
Hilles AR, Mahmood S, Widodo RT, Azemi AK, Nordin AH, Mohamad Norpi AS, Abd Jalil MF, Sofian ZM, Mohamed M, Hambali KA, Mohd Sani NI, Rashid R, Nordin ML. Cell membrane vaccine delivery system: A review of the recent advances against breast cancer. Int J Pharm 2025; 681:125849. [PMID: 40516769 DOI: 10.1016/j.ijpharm.2025.125849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 06/02/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Natural cells exhibit unique properties, such as homotypic targeting and prolonged circulation in the bloodstream, making them attractive candidates for breast cancer therapy vaccine delivery systems (VDSs). To improve delivery efficiency, these cells can be integrated with nanocarriers, paving the way for next-generation VDSs in the form of biomimetic nanomedicines. This review highlights recent progress in cell membrane-based vaccine delivery systems (CVDSs) and other types of vaccine delivery systems as a potential for breast cancer vaccine development. Cell membrane sources, including red blood cells (RBCs), cancer cells, and platelets are explored. Additionally, the methods for extracting these membranes and the mechanisms driving the functionality of biomimetic CVDSs are detailed. This review also highlights recent advancements and cutting-edge strategies of cell membrane vaccine delivery systems, with a particular focus on the use of cell-derived biomimetic nanotechnology for breast cancer. It also highlights breast cancer vaccines under development encompass protein-based (e.g., peptide vaccines), nucleic acid-based (DNA and RNA vaccines), immune cell-based (including dendritic cell and autologous tumor cell vaccines), and bacterial/viral vector-based vaccines. Each platform aims to elicit targeted anti-tumor immune responses.
Collapse
Affiliation(s)
- Ayah Rebhi Hilles
- INHART, International Islamic University Malaysia, Jalan Gombak, 53100 Kuala Lumpur, Malaysia
| | - Syed Mahmood
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Universiti Malaya Research Centre for Pharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Centre of Advanced Materials (CAM), Faculty of Engineering, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathum Wan, Bangkok 10330, Thailand
| | - Riyanto Teguh Widodo
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Universiti Malaya Research Centre for Pharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ahmad Khusairi Azemi
- Institute of Climate Change Adaptation and Marine Biotechnology, Universiti Malaysia Terengganu 21030 Kuala Terengganu, Terengganu, Malaysia
| | - Abu Hassan Nordin
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM), Campus Arau, 02600 Perlis, Malaysia
| | - Abdin Shakirin Mohamad Norpi
- Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur, Royal College of Medicine Perak, Perak 30450, Malaysia
| | - Muhammad Fauzi Abd Jalil
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Zarif Mohamed Sofian
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Universiti Malaya Research Centre for Pharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mazlan Mohamed
- Faculty of Data Science and Computing, Universiti Malaysia Kelantan, City Campus Pengkalan Chepa, Kota Bharu 16100 Kelantan, Malaysia
| | - Kamarul Ariffin Hambali
- Faculty of Earth Science, Universiti Malaysia Kelantan, Jeli 17600 Kelantan, Malaysia; Animal and Wildlife Research Group, Faculty of Earth Science, Jeli Campus, Universiti Malaysia Kelantan, Jeli 17600 Kelantan, Malaysia
| | - Nani Izreen Mohd Sani
- Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa 16100 Kelantan, Malaysia
| | - Roslina Rashid
- Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa 16100 Kelantan, Malaysia
| | - Muhammad Luqman Nordin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia; Universiti Malaya Research Centre for Pharmaceuticals and Advanced Therapeutics (UBAT), Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
3
|
Ghazizadeh Y, Salehi Shadkami H, Madani F, Niknam S, Adabi M. Advances in cancer nanovaccines: a focus on colorectal cancer. Nanomedicine (Lond) 2025; 20:1029-1041. [PMID: 40186876 PMCID: PMC12051617 DOI: 10.1080/17435889.2025.2486930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Nanotechnology has revolutionized cancer treatment by providing innovative solutions through nanocancer therapies, nanovaccines, and nanoparticles. This review focuses on the application of these technologies in colorectal cancer (CRC), highlighting their progression from preclinical studies to clinical trials. Nanoparticles, including liposomes, silica, gold, and lipid nanoparticles, possess unique properties that enhance drug delivery, improve therapeutic efficacy, and minimize systemic toxicity. Additionally, nanovaccines are being developed to elicit robust immune responses against CRC cells. This paper offers a comprehensive overview of the current state of nanotechnology-based treatments for CRC, emphasizing key preclinical studies and clinical trials that demonstrate their potential. Furthermore, the review discusses the challenges faced in this field. It outlines future directions for research, underscoring the need for ongoing efforts to translate these promising technologies into practical clinical applications.
Collapse
Affiliation(s)
- Yalda Ghazizadeh
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi Shadkami
- Nanomedicine Student Association (NMA), Student’s Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Madani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedigheh Niknam
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan, Iran
| | - Mahdi Adabi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Food Microbiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Ding B, Li J, Tan J, Chen H, Zheng P, Ma P, Lin J. Accelerating Tumor Immunotherapy Through a Synergistic Strategy of Increasing Throttle and Relaxing Brake. Angew Chem Int Ed Engl 2025; 64:e202422502. [PMID: 39814603 DOI: 10.1002/anie.202422502] [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: 11/19/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/18/2025]
Abstract
Tumor immunotherapy has been widely used clinically, but it is still hindered by weak antitumor immunity and immunosuppressive tumor microenvironment (TME). Here, a kind of simple disodium hydrogen phosphate nanoparticle (Na2HPO4 NP) is prepared to "accelerate" tumor immunotherapy by "increasing throttle" and "relaxing brake" simultaneously. The obtained Na2HPO4 NPs release a large amount of Na+ and HPO4 2- ions within tumor cells, thereby activating the caspase 1/GSDMD-mediated pyroptosis pathway to achieve immune activation. Meanwhile, alkalescent Na2HPO4 NPs can further consume lactic acid through acid-base neutralization, and regulate adenosine (Ado) metabolism via nanomaterial-induced biocatalytic process to relieve two-tier immunosuppression. Collectively, Na2HPO4 NPs effectively activate the antitumor immune process in vivo, and dramatically inhibit primary and distal tumor growth. This work will provide unique inspiration and strategy for the regulation of both positive and negative directions in immunotherapy.
Collapse
Affiliation(s)
- Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Superlight Materials & Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
5
|
Zhao Y, Tian M, Tong X, Yang X, Gan L, Yong T. Emerging strategies in lymph node-targeted nano-delivery systems for tumor immunotherapy. Essays Biochem 2025; 69:EBC20253008. [PMID: 40159756 DOI: 10.1042/ebc20253008] [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: 01/13/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
The emergence of immunotherapy has led to the clinical approval of several related drugs. However, their efficacy against solid tumors remains limited. As the hub of immune activation, lymph nodes (LNs) play a critical role in tumor immunotherapy by initiating and amplifying immune responses. Nevertheless, the intricate physiological structure and barriers within LNs, combined with the immunosuppressive microenvironment induced by tumor cells, significantly impede the therapeutic efficacy of immunotherapy. Engineered nanoparticles (NPs) have shown great potential in overcoming these challenges by facilitating targeted drug transport to LNs and directly or indirectly activating T cells. This review systematically examines the structural features of LNs, key factors influencing the targeting efficiency of NPs, and current strategies for remodeling the immunosuppressive microenvironment of LNs. Additionally, it discusses future opportunities for optimizing NPs to enhance tumor immunotherapy, addressing challenges in clinical translation and safety evaluation.
Collapse
Affiliation(s)
- Yaoli Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Muzi Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Tong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
6
|
Mbaye EHA, Scott EA, Burke JA. From Edmonton to Lantidra and beyond: immunoengineering islet transplantation to cure type 1 diabetes. FRONTIERS IN TRANSPLANTATION 2025; 4:1514956. [PMID: 40182604 PMCID: PMC11965681 DOI: 10.3389/frtra.2025.1514956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing β cells within pancreatic islets, the specialized endocrine cell clusters of the pancreas. Islet transplantation has emerged as a β cell replacement therapy, involving the infusion of cadaveric islets into a patient's liver through the portal vein. This procedure offers individuals with T1D the potential to restore glucose control, reducing or even eliminating the need for exogenous insulin therapy. However, it does not address the underlying autoimmune condition responsible for T1D. The need for systemic immunosuppression remains the primary barrier to making islet transplantation a more widespread therapy for patients with T1D. Here, we review recent progress in addressing the key limitations of islet transplantation as a viable treatment for T1D. Concerns over systemic immunosuppression arise from its potential to cause severe side effects, including opportunistic infections, malignancies, and toxicity to transplanted islets. Recognizing the risks, the Edmonton protocol (2000) marked a shift away from glucocorticoids to prevent β cell damage specifically. This transition led to the development of combination immunosuppressive therapies and the emergence of less toxic immunosuppressive and anti-inflammatory drugs. More recent advances in islet transplantation derive from islet encapsulation devices, biomaterial platforms releasing immunomodulatory compounds or surface-modified with immune regulating ligands, islet engineering and co-transplantation with accessory cells. While most of the highlighted studies in this review remain at the preclinical stage using mouse and non-human primate models, they hold significant potential for clinical translation if a transdisciplinary research approach is prioritized.
