1
|
Yin C, Xing Y, Zhao P, Yin Y, Yao H, Xue J, Gu W. Tetradecanol-wrapped, CpG-loaded porous Prussian blue nanoimmunomodulator for photothermal-responsive in situ anti-tumor vaccine-like immunotherapy. BIOMATERIALS ADVANCES 2024; 164:213996. [PMID: 39146604 DOI: 10.1016/j.bioadv.2024.213996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Therapeutic vaccine becomes a promising strategy to fight cancer by enhancing and sustaining specific anti-tumor immune responses. However, its efficacy is often impeded by low immunogenicity, the immunosuppressive tumor microenvironment (TME), and immune-related adverse events. Herein, we introduce 1-tetradecanol (TD)-wrapped, CpG-loaded porous Prussian blue nanoparticles (pPBNPs-CpG@TD) as a nanoimmunomodulator to initiate photothermal-induced immunogenic cell death (ICD) and photothermal-responsive release of CpG for augmenting the ICD effect. It was revealed that the dual-photothermal action significantly potentiated the in situ anti-tumor vaccine-like immunotherapy in terms of enhanced immunogenicity, promoted dendritic cell maturation, and increased T lymphocyte infiltration, consequently eliciting a robust immune response for inhibiting both primary and rechallenge tumors on a subcutaneous 4T1 tumor-bearing mouse model. The development and use of photoactive nanoimmunomodulators represents a novel and effective strategy to boost immunogenicity and counteract immunosuppressive TME, marking a significant advancement in the realm of ICD-driven in situ anti-tumor vaccine-like immunotherapy.
Collapse
Affiliation(s)
- Chenlu Yin
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Yixin Xing
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Peng Zhao
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Yuying Yin
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Hanye Yao
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Jingqiang Xue
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Wei Gu
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China.
| |
Collapse
|
2
|
Rao Z, Zhu Y, Chen Z, Luo Y, Yang Z, Liu W, Qiao C, Xia Y, Yang P, Ye DM, Wang Z. Injectable Autocatalytic Hydrogel Triggers Pyroptosis to Stimulate Anticancer Immune Response for Preventing Postoperative Tumor Recurrence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408415. [PMID: 39465669 DOI: 10.1002/advs.202408415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/14/2024] [Indexed: 10/29/2024]
Abstract
Modulating immunosuppression while eliminating residual microscopic tumors is critical for inhibiting the postoperative recurrence of triple-negative breast cancer (TNBC). Although immunotherapy has shown potential in achieving this goal, due to multiple immunosuppression and poor immunogenicity of apoptosis, a satisfactory anti-recurrence effect still faces the challenge. Herein, an injectable hydrogel-encapsulated autocatalytic copper peroxide (CP@Gel) therapeutic platform is designed and combine it with the clinical-grade DNA methyltransferase inhibitor decitabine (DAC) to effectively inhibit TNBC growth and postoperative recurrence via pyroptosis, killing residual cancer cells that bypass apoptosis resistance while also improving immunogenicity and modulating immunosuppression to achieve an intense anti-tumor immune response. Following injection of the CP@Gel, the sustained release of CP leads to the autocatalytic generation of reactive oxygen species, resulting in caspase-3 activation, and the pre-administered DAC inhibits the methylation of Gsdme to elevate the GSDME protein levels, leading to intense pyroptosis and anti-tumor immune responses. The in vivo results show a 67% elimination of local tumor recurrence via treatment with DAC+CP@Gel, suggesting the successful integration of sustained drug release with autocatalysis and epigenetic modification. The results thus suggest great potential for pyroptosis-based and injectable hydrogel-aided strategies for preventing the postoperative recurrence of TNBC.
Collapse
Affiliation(s)
- Zhiping Rao
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Yutong Zhu
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
- Medical college, Xi'an International University, Xi'an, Shaanxi, 710077, P. R. China
| | - Zhuang Chen
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Yi Luo
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Zuo Yang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Weijing Liu
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Chaoqiang Qiao
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Yuqiong Xia
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Peng Yang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| | - Dong-Man Ye
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, P. R. China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM), Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment, Xi'an, Shaanxi, 710126, P. R. China
| |
Collapse
|
3
|
Ding Y, Jia Q, Su Z, Chen H, Ye J, Xie D, Wu Y, He H, Peng Y, Ni Y. Homologous cell membrane-based hydrogel creates spatiotemporal niches to improve outcomes of dysregulated chronic wound healing. Mater Today Bio 2024; 28:101243. [PMID: 39315394 PMCID: PMC11419813 DOI: 10.1016/j.mtbio.2024.101243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/25/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
The (M2M + TGF-β)@HAMA hydrogel dressing improves the outcomes of dysregulated chronic wound healing by protecting the open wound from repeated bacterial infections, reprogramming endogenous monocytes and M1 macrophages into an M2-phenotype, as well as enhancing fibroblastic proliferation and migration for matrix remodeling and granulation tissue formation.Image 1.
Collapse
Affiliation(s)
| | | | - Ziwen Su
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Heying Chen
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jialing Ye
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Dafeng Xie
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yubo Wu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Haiyan He
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yanlin Peng
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Yilu Ni
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
4
|
Luo S, Wang J, Gao M. Sodium alginate hydrogel encapsulating microglia cell lysate subjected to serum starvation for mitigating glioma cells. J Biomater Appl 2024; 39:396-405. [PMID: 39075851 DOI: 10.1177/08853282241268694] [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: 07/31/2024]
Abstract
Glioma is the most common malignant tumor in the brain, accounting for over 80% of all primary intracranial tumors. The current clinical treatment has shown certain limitations. Although M1 type microglia can secrete various pro-inflammatory cytokines and are expected to be used for glioma treatment, direct use of microglia may lead to overactivation and trigger immune storms. Therefore, we first found that serum starvation can stimulate the transformation of microglia into M1 type. Subsequently, we found through comparative experiments that the inhibitory effect of microglial cell lysis medium on glioma cells was stronger than that of microglial cell culture medium. Finally, we successfully prepared sodium alginate hydrogel loaded with microglia lysis solution to achieve sustained inhibitory effect on the growth of glioma and avoid its proliferation.
Collapse
Affiliation(s)
- Shenzhong Luo
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Meng Gao
- Department of Gastroenterology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Li W, Ye Q, Jiang Z, Xia D, Yan Z, Wang D, Chen Y, Cao T, Wang J, Lin C, Yang H, Deng J, Lin J. A cross-linked macropore hydrogel based on M1 macrophage lysate and alginate regulates tumor-associated macrophages for the treatment of melanoma. Int J Biol Macromol 2024; 269:132089. [PMID: 38705331 DOI: 10.1016/j.ijbiomac.2024.132089] [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: 10/24/2023] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Pro-inflammatory M1 macrophages possess the ability to change the immunosuppressive tumor microenvironment by releasing various inflammatory factors simultaneously, which can effectively inhibit tumor progression and relapse. Promoting macrophage polarization towards M1 may be an effective way to treat Melanoma. However, the risk of cytokine storm caused by the proliferation and excessive activation of M1 macrophages greatly limits it as a biosafety therapeutic strategy in anti-tumor immunotherapy. Therefore, how to engineer natural M1 macrophage to a biocompatible biomaterial that maintains the duration time of tumor suppressive property duration time still remains a huge challenge. To achieve this goal, we developed an injectable macroporous hydrogel (M1LMHA) using natural M1 macrophage lysates and alginate as raw materials. M1LMHA had excellent biocompatibility, adjustable degradation rate and could sustainably release varieties of natural inflammatory factors, such as tumor necrosis factor-α (TNF-α), interferon-gamma (IFN-γ), and interleukin-12 (IL-12), etc. M1LMHA could repolarize anti-inflammatory M2 macrophages to M1 macrophages by the synergistic effect of released tiny inflammatory factors via the NF-κB pathway. This study supported that M1LMHA might be an effective and safe tool to activate tumor-associated immune cells, improving the efficiency of anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Wanyu Li
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524003, China; Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Qingbang Ye
- The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Zhonghao Jiang
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524003, China; Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Dong Xia
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524003, China; Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhuo Yan
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524003, China; Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Dan Wang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yanan Chen
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Tianshou Cao
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jilong Wang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Chihao Lin
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Huiling Yang
- Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Junjie Deng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China.