Collapse
Affiliation(s)
- El Hadji Arona Mbaye
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
7
|
So Y, Yim D, Kim HK, Lee S, Lee H, Yu Y, Choi C, Choi Y, Kim H, Yang CS, Kim JH. Functional Nanosheet Immunoswitches Reprogramming Innate Macrophages for Immunotherapy of Colorectal Cancer and Sepsis. ACS NANO 2025; 19:5165-5177. [PMID: 39898465 DOI: 10.1021/acsnano.4c08828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Macrophages are involved in the immunopathogenesis of cancer and inflammatory diseases and are a primary target for immunotherapy to reprogram the M1 and M2 phenotypes in tumor and inflammatory microenvironments. Herein, functional nanosheet immunoswitches that can bidirectionally polarize macrophages in tumor and inflammatory microenvironments are designed for effective immunotherapy of colorectal cancer and sepsis. WSe2 nanosheets are functionalized with palmitic acid to obtain an M1 immunoswitch (PA-WSe2) that promotes the polarization of macrophages toward the M1 phenotype in the tumor microenvironment by activating the STAT1 signaling pathway. WS2 nanosheets bearing linoleic acid are synthesized as an M2 immunoswitch (LA-WS2) that effectively polarizes macrophages to the M2 phenotype in the septic microenvironment by activating the STAT3 signaling pathway. The PA-WSe2 M1 immunoswitch upregulates the secretion of pro-inflammatory cytokines and reactive oxygen and nitrogen species (ROS and RNS) via M1 polarization, leading to the effective immunotherapy for colorectal cancer in vivo. In contrast, the LA-WS2 M2 immunoswitch induces the elevated production of anti-inflammatory cytokines and scavenging of ROS and RNS through M2 polarization, resulting in superior immunotherapy for severe sepsis in mice. These nanosheet immunoswitches can provide a route to immunotherapy for various cancers and inflammatory diseases.
Collapse
Affiliation(s)
- Yoonhee So
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - DaBin Yim
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720, United States
| | - Hyo Keun Kim
- Department of Molecular and Life Science, and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Sin Lee
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Hyunji Lee
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Yejoo Yu
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Chanhee Choi
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Yujin Choi
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Hongwon Kim
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, Republic of Korea
| | - Jong-Ho Kim
- Department of Materials Science and Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
8
|
Lv Y, Chen Z, Wang S, Zou M. Macrophage-Mediated Liquid Metal Nanoparticles for Enhanced Tumor Accumulation and Inhibition. ACS Biomater Sci Eng 2025; 11:903-915. [PMID: 39855913 DOI: 10.1021/acsbiomaterials.4c01130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
In most studies, the penetration of nanoparticles into tumors was mainly dependent on the enhanced permeability and retention (ERP) effect. However, the penetration of nanoparticles would be limited by tumor-dense structure, immune system, and other factors. To solve these problems, macrophages with active tropism to tumor tissues, loaded nanoparticles with photothermal therapy, and chemotherapy were designed. In detail, liquid metal (gallium indium alloy) nanoparticles were modified with mesoporous silica and then embedded with the chemotherapeutic drug sorafenib (LM@Si/SO) for photothermal therapy and chemotherapy. After that, the LM@Si/SO nanoparticles were carried by the mouse macrophage RAW264.7 cell line (LM@Si/SO@R) to increase the accumulation of the nanoparticles in the tumor site and improve the tumor immune microenvironment. With the enhanced tumor accumulation, LM@Si/SO@R exhibited excellent antitumor ability in vitro and in vivo. Thus, these strategies via the cell carrier to enhance tumor therapeutic efficiency had the potential for the improvement of tumor therapy.
Collapse
Affiliation(s)
- Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China
| | - Zhenghang Chen
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Shuai Wang
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Meizhen Zou
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China
| |
Collapse
|
9
|
Lu T, Lin W, Guo Y, Shao M, Bai Y, Tommaso DD, Wang X, Zhang X. Metal nanoparticles encapsulation within multi-shell spongy-core porous microspheres for efficient tandem catalysis. J Colloid Interface Sci 2025; 679:705-713. [PMID: 39388956 DOI: 10.1016/j.jcis.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
The "one-pot" cascade process involves multiple catalytic conversions followed by a single workup stage. This method has the capability to optimize catalytic efficiency by reducing chemical processes. The key to achieving cascade reactions lies in designing cascade catalysts with well-dispersed, stably immobilized, and accessible noble metal nanoparticles for multiple catalytic conversions. This work presents a strategy for creating long-lasting cascade catalysts by encapsulating Ru and Pd nanoparticles within multi-shell spongy-core porous microspheres (MS-SC-PMs). This cascade catalyst strategy enables the continuous hydrogenation of nitrobenzene to aniline and further to cyclohexylamine, demonstrating both high selectivity and conversion rates. Notably, this approach overcomes the typical challenges associated with noble metal nanoparticles, such as poor stability and recyclability, as it maintains its performance over ten consecutive cycles. Additionally, the MS-SC-PMs have the versatility to encapsulate various metal nanoparticles, providing catalytic versatility, scalability, and a promising avenue for designing long-lasting catalysts loaded with nanoparticles.
Collapse
Affiliation(s)
- Tao Lu
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Wuyang Lin
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Yingchun Guo
- Huzhou Key Laboratory of Environmental Functional Materials and Pollution Control, Huzhou University, Huzhou 313000, China
| | - Mengliu Shao
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Yuanyuan Bai
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Devis Di Tommaso
- Department of Chemistry, School of Physical and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, UK; Digital Environment Research Institute, Queen Mary University of London, Empire House, 67-75 New Road, London E1 1HH, UK.
| | - Xiaomei Wang
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China.
| | - Xu Zhang
- Hebei Key Laboratory of Functional Polymers, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China; National-Local Joint Engineering Laboratory for Energy Conservation of Chemical Process Integration and Resources Utilization, Hebei University of Technology, China.
| |
Collapse
|
10
|
Saleh M, El-Moghazy A, Elgohary AH, Saber WIA, Helmy YA. Revolutionizing Nanovaccines: A New Era of Immunization. Vaccines (Basel) 2025; 13:126. [PMID: 40006673 PMCID: PMC11860605 DOI: 10.3390/vaccines13020126] [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: 11/16/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Infectious diseases continue to pose a significant global health threat. To combat these challenges, innovative vaccine technologies are urgently needed. Nanoparticles (NPs) have unique properties and have emerged as a promising platform for developing next-generation vaccines. Nanoparticles are revolutionizing the field of vaccine development, offering a new era of immunization. They allow the creation of more effective, stable, and easily deliverable vaccines. Various types of NPs, including lipid, polymeric, metal, and virus-like particles, can be employed to encapsulate and deliver vaccine components, such as mRNA or protein antigens. These NPs protect antigens from degradation, target them to specific immune cells, and enhance antigen presentation, leading to robust and durable immune responses. Additionally, NPs can simultaneously deliver multiple vaccine components, including antigens, and adjuvants, in a single formulation, simplifying vaccine production and administration. Nanovaccines offer a promising approach to combat food- and water-borne bacterial diseases, surpassing traditional formulations. Further research is needed to address the global burden of these infections. This review highlights the potential of NPs to revolutionize vaccine platforms. We explore their mechanisms of action, current applications, and emerging trends. The review discusses the limitations of nanovaccines, innovative solutions and the potential role of artificial intelligence in developing more effective and accessible nanovaccines to combat infectious diseases.
Collapse
Affiliation(s)
- Mohammed Saleh
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Ahmed El-Moghazy
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA 92521, USA
| | - Adel H. Elgohary
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - WesamEldin I. A. Saber
- Microbial Activity Unit, Department of Microbiology, Soils, Water and Environment Research Institute, Agricultural Research Center, Giza 12619, Egypt
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food, and Environment, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|
11
|
Xu S, Zhao Z, Sun C, Ji Y, Luan Q, Zhang Q, Jin Z, Zhao K. Immunoprotective effect of chitosan nanoparticles with different particle sizes against H9N2 avian influenza infection. Poult Sci 2025; 104:104559. [PMID: 39603189 PMCID: PMC11635735 DOI: 10.1016/j.psj.2024.104559] [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: 06/15/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
H9N2 is the most common avian influenza virus (AIV), which causes significant losses in chickens. Safe and effective vaccines are crucial for the prevention of H9N2 AIVs. Chitosan nanoparticles, as novel adjuvants, enhance vaccine immunity and biocompatibility; however, the impact of particle size on the immunological effects remains underexplored. To solve these problems and to prepare an efficient novel H9N2 vaccine, we constructed four N-2-HACC/CMCS NPs (NHC NPs) of different particle sizes (165.6 ± 12.0 nm, 272.5 ± 7.0 nm, 343.2 ± 8.0 nm, and 443.5 ± 15.0 nm). Subsequent in vivo immunogenicity screening revealed that H9N2 with the 272.5 ± 7.0 nm NHC NPs vaccine group induced higher levels of neutralizing antibodies in the early stage of the immune response, while the 343.2 ± 8.0 nm NHC NPs vaccine group induced higher levels of neutralizing antibodies in the late stages of the immune response. Subsequently, the results of the optimal particle size combination screening revealed that more neutralizing antibodies were induced when the NHC NPs particle size combination of 272.5 ± 7.0 nm:343.2 ± 8.0 nm ratio was 1.5:1. This optimal particle size combination for NP vaccines promoted lymphocyte proliferation, induced higher IgG2a/IgG1 ratios, and promoted the production of cytokines (i.e., IL-2, IL-4, and IFN-γ). Moreover, a mechanistic analysis revealed that the optimal NHC NPs combination triggered the activation of antigen presenting cells via TLR4 and participated in immune responses through the production of NO and TNF-α. Taken together, our study revealed that the optimal combination of NHC NPs may be a promising strategy against influenza viruses.
Collapse
Affiliation(s)
- Shangen Xu
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Zhi Zhao
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Chenxi Sun
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Yile Ji
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Qingshuang Luan
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Qihong Zhang
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Zheng Jin
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China
| | - Kai Zhao
- Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Institute of Nanobiomaterials and Immunology, School of Life Sciences, Taizhou University, Taizhou Zhejiang 318000, China.
| |
Collapse
|
12
|
Bixenmann L, Ahmad T, Stephan F, Nuhn L. End-Group Dye-Labeled Poly(hemiacetal ester) Block Copolymers: Enhancing Hydrolytic Stability and Loading Capacity for Micellar (Immuno-)Drug Delivery. Biomacromolecules 2024; 25:7958-7974. [PMID: 39509250 DOI: 10.1021/acs.biomac.4c01229] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Polymers with hemiacetal esters integrated in their backbone provide beneficial degradation profiles for (immuno-) drug delivery. However, their fast hydrolysis and low drug loading capacity have limited their applications so far. Therefore, this study focuses on the stability and loading capacity of hemiacetal ester polymers. The hydrophobicity of the micellar core has a tremendous effect on the hemiacetal ester stability. For that purpose, we introduce a new monomer with a phenyl moiety for stabilizing the micellar core and improving drug loading. The carrier functionality can further be expanded by post-polymerization modifications via activated ester groups at the polymer chain end. This allows for covalent dye labeling, which provides substantial insights into the polymers' in vitro performance. Flow cytometric analyses on RAW dual macrophages revealed intact micelles exhibiting significantly higher cellular uptake compared to degraded species, thus, highlighting the potential of end group functionalized poly(hemiacetal ester)s for (immuno)drug delivery purposes.