| | - Jiantao Lin
- The Second Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang 524003, China; Dongguan Key Laboratory of Traditional Chinese Medicine and New Pharmaceutical Development, The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
6
|
Bao P, Gu HY, Jiang YC, Wang JW, Wu M, Yu A, Zhong Z, Zhang XZ. In Situ Sprayed Exosome-Cross-Linked Gel as Artificial Lymph Nodes for Postoperative Glioblastoma Immunotherapy. ACS NANO 2024; 18:13266-13276. [PMID: 38709874 DOI: 10.1021/acsnano.4c02425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
One key challenge in postoperative glioblastoma immunotherapy is to guarantee a potent and durable T-cell response, which is restricted by the immunosuppressive microenvironment within the lymph nodes (LNs). Here, we develop an in situ sprayed exosome-cross-linked gel that acts as an artificial LN structure to directly activate the tumor-infiltrating T cells for prevention of glioma recurrence. Briefly, this gel is generated by a bio-orthogonal reaction between azide-modified chimeric exosomes and alkyne-modified alginate polymers. Particularly, these chimeric exosomes are generated from dendritic cell (DC)-tumor hybrid cells, allowing for direct and robust T-cell activation. The gel structure with chimeric exosomes as cross-linking points avoids the quick clearance by the immune system and thus prolongs the durability of antitumor T-cell immunity. Importantly, this exosome-containing immunotherapeutic gel provides chances for ameliorating functions of antigen-presenting cells (APCs) through accommodating different intracellular-acting adjuvants, such as stimulator of interferon genes (STING) agonists. This further enhances the antitumor T-cell response, resulting in the almost complete elimination of residual lesions after surgery. Our findings provide a promising strategy for postsurgical glioma immunotherapy that warrants further exploration in the clinical arena.
Collapse
Affiliation(s)
- Peng Bao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Hui-Yun Gu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Yuan-Cheng Jiang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Jia-Wei Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Aixi Yu
- Department of Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| | - Zhenlin Zhong
- 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 Orthopedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
7
|
Peng Y, Liang S, Meng QF, Liu D, Ma K, Zhou M, Yun K, Rao L, Wang Z. Engineered Bio-Based Hydrogels for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313188. [PMID: 38362813 DOI: 10.1002/adma.202313188] [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: 12/05/2023] [Revised: 02/01/2024] [Indexed: 02/17/2024]
Abstract
Immunotherapy represents a revolutionary paradigm in cancer management, showcasing its potential to impede tumor metastasis and recurrence. Nonetheless, challenges including limited therapeutic efficacy and severe immune-related side effects are frequently encountered, especially in solid tumors. Hydrogels, a class of versatile materials featuring well-hydrated structures widely used in biomedicine, offer a promising platform for encapsulating and releasing small molecule drugs, biomacromolecules, and cells in a controlled manner. Immunomodulatory hydrogels present a unique capability for augmenting immune activation and mitigating systemic toxicity through encapsulation of multiple components and localized administration. Notably, hydrogels based on biopolymers have gained significant interest owing to their biocompatibility, environmental friendliness, and ease of production. This review delves into the recent advances in bio-based hydrogels in cancer immunotherapy and synergistic combinatorial approaches, highlighting their diverse applications. It is anticipated that this review will guide the rational design of hydrogels in the field of cancer immunotherapy, fostering clinical translation and ultimately benefiting patients.
Collapse
Affiliation(s)
- Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qian-Fang Meng
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kaiqing Yun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
8
|
Yu L, Zhou A, Jia J, Wang J, Ji X, Deng Y, Lin X, Wang F. Immunoactivity of a hybrid membrane biosurface on nanoparticles: enhancing interactions with dendritic cells to augment anti-tumor immune responses. Biomater Sci 2024; 12:1016-1030. [PMID: 38206081 DOI: 10.1039/d3bm01628e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Nano-biointerfaces play a pivotal role in determining the functionality of engineered therapeutic nanoparticles, particularly in the context of designing nanovaccines to effectively activate immune cells for cancer immunotherapy. Unlike involving chemical reactions by conventional surface decorating strategies, cell membrane-coating technology offers a straightforward approach to endow nanoparticles with natural biosurfaces, enabling them to mimic and integrate into the intricate biosystems of the body to interact with specific cells under physiological conditions. In this study, cell membranes, in a hybrid formulation, derived from cancer and activated macrophage cells were found to enhance the interaction of nanoparticles (HMP) with dendritic cells (DCs) and T cells among the mixed immune cells from lymph nodes (LNs), which could be leveraged in the development of nanovaccines for anti-tumor therapy. After loading with an adjuvant (R837), the nanoparticles coated with a hybrid membrane (HMPR) demonstrated effectiveness in priming DCs both in vitro and in vivo, resulting in amplified anti-tumor immune responses compared to those of nanoparticles coated with a single type of membrane or those lacking a membrane coating. The elevated immunoactivity of nanoparticles achieved by incorporating a hybrid membrane biosurface provides us a more profound comprehension of the nano-immune interaction, which may significantly benefit the development of bioactive nanomaterials for advanced therapy.
Collapse
Affiliation(s)
- Luying Yu
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Ao Zhou
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Jingyan Jia
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Jieting Wang
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Xueyang Ji
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Yu Deng
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Xinhua Lin
- Key Laboratory of Nanomedical Technology (Education Department of Fujian Province), Nanomedical Technology Research Institute, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| | - Fang Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
9
|
Bao P, Gu H, Ye J, He J, Zhong Z, Yu A, Zhang X. Chimeric Exosomes Functionalized with STING Activation for Personalized Glioblastoma Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306336. [PMID: 38072677 PMCID: PMC10853748 DOI: 10.1002/advs.202306336] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/13/2023] [Indexed: 02/10/2024]
Abstract
A critical challenge of existing cancer vaccines is to orchestrate the demands of antigen-enriched furnishment and optimal antigen-presentation functionality within antigen-presenting cells (APCs). Here, a complementary immunotherapeutic strategy is developed using dendritic cell (DC)-tumor hybrid cell-derived chimeric exosomes loaded with stimulator of interferon genes (STING) agonists (DT-Exo-STING) for maximized tumor-specific T-cell immunity. These chimeric carriers are furnished with broad-spectrum antigen complexes to elicit a robust T-cell-mediated inflammatory program through direct self-presentation and indirect DC-to-T immunostimulatory pathway. This chimeric exosome-assisted delivery strategy possesses the merits versus off-the-shelf cyclic dinucleotide (CDN) delivery techniques in both the brilliant tissue-homing capacity, even across the intractable blood-brain barrier (BBB), and the desired cytosolic entry for enhanced STING-activating signaling. The improved antigen-presentation performance with this nanovaccine-driven STING activation further enhances tumor-specific T-cell immunoresponse. Thus, DT-Exo-STING reverses immunosuppressive glioblastoma microenvironments to pro-inflammatory, tumoricidal states, leading to an almost obliteration of intracranial primary lesions. Significantly, an upscaling option that harnesses autologous tumor tissues for personalized DT-Exo-STING vaccines increases sensitivity to immune checkpoint blockade (ICB) therapy and exerts systemic immune memory against post-operative glioma recrudesce. These findings represent an emerging method for glioblastoma immunotherapy, warranting further exploratory development in the clinical realm.
Collapse
Affiliation(s)
- Peng Bao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Hui‐Yun Gu
- Department of Orthopedic Trauma and MicrosurgeryZhongnan Hospital of Wuhan UniversityWuhan430071P. R. China
| | - Jing‐Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Jin‐Lian He
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Zhenlin Zhong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Ai‐Xi Yu
- Department of Orthopedic Trauma and MicrosurgeryZhongnan Hospital of Wuhan UniversityWuhan430071P. R. China
| | - Xian‐Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of ChemistryWuhan UniversityWuhan430072P. R. China
- Department of Orthopedic Trauma and MicrosurgeryZhongnan Hospital of Wuhan UniversityWuhan430071P. R. China
| |
Collapse
|
10
|
Silva AJD, de Moura IA, da Gama MATM, Leal LRS, de Pinho SS, Espinoza BCF, dos Santos DL, Santos VEP, Sena MGAMD, Invenção MDCV, de Macêdo LS, de França Neto PL, de Freitas AC. Advancing Immunotherapies for HPV-Related Cancers: Exploring Novel Vaccine Strategies and the Influence of Tumor Microenvironment. Vaccines (Basel) 2023; 11:1354. [PMID: 37631922 PMCID: PMC10458729 DOI: 10.3390/vaccines11081354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/27/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
The understanding of the relationship between immunological responses and cancers, especially those related to HPV, has allowed for the study and development of therapeutic vaccines against these neoplasias. There is a growing number of studies about the composition and influence of the tumor microenvironment (TME) in the progression or establishment of the most varied types of cancer. Hence, it has been possible to structure immunotherapy approaches based on therapeutic vaccines that are even more specific and directed to components of TME and the immune response associated with tumors. Among these components are dendritic cells (DCs), which are the main professional antigen-presenting cells (APCs) already studied in therapy strategies for HPV-related cancers. On the other hand, tumor-associated macrophages are also potential targets since the profile present in tumor infiltrates, M1 or M2, influences the prognosis of some types of cancer. These two cell types can be targets for therapy or immunomodulation. In this context, our review aims to provide an overview of immunotherapy strategies for HPV-positive tumors, such as cervical and head and neck cancers, pointing to TME immune cells as promising targets for these approaches. This review also explores the potential of immunotherapy in cancer treatment, including checkpoint inhibitors, cytokine immunotherapies, immunotherapy vaccines, and cell therapies. Furthermore, it highlights the importance of understanding the TME and its effect on the design and achievement of immunotherapeutic methods.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (A.J.D.S.); (I.A.d.M.); (M.A.T.M.d.G.); (L.R.S.L.); (S.S.d.P.); (B.C.F.E.); (D.L.d.S.); (V.E.P.S.); (M.G.A.M.D.S.); (M.D.C.V.I.); (L.S.d.M.); (P.L.d.F.N.)