Collapse
Affiliation(s)
- Leon Bixenmann
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, 97070 Würzburg, Germany
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Taufiq Ahmad
- Institute of Functional Materials and Biofabrication, Department of Functional Materials in Medicine and Dentistry, University Hospital of Würzburg, 97070 Würzburg, Germany
| | - Fabian Stephan
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Lutz Nuhn
- Institute of Functional Materials and Biofabrication, Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, 97070 Würzburg, Germany
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| |
Collapse
|
13
|
Heck AG, Medina-Montano C, Zhong Z, Deswarte K, Eigen K, Stickdorn J, Kockelmann J, Scherger M, Sanders NN, Lienenklaus S, Lambrecht BN, Grabbe S, De Geest BG, Nuhn L. PH-Triggered, Lymph Node Focused Immunodrug Release by Polymeric 2-Propionic-3-Methyl-maleic Anhydrides with Cholesteryl End Groups. Adv Healthc Mater 2024; 13:e2402875. [PMID: 39313985 DOI: 10.1002/adhm.202402875] [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: 08/17/2024] [Indexed: 09/25/2024]
Abstract
Gaining spatial control over innate immune activation is of great relevance during vaccine delivery and anticancer therapy, where one aims at activating immune cells at draining lymphoid tissue while avoiding systemic off-target innate immune activation. Lipid-polymer amphiphiles show high tendency to drain to lymphoid tissue upon local administration. Here, pH-sensitive, cholesteryl end group functionalized polymers as stimuli-responsive carriers are introduced for controlled immunoactivation of draining lymph nodes. Methacrylamide-based monomers bearing pendant 2-propionic-3-methylmaleic anhydride groups are polymerized by Reversible Addition-Fragmentation Chain Transfer (RAFT) polymerization using a cholesterol chain-transfer agent (chol-CTA). The amine-reactive anhydrides are conjugated with various amines, however, while primary amines afforded irreversible imides, secondary amines provided pH-responsive conjugates that are released upon acidification. This can be applied to fluorescent dyes for irreversibly carrier labeling or immunostimulatory Toll-like receptor (TLR) 7/8 agonists as cargos for pH-responsive delivery. Hydrophilization of remaining anhydride repeating units with short PEG-chains yielded cholesteryl-polymer amphiphiles that showed efficient cellular uptake and increased drug release at endosomal pH. Moreover, reversibly conjugated TLR 7/8 agonist amphiphiles efficiently drained to lymph nodes and increased the number of effectively maturated antigen-presenting cells after subcutaneous injection in vivo. Consequently, cholesteryl-linked methacrylamide-based polymers with pH-sensitive 2-propionic-3-methylmaleic anhydride side groups provide ideal features for immunodrug delivery.
Collapse
Affiliation(s)
- Alina G Heck
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Zifu Zhong
- Department of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, 9000, Belgium
| | - Kim Deswarte
- Department of Internal Medicine and Pediatrics, VIB Center for Inflammation Research, Ghent University, Ghent, 9052, Belgium
| | - Katharina Eigen
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
| | - Judith Stickdorn
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Johannes Kockelmann
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
| | | | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Ghent University, Merelbeke, 9820, Belgium
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science and Institute of Immunology, Hannover Medical School, 30625, Hanover, Germany
| | - Bart N Lambrecht
- Department of Internal Medicine and Pediatrics, VIB Center for Inflammation Research, Ghent University, Ghent, 9052, Belgium
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center (UMC) of the Johannes Gutenberg-University Mainz, 55131, Mainz, Germany
| | - Bruno G De Geest
- Department of Pharmaceutics and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, 9000, Belgium
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Julius-Maximilians-Universität Würzburg, 97070, Würzburg, Germany
- Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| |
Collapse
|
14
|
Wang J, Zhao Z, Wang Q, Shi J, Wong DWC, Cheung JCW. Advancements in Nanoparticle-Based Adjuvants for Enhanced Tuberculosis Vaccination: A Review. Vaccines (Basel) 2024; 12:1335. [PMID: 39771997 PMCID: PMC11680411 DOI: 10.3390/vaccines12121335] [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: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Tuberculosis (TB) remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective vaccines. Nanoparticle-based adjuvants represent a promising approach to enhancing tuberculosis vaccine efficacy. This review focuses on the advantages of nanoparticulate-loaded vaccines, emphasizing their ability to improve antigen delivery, safety, and immunogenicity. We discuss the various types of nanoparticles and their unique physicochemical properties that contribute to improved antigen delivery and sustained immune activation. Additionally, we highlight the advantages of nanoparticle-based adjuvants in inducing strong cellular and humoral immunity, enhancing vaccine stability, and reducing adverse effects. Finally, we address current challenges and future perspectives in the application of these novel adjuvants, emphasizing their potential to transform TB vaccine strategies and ultimately contribute to better global health outcomes.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430073, China
| | - Zian Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Quan Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jingyu Shi
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Duo Wai-Chi Wong
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - James Chung-Wai Cheung
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
15
|
Bridgeman CJ, Shen R, McIlvaine RA, Edwards C, Ackun-Farmmer MA, Jewell CM. Synthetic organic materials for targeting immunotherapies to lymph nodes. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2024; 36:9031-9045. [PMID: 40405914 PMCID: PMC12094523 DOI: 10.1021/acs.chemmater.4c00947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Immunotherapies have yielded tremendous advances over the last three decades. However even the most promising therapies, for example monoclonal antibodies, require systemic infusion that can limit dosing and lead to off target immunotoxicity. To address such challenges and improve immunotherapy, the field is investing in synthetic biomaterials to target lymph nodes (LNs) - sites of coordinated immune activation and suppression. These synthetic materials allow enhanced targeting, retention, and control over the signals that are required to elicit desired immune processes during immunotherapy. Two broad classes of materials that have been employed for LN targeting include synthetic lipids and polymers. This review will discuss how the chemistries of these materials can be leveraged to improve lymph node targeting of immunotherapies to treat disease. We will also provide commentary on translational barriers to the clinic, an outlook on current therapies that are clinically used, and a forward-looking perspective.
Collapse
Affiliation(s)
- Christopher J. Bridgeman
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, College Park, Maryland, 20742, United States
| | - Ruochen Shen
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, College Park, Maryland, 20742, United States
| | - Ryan A. McIlvaine
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, College Park, Maryland, 20742, United States
| | - Camilla Edwards
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, College Park, Maryland, 20742, United States
| | - Marian A. Ackun-Farmmer
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, College Park, Maryland, 20742, United States
| | - Christopher M. Jewell
- Robert E Fischell Institute of Biomedical Devices, University of Maryland College Park, College Park, Maryland, 20742, United States
- United States Department of Veterans Affairs, Baltimore, Maryland, 21201, United States
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, Maryland, 21205, United States
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, Maryland, 21201, United States
| |
Collapse
|
16
|
Gonzalez-Melero L, Santos-Vizcaino E, Varela-Calvino R, Gomez-Tourino I, Asumendi A, Boyano MD, Igartua M, Hernandez RM. PLGA-PEI nanoparticle covered with poly(I:C) for personalised cancer immunotherapy. Drug Deliv Transl Res 2024; 14:2788-2803. [PMID: 38427275 PMCID: PMC11525302 DOI: 10.1007/s13346-024-01557-2] [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] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Melanoma is the main cause of death among skin cancers and its incidence worldwide has been experiencing an appalling increase. However, traditional treatments lack effectiveness in advanced or metastatic patients. Immunotherapy, meanwhile, has been shown to be an effective treatment option, but the rate of cancers responding remains far from ideal. Here we have developed a personalized neoantigen peptide-based cancer vaccine by encapsulating patient derived melanoma neoantigens in polyethylenimine (PEI)-functionalised poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) and coating them with polyinosinic:polycytidylic acid (poly(I:C)). We found that PLGA NPs can be effectively modified to be coated with the immunoadjuvant poly(I:C), as well as to encapsulate neoantigens. In addition, we found that both dendritic cells (DCs) and lymphocytes were effectively stimulated. Moreover, the developed NP was found to have a better immune activation profile than NP without poly(I:C) or without antigen. Our results demonstrate that the developed vaccine has a high capacity to activate the immune system, efficiently maturing DCs to present the antigen of choice and promoting the activity of lymphocytes to exert their cytotoxic function. Therefore, the immune response generated is optimal and specific for the elimination of melanoma tumour cells.
Collapse
Affiliation(s)
- Lorena Gonzalez-Melero
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain
| | - Ruben Varela-Calvino
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Santiago de Compostela, Santiago, Spain
| | - Iria Gomez-Tourino
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago, Spain
| | - Aintzane Asumendi
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Maria Dolores Boyano
- Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
- Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
17
|
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
|
18
|
Wei F, Liu H, Wang Y, Li Y, Han S. Engineering macrophages and their derivatives: A new hope for antitumor therapy. Biomed Pharmacother 2024; 177:116925. [PMID: 38878637 DOI: 10.1016/j.biopha.2024.116925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 07/28/2024] Open
Abstract
Macrophages are central to the immune system and are found in nearly all tissues. Recently, the development of therapies based on macrophages has attracted significant interest. These therapies utilize macrophages' key roles in immunity, their ability to navigate biological barriers, and their tendency to accumulate in tumors. This review explores the advancement of macrophage-based treatments. We discuss the bioengineering of macrophages for improved anti-tumor effects, the use of CAR macrophage therapy for targeting cancer cells, and macrophages as vehicles for therapeutic delivery. Additionally, we examine engineered macrophage products, like extracellular vesicles and membrane-coated nanoparticles, for their potential in precise and less toxic tumor therapy. Challenges in moving these therapies from research to clinical practice are also highlighted. The aim is to succinctly summarize the current status, challenges, and future directions of engineered macrophages in cancer therapy.