| |
Collapse
|
11
|
Wu G, Liu Z, Mu C, Song D, Wang J, Meng X, Li Z, Qing H, Dong Y, Xie HY, Pang DW. Enhanced Proliferation of Visualizable Mesenchymal Stem Cell-Platelet Hybrid Cell for Versatile Intracerebral Hemorrhage Treatment. ACS NANO 2023; 17:7352-7365. [PMID: 37037487 DOI: 10.1021/acsnano.2c11329] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The intrinsic features and functions of platelets and mesenchymal stem cells (MSCs) indicate their great potential in the treatment of intracerebral hemorrhage (ICH). However, neither of them can completely overcome ICH because of the stealth process and the complex pathology of ICH. Here, we fabricate hybrid cells for versatile and highly efficient ICH therapy by fusing MSCs with platelets and loading with lysophosphatidic acid-modified PbS quantum dots (LPA-QDs). The obtained LPA-QDs@FCs (FCs = fusion cells) not only inherit the capabilities of both platelets and MSCs but also exhibit clearly enhanced proliferation activated by LPA. After systemic administration, many proliferating LPA-QDs@FCs rapidly accumulate in ICH areas for responding to the vascular damage and inflammation and then efficiently prevent both the primary and secondary injuries of ICH but with no obvious side effects. Moreover, the treatment process can be tracked by near-infrared II fluorescence imaging with highly spatiotemporal resolution, providing a promising solution for ICH therapy.
Collapse
Affiliation(s)
- Guanghao Wu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhenya Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
| | - Changwen Mu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Da Song
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaxin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100083, P. R. China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, Beijing 100142, P. R. China
| | - Ziyuan Li
- Department of Biomedical Engineering, Peking University, Beijing 100871, P. R. China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yuping Dong
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
| |
Collapse
|
12
|
Lyu X, Cui F, Zhou H, Cao B, Zhang X, Cai M, Yang S, Sun B, Li G. 3D co-culture of macrophages and fibroblasts in a sessile drop array for unveiling the role of macrophages in skin wound-healing. Biosens Bioelectron 2023; 225:115111. [PMID: 36731395 DOI: 10.1016/j.bios.2023.115111] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023]
Abstract
Three-dimensional (3D) heterotypic multicellular spheroid models play important roles in researches of the proliferation and remodeling phases in wound healing. This study aimed to develop a sessile drop array to cultivate 3D spheroids and simulate wound healing stage in vitro using NIH-3T3 fibroblasts and M2-type macrophages. By the aid of the offset of surface tension and gravity, the sessile drop array is able to transfer cell suspensions to spheroids via the superhydrophobic surface of each microwell. Meanwhile, each microwell has a cylinder hole at its bottom that provides adequate oxygen to the spheroid. It demonstrated that the NIH-3T3 fibroblast spheroid and the 3T3 fibroblast/M2-type macrophage heterotypic multicellular spheroid can form and maintain physiological activities within nine days. In order to further investigate the structure without destroying the entire spheroid, we reconstructed its 3D morphology using transparent processing technology and the Z-stack function of confocal microscopy. Additionally, a nano antibody-based 3D immunostaining assay was used to analyze the proliferation and differentiation characteristics of these cells. It found that M2-type macrophages were capable of promoting the differentiation of 3T3 fibroblast spheroid. In this study, a novel, inexpensive platform was constructed for developing spheroids, as well as a 3D immunofluorescence method for investigating the macrophage-associated wound healing microenvironment.