Collapse
Affiliation(s)
- Fang Wei
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Haiyang Liu
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Yuxiao Wang
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China
| | - Yan Li
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China.
| | - Shuo Han
- Department of Cardiology, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province 110032, China.
| |
Collapse
|
19
|
Hazra R, Chattopadhyay S, Mallick A, Gayen S, Roy S. Revealing the therapeutic properties of gut microbiota: transforming cancer immunotherapy from basic to clinical approaches. Med Oncol 2024; 41:175. [PMID: 38874788 DOI: 10.1007/s12032-024-02416-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
The immune system plays a pivotal role in the battle against cancer, serving as a formidable guardian in the ongoing fight against malignant cells. To combat these malignant cells, immunotherapy has emerged as a prevalent approach leveraging antibodies and peptides such as anti-PD-1, anti-PD-L1, and anti-CTLA-4 to inhibit immune checkpoints and activate T lymphocytes. The optimization of gut microbiota plays a significant role in modulating the defense system in the body. This study explores the potential of certain gut-resident bacteria to amplify the impact of immunotherapy. Contemporary antibiotic treatments, which can impair gut flora, may diminish the efficacy of immune checkpoint blockers. Conversely, probiotics or fecal microbiota transplantation can help re-establish intestinal microflora equilibrium. Additionally, the gut microbiome has been implicated in various strategies to counteract immune resistance, thereby enhancing the success of cancer immunotherapy. This paper also acknowledges cutting-edge technologies such as nanotechnology, CAR-T therapy, ACT therapy, and oncolytic viruses in modulating gut microbiota. Thus, an exhaustive review of literature was performed to uncover the elusive link that could potentiate the gut microbiome's role in augmenting the success of cancer immunotherapy.
Collapse
Affiliation(s)
- Rudradeep Hazra
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, Kolkata-Group of Institutions, NSHM Knowledge Campus, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
20
|
Özcolak B, Erenay B, Odabaş S, Jandt KD, Garipcan B. Effects of bone surface topography and chemistry on macrophage polarization. Sci Rep 2024; 14:12721. [PMID: 38830871 PMCID: PMC11148019 DOI: 10.1038/s41598-024-62484-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
Surface structure plays a crucial role in determining cell behavior on biomaterials, influencing cell adhesion, proliferation, differentiation, as well as immune cells and macrophage polarization. While grooves and ridges stimulate M2 polarization and pits and bumps promote M1 polarization, these structures do not accurately mimic the real bone surface. Consequently, the impact of mimicking bone surface topography on macrophage polarization remains unknown. Understanding the synergistic sequential roles of M1 and M2 macrophages in osteoimmunomodulation is crucial for effective bone tissue engineering. Thus, exploring the impact of bone surface microstructure mimicking biomaterials on macrophage polarization is critical. In this study, we aimed to sequentially activate M1 and M2 macrophages using Poly-L-Lactic acid (PLA) membranes with bone surface topographical features mimicked through the soft lithography technique. To mimic the bone surface topography, a bovine femur was used as a model surface, and the membranes were further modified with collagen type-I and hydroxyapatite to mimic the bone surface microenvironment. To determine the effect of these biomaterials on macrophage polarization, we conducted experimental analysis that contained estimating cytokine release profiles and characterizing cell morphology. Our results demonstrated the potential of the hydroxyapatite-deposited bone surface-mimicked PLA membranes to trigger sequential and synergistic M1 and M2 macrophage polarizations, suggesting their ability to achieve osteoimmunomodulatory macrophage polarization for bone tissue engineering applications. Although further experimental studies are required to completely investigate the osteoimmunomodulatory effects of these biomaterials, our results provide valuable insights into the potential advantages of biomaterials that mimic the complex microenvironment of bone surfaces.
Collapse
Affiliation(s)
- Birgün Özcolak
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Berkay Erenay
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey
| | - Sedat Odabaş
- Biomaterials and Tissue Engineering Laboratory (bteLAB), Department of Chemistry, Faculty of Science, Ankara University, 06560, Ankara, Turkey
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, 06560, Ankara, Turkey
| | - Klaus D Jandt
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research, Faculty of Physics and Astronomy, Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
| | - Bora Garipcan
- Biomimetic and Bioinspired Biomaterials Research Laboratory, Institute of Biomedical Engineering, Boğaziçi University, 34684, Istanbul, Turkey.
| |
Collapse
|
21
|
Jungcharoen P, Panaampon J, Imemkamon T, Saengboonmee C. Magnetic nanoparticles: An emerging nanomedicine for cancer immunotherapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:183-214. [PMID: 39461752 DOI: 10.1016/bs.pmbts.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Cancer immunotherapy is a revolutionised strategy that strikingly improves cancer treatment in recent years. However, like other therapeutic modalities, immunotherapy faces several challenges and limitations. Many methods have been developed to overcome those limitations; thus, nanomedicine is one of the emerging fields with a highly promising application. Magnetite nanoparticles (MNPs) have long been used for medical applications, for example, as a contrast medium, and are being investigated as a tool for boosting and synergizing the effects of immunotherapy. With known physicochemical properties and the interaction with the surroundings in biological systems, MNPs are used to improve the efficacy of immunotherapy in both cell-based and antibody-based treatment. This chapter reviews and discusses state-of-the-art MNPs as a tool to advance cancer immunotherapy as well as its limitations that need further investigation for a better therapeutic outcome in preclinical and clinical settings.
Collapse
Affiliation(s)
- Phoomipat Jungcharoen
- Department of Environmental Engineering, Faculty of Engineering, Khon Kaen University, Khon Kaen, Thailand
| | - Jutatip Panaampon
- Division of Hematologic Neoplasm, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection Kumamoto University, Kumamoto, Japan
| | - Thanit Imemkamon
- Division of Medical Oncology, Department of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
22
|
Kane G, Lusi C, Brassil M, Atukorale P. Engineering approaches for innate immune-mediated tumor microenvironment remodeling. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 21:100406. [PMID: 38213392 PMCID: PMC10777078 DOI: 10.1016/j.iotech.2023.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Cancer immunotherapy offers transformative promise particularly for the treatment of lethal cancers, since a correctly trained immune system can comprehensively orchestrate tumor clearance with no need for continued therapeutic intervention. Historically, the majority of immunotherapies have been T cell-focused and have included immune checkpoint inhibitors, chimeric antigen receptor T cells, and T-cell vaccines. Unfortunately T-cell-focused therapies have failed to achieve optimal efficacy in most solid tumors largely because of a highly immunosuppressed 'cold' or immune-excluded tumor microenvironment (TME). Recently, a rapidly growing treatment paradigm has emerged that focuses on activation of tumor-resident innate antigen-presenting cells, such as dendritic cells and macrophages, which can drive a proinflammatory immune response to remodel the TME from 'cold' or immune-excluded to 'hot'. Early strategies for TME remodeling centered on free cytokines and agonists, but these approaches have faced significant hurdles in both delivery and efficacy. Systemic toxicity from off-target inflammation is a paramount concern in these therapies. To address this critical gap, engineering approaches have provided the opportunity to add 'built-in' capabilities to cytokines, agonists, and other therapeutic agents to mediate improved delivery and efficacy. Such capabilities have included protective encapsulation to shield them from degradation, targeting to direct them with high specificity to tumors, and co-delivery strategies to harness synergistic proinflammatory pathways. Here, we review innate immune-mediated TME remodeling engineering approaches that focus on cytokines, innate immune agonists, immunogenic viruses, and cell-based methods, highlighting emerging preclinical approaches and strategies that are either being tested in clinical trials or already Food and Drug Administration approved.
Collapse
Affiliation(s)
- G.I. Kane
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - C.F. Lusi
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - M.L. Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
| | - P.U. Atukorale
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst
- University of Massachusetts Cancer Center, Worcester
- Division of Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, USA
| |
Collapse
|
23
|
García-Domínguez DJ, López-Enríquez S, Alba G, Garnacho C, Jiménez-Cortegana C, Flores-Campos R, de la Cruz-Merino L, Hajji N, Sánchez-Margalet V, Hontecillas-Prieto L. Cancer Nano-Immunotherapy: The Novel and Promising Weapon to Fight Cancer. Int J Mol Sci 2024; 25:1195. [PMID: 38256268 PMCID: PMC10816838 DOI: 10.3390/ijms25021195] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer is a complex disease that, despite advances in treatment and the greater understanding of the tumor biology until today, continues to be a prevalent and lethal disease. Chemotherapy, radiotherapy, and surgery are the conventional treatments, which have increased the survival for cancer patients. However, the complexity of this disease together with the persistent problems due to tumor progression and recurrence, drug resistance, or side effects of therapy make it necessary to explore new strategies that address the challenges to obtain a positive response. One important point is that tumor cells can interact with the microenvironment, promoting proliferation, dissemination, and immune evasion. Therefore, immunotherapy has emerged as a novel therapy based on the modulation of the immune system for combating cancer, as reflected in the promising results both in preclinical studies and clinical trials obtained. In order to enhance the immune response, the combination of immunotherapy with nanoparticles has been conducted, improving the access of immune cells to the tumor, antigen presentation, as well as the induction of persistent immune responses. Therefore, nanomedicine holds an enormous potential to enhance the efficacy of cancer immunotherapy. Here, we review the most recent advances in specific molecular and cellular immunotherapy and in nano-immunotherapy against cancer in the light of the latest published preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Daniel J. García-Domínguez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
| | - Soledad López-Enríquez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
| | - Gonzalo Alba
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
| | - Carmen Garnacho
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain;
| | - Carlos Jiménez-Cortegana
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
| | - Rocío Flores-Campos
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Department of Medicine, University of Seville, 41009 Seville, Spain
| | - Nabil Hajji
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Cancer Division, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; (D.J.G.-D.); (S.L.-E.); (G.A.); (C.J.-C.); (R.F.-C.); (N.H.)
- Institute of Biomedicine of Seville, IBiS, 41013 Seville, Spain;
- Oncology Service, Department of Medicines, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Clinical Biochemistry Service, Hospital Universitario Virgen Macarena, University of Seville, 41009 Seville, Spain
| |
Collapse
|
24
|
Cao W, Jin M, Zhou W, Yang K, Cheng Y, Chen J, Cao G, Xiong M, Chen B. Forefronts and hotspots evolution of the nanomaterial application in anti-tumor immunotherapy: a scientometric analysis. J Nanobiotechnology 2024; 22:30. [PMID: 38218872 PMCID: PMC10788038 DOI: 10.1186/s12951-023-02278-3] [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: 06/20/2023] [Accepted: 12/17/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Tumor immunotherapy can not only eliminate the primary lesion, but also produce long-term immune memory, effectively inhibiting tumor metastasis and recurrence. However, immunotherapy also showed plenty of limitations in clinical practice. In recent years, the combination of nanomaterials and immunotherapy has brought new light for completely eliminating tumors with its fabulous anti-tumor effects and negligible side effects. METHODS The Core Collection of Web of Science (WOSCC) was used to retrieve and obtain relevant literatures on antitumor nano-immunotherapy since the establishment of the WOSCC. Bibliometrix, VOSviewer, CiteSpace, GraphPad Prism, and Excel were adopted to perform statistical analysis and visualization. The annual output, active institutions, core journals, main authors, keywords, major countries, key documents, and impact factor of the included journals were evaluated. RESULTS A total of 443 related studies were enrolled from 2004 to 2022, and the annual growth rate of articles reached an astonishing 16.85%. The leading countries in terms of number of publications were China and the United States. Journal of Controlled Release, Biomaterials, Acta Biomaterialia, Theranostics, Advanced Materials, and ACS Nano were core journals publishing high-quality literature on the latest advances in the field. Articles focused on dendritic cells and drug delivery accounted for a large percentage in this field. Key words such as regulatory T cells, tumor microenvironment, immune checkpoint blockade, drug delivery, photodynamic therapy, photothermal therapy, tumor-associated macrophages were among the hottest themes with high maturity. Dendritic cells, vaccine, and T cells tend to become the popular and emerging research topics in the future. CONCLUSIONS The combined treatment of nanomaterials and antitumor immunotherapy, namely antitumor nano-immunotherapy has been paid increasing attention. Antitumor nano-immunotherapy is undergoing a transition from simple to complex, from phenotype to mechanism.
Collapse
Affiliation(s)
- Wei Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Mengyao Jin
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Weiguo Zhou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Kang Yang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
- Department of General Surgery, Anhui Public Health Clinical Center, Hefei, 230011, People's Republic of China
| | - Yixian Cheng
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Junjie Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Guodong Cao
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Maoming Xiong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Bo Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
- Department of Surgery, The People's Hospital of Hanshan County, Ma'anshan, 238101, People's Republic of China.
| |
Collapse
|
25
|
Tan J, Ding B, Chen H, Meng Q, Li J, Yang C, Zhang W, Li X, Han D, Zheng P, Ma P, Lin J. Effects of Skeleton Structure of Mesoporous Silica Nanoadjuvants on Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305567. [PMID: 37702141 DOI: 10.1002/smll.202305567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/20/2023] [Indexed: 09/14/2023]
Abstract
Mesoporous silica nanoparticles (MSNs) have been widely praised as nanoadjuvants in vaccine/tumor immunotherapy thanks to their excellent biocompatibility, easy-to-modify surface, adjustable particle size, and remarkable immuno-enhancing activity. However, the application of MSNs is still greatly limited by some severe challenges including the unclear and complicated relationships of structure and immune effect. Herein, three commonly used MSNs with different skeletons including MSN with tetrasulfide bonds (TMSN), MSN containing ethoxy framework (EMSN), and pure -Si-O-Si- framework of MSN (MSN) are comprehensively compared to study the impact of chemical construction on immune effect. The results fully demonstrate that the three MSNs have great promise in improving cellular immunity for tumor immunotherapy. Moreover, the TMSN performs better than the other two MSNs in antigen loading, cellular uptake, reactive oxygen species (ROS) generation, lymph node targeting, immune activation, and therapeutic efficiency. The findings provide a new paradigm for revealing the structure-function relationship of mesoporous silica nanoadjuvants, paving the way for their future clinical application.
Collapse
Affiliation(s)
- Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Chunzheng Yang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Wenying Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Xinyang Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Di Han
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Superlight Materials & Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
26
|
Xie C, You X, Zhang H, Li J, Wang L, Liu Y, Wang Z, Yao R, Tong T, Li M, Wang X, Cui L, Zhang H, Guo H, Li C, Wu J, Xia X. A Nanovaccine Based on Adjuvant Peptide FK-13 and l-Phenylalanine Poly(ester amide) Enhances CD8 + T Cell-Mediated Antitumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300418. [PMID: 37162249 PMCID: PMC10369282 DOI: 10.1002/advs.202300418] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/17/2023] [Indexed: 05/11/2023]
Abstract
Cancer vaccines have shown promise as effective means of antitumor immunotherapy by inducing tumor antigen-specific T cell immunity. In this study, a novel peptide-based tumor nanovaccine that boosts antigen presentation and elicits effective antitumor immunity is developed. The adjuvant characteristics of an antimicrobial peptide-derived core peptide, FK-13, are investigated and used it to generate a fusion peptide named FK-33 with tumor antigen epitopes. l-phenylalanine-based poly(ester amide) (Phe-PEA), 8p4, is also identified as a competent delivery vehicle for the fusion peptide FK-33. Notably, the vaccination of 8p4 + FK-33 nanoparticles (8FNs) in vivo induces dendritic cell activation in the lymph nodes and elicits robust tumor antigen-specific CD8+ T cell response. The nanovaccine 8FNs demonstrate significant therapeutic and prophylactic efficacy against in situ tumor growth, effectively inhibit tumor metastasis, and significantly prolong the survival of tumor-bearing mice. Moreover, 8FNs can incorporate different tumor antigens and exhibit a synergistic therapeutic effect with antiprogrammed cell death protein 1 (PD-1) therapy. In summary, 8FNs represent a promising platform for personalized cancer vaccines and may serve as a potential combinational modality to improve current immunotherapy.
Collapse
Affiliation(s)
- Chunyuan Xie
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Xinru You
- Center for Nanomedicine and Department of AnesthesiologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Hongxia Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Jiahui Li
- School of Food Science and TechnologyNational Engineering Research Center of SeafoodDalian Polytechnic UniversityDalian116024China
| | - Liying Wang
- School of Biomedical EngineeringSun Yat‐sen University66 Gongchang RoadShenzhen518107China
| | - Yongxiang Liu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Zining Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Ruhui Yao
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Tong Tong
- School of Biomedical EngineeringSun Yat‐sen University66 Gongchang RoadShenzhen518107China
| | - Mengyun Li
- State Key Laboratory of BiocontrolSchool of Life ScienceSun Yat‐sen University135 Xingang West RoadGuangzhou510275China
| | - Xiaojuan Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Lei Cui
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Huanling Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Hui Guo
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Chunwei Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| | - Jun Wu
- Bioscience and Biomedical Engineering ThrustThe Hong Kong University of Science and Technology (Guangzhou)NanshaGuangzhou511400China
- Division of Life ScienceThe Hong Kong University of Science and TechnologyHong Kong SAR999077China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer Center651 Dongfeng East RoadGuangzhou510060China
| |
Collapse
|
27
|
Wu N, Chen Q, Zou Y, Miao C, Ma G, Wu J. Chitosan particle-emulsion complex adjuvants: The effect of particle distribution on the immune intensity and response type. Carbohydr Polym 2023; 309:120673. [PMID: 36906359 DOI: 10.1016/j.carbpol.2023.120673] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Particle-emulsion complex adjuvants as a new trend in the research of vaccine formulation, can improve the immune strength and balance the immune type. However, the location of the particle in the formulation is a key factor that has not been investigated extensively and its type of immunity. In order to investigate the effect of different combining modes of emulsion and particle on the immune response, three types of particle-emulsion complex adjuvant formulations were designed with the combination of chitosan nanoparticles (CNP) and an o/w emulsion with squalene as the oil phase. The complex adjuvants included the CNP-I group (particle inside the emulsion droplet), CNP-S group (particle on the surface of emulsion droplet) and CNP-O group (particle outside the emulsion droplet), respectively. The formulations with different particle locations behaved with different immunoprotective effects and immune-enhancing mechanisms. Compared with CNP-O, CNP-I and CNP-S significantly improve humoral and cellular immunity. CNP-O was more like two independent systems for immune enhancement. As a result, CNP-S triggered a Th1-type immune bias and CNP-I had more of a Th2-type of the immune response. These data highlight the key influence of the subtle difference of particle location in the droplets for immune response.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Qiuting Chen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Yongjuan Zou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Chunyu Miao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Jie Wu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
28
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Li Q, Hatakeyama M, Kitaoka T. Polysaccharide Nanofiber-Stabilized Pickering Emulsion Microparticles Induce Pyroptotic Cell Death in Hepatocytes and Kupffer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2207433. [PMID: 36978239 DOI: 10.1002/smll.202207433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/10/2023] [Indexed: 06/18/2023]
Abstract
The intracellular uptake and interaction behavior of emulsion microparticles in liver cells critical to host defense and inflammation is significant to understanding their potential cytotoxicity and biomedical applications. In this study, the cell death responses of fibroblastic, hepatocyte, and Kupffer cells (KCs) induced by four types of emulsion particles that are stabilized by polysaccharide nanofibers (cellulose or chitin), an inorganic nanoparticle (β-tricalcium phosphate), or surfactants are compared. Pickering emulsion (PE) microparticles stabilized by polysaccharide nanofibers or inorganic nanoparticles have a droplet size of 1-3 µm, while the surfactant-stabilized emulsion has a diameter of ≈190 nm. Polysaccharide nanofiber-stabilized PEs (PPEs) markedly induce lactate dehydrogenase release in all cell types. Additionally, characteristic pyroptotic cell death, which is accompanied by cell swelling, membrane blebbing, and caspase-1 activation, occurs in hepatocytes and KCs. These PE microparticles are co-cultured with lipopolysaccharide-primed KCs associated with cytokine interleukin-1β release, and the PPEs demonstrate biological activity as a mediator of the inflammation response. Well-designed PPE microparticles induce pyroptosis of liver cells, which may provide new insight into regulating inflammation-related diseases for designing potent anticancer drugs and vaccine adjuvants.