Collapse
Affiliation(s)
- Xiaoyan Lyu
- Department of Dermatology, Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Feiyun Cui
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Hang Zhou
- The Ministry of Education Key Laboratory of Clinical Diagnostics, School of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Bo Cao
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Xiaolan Zhang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Minghui Cai
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Shulong Yang
- Department of Pediatric Surgery, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Bangyong Sun
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing, 400044, China
| | - Gang Li
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
13
|
Yi Y, Yu M, Li W, Zhu D, Mei L, Ou M. Vaccine-like nanomedicine for cancer immunotherapy. J Control Release 2023; 355:760-778. [PMID: 36822241 DOI: 10.1016/j.jconrel.2023.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The successful clinical application of immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) therapeutics has attracted extensive attention to immunotherapy, however, their drawbacks such as limited specificity, persistence and toxicity haven't met the high expectations on efficient cancer treatments. Therapeutic cancer vaccines which instruct the immune system to capture tumor specific antigens, generate long-term immune memory and specifically eliminate cancer cells gradually become the most promising strategies to eradicate tumor. However, the disadvantages of some existing vaccines such as weak immunogenicity and in vivo instability have restricted their development. Nanotechnology has been recently incorporated into vaccine fabrication and exhibited promising results for cancer immunotherapy. Nanoparticles promote the stability of vaccines, as well as enhance antigen recognition and presentation owing to their nanometer size which promotes internalization of antigens by phagocytic cells. The surface modification with targeting units further permits the delivery of vaccines to specific cells. Meanwhile, nanocarriers with adjuvant effect can improve the efficacy of vaccines. In addition to classic vaccines composed of antigens and adjuvants, the nanoparticle-mediated chemotherapy, radiotherapy and certain other therapeutics could induce the release of tumor antigens in situ, which therefore effectively simulate antitumor immune responses. Such vaccine-like nanomedicine not only kills primary tumors, but also prevents tumor recurrence and helps eliminate metastatic tumors. Herein, we introduce recent developments in nanoparticle-based delivery systems for antigen delivery and in situ antitumor vaccination. We will also discuss the remaining opportunities and challenges of nanovaccine in clinical translation towards cancer treatment.
Collapse
Affiliation(s)
- Yunfei Yi
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Meitong Ou
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
14
|
Li P, Cui F, Chen H, Yang Y, Li G, Mao H, Lyu X. A Microfluidic Cell Co-Culture Chip for the Monitoring of Interactions between Macrophages and Fibroblasts. BIOSENSORS 2022; 13:bios13010070. [PMID: 36671905 PMCID: PMC9855520 DOI: 10.3390/bios13010070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/24/2022] [Accepted: 12/25/2022] [Indexed: 05/28/2023]
Abstract
Macrophages and fibroblasts are two types of important cells in wound healing. The development of novel platforms for studying the interrelationship between these two cells is crucial for the exploration of wound-healing mechanisms and drug development. In this study, a microfluidic chip composed of two layers was designed for the co-culturing of these two cells. An air valve was employed to isolate fibroblasts to simulate the wound-healing microenvironment. The confluence rate of fibroblasts in the co-culture system with different macrophages was explored to reflect the role of different macrophages in wound healing. It was demonstrated that M2-type macrophages could promote the activation and migration of fibroblasts and it can be inferred that they could promote the wound-healing process. The proposed microfluidic co-culture system was designed for non-contact cell-cell interactions, which has potential significance for the study of cell-cell interactions in biological processes such as wound healing, tumor microenvironment, and embryonic development.
Collapse
Affiliation(s)
- Pengcheng Li
- Department of Orthopedics, West China Hospital, West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Feiyun Cui
- School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China
| | - Heying Chen
- The Ministry of Education Key Laboratory of Clinical Diagnostics, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yao Yang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Li
- Key Laboratory of Optoelectronic Technology and Systems, Ministry of Education, Defense Key Disciplines Lab of Novel Micro-Nano Devices and System Technology, Chongqing University, Chongqing 400044, China
| | - Hongju Mao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Xiaoyan Lyu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Lou K, Feng S, Luo H, Zou J, Zhang G, Zou X. Extracellular vesicles derived from macrophages: Current applications and prospects in tumors. Front Bioeng Biotechnol 2022; 10:1097074. [PMID: 36588947 PMCID: PMC9797603 DOI: 10.3389/fbioe.2022.1097074] [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: 11/13/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Macrophages (Mφs) are significant innate immune cells that perform a variety of tasks in response to different pathogens or stimuli. They are widely engaged in the pathological processes of various diseases and can contribute to tumorigenesis, progression and metastasis by regulating the tumor microenvironment and cancer cells. They are also the basis of chemoresistance. In turn, the tumor microenvironment and the metabolism of cancer cells can limit the differentiation, polarization, mobilization and the ability of Mφs to initiate an effective anti-tumor response. Extracellular vesicles (EVs) are small vesicles released by live cells that serve as crucial mediators of intercellular cell communication as well as a potential promising drug carrier. A growing number of studies have demonstrated that Mφs-EVs are not only important mediators in the pathological processes of various diseases such as inflammatory disorders, fibrosis and cancer, but also show significant potential in immunological modulation, cancer therapy, infectious defense and tissue repair. These natural nanoparticles (NPs) derived from Mφs are believed to be pleiotropic, stable, biocompatible and low immunogenic, providing novel alternatives for cancer treatment. This review provides an update on the pathological and therapeutic roles of Mφs-EVs in cancer, as well as their potential clinical applications and prospects.