Collapse
Affiliation(s)
- Qi Li
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Mayumi Hatakeyama
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Takuya Kitaoka
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| |
Collapse
|
30
|
Ding B, Chen H, Tan J, Meng Q, Zheng P, Ma P, Lin J. ZIF-8 Nanoparticles Evoke Pyroptosis for High-Efficiency Cancer Immunotherapy. Angew Chem Int Ed Engl 2023; 62:e202215307. [PMID: 36629270 DOI: 10.1002/anie.202215307] [Citation(s) in RCA: 137] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Although zeolitic imidazolate framework-8 (ZIF-8) has been applied in various tumor therapies, the intrinsic immunogenicity remains unclear. Here, we initiatively discover that ZIF-8 nanoparticles (NPs) can intrinsically induce pyroptosis by a caspase-1/gasdermin D (GSDMD)-dependent pathway. The pyroptotic cell death is accompanied by necrosis and immunogenic cell death (ICD) simultaneously for efficient in situ immunity initiation. Meanwhile, carbonyl cyanide m-chlorophenyl hydrazone (CCCP), a mitochondrial depolarizing agent, is successfully loaded into ZIF-8 NPs and found to further enhance the pyroptosis process. Collectively, the obtained Pluronic F127-modified CCCP-incorporated ZIF-8 NPs (F127 ZIF-8CCCP NPs) activate antitumor immunity and reprogram immunosuppressive tumor microenvironment (TME), realizing high-efficiency tumor growth inhibition. This work will facilitate biomedicine applications of ZIF-8 and provide good inspiration for pyroptosis-induced cancer therapy.
Collapse
Affiliation(s)
- Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jia Tan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Qi Meng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Institute of Frontier and Interdisciplinarity Science and Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, China
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
31
|
Cao D, Chen L, Zhang Z, Luo Y, Zhao L, Yuan C, Lu J, Liu X, Li J. Biodegradable nanomaterials for diagnosis and therapy of tumors. J Mater Chem B 2023; 11:1829-1848. [PMID: 36786439 DOI: 10.1039/d2tb02591d] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although degradable nanomaterials have been widely designed and applied for cancer bioimaging and various cancer treatments, few reviews of biodegradable nanomaterials have been reported. Herein, we have summarized the representative research advances of biodegradable nanomaterials with respect to the mechanism of degradation and their application in tumor imaging and therapy. First, four kinds of tumor microenvironment (TME) responsive degradation are presented, including pH, glutathione (GSH), hypoxia and matrix metalloproteinase (MMP) responsive degradation. Second, external stimulation degradation is summarized briefly. Next, we have outlined the applications of nanomaterials in bioimaging. Finally, we have focused on some typical examples of biodegradable nanomaterials in radiotherapy (RT), photothermal therapy (PTT), starvation therapy, photodynamic therapy (PDT), chemotherapy, chemodynamic therapy (CDT), sonodynamic therapy (SDT), gene therapy, immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Dongmiao Cao
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Ziwen Zhang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Yu Luo
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Linjing Zhao
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Chunping Yuan
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Jie Lu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Xijian Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
32
|
Zhu R, Niu Y, Zhou W, Wang S, Mao J, Guo Y, Lei Y, Xiong X, Li Y, Guo L. Effect of nanoparticles on gouty arthritis: a systematic review and meta-analysis. BMC Musculoskelet Disord 2023; 24:124. [PMID: 36788552 PMCID: PMC9926759 DOI: 10.1186/s12891-023-06186-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
OBJECTIVE The purpose of this study was to explore the effects of nanoparticles on gouty arthritis, and to provide evidence for the preclinical application of nanoparticles in gouty arthritis and ideas for nanomedicine improvement for nanoparticle researchers. METHODS Five databases including the Cochrane Library, PubMed, Scopus, Web of Science, and Embase were searched for eligible studies until April 2022. The quality of the selected studies was assessed by SYRCLE's risk of bias (RoB) tool, and the random-effects model was used to calculate the overall effect sizes of weighted mean differences (WMD). RESULTS Ten studies met the inclusion criteria. Results showed that nanoparticles were effective in reducing uric acid levels (WMD: -4.91; 95% confidence interval (CI): - 5.41 to - 4.41; p < 0.001), but were not better than allopurinol (WMD: -0.20; 95% CI: - 0.42 to 0.02; p = 0.099). It was worth noting that the nanoparticles were safer than allopurinol. Subgroup analyses indicated that nanoparticle encapsulated substance, animal species, nanoparticle dosage, animal quantity, and animal gender were all sources of heterogeneity. CONCLUSION The nanoparticles are safe medications for gouty arthritis which can effectively reduce uric acid levels in rodents. Although the results are still uncertain, it is expected to have certain clinical application value. The nanoparticles may be the preclinical medications for gouty arthritis in the future.
Collapse
Affiliation(s)
- Ruiting Zhu
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| | - Yirou Niu
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| | - Wei Zhou
- The First Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Saikun Wang
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| | - Jing Mao
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| | - Yingze Guo
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| | - Yangyang Lei
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| | - Xuance Xiong
- Medical College, Beihua University, Jilin, 132013 Jilin China
| | - Yingzhi Li
- Orthpoeadic Medical Center, Jilin University Second Hospital, Changchun, 130041 Jilin China
| | - Lirong Guo
- School of Nursing, Jilin University, Changchun, 130021 Jilin China
| |
Collapse
|
33
|
Choi A, Javius-Jones K, Hong S, Park H. Cell-Based Drug Delivery Systems with Innate Homing Capability as a Novel Nanocarrier Platform. Int J Nanomedicine 2023; 18:509-525. [PMID: 36742991 PMCID: PMC9893846 DOI: 10.2147/ijn.s394389] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023] Open
Abstract
Nanoparticle-based drug delivery systems have been designed to treat various diseases. However, many problems remain, such as inadequate tumor targeting and poor therapeutic outcomes. To overcome these obstacles, cell-based drug delivery systems have been developed. Candidates for cell-mediated drug delivery include blood cells, immune cells, and stem cells with innate tumor tropism and low immunogenicity; they act as a disguise to deliver the therapeutic payload. In drug delivery systems, therapeutic agents are encapsulated intracellularly or attached to the surface of the plasma membrane and transported to the desired site. Here, we review the pros and cons of cell-based therapies and discuss their homing mechanisms in the tumor microenvironment. In addition, different strategies to load therapeutic agents inside or on the surface of circulating cells and the current applications for a wide range of disease treatments are summarized.
Collapse
Affiliation(s)
- Anseo Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea,Correspondence: Hansoo Park; Seungpyo Hong, School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea, Tel +82-2 820 5804, Fax +82-2 813 8159, Email ;
| |
Collapse
|
34
|
Muñoz-Wolf N, Ward RW, Hearnden CH, Sharp FA, Geoghegan J, O’Grady K, McEntee CP, Shanahan KA, Guy C, Bowie AG, Campbell M, Roces C, Anderluzzi G, Webb C, Perrie Y, Creagh E, Lavelle EC. Non-canonical inflammasome activation mediates the adjuvanticity of nanoparticles. Cell Rep Med 2023; 4:100899. [PMID: 36652908 PMCID: PMC9873954 DOI: 10.1016/j.xcrm.2022.100899] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/24/2022] [Accepted: 12/19/2022] [Indexed: 01/19/2023]
Abstract
The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens.
Collapse
Affiliation(s)
- Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland,Translational & Respiratory Immunology Lab, Department of Clinical Medicine, School of Medicine, Trinity Biomedical Sciences Institute, Dublin D02 R590, Ireland,Clinical Medicine Tallaght University Hospital, Dublin D24 NR04, Ireland
| | - Ross W. Ward
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Claire H. Hearnden
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Fiona A. Sharp
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Joan Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland,Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Katie O’Grady
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Craig P. McEntee
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland
| | - Katharine A. Shanahan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Coralie Guy
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Andrew G. Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Carla.B. Roces
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Giulia Anderluzzi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Cameron Webb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Emma Creagh
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin D02 R590, Ireland
| | - Ed C. Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 R590, Ireland,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin D02 PN40, Ireland,Corresponding author
| |
Collapse
|
35
|
Effects of Magnetic Nanoparticles on the Functional Activity of Human Monocytes and Dendritic Cells. Int J Mol Sci 2023; 24:ijms24021358. [PMID: 36674876 PMCID: PMC9864373 DOI: 10.3390/ijms24021358] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
The use of nanoparticles in medicine is sometimes hampered by their potential to activate immune cells, eliciting inflammation or allergy. We investigated whether magnetic nanoparticles (MNPs) or biomimetic magnetic nanoparticles (BMNPs) affect relevant activities of human monocytes. We found that the nanoparticles neither elicited the production of pro-inflammatory mediators IL-6 and TNFα by resting monocytes (when BMNP dose < 300 μg/mL) nor enhanced their secretion induced by R848, a molecule engaging virus-recognizing receptors, or bacterial lipopolysaccharide (LPS). MNPs and BMNPs neither induced the generation of reactive oxygen species (ROS), nor affected the ROS production elicited by the NADPH oxidase activator phorbol myristate acetate (PMA) or the fungal derivative β-glucan. BMNPs, but not MNPs, caused an up-regulation of the maturation markers CD80, CD83, and CD86 in immature monocyte-derived dendritic cells (DCs), whereas both nanoparticles did not affect the LPS-induced expression of these markers. Moreover, the nanoparticles were greedily ingested by monocytes and DCs without altering their viability. Therefore, these nanoparticles are candidates for medical applications because they do not activate pro-inflammatory activities of monocytes. Furthermore, their ability to stimulate DC maturation could be used for the design of vaccines. Moreover, harmlessly engulfed nanoparticles could be vehicles to carry molecules inside the immune cells to regulate the immune response.
Collapse
|
36
|
Achmad H, Saleh Ibrahim Y, Mohammed Al-Taee M, Gabr GA, Waheed Riaz M, Hamoud Alshahrani S, Alexis Ramírez-Coronel A, Turki Jalil A, Setia Budi H, Sawitri W, Elena Stanislavovna M, Gupta J. Nanovaccines in cancer immunotherapy: Focusing on dendritic cell targeting. Int Immunopharmacol 2022; 113:109434. [DOI: 10.1016/j.intimp.2022.109434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
|
37
|
Li W, Fan JX, Zheng DW, Zhang XZ. Tumor Antigen Loaded Nanovaccine Induced NIR-Activated Inflammation for Enhanced Antigen Presentation During Immunotherapy of Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2205193. [PMID: 36285774 DOI: 10.1002/smll.202205193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/03/2022] [Indexed: 06/16/2023]
Abstract
Although anticancer vaccines have achieved certain effects in early clinical practice, T cell immunity as the most common responsive pattern of anticancer vaccines is still limited by unsatisfied tumor recognition and inhibition efficiency. As the critical step of T cell immunity, uptake and presentation of specific antigen by antigen-presenting cells (APC) can be activated by inflammation for enhancing the response of T cells to the antigen source. Here, a hybrid nanovaccine named PTh/MnO2 @M activated with a near-infrared ray (NIR) is prepared by coating an autologous tumor cell membrane on the surface of a polythiophene/MnO2 composite core. The photoelectrical material polythiophene can produce local microinflammation under NIR radiation and activate specific T cell antitumor immunity by promoting APC maturation and autologous tumor antigens presentation. Moreover, the synthesized nanovaccine PTh/MnO2 @M is shown to induce a significant antitumor immune response, effectively inhibit the progression of melanoma in mice, and significantly prolong the survival time of mice in vivo. This strategy aims to enhance T-cell immune responses by promoting antigen presentation, leading to effective and specific cancer therapy.
Collapse
Affiliation(s)
- Wen Li
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Jin-Xuan Fan
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Di-Wei Zheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Traditional Chinese Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
| |
Collapse
|
38
|
Dai H, Huang Y, Guo J, Li L, Ke Y, Cen L, Meng F, Chen X, Liu B, Qian X. Engineering a HemoMap Nanovaccine for Inducing Immune Responses against Melanoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:52634-52642. [PMID: 36383430 DOI: 10.1021/acsami.2c14379] [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: 06/16/2023]
Abstract
Neoantigen vaccines have opened a new paradigm for cancer immunotherapy. Here, we constructed a neoantigen nanovaccine-HemoMap, with the ability to target lymph nodes and activate immune cells. We propose a HemoMap nanovaccine consisting of the mouse melanoma highly expressed antigenic peptide Tyrp1 and a magnesium nanoadjuvant-HemoM. By immunofluorescence labeling of the nanovaccine, the lymph node targeting of the vaccine was observed and verified by a mouse near-infrared imaging system. About two-fold higher effective retention of HemoMap induces the internalization of Tyrp1 in DCs than that of free Tyrp1 in draining lymph nodes (DLNs) for 48 h. A mouse melanoma subcutaneous model was established to evaluate neoantigen-specific antitumor immune responses. In comparison to the control group, the tumor growth rate was dramatically slowed down by HemoMap treatment, and the median survival time was extended by 7 days. We discovered that effective co-delivery of Tyrp1 antigen and magnesium (Mg2+) to lymph nodes (LNs) boosted cellular internalization and activated immune cells, such as CD11c+ DCs and CD8+ T lymphocytes. Spleen lymphocytes from the HemoMap group displayed much more antitumor activity than those from the other groups. Our findings highlight that HemoMap is promising to trigger T cell responses and to provide novel nanoadjuvants strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Hengheng Dai
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Ying Huang
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing 210008, China
| | - Jingyi Guo
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Lin Li
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Yaohua Ke
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Lanqi Cen
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Xinjie Chen
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Xiaoping Qian
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| |
Collapse
|
39
|
Mamuti M, Chen W, Jiang X. Nanotechnology‐Assisted Immunoengineering for Cancer Vaccines. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Muhetaerjiang Mamuti
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| |
Collapse
|
40
|
Liu Q, Chu Y, Shao J, Qian H, Yang J, Sha H, Cen L, Tian M, Xu Q, Chen F, Yang Y, Wang W, Wang K, Yu L, Wei J, Liu B. Benefits of an Immunogenic Personalized Neoantigen Nanovaccine in Patients with High-Risk Gastric/Gastroesophageal Junction Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2203298. [PMID: 36351249 PMCID: PMC9811442 DOI: 10.1002/advs.202203298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/17/2022] [Indexed: 06/16/2023]
Abstract
Personalized neoantigen vaccines have shown strong immunogenicity in clinical trial, but still face various challenges in facilitating an efficient antitumor immune response. Here, a personalized neoantigen nanovaccine (PNVAC) platform for adjuvant cancer immunotherapy is generated. PNVAC triggers superior protective efficacy against tumor recurrence and promotes longer survival than free neoantigens, especially when combined with anti-PD-1 treatment in a murine tumor model. A phase I clinical trial (ChiCTR1800017319) is initiated to evaluate the safety, immunogenicity, and prophylactic effect of PNVAC on preventing tumor recurrence in patients with high-risk gastric/gastroesophageal junction cancer after adjuvant chemotherapy of postsurgical resection. The one- and two-year disease-free survival rates are significantly higher than historical record. PNVAC induces both CD4+ and CD8+ T cell responses as well as antigen-experienced memory T cell phenotype. Furthermore, the immune response is persistent and remains evident one year after the vaccination. This work provides a safe and feasible strategy for developing neoantigen vaccines to delay gastric cancer recurrence after surgery.
Collapse
Affiliation(s)
- Qin Liu
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Yanhong Chu
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Jie Shao
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Ju Yang
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Huizi Sha
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Lanqi Cen
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Manman Tian
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Qiuping Xu
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Fangjun Chen
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Yang Yang
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | | | | | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Jia Wei
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower HospitalMedical School of Nanjing University and Clinical Cancer Institute of Nanjing UniversityNanjing210008China
| |
Collapse
|
41
|
Advancements in clinical translation of flavonoid nanoparticles for cancer treatment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100074] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
42
|
Escriche‐Navarro B, Escudero A, Lucena‐Sánchez E, Sancenón F, García‐Fernández A, Martínez‐Máñez R. Mesoporous Silica Materials as an Emerging Tool for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200756. [PMID: 35866466 PMCID: PMC9475525 DOI: 10.1002/advs.202200756] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/16/2022] [Indexed: 05/16/2023]
Abstract
Cancer immunotherapy has emerged in the past decade as a promising strategy for treating many forms of cancer by stimulating the patient's immune system. Although immunotherapy has achieved some promising results in clinics, more efforts are required to improve the limitations of current treatments related to lack of effective and targeted cancer antigens delivery to immune cells, dose-limiting toxicity, and immune-mediated adverse effects, among others. In recent years, the use of nanomaterials has proven promising to enhance cancer immunotherapy efficacy and reduce side effects. Among nanomaterials, attention has been recently paid to mesoporous silica nanoparticles (MSNs) as a potential multiplatform for enhancing cancer immunotherapy by considering their unique properties, such as high porosity, and good biocompatibility, facile surface modification, and self-adjuvanticity. This review explores the role of MSN and other nano/micro-materials as an emerging tool to enhance cancer immunotherapy, and it comprehensively summarizes the different immunotherapeutic strategies addressed to date by using MSN.
Collapse
Affiliation(s)
- Blanca Escriche‐Navarro
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Joint Unit of Nanomedicine and Sensors, Polytechnic University of Valencia, IIS La FeAv. Fernando Abril Martorell, 106Valencia46026Spain
| | - Andrea Escudero
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
| | - Elena Lucena‐Sánchez
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
| | - Félix Sancenón
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Joint Unit of Nanomedicine and Sensors, Polytechnic University of Valencia, IIS La FeAv. Fernando Abril Martorell, 106Valencia46026Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)Av. Monforte de Lemos, 3–5. Pabellón 11., Planta 0Madrid28029Spain
| | - Alba García‐Fernández
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)Av. Monforte de Lemos, 3–5. Pabellón 11., Planta 0Madrid28029Spain
| | - Ramón Martínez‐Máñez
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM) Polytechnic University of Valencia‐University of ValenciaCamino de Vera s/nValencia46022Spain
- Universitat Politècnica de ValènciaJoint Unit UPV‐CIPF of Developmental Biology and Disease Models and Nanomedicine, Polytechnic University of Valencia (UPV)‐Príncipe Felipe Research Center Foundation (CIPF)C/ Eduardo Primo Yúfera 3.Valencia46012Spain
- Joint Unit of Nanomedicine and Sensors, Polytechnic University of Valencia, IIS La FeAv. Fernando Abril Martorell, 106Valencia46026Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)Av. Monforte de Lemos, 3–5. Pabellón 11., Planta 0Madrid28029Spain
| |
Collapse
|
43
|
Luo T, Nash GT, Jiang X, Feng X, Mao J, Liu J, Juloori A, Pearson AT, Lin W. A 2D Nanoradiosensitizer Enhances Radiotherapy and Delivers STING Agonists to Potentiate Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110588. [PMID: 35952624 PMCID: PMC9529854 DOI: 10.1002/adma.202110588] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/27/2022] [Indexed: 05/11/2023]
Abstract
Despite potent preclinical antitumor activity, activation of stimulator of interferon genes (STING) has shown modest therapeutic effects in clinical studies. Many STING agonists, including 2',3'-cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), show poor pharmacokinetic properties for sustaining STING activation in tumors and achieving optimal antitumor efficacy. Improved delivery of STING agonists and their effective combination with other treatments are needed to enhance their therapeutic effects. Herein, a 2D nanoplatform, cGAMP/MOL, is reported via conjugating cGAMP to a nanoscale metal-organic layer (MOL) for simultaneous STING activation and radiosensitization. The MOL not only exhibits strong radiosensitization effects for enhanced cancer killing and induction of immunogenic cell death, but also retains cGAMP in tumors for sustained STING activation. Compared to free cGAMP, cGAMP/MOL elicits stronger STING activation and regresses local tumors upon X-ray irradiation. Further combination with an immune checkpoint inhibitor bridges innate and adaptive immune systems by activating the tumor microenvironment to elicit systemic antitumor responses.