Collapse
Affiliation(s)
- Kecheng Lou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shangzhi Feng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hui Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China
| | - Guoxi Zhang
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China
| | - Xiaofeng Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,Institute of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou, Jiangxi, China,*Correspondence: Xiaofeng Zou,
| |
Collapse
|
16
|
Wang D, Chen H, Lei L, Chen J, Gao J, Liu J, Li Q, Xie Y, Hu Y, Ni Y. Biofabricated macrophage and fibroblast membranes synergistically promote skin wound healing. Bioeng Transl Med 2022; 7:e10344. [PMID: 36176601 PMCID: PMC9472019 DOI: 10.1002/btm2.10344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/21/2022] [Accepted: 05/14/2022] [Indexed: 12/11/2022] Open
Abstract
Effective skin wound healing is a complex process involving anti-inflammation, fibrosis, matrix reconstruction, and angiogenesis. This work aimed to integrate the macrophage-mediated anti-inflammation and fibroblast-assisted matrix reconstruction for enhanced skin wound healing. Herein, we utilized the cytomembranes derived from repolarized M2 macrophages and fibroblasts to prepare the natural biologics. Results showed that the inflammatory M1 macrophages were repolarized to M2 phenotype by the M2 macrophage cytomembranes. As a consequence, the cytomembranes of M2 macrophage could facilitate the wound closure in mice. Furthermore, the addition of fibroblast membranes to the macrophage cytomembranes contributed to a better matrix reconstruction, neovascularization and angiogenesis. Next, we used a transforming growth factor-β (TGF-β) inhibitor to attenuate cutaneous scar formation. Therefore, our modality could promote skin wound healing and effectively suppress scar formation in the preclinical murine skin wounds. The cytomembrane biologics might provide a biocompatible and versatile tool for wound healing.
Collapse
Affiliation(s)
- Dongqing Wang
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| | - Heying Chen
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| | - Li Lei
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS)BeijingChina
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Laboratory Medicine and Life ScienceWenzhou Medical University, University TownWenzhouZhejiangChina
| | - Jiahe Liu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| | - Qianyin Li
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| | - Yajun Xie
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| | - Yi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyInstitute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS)BeijingChina
| | - Yilu Ni
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory MedicineChongqing Medical UniversityYuzhong DistrictChongqingChina
| |
Collapse
|
17
|
Huo W, Yang X, Wang B, Cao L, Fang Z, Li Z, Liu H, Liang XJ, Zhang J, Jin Y. Biomineralized hydrogel DC vaccine for cancer immunotherapy: A boosting strategy via improving immunogenicity and reversing immune-inhibitory microenvironment. Biomaterials 2022; 288:121722. [DOI: 10.1016/j.biomaterials.2022.121722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022]
|
18
|
Jorquera-Cordero C, Lara P, Cruz LJ, Schomann T, van Hofslot A, de Carvalho TG, Guedes PMDM, Creemers L, Koning RI, Chan AB, de Araujo Junior RF. Extracellular Vesicles from M1-Polarized Macrophages Combined with Hyaluronic Acid and a β-Blocker Potentiate Doxorubicin’s Antitumor Activity by Downregulating Tumor-Associated Macrophages in Breast Cancer. Pharmaceutics 2022; 14:pharmaceutics14051068. [PMID: 35631654 PMCID: PMC9143936 DOI: 10.3390/pharmaceutics14051068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/21/2022] Open
Abstract
One of the main reasons for cancer’s low clinical response to chemotherapeutics is the highly immunosuppressive tumor microenvironment (TME). Tumor-ass ociated M2 macrophages (M2-TAMs) orchestrate the immunosuppression, which favors tumor progression. Extracellular vesicles (EVs) have shown great potential for targeted therapies as, depending on their biological origin, they can present different therapeutic properties, such as enhanced accumulation in the target tissue or modulation of the immune system. In the current study, EVs were isolated from M1-macrophages (M1-EVs) pre-treated with hyaluronic acid (HA) and the β-blocker carvedilol (CV). The resulting modulated-M1 EVs (MM1-EVs) were further loaded with doxorubicin (MM1-DOX) to assess their effect in a mouse model of metastatic tumor growth. The cell death and cell migration profile were evaluated in vitro in 4T1 cells. The polarization of the RAW 264.7 murine macrophage cell line was also analyzed to evaluate the effects on the TME. Tumors were investigated by qRT-PCR and immunohistochemistry. MM1-DOX reduced the primary tumor size and metastases. NF-κB was the major gene downregulated by MM1-DOX. Furthermore, MM1-DOX reduced the expression of M2-TAM (CD-163) in tumors, which resulted in increased apoptosis (FADD) as well as decreased expression of MMP-2 and TGF-β. These results suggest a direct effect in tumors and an upregulation in the TME immunomodulation, which corroborate with our in vitro data that showed increased apoptosis, modulation of macrophage polarization, and reduced cell migration after treatment with M1-EVs combined with HA and CV. Our results indicate that the M1-EVs enhanced the antitumor effects of DOX, especially if combined with HA and CV in an animal model of metastatic cancer.