Collapse
Affiliation(s)
- Taokun Luo
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Geoffrey T. Nash
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaomin Jiang
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Xuanyu Feng
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Jianming Mao
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Jianqiao Liu
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Aditya Juloori
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| | - Alexander T. Pearson
- Department of Pathology & University of Chicago Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
44
|
Exploring dendrimer-based drug delivery systems and their potential applications in cancer immunotherapy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111471] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
45
|
Mamuti M, Wang Y, Zhao YD, Wang JQ, Wang J, Fan YL, Xiao WY, Hou DY, Yang J, Zheng R, An HW, Wang H. A Polyvalent Peptide CD40 Nanoagonist for Targeted Modulation of Dendritic Cells and Amplified Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109432. [PMID: 35426184 DOI: 10.1002/adma.202109432] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/06/2022] [Indexed: 06/14/2023]
Abstract
Targeted immunomodulation through biomolecule-based nanostructures, especially to dendritic cells (DCs), holds great promise for effective cancer therapy. However, construction of high-performance agonist by mimicking natural ligand to activate immune cell signaling is a great challenge so far. Here, a peptide-based nanoagonist toward CD40 (PVA-CD40) with preorganized interfacial topological structure that activates lymph node DCs efficiently and persistently, achieving amplified immune therapeutic efficacy is described. The on-site fabrication of PVA-CD40 is realized through the click conjugation of two functional peptides including the "CD40 anchoring arm" and the "assembly-driving motor." The resultant polyvalent interface rapidly triggers the receptor oligomerization and downstream signaling. Strikingly, one shot administration of PVA-CD40 elicits maturation period of DCs up to 2.3-fold comparing to that of CD40 antibody. Finally, combining the PVA-CD40 with anti-PD-1 antibody results in subsequent inhibition of tumor growth in both B16F10 and 4T1 mice tumor models with survival rate up to 37%, while none of the mice survives in the clinically relevant CD40 mAb and anti-PD-1 combination-treated group. It is envisioned that the fabrication of antibody-like superstructures in vivo provides an efficient platform for modulating the duration of immune response to achieve optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Muhetaerjiang Mamuti
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yong-Dan Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jia-Qi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Jie Wang
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yan-Lei Fan
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Wu-Yi Xiao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
| | - Jia Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Rui Zheng
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Haidian District, Beijing, 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
46
|
Ngo W, Ahmed S, Blackadar C, Bussin B, Ji Q, Mladjenovic SM, Sepahi Z, Chan WC. Why nanoparticles prefer liver macrophage cell uptake in vivo. Adv Drug Deliv Rev 2022; 185:114238. [PMID: 35367524 DOI: 10.1016/j.addr.2022.114238] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/26/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023]
Abstract
Effective delivery of therapeutic and diagnostic nanoparticles is dependent on their ability to accumulate in diseased tissues. However, most nanoparticles end up in liver macrophages regardless of nanoparticle design after administration. In this review, we describe the interactions of liver macrophages with nanoparticles. Liver macrophages have significant advantages in interacting with circulating nanoparticles over most target cells and tissues in the body. We describe these advantages in this article. Understanding these advantages will enable the development of strategies to overcome liver macrophages and deliver nanoparticles to targeted diseased tissues effectively. Ultimately, these approaches will increase the therapeutic efficacy and diagnostic signal of nanoparticles.
Collapse
|
47
|
Wong NKY, Dong X, Lin YY, Xue H, Wu R, Lin D, Collins C, Wang Y. Framework of Intrinsic Immune Landscape of Dormant Prostate Cancer. Cells 2022; 11:cells11091550. [PMID: 35563856 PMCID: PMC9105276 DOI: 10.3390/cells11091550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Androgen deprivation therapy (ADT) is the standard therapy for men with advanced prostate cancer (PCa). PCa often responds to ADT and enters a dormancy period, which can be recognized clinically as a minimal residual disease. However, the majority of these patients will eventually experience a relapse in the form of castration-resistant PCa with poor survival. Therefore, ADT-induced dormancy is a unique time window for treatment that can provide a cure. The study of this well-recognized phase of prostate cancer progression is largely hindered by the scarcity of appropriate clinical tissue and clinically relevant preclinical models. Here, we report the utility of unique and clinically relevant patient-derived xenograft models in the study of the intrinsic immune landscape of dormant PCa. Using data from RNA sequencing, we have reconstructed the immune evasion mechanisms that can be utilized by dormant PCa cells. Since dormant PCa cells need to evade the host immune surveillance for survival, our results provide a framework for further study and for devising immunomodulatory mechanisms that can eliminate dormant PCa cells.
Collapse
Affiliation(s)
- Nelson K. Y. Wong
- Department of Experimental Therapeutics, BC Cancer, 675 W 10th Ave, Vancouver, BC V5Z 1L3 Canada; (N.K.Y.W.); (X.D.); (H.X.); (R.W.); (D.L.)
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer, 675 W 10th Ave, Vancouver, BC V5Z 1L3 Canada; (N.K.Y.W.); (X.D.); (H.X.); (R.W.); (D.L.)
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (Y.-Y.L.); (C.C.)
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer, 675 W 10th Ave, Vancouver, BC V5Z 1L3 Canada; (N.K.Y.W.); (X.D.); (H.X.); (R.W.); (D.L.)
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer, 675 W 10th Ave, Vancouver, BC V5Z 1L3 Canada; (N.K.Y.W.); (X.D.); (H.X.); (R.W.); (D.L.)
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer, 675 W 10th Ave, Vancouver, BC V5Z 1L3 Canada; (N.K.Y.W.); (X.D.); (H.X.); (R.W.); (D.L.)
| | - Colin Collins
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (Y.-Y.L.); (C.C.)
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer, 675 W 10th Ave, Vancouver, BC V5Z 1L3 Canada; (N.K.Y.W.); (X.D.); (H.X.); (R.W.); (D.L.)
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (Y.-Y.L.); (C.C.)
- Correspondence: ; Tel.: +1-604-675-8013
| |
Collapse
|
48
|
Ruan S, Huang Y, He M, Gao H. Advanced Biomaterials for Cell-Specific Modulation and Restore of Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200027. [PMID: 35343112 PMCID: PMC9165523 DOI: 10.1002/advs.202200027] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/18/2022] [Indexed: 05/09/2023]
Abstract
The past decade has witnessed the explosive development of cancer immunotherapies. Nevertheless, low immunogenicity, limited specificity, poor delivery efficiency, and off-target side effects remain to be the major limitations for broad implementation of cancer immunotherapies to patient bedside. Encouragingly, advanced biomaterials offering cell-specific modulation of immunological cues bring new solutions for improving the therapeutic efficacy while relieving side effect risks. In this review, focus is given on how functional biomaterials can enable cell-specific modulation of cancer immunotherapy within the cancer-immune cycle, with particular emphasis on antigen-presenting cells (APCs), T cells, and tumor microenvironment (TME)-resident cells. By reviewing the current progress in biomaterial-based cancer immunotherapy, here the aim is to provide a better understanding of biomaterials' role in targeting modulation of antitumor immunity step-by-step and guidelines for rationally developing targeting biomaterials for more personalized cancer immunotherapy. Moreover, the current challenge and future perspective regarding the potential application and clinical translation will also be discussed.
Collapse
Affiliation(s)
- Shaobo Ruan
- Advanced Research Institute of Multidisciplinary ScienceBeijing Institute of TechnologyBeijing100081China
| | - Yuanyu Huang
- Advanced Research Institute of Multidisciplinary ScienceBeijing Institute of TechnologyBeijing100081China
| | - Mei He
- College of PharmacyUniversity of FloridaGainesvilleFL32610USA
| | - Huile Gao
- West China School of PharmacySichuan UniversityChengdu610041China
| |
Collapse
|
49
|
Chen H, Liu H, Liu L, Chen Y. Fabrication of subunit nanovaccines by physical interaction. SCIENCE CHINA. TECHNOLOGICAL SCIENCES 2022; 65:989-999. [PMID: 35432491 PMCID: PMC9004205 DOI: 10.1007/s11431-021-2011-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/09/2022] [Indexed: 06/14/2023]
Abstract
Vaccines can improve the quality of human life by preventing the burden of infectious diseases. Also, vaccination is becoming a powerful medication for preventing and treating tumors. Various vaccines have been developed based on the origin of the antigens. Herein, we focus on the subunit vaccines whose antigens are proteins or peptides. The advantage of subunit vaccines is safety for recipients; however, the immunogenicity of subunit antigens is relatively low. Nanoparticular delivery systems have been applied to improve the immunocompetence of subunit vaccines by targeting lymph nodes, and effectively present antigens to immune cells. Moreover, adding appropriate molecular adjuvants may strengthen the antigens to elicit immune response. In this perspective article, we first elucidate the characteristics of immunity induced by subunit nanovaccines and then summarize the strategies to fabricate subunit nanovaccines with delivering materials. Herein we highlight non-covalent interaction to fabricate nanoparticular subunit vaccines.
Collapse
Affiliation(s)
- HaoLin Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275 China
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Jinan University, Zhuhai, 519000 China
| | - LiXin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275 China
| | - YongMing Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, 510275 China
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 China
| |
Collapse
|
50
|
Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Semin Cancer Biol 2022; 86:624-644. [DOI: 10.1016/j.semcancer.2022.03.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/16/2022]
|