Collapse
Affiliation(s)
- Carla Jorquera-Cordero
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (C.J.-C.); (L.C.); (A.B.C.)
- Percuros B.V., 2333 CL Leiden, The Netherlands; (T.S.); (T.G.d.C.)
| | - Pablo Lara
- Percuros B.V., 2333 CL Leiden, The Netherlands; (T.S.); (T.G.d.C.)
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (L.J.C.); (A.v.H.)
- Correspondence: (P.L.); (R.F.d.A.J.); Tel.: +31-06-21180677 (P.L.); +31-65-562-0247 (R.F.d.A.J.)
| | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (L.J.C.); (A.v.H.)
| | - Timo Schomann
- Percuros B.V., 2333 CL Leiden, The Netherlands; (T.S.); (T.G.d.C.)
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (L.J.C.); (A.v.H.)
| | - Anna van Hofslot
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (L.J.C.); (A.v.H.)
| | - Thaís Gomes de Carvalho
- Percuros B.V., 2333 CL Leiden, The Netherlands; (T.S.); (T.G.d.C.)
- Postgraduate Program in Health Science, Health Science Department, Federal University of Rio Grande do Norte (UFRN), Natal 59078 970, RN, Brazil
- Cancer and Inflammation Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte, Natal 59078 970, RN, Brazil
| | - Paulo Marcos Da Matta Guedes
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal 59078 970, RN, Brazil;
| | - Laura Creemers
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (C.J.-C.); (L.C.); (A.B.C.)
| | - Roman I. Koning
- Electron Microscopy, Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Alan B. Chan
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (C.J.-C.); (L.C.); (A.B.C.)
- Percuros B.V., 2333 CL Leiden, The Netherlands; (T.S.); (T.G.d.C.)
| | - Raimundo Fernandes de Araujo Junior
- Percuros B.V., 2333 CL Leiden, The Netherlands; (T.S.); (T.G.d.C.)
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (L.J.C.); (A.v.H.)
- Postgraduate Program in Health Science, Health Science Department, Federal University of Rio Grande do Norte (UFRN), Natal 59078 970, RN, Brazil
- Cancer and Inflammation Research Laboratory, Department of Morphology, Federal University of Rio Grande do Norte, Natal 59078 970, RN, Brazil
- Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59078 970, RN, Brazil
- Correspondence: (P.L.); (R.F.d.A.J.); Tel.: +31-06-21180677 (P.L.); +31-65-562-0247 (R.F.d.A.J.)
| |
Collapse
|
19
|
Xu T, Yu S, Zhang J, Wu S. Dysregulated tumor-associated macrophages in carcinogenesis, progression and targeted therapy of gynecological and breast cancers. J Hematol Oncol 2021; 14:181. [PMID: 34717710 PMCID: PMC8557603 DOI: 10.1186/s13045-021-01198-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Gynecological and breast cancers are a group of heterogeneous malignant tumors. Although existing treatment strategies have ameliorated the clinical outcomes of patients, the overall survival rate of advanced diseases remains unsatisfactory. Increasing evidence has indicated that the development and prognosis of tumors are closely related to the tumor microenvironment (TME), which restricts the immune response and provokes malignant progression. Tumor-associated macrophages (TAMs) are the main component of TME and act as a key regulator in tumor metastasis, immunosuppression and therapeutic resistance. Several preclinical trials have studied potential drugs that target TAMs to achieve potent anticancer therapy. This review focuses on the various functions of TAMs and how they influence the carcinogenesis of gynecological and breast cancers through regulating cancer cell proliferation, tumor angiogenesis and tumor-related immunosuppression. Besides, we also discuss the potential application of disabling TAMs signaling as a part of cancer therapeutic strategies, as well as CAR macrophages, TAMs-based vaccines and TAMs nanobiotechnology. These research advances support that targeting TAMs combined with conventional therapy might be used as effective therapeutics for gynecological and breast cancers in the future.
Collapse
Affiliation(s)
- Tianhan Xu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Sihui Yu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China. .,Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Sufang Wu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